Allowed Publications
Slot System
Featured Buckets
Featured Buckets Admin

Aquatic Antagonists: Dermatologic Injuries From Sea Urchins (Echinoidea)

Article Type
Changed
Fri, 06/14/2024 - 12:41
Display Headline
Aquatic Antagonists: Dermatologic Injuries From Sea Urchins (Echinoidea)

Sea urchins—members of the phylum Echinodermata and the class Echinoidea—are spiny marine invertebrates. Their consumption of fleshy algae makes them essential players in maintaining reef ecosystems.1,2 Echinoids, a class that includes heart urchins and sand dollars, are ubiquitous in benthic marine environments, both free floating and rock boring, and inhabit a wide range of latitudes spanning from polar oceans to warm seas.3 Despite their immobility and nonaggression, sea urchin puncture wounds are common among divers, snorkelers, swimmers, surfers, and fishers who accidentally come into contact with their sharp spines. Although the epidemiology of sea urchin exposure and injury is difficult to assess, the American Association of Poison Control Centers’ most recent annual report in 2022 documents approximately 1426 annual aquatic bites and/or envenomations.4

Sea Urchin Morphology and Toxicity

Echinoderms (a term of Greek origin meaning spiny skin) share a radially symmetric calcium carbonate skeleton (termed stereom) that is supported by collagenous ligaments.1 Sea urchins possess spines composed of calcite crystals, which radiate from their body and play a role in locomotion and defense against predators—namely sea otters, starfish/sea stars, wolf eels, and triggerfish, among others (Figure).5 These brittle spines can easily penetrate human skin and subsequently break off the sea urchin body. Most species of sea urchins possess solid spines, but a small percentage (80 of approximately 700 extant species) have hollow spines containing various toxic substances.6 Penetration and systemic absorption of the toxins within these spines can generate severe systemic responses.

The venomous flower urchin (Toxopneustes pileolus), found in the Indian and Pacific oceans, is one of the more common species known to produce a systemic reaction involving neuromuscular blockage.7-9 The most common species harvested off the Pacific coast of the United States—Strongylocentrotus purpuratus (purple sea urchin) and Strongylocentrotus franciscanus (red sea urchins)—are not inherently venomous.8

bucogelethitriboshat
%3Cp%3EPurple%20sea%20urchin%20(%3Cem%3EStrongylocentrotus%20purpuratus%3C%2Fem%3E).%20Photograph%20courtesy%20of%20the%20South%20Carolina%20Aquarium%20(Charleston%2C%20South%20Carolina).%3C%2Fp%3E


Both the sea urchin body and spines are covered in a unique epithelium thought to be responsible for the majority of their proinflammatory and pronociceptive properties. Epithelial compounds identified include serotonin, histamines, steroids, glycosides, hemolysins, proteases, and bradykininlike and cholinergic substances.5,7 Additionally, certain sea urchin species possess 3-pronged pincerlike organs at the base of spines called pedicellariae, which are used in feeding.10 Skin penetration by the pedicellariae is especially dangerous, as they tightly adhere to wounds and contain venom-producing organs that allow them to continue injecting toxins after their detachment from the sea urchin body.11

Presentation and Diagnosis of Sea Urchin Injuries

Sea urchin injuries have a wide range of manifestations depending on the number of spines involved, the presence of venom, the depth and location of spine penetration, the duration of spine retention in the skin, and the time before treatment initiation. The most common site of sea urchin injury unsurprisingly is the lower extremities and feet, often in the context of divers and swimmers walking across the sea floor. The hands are another frequently injured site, along with the legs, arms, back, scalp, and even oral mucosa.11

Although clinical history and presentation frequently reveal the mechanism of aquatic injury, patients often are unsure of the agent to which they were exposed and may be unaware of retained foreign bodies. Dermoscopy can distinguish the distinct lines radiating from the core of sea urchin spines from other foreign bodies lodged within the skin.6 It also can be used to locate spines for removal or for their analysis following punch biopsy.6,12 The radiopaque nature of sea urchin spines makes radiography and magnetic resonance imaging useful tools in assessment of periarticular soft-tissue damage and spine removal.8,11,13 Ultrasonography can reveal spines that no longer appear on radiography due to absorption by human tissue.14

Immediate Dermatologic Effects

Sea urchin injuries can be broadly categorized into immediate and delayed reactions. Immediate manifestations of contact with sea urchin spines include localized pain, bleeding, erythema, myalgia, and edema at the site of injury that can last from a few hours to 1 week without proper wound care and spine removal.5 Systemic symptoms ranging from dizziness, lightheadedness, paresthesia, aphonia, paralysis, coma, and death generally are only seen following injuries from venomous species, attachment of pedicellariae, injuries involving neurovascular structures, or penetration by more than 15 spines.7,11

Initial treatment includes soaking the wound in hot water (113 °F [45 °C]) for 30 to 90 minutes and subsequently removing spines and pedicellariae to prevent development of delayed reactions.5,15,16 The compounds in the sea urchin epithelium are heat labile and will be inactivated upon soaking in hot water.16 Extraction of spines can be difficult, as they are brittle and easily break in the skin. Successful removal has been reported using forceps and a hypodermic needle as well as excision; both approaches may require local anesthesia.8,17 Another technique involves freezing the localized area with liquid nitrogen to allow easier removal upon skin blistering.18 Punch biopsy also has been utilized as an effective means of ensuring all spiny fragments are removed.9,19,20 These spines often cause black or purple tattoolike staining at the puncture site, which can persist for a few days after spine extraction.8 Ablation using the erbium-doped:YAG laser may be helpful for removal of associated pigment.21,22

Delayed Dermatologic Effects

Delayed reactions to sea urchin injuries often are attributable to prolonged retention of spines in the skin. Granulomatous reactions typically manifest 2 weeks after injury as firm nonsuppurative nodules with central umbilication and a hyperkeratotic surface.7 These nodules may or may not be painful. Histopathology most often reveals foreign body and sarcoidal-type granulomatous reactions. However, tuberculoid, necrobiotic, and suppurative granulomas also may develop.13 Other microscopic features include inflammatory reactions, suppurative dermatitis, focal necrosis, and microabscesses.23 Wounds with progression to granulomatous disease often require surgical debridement.

Other more serious sequalae can result from involvement of joint capsules, especially in the hands and feet. Sea urchin injury involving joint spaces should be treated aggressively, as progression to inflammatory or infectious synovitis and tenosynovitis can cause irreversible loss of joint function. Inflammatory synovitis occurs 1 to 2 months on average after injury following a period of minimal symptoms and begins as a gradual increase in joint swelling and decrease in range of motion.8 Infectious tenosynovitis manifests quite similarly. Although suppurative etiologies generally progress with a more acute onset, certain infectious organisms (eg, Mycobacterium) take on an indolent course and should not be overlooked as a cause of delayed symptoms.8 The Kavanel cardinal signs are a sensitive tool used in the diagnosis of infectious flexor sheath tenosynovitis.8,24 If suspicion for joint infection is high, emergency referral should be made for debridement and culture-guided antibiotic therapy. Left untreated, infectious tenosynovitis can result in tendon necrosis or rupture, digit necrosis, and systemic infection.24 Patients with joint involvement should be referred to specialty care (eg, hand surgeon), as they often require synovectomy and surgical removal of foreign material.8

From 1 month to 1 year after injury, prolonged granulomatous synovitis of the hand may eventually lead to joint destruction known as “sea urchin arthritis.” These patients present with decreased range of motion and numerous nodules on the hand with a hyperkeratotic surface. Radiography reveals joint space narrowing, osteolysis, subchondral sclerosis, and periosteal reaction. Synovectomy and debridement are necessary to prevent irreversible joint damage or the need for arthrodesis and bone grafting.24

Other Treatment Considerations

Other important considerations in the care of sea urchin spine injuries include assessment of tetanus immunization status and administration of necessary prophylaxis as soon as possible, even in delayed presentations (Table).16,25 Cultures should be taken only if infection is suspected. Prophylactic antibiotics are not recommended unless the patient is immunocompromised or otherwise has impaired wound healing. If a patient presents with systemic symptoms, they should be referred to an emergency care facility for further management.

Final Thoughts

Sea urchin injuries can lead to serious complications if not diagnosed quickly and treated properly. Retention of sea urchin spines in the deep tissues and joint spaces may lead to granulomas, inflammatory and infectious tenosynovitis (including mycobacterial infection), and sea urchin arthritis requiring surgical debridement and possible irreversible joint damage, up to a year after initial injury. Patients should be educated on the possibility of developing these delayed reactions and instructed to seek immediate care. Joint deformities, range-of-motion deficits, and involvement of neurovascular structures should be considered emergent and referred for proper management. Shoes and diving gear offer some protection but are easily penetrable by sharp sea urchin spines. Preventive focus should be aimed at educating patients and providers on the importance of prompt spine removal upon injury. Although dermatologic and systemic manifestations vary widely, a thorough history, physical examination, and appropriate use of imaging modalities can facilitate accurate diagnosis and guide treatment.

midetawucrislasastumitheprutradreprohijupedupuspeshaciphomistemicruslifriclupridiwritoslafrophajaheswecretruchechewefrowrouinemasohubrotuphiphutaslothuwrisow

References
  1. Amemiya CT, Miyake T, Rast JP. Echinoderms. Curr Biol. 2005;15:R944-R946. doi:10.1016/j.cub.2005.11.026
  2. Koch NM, Coppard SE, Lessios HA, et al. A phylogenomic resolution of the sea urchin tree of life. BMC Evol Biol. 2018;18:189. doi:10.1186/s12862-018-1300-4
  3. Amir Y, Insler M, Giller A, et al. Senescence and longevity of sea urchins. Genes (Basel). 2020;11:573. doi:10.3390/genes11050573
  4. Gummin DD, Mowry JB, Beuhler MC, et al. 2022 Annual Report of the National Poison Data System® (NPDS) from America's Poison Centers®: 40th annual report. Clin Toxicol (Phila). 2023;61:717-939. doi:10.1080/15563650.2023.2268981
  5. Gelman Y, Kong EL, Murphy-Lavoie HM. Sea urchin toxicity. In: StatPearls [Internet]. StatPearls Publishing; 2021.
  6. Suarez-Conde MF, Vallone MG, González VM, et al. Sea urchin skin lesions: a case report. Dermatol Pract Concept. 2021;11:E2021009. doi:10.5826/dpc.1102a09
  7. Al-Kathiri L, Al-Najjar T, Sulaiman I. Sea urchin granuloma of the hands: a case report. Oman Med J. 2019;34:350-353. doi:10.5001/omj.2019.68
  8. Dahl WJ, Jebson P, Louis DS. Sea urchin injuries to the hand: a case report and review of the literature. Iowa Orthop J. 2010;30:153-156.
  9. Hatakeyama T, Ichise A, Unno H, et al. Carbohydrate recognition by the rhamnose-binding lectin SUL-I with a novel three-domain structure isolated from the venom of globiferous pedicellariae of the flower sea urchin Toxopneustes pileolus. Protein Sci. 2017;26:1574-1583. doi:10.1002/pro.3185
  10. Balhara KS, Stolbach A. Marine envenomations. Emerg Med Clin North Am. 2014;32:223-243. doi:10.1016/j.emc.2013.09.009
  11. Schwartz Z, Cohen M, Lipner SR. Sea urchin injuries: a review and clinical approach algorithm. J Dermatolog Treat. 2021;32:150-156. doi:10.1080/09546634.2019.1638884
  12. Park SJ, Park JW, Choi SY, et al. Use of dermoscopy after punch removal of a veiled sea urchin spine. Dermatol Ther. 2021;34:E14947. doi:10.1111/dth.14947
  13. Wada T, Soma T, Gaman K, et al. Sea urchin spine arthritis of the hand. J Hand Surg Am. 2008;33:398-401. doi:10.1016/j.jhsa.2007.11.016
  14. Groleau S, Chhem RK, Younge D, et al. Ultrasonography of foreign-body tenosynovitis. Can Assoc Radiol J. 1992;43:454-456. 
  15. Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337. doi:10.1016/j.emc.2016.12.004
  16. Noonburg GE. Management of extremity trauma and related infections occurring in the aquatic environment. J Am Acad Orthop Surg. 2005;13:243-253. doi:10.5435/00124635-200507000-00004
  17. Haddad Junior V. Observation of initial clinical manifestations and repercussions from the treatment of 314 human injuries caused by black sea urchins (Echinometra lucunter) on the southeastern Brazilian coast. Rev Soc Bras Med Trop. 2012;45:390-392. doi:10.1590/s0037-86822012000300021
  18. Gargus MD, Morohashi DK. A sea-urchin spine chilling remedy. N Engl J Med. 2012;367:1867-1868. doi:10.1056/NEJMc1209382
  19. Sjøberg T, de Weerd L. The usefulness of a skin biopsy punch to remove sea urchin spines. ANZ J Surg. 2010;80:383. doi:10.1111/j.1445-2197.2010.05296.x
  20. Cardenas-de la Garza JA, Cuellar-Barboza A, Ancer-Arellano J, et al. Classic dermatological tools: foreign body removal with punch biopsy.J Am Acad Dermatol. 2019;81:E93-E94. doi:10.1016/j.jaad.2018.10.038
  21. Gungor S, Tarikçi N, Gokdemir G. Removal of sea urchin spines using erbium-doped yttrium aluminum garnet ablation. Dermatol Surg. 2012;38:508-510. doi:10.1111/j.1524-4725.2011.02259.x
  22. Böer A, Ochsendorf FR, Beier C, et al. Effective removal of sea-urchin spines by erbium:YAG laser ablation. Br J Dermatol. 2001;145:169-170. doi:10.1046/j.1365-2133.2001.04306.x
  23. De La Torre C, Toribio J. Sea-urchin granuloma: histologic profile. a pathologic study of 50 biopsies. J Cutan Pathol. 2001;28:223-228. doi:10.1034/j.1600-0560.2001.028005223.x
  24. Yi A, Kennedy C, Chia B, et al. Radiographic soft tissue thickness differentiating pyogenic flexor tenosynovitis from other finger infections. J Hand Surg Am. 2019;44:394-399. doi:10.1016/j.jhsa.2019.01.013
  25. Callison C, Nguyen H. Tetanus prophylaxis. In: StatPearls [Internet]. StatPearls Publishing; 2022.
Article PDF
Author and Disclosure Information

 

From the Medical University of South Carolina, Charleston. Dr. Brailsford is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Caroline J. Brailsford, MD, Medical University of South Carolina, 135 Rutledge Ave, 11th Floor, Charleston, SC 29425-5780 (cjbrailsford@gmail.com).

Cutis. 2024 June;113(6):255-257. doi:10.12788/cutis.1034

Issue
Cutis - 113(6)
Publications
Topics
Page Number
255-257
Sections
Author and Disclosure Information

 

From the Medical University of South Carolina, Charleston. Dr. Brailsford is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Caroline J. Brailsford, MD, Medical University of South Carolina, 135 Rutledge Ave, 11th Floor, Charleston, SC 29425-5780 (cjbrailsford@gmail.com).

Cutis. 2024 June;113(6):255-257. doi:10.12788/cutis.1034

Author and Disclosure Information

 

From the Medical University of South Carolina, Charleston. Dr. Brailsford is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Caroline J. Brailsford, MD, Medical University of South Carolina, 135 Rutledge Ave, 11th Floor, Charleston, SC 29425-5780 (cjbrailsford@gmail.com).

Cutis. 2024 June;113(6):255-257. doi:10.12788/cutis.1034

Article PDF
Article PDF

Sea urchins—members of the phylum Echinodermata and the class Echinoidea—are spiny marine invertebrates. Their consumption of fleshy algae makes them essential players in maintaining reef ecosystems.1,2 Echinoids, a class that includes heart urchins and sand dollars, are ubiquitous in benthic marine environments, both free floating and rock boring, and inhabit a wide range of latitudes spanning from polar oceans to warm seas.3 Despite their immobility and nonaggression, sea urchin puncture wounds are common among divers, snorkelers, swimmers, surfers, and fishers who accidentally come into contact with their sharp spines. Although the epidemiology of sea urchin exposure and injury is difficult to assess, the American Association of Poison Control Centers’ most recent annual report in 2022 documents approximately 1426 annual aquatic bites and/or envenomations.4

Sea Urchin Morphology and Toxicity

Echinoderms (a term of Greek origin meaning spiny skin) share a radially symmetric calcium carbonate skeleton (termed stereom) that is supported by collagenous ligaments.1 Sea urchins possess spines composed of calcite crystals, which radiate from their body and play a role in locomotion and defense against predators—namely sea otters, starfish/sea stars, wolf eels, and triggerfish, among others (Figure).5 These brittle spines can easily penetrate human skin and subsequently break off the sea urchin body. Most species of sea urchins possess solid spines, but a small percentage (80 of approximately 700 extant species) have hollow spines containing various toxic substances.6 Penetration and systemic absorption of the toxins within these spines can generate severe systemic responses.

The venomous flower urchin (Toxopneustes pileolus), found in the Indian and Pacific oceans, is one of the more common species known to produce a systemic reaction involving neuromuscular blockage.7-9 The most common species harvested off the Pacific coast of the United States—Strongylocentrotus purpuratus (purple sea urchin) and Strongylocentrotus franciscanus (red sea urchins)—are not inherently venomous.8

bucogelethitriboshat
%3Cp%3EPurple%20sea%20urchin%20(%3Cem%3EStrongylocentrotus%20purpuratus%3C%2Fem%3E).%20Photograph%20courtesy%20of%20the%20South%20Carolina%20Aquarium%20(Charleston%2C%20South%20Carolina).%3C%2Fp%3E


Both the sea urchin body and spines are covered in a unique epithelium thought to be responsible for the majority of their proinflammatory and pronociceptive properties. Epithelial compounds identified include serotonin, histamines, steroids, glycosides, hemolysins, proteases, and bradykininlike and cholinergic substances.5,7 Additionally, certain sea urchin species possess 3-pronged pincerlike organs at the base of spines called pedicellariae, which are used in feeding.10 Skin penetration by the pedicellariae is especially dangerous, as they tightly adhere to wounds and contain venom-producing organs that allow them to continue injecting toxins after their detachment from the sea urchin body.11

Presentation and Diagnosis of Sea Urchin Injuries

Sea urchin injuries have a wide range of manifestations depending on the number of spines involved, the presence of venom, the depth and location of spine penetration, the duration of spine retention in the skin, and the time before treatment initiation. The most common site of sea urchin injury unsurprisingly is the lower extremities and feet, often in the context of divers and swimmers walking across the sea floor. The hands are another frequently injured site, along with the legs, arms, back, scalp, and even oral mucosa.11

Although clinical history and presentation frequently reveal the mechanism of aquatic injury, patients often are unsure of the agent to which they were exposed and may be unaware of retained foreign bodies. Dermoscopy can distinguish the distinct lines radiating from the core of sea urchin spines from other foreign bodies lodged within the skin.6 It also can be used to locate spines for removal or for their analysis following punch biopsy.6,12 The radiopaque nature of sea urchin spines makes radiography and magnetic resonance imaging useful tools in assessment of periarticular soft-tissue damage and spine removal.8,11,13 Ultrasonography can reveal spines that no longer appear on radiography due to absorption by human tissue.14

Immediate Dermatologic Effects

Sea urchin injuries can be broadly categorized into immediate and delayed reactions. Immediate manifestations of contact with sea urchin spines include localized pain, bleeding, erythema, myalgia, and edema at the site of injury that can last from a few hours to 1 week without proper wound care and spine removal.5 Systemic symptoms ranging from dizziness, lightheadedness, paresthesia, aphonia, paralysis, coma, and death generally are only seen following injuries from venomous species, attachment of pedicellariae, injuries involving neurovascular structures, or penetration by more than 15 spines.7,11

Initial treatment includes soaking the wound in hot water (113 °F [45 °C]) for 30 to 90 minutes and subsequently removing spines and pedicellariae to prevent development of delayed reactions.5,15,16 The compounds in the sea urchin epithelium are heat labile and will be inactivated upon soaking in hot water.16 Extraction of spines can be difficult, as they are brittle and easily break in the skin. Successful removal has been reported using forceps and a hypodermic needle as well as excision; both approaches may require local anesthesia.8,17 Another technique involves freezing the localized area with liquid nitrogen to allow easier removal upon skin blistering.18 Punch biopsy also has been utilized as an effective means of ensuring all spiny fragments are removed.9,19,20 These spines often cause black or purple tattoolike staining at the puncture site, which can persist for a few days after spine extraction.8 Ablation using the erbium-doped:YAG laser may be helpful for removal of associated pigment.21,22

Delayed Dermatologic Effects

Delayed reactions to sea urchin injuries often are attributable to prolonged retention of spines in the skin. Granulomatous reactions typically manifest 2 weeks after injury as firm nonsuppurative nodules with central umbilication and a hyperkeratotic surface.7 These nodules may or may not be painful. Histopathology most often reveals foreign body and sarcoidal-type granulomatous reactions. However, tuberculoid, necrobiotic, and suppurative granulomas also may develop.13 Other microscopic features include inflammatory reactions, suppurative dermatitis, focal necrosis, and microabscesses.23 Wounds with progression to granulomatous disease often require surgical debridement.

Other more serious sequalae can result from involvement of joint capsules, especially in the hands and feet. Sea urchin injury involving joint spaces should be treated aggressively, as progression to inflammatory or infectious synovitis and tenosynovitis can cause irreversible loss of joint function. Inflammatory synovitis occurs 1 to 2 months on average after injury following a period of minimal symptoms and begins as a gradual increase in joint swelling and decrease in range of motion.8 Infectious tenosynovitis manifests quite similarly. Although suppurative etiologies generally progress with a more acute onset, certain infectious organisms (eg, Mycobacterium) take on an indolent course and should not be overlooked as a cause of delayed symptoms.8 The Kavanel cardinal signs are a sensitive tool used in the diagnosis of infectious flexor sheath tenosynovitis.8,24 If suspicion for joint infection is high, emergency referral should be made for debridement and culture-guided antibiotic therapy. Left untreated, infectious tenosynovitis can result in tendon necrosis or rupture, digit necrosis, and systemic infection.24 Patients with joint involvement should be referred to specialty care (eg, hand surgeon), as they often require synovectomy and surgical removal of foreign material.8

From 1 month to 1 year after injury, prolonged granulomatous synovitis of the hand may eventually lead to joint destruction known as “sea urchin arthritis.” These patients present with decreased range of motion and numerous nodules on the hand with a hyperkeratotic surface. Radiography reveals joint space narrowing, osteolysis, subchondral sclerosis, and periosteal reaction. Synovectomy and debridement are necessary to prevent irreversible joint damage or the need for arthrodesis and bone grafting.24

Other Treatment Considerations

Other important considerations in the care of sea urchin spine injuries include assessment of tetanus immunization status and administration of necessary prophylaxis as soon as possible, even in delayed presentations (Table).16,25 Cultures should be taken only if infection is suspected. Prophylactic antibiotics are not recommended unless the patient is immunocompromised or otherwise has impaired wound healing. If a patient presents with systemic symptoms, they should be referred to an emergency care facility for further management.

Final Thoughts

Sea urchin injuries can lead to serious complications if not diagnosed quickly and treated properly. Retention of sea urchin spines in the deep tissues and joint spaces may lead to granulomas, inflammatory and infectious tenosynovitis (including mycobacterial infection), and sea urchin arthritis requiring surgical debridement and possible irreversible joint damage, up to a year after initial injury. Patients should be educated on the possibility of developing these delayed reactions and instructed to seek immediate care. Joint deformities, range-of-motion deficits, and involvement of neurovascular structures should be considered emergent and referred for proper management. Shoes and diving gear offer some protection but are easily penetrable by sharp sea urchin spines. Preventive focus should be aimed at educating patients and providers on the importance of prompt spine removal upon injury. Although dermatologic and systemic manifestations vary widely, a thorough history, physical examination, and appropriate use of imaging modalities can facilitate accurate diagnosis and guide treatment.

midetawucrislasastumitheprutradreprohijupedupuspeshaciphomistemicruslifriclupridiwritoslafrophajaheswecretruchechewefrowrouinemasohubrotuphiphutaslothuwrisow

Sea urchins—members of the phylum Echinodermata and the class Echinoidea—are spiny marine invertebrates. Their consumption of fleshy algae makes them essential players in maintaining reef ecosystems.1,2 Echinoids, a class that includes heart urchins and sand dollars, are ubiquitous in benthic marine environments, both free floating and rock boring, and inhabit a wide range of latitudes spanning from polar oceans to warm seas.3 Despite their immobility and nonaggression, sea urchin puncture wounds are common among divers, snorkelers, swimmers, surfers, and fishers who accidentally come into contact with their sharp spines. Although the epidemiology of sea urchin exposure and injury is difficult to assess, the American Association of Poison Control Centers’ most recent annual report in 2022 documents approximately 1426 annual aquatic bites and/or envenomations.4

Sea Urchin Morphology and Toxicity

Echinoderms (a term of Greek origin meaning spiny skin) share a radially symmetric calcium carbonate skeleton (termed stereom) that is supported by collagenous ligaments.1 Sea urchins possess spines composed of calcite crystals, which radiate from their body and play a role in locomotion and defense against predators—namely sea otters, starfish/sea stars, wolf eels, and triggerfish, among others (Figure).5 These brittle spines can easily penetrate human skin and subsequently break off the sea urchin body. Most species of sea urchins possess solid spines, but a small percentage (80 of approximately 700 extant species) have hollow spines containing various toxic substances.6 Penetration and systemic absorption of the toxins within these spines can generate severe systemic responses.

The venomous flower urchin (Toxopneustes pileolus), found in the Indian and Pacific oceans, is one of the more common species known to produce a systemic reaction involving neuromuscular blockage.7-9 The most common species harvested off the Pacific coast of the United States—Strongylocentrotus purpuratus (purple sea urchin) and Strongylocentrotus franciscanus (red sea urchins)—are not inherently venomous.8

bucogelethitriboshat
%3Cp%3EPurple%20sea%20urchin%20(%3Cem%3EStrongylocentrotus%20purpuratus%3C%2Fem%3E).%20Photograph%20courtesy%20of%20the%20South%20Carolina%20Aquarium%20(Charleston%2C%20South%20Carolina).%3C%2Fp%3E


Both the sea urchin body and spines are covered in a unique epithelium thought to be responsible for the majority of their proinflammatory and pronociceptive properties. Epithelial compounds identified include serotonin, histamines, steroids, glycosides, hemolysins, proteases, and bradykininlike and cholinergic substances.5,7 Additionally, certain sea urchin species possess 3-pronged pincerlike organs at the base of spines called pedicellariae, which are used in feeding.10 Skin penetration by the pedicellariae is especially dangerous, as they tightly adhere to wounds and contain venom-producing organs that allow them to continue injecting toxins after their detachment from the sea urchin body.11

Presentation and Diagnosis of Sea Urchin Injuries

Sea urchin injuries have a wide range of manifestations depending on the number of spines involved, the presence of venom, the depth and location of spine penetration, the duration of spine retention in the skin, and the time before treatment initiation. The most common site of sea urchin injury unsurprisingly is the lower extremities and feet, often in the context of divers and swimmers walking across the sea floor. The hands are another frequently injured site, along with the legs, arms, back, scalp, and even oral mucosa.11

Although clinical history and presentation frequently reveal the mechanism of aquatic injury, patients often are unsure of the agent to which they were exposed and may be unaware of retained foreign bodies. Dermoscopy can distinguish the distinct lines radiating from the core of sea urchin spines from other foreign bodies lodged within the skin.6 It also can be used to locate spines for removal or for their analysis following punch biopsy.6,12 The radiopaque nature of sea urchin spines makes radiography and magnetic resonance imaging useful tools in assessment of periarticular soft-tissue damage and spine removal.8,11,13 Ultrasonography can reveal spines that no longer appear on radiography due to absorption by human tissue.14

Immediate Dermatologic Effects

Sea urchin injuries can be broadly categorized into immediate and delayed reactions. Immediate manifestations of contact with sea urchin spines include localized pain, bleeding, erythema, myalgia, and edema at the site of injury that can last from a few hours to 1 week without proper wound care and spine removal.5 Systemic symptoms ranging from dizziness, lightheadedness, paresthesia, aphonia, paralysis, coma, and death generally are only seen following injuries from venomous species, attachment of pedicellariae, injuries involving neurovascular structures, or penetration by more than 15 spines.7,11

Initial treatment includes soaking the wound in hot water (113 °F [45 °C]) for 30 to 90 minutes and subsequently removing spines and pedicellariae to prevent development of delayed reactions.5,15,16 The compounds in the sea urchin epithelium are heat labile and will be inactivated upon soaking in hot water.16 Extraction of spines can be difficult, as they are brittle and easily break in the skin. Successful removal has been reported using forceps and a hypodermic needle as well as excision; both approaches may require local anesthesia.8,17 Another technique involves freezing the localized area with liquid nitrogen to allow easier removal upon skin blistering.18 Punch biopsy also has been utilized as an effective means of ensuring all spiny fragments are removed.9,19,20 These spines often cause black or purple tattoolike staining at the puncture site, which can persist for a few days after spine extraction.8 Ablation using the erbium-doped:YAG laser may be helpful for removal of associated pigment.21,22

Delayed Dermatologic Effects

Delayed reactions to sea urchin injuries often are attributable to prolonged retention of spines in the skin. Granulomatous reactions typically manifest 2 weeks after injury as firm nonsuppurative nodules with central umbilication and a hyperkeratotic surface.7 These nodules may or may not be painful. Histopathology most often reveals foreign body and sarcoidal-type granulomatous reactions. However, tuberculoid, necrobiotic, and suppurative granulomas also may develop.13 Other microscopic features include inflammatory reactions, suppurative dermatitis, focal necrosis, and microabscesses.23 Wounds with progression to granulomatous disease often require surgical debridement.

Other more serious sequalae can result from involvement of joint capsules, especially in the hands and feet. Sea urchin injury involving joint spaces should be treated aggressively, as progression to inflammatory or infectious synovitis and tenosynovitis can cause irreversible loss of joint function. Inflammatory synovitis occurs 1 to 2 months on average after injury following a period of minimal symptoms and begins as a gradual increase in joint swelling and decrease in range of motion.8 Infectious tenosynovitis manifests quite similarly. Although suppurative etiologies generally progress with a more acute onset, certain infectious organisms (eg, Mycobacterium) take on an indolent course and should not be overlooked as a cause of delayed symptoms.8 The Kavanel cardinal signs are a sensitive tool used in the diagnosis of infectious flexor sheath tenosynovitis.8,24 If suspicion for joint infection is high, emergency referral should be made for debridement and culture-guided antibiotic therapy. Left untreated, infectious tenosynovitis can result in tendon necrosis or rupture, digit necrosis, and systemic infection.24 Patients with joint involvement should be referred to specialty care (eg, hand surgeon), as they often require synovectomy and surgical removal of foreign material.8

From 1 month to 1 year after injury, prolonged granulomatous synovitis of the hand may eventually lead to joint destruction known as “sea urchin arthritis.” These patients present with decreased range of motion and numerous nodules on the hand with a hyperkeratotic surface. Radiography reveals joint space narrowing, osteolysis, subchondral sclerosis, and periosteal reaction. Synovectomy and debridement are necessary to prevent irreversible joint damage or the need for arthrodesis and bone grafting.24

Other Treatment Considerations

Other important considerations in the care of sea urchin spine injuries include assessment of tetanus immunization status and administration of necessary prophylaxis as soon as possible, even in delayed presentations (Table).16,25 Cultures should be taken only if infection is suspected. Prophylactic antibiotics are not recommended unless the patient is immunocompromised or otherwise has impaired wound healing. If a patient presents with systemic symptoms, they should be referred to an emergency care facility for further management.

Final Thoughts

Sea urchin injuries can lead to serious complications if not diagnosed quickly and treated properly. Retention of sea urchin spines in the deep tissues and joint spaces may lead to granulomas, inflammatory and infectious tenosynovitis (including mycobacterial infection), and sea urchin arthritis requiring surgical debridement and possible irreversible joint damage, up to a year after initial injury. Patients should be educated on the possibility of developing these delayed reactions and instructed to seek immediate care. Joint deformities, range-of-motion deficits, and involvement of neurovascular structures should be considered emergent and referred for proper management. Shoes and diving gear offer some protection but are easily penetrable by sharp sea urchin spines. Preventive focus should be aimed at educating patients and providers on the importance of prompt spine removal upon injury. Although dermatologic and systemic manifestations vary widely, a thorough history, physical examination, and appropriate use of imaging modalities can facilitate accurate diagnosis and guide treatment.

midetawucrislasastumitheprutradreprohijupedupuspeshaciphomistemicruslifriclupridiwritoslafrophajaheswecretruchechewefrowrouinemasohubrotuphiphutaslothuwrisow

References
  1. Amemiya CT, Miyake T, Rast JP. Echinoderms. Curr Biol. 2005;15:R944-R946. doi:10.1016/j.cub.2005.11.026
  2. Koch NM, Coppard SE, Lessios HA, et al. A phylogenomic resolution of the sea urchin tree of life. BMC Evol Biol. 2018;18:189. doi:10.1186/s12862-018-1300-4
  3. Amir Y, Insler M, Giller A, et al. Senescence and longevity of sea urchins. Genes (Basel). 2020;11:573. doi:10.3390/genes11050573
  4. Gummin DD, Mowry JB, Beuhler MC, et al. 2022 Annual Report of the National Poison Data System® (NPDS) from America's Poison Centers®: 40th annual report. Clin Toxicol (Phila). 2023;61:717-939. doi:10.1080/15563650.2023.2268981
  5. Gelman Y, Kong EL, Murphy-Lavoie HM. Sea urchin toxicity. In: StatPearls [Internet]. StatPearls Publishing; 2021.
  6. Suarez-Conde MF, Vallone MG, González VM, et al. Sea urchin skin lesions: a case report. Dermatol Pract Concept. 2021;11:E2021009. doi:10.5826/dpc.1102a09
  7. Al-Kathiri L, Al-Najjar T, Sulaiman I. Sea urchin granuloma of the hands: a case report. Oman Med J. 2019;34:350-353. doi:10.5001/omj.2019.68
  8. Dahl WJ, Jebson P, Louis DS. Sea urchin injuries to the hand: a case report and review of the literature. Iowa Orthop J. 2010;30:153-156.
  9. Hatakeyama T, Ichise A, Unno H, et al. Carbohydrate recognition by the rhamnose-binding lectin SUL-I with a novel three-domain structure isolated from the venom of globiferous pedicellariae of the flower sea urchin Toxopneustes pileolus. Protein Sci. 2017;26:1574-1583. doi:10.1002/pro.3185
  10. Balhara KS, Stolbach A. Marine envenomations. Emerg Med Clin North Am. 2014;32:223-243. doi:10.1016/j.emc.2013.09.009
  11. Schwartz Z, Cohen M, Lipner SR. Sea urchin injuries: a review and clinical approach algorithm. J Dermatolog Treat. 2021;32:150-156. doi:10.1080/09546634.2019.1638884
  12. Park SJ, Park JW, Choi SY, et al. Use of dermoscopy after punch removal of a veiled sea urchin spine. Dermatol Ther. 2021;34:E14947. doi:10.1111/dth.14947
  13. Wada T, Soma T, Gaman K, et al. Sea urchin spine arthritis of the hand. J Hand Surg Am. 2008;33:398-401. doi:10.1016/j.jhsa.2007.11.016
  14. Groleau S, Chhem RK, Younge D, et al. Ultrasonography of foreign-body tenosynovitis. Can Assoc Radiol J. 1992;43:454-456. 
  15. Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337. doi:10.1016/j.emc.2016.12.004
  16. Noonburg GE. Management of extremity trauma and related infections occurring in the aquatic environment. J Am Acad Orthop Surg. 2005;13:243-253. doi:10.5435/00124635-200507000-00004
  17. Haddad Junior V. Observation of initial clinical manifestations and repercussions from the treatment of 314 human injuries caused by black sea urchins (Echinometra lucunter) on the southeastern Brazilian coast. Rev Soc Bras Med Trop. 2012;45:390-392. doi:10.1590/s0037-86822012000300021
  18. Gargus MD, Morohashi DK. A sea-urchin spine chilling remedy. N Engl J Med. 2012;367:1867-1868. doi:10.1056/NEJMc1209382
  19. Sjøberg T, de Weerd L. The usefulness of a skin biopsy punch to remove sea urchin spines. ANZ J Surg. 2010;80:383. doi:10.1111/j.1445-2197.2010.05296.x
  20. Cardenas-de la Garza JA, Cuellar-Barboza A, Ancer-Arellano J, et al. Classic dermatological tools: foreign body removal with punch biopsy.J Am Acad Dermatol. 2019;81:E93-E94. doi:10.1016/j.jaad.2018.10.038
  21. Gungor S, Tarikçi N, Gokdemir G. Removal of sea urchin spines using erbium-doped yttrium aluminum garnet ablation. Dermatol Surg. 2012;38:508-510. doi:10.1111/j.1524-4725.2011.02259.x
  22. Böer A, Ochsendorf FR, Beier C, et al. Effective removal of sea-urchin spines by erbium:YAG laser ablation. Br J Dermatol. 2001;145:169-170. doi:10.1046/j.1365-2133.2001.04306.x
  23. De La Torre C, Toribio J. Sea-urchin granuloma: histologic profile. a pathologic study of 50 biopsies. J Cutan Pathol. 2001;28:223-228. doi:10.1034/j.1600-0560.2001.028005223.x
  24. Yi A, Kennedy C, Chia B, et al. Radiographic soft tissue thickness differentiating pyogenic flexor tenosynovitis from other finger infections. J Hand Surg Am. 2019;44:394-399. doi:10.1016/j.jhsa.2019.01.013
  25. Callison C, Nguyen H. Tetanus prophylaxis. In: StatPearls [Internet]. StatPearls Publishing; 2022.
References
  1. Amemiya CT, Miyake T, Rast JP. Echinoderms. Curr Biol. 2005;15:R944-R946. doi:10.1016/j.cub.2005.11.026
  2. Koch NM, Coppard SE, Lessios HA, et al. A phylogenomic resolution of the sea urchin tree of life. BMC Evol Biol. 2018;18:189. doi:10.1186/s12862-018-1300-4
  3. Amir Y, Insler M, Giller A, et al. Senescence and longevity of sea urchins. Genes (Basel). 2020;11:573. doi:10.3390/genes11050573
  4. Gummin DD, Mowry JB, Beuhler MC, et al. 2022 Annual Report of the National Poison Data System® (NPDS) from America's Poison Centers®: 40th annual report. Clin Toxicol (Phila). 2023;61:717-939. doi:10.1080/15563650.2023.2268981
  5. Gelman Y, Kong EL, Murphy-Lavoie HM. Sea urchin toxicity. In: StatPearls [Internet]. StatPearls Publishing; 2021.
  6. Suarez-Conde MF, Vallone MG, González VM, et al. Sea urchin skin lesions: a case report. Dermatol Pract Concept. 2021;11:E2021009. doi:10.5826/dpc.1102a09
  7. Al-Kathiri L, Al-Najjar T, Sulaiman I. Sea urchin granuloma of the hands: a case report. Oman Med J. 2019;34:350-353. doi:10.5001/omj.2019.68
  8. Dahl WJ, Jebson P, Louis DS. Sea urchin injuries to the hand: a case report and review of the literature. Iowa Orthop J. 2010;30:153-156.
  9. Hatakeyama T, Ichise A, Unno H, et al. Carbohydrate recognition by the rhamnose-binding lectin SUL-I with a novel three-domain structure isolated from the venom of globiferous pedicellariae of the flower sea urchin Toxopneustes pileolus. Protein Sci. 2017;26:1574-1583. doi:10.1002/pro.3185
  10. Balhara KS, Stolbach A. Marine envenomations. Emerg Med Clin North Am. 2014;32:223-243. doi:10.1016/j.emc.2013.09.009
  11. Schwartz Z, Cohen M, Lipner SR. Sea urchin injuries: a review and clinical approach algorithm. J Dermatolog Treat. 2021;32:150-156. doi:10.1080/09546634.2019.1638884
  12. Park SJ, Park JW, Choi SY, et al. Use of dermoscopy after punch removal of a veiled sea urchin spine. Dermatol Ther. 2021;34:E14947. doi:10.1111/dth.14947
  13. Wada T, Soma T, Gaman K, et al. Sea urchin spine arthritis of the hand. J Hand Surg Am. 2008;33:398-401. doi:10.1016/j.jhsa.2007.11.016
  14. Groleau S, Chhem RK, Younge D, et al. Ultrasonography of foreign-body tenosynovitis. Can Assoc Radiol J. 1992;43:454-456. 
  15. Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337. doi:10.1016/j.emc.2016.12.004
  16. Noonburg GE. Management of extremity trauma and related infections occurring in the aquatic environment. J Am Acad Orthop Surg. 2005;13:243-253. doi:10.5435/00124635-200507000-00004
  17. Haddad Junior V. Observation of initial clinical manifestations and repercussions from the treatment of 314 human injuries caused by black sea urchins (Echinometra lucunter) on the southeastern Brazilian coast. Rev Soc Bras Med Trop. 2012;45:390-392. doi:10.1590/s0037-86822012000300021
  18. Gargus MD, Morohashi DK. A sea-urchin spine chilling remedy. N Engl J Med. 2012;367:1867-1868. doi:10.1056/NEJMc1209382
  19. Sjøberg T, de Weerd L. The usefulness of a skin biopsy punch to remove sea urchin spines. ANZ J Surg. 2010;80:383. doi:10.1111/j.1445-2197.2010.05296.x
  20. Cardenas-de la Garza JA, Cuellar-Barboza A, Ancer-Arellano J, et al. Classic dermatological tools: foreign body removal with punch biopsy.J Am Acad Dermatol. 2019;81:E93-E94. doi:10.1016/j.jaad.2018.10.038
  21. Gungor S, Tarikçi N, Gokdemir G. Removal of sea urchin spines using erbium-doped yttrium aluminum garnet ablation. Dermatol Surg. 2012;38:508-510. doi:10.1111/j.1524-4725.2011.02259.x
  22. Böer A, Ochsendorf FR, Beier C, et al. Effective removal of sea-urchin spines by erbium:YAG laser ablation. Br J Dermatol. 2001;145:169-170. doi:10.1046/j.1365-2133.2001.04306.x
  23. De La Torre C, Toribio J. Sea-urchin granuloma: histologic profile. a pathologic study of 50 biopsies. J Cutan Pathol. 2001;28:223-228. doi:10.1034/j.1600-0560.2001.028005223.x
  24. Yi A, Kennedy C, Chia B, et al. Radiographic soft tissue thickness differentiating pyogenic flexor tenosynovitis from other finger infections. J Hand Surg Am. 2019;44:394-399. doi:10.1016/j.jhsa.2019.01.013
  25. Callison C, Nguyen H. Tetanus prophylaxis. In: StatPearls [Internet]. StatPearls Publishing; 2022.
Issue
Cutis - 113(6)
Issue
Cutis - 113(6)
Page Number
255-257
Page Number
255-257
Publications
Publications
Topics
Article Type
Display Headline
Aquatic Antagonists: Dermatologic Injuries From Sea Urchins (Echinoidea)
Display Headline
Aquatic Antagonists: Dermatologic Injuries From Sea Urchins (Echinoidea)
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Brailsford</fileName> <TBEID>0C02F821.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F821</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Brailsford</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240614T092858</firstPublished> <LastPublished>20240614T092858</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240614T092857</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Caroline J. Brailsford, MD; Dirk M. Elston, MD</byline> <bylineText>Caroline J. Brailsford, MD; Dirk M. Elston, MD</bylineText> <bylineFull>Caroline J. Brailsford, MD; Dirk M. Elston, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>255-257</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Sea urchins—members of the phylum Echinodermata and the class Echinoidea—are spiny marine invertebrates. Their consumption of fleshy algae makes them essential </metaDescription> <articlePDF>301780</articlePDF> <teaserImage/> <title>Aquatic Antagonists: Dermatologic Injuries From Sea Urchins (Echinoidea)</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>June</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>6</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2159</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>June 2024</pubIssueName> <pubArticleType>Departments | 2159</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">60</term> </sections> <topics> <term canonical="true">27442</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/1800274b.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Aquatic Antagonists: Dermatologic Injuries From Sea Urchins (Echinoidea)</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Sea urchin injuries are common following accidental contact with sharp sea urchin spines. Immediate manifestations of injury include local erythema, pain, and myalgia. Failure to remove the spines from the skin may result in delayed systemic reactions, secondary infection, granulomas, and—if joint spaces are involved—inflammatory or infectious synovitis and arthritis. The majority of severe complications can be avoided if the spines are fully removed from the skin soon after injury, which can be difficult. This article aims to bring awareness to the myriad complications from sea urchin injuries as well as the mechanisms for successful spine removal.</p> <p>Sea urchins—members of the phylum Echinodermata and the class Echinoidea—are spiny marine invertebrates. Their consumption of fleshy algae makes them essential players in maintaining reef ecosystems.<sup>1,2</sup> Echinoids, a class that includes heart urchins and sand dollars, are ubiquitous in benthic marine environments, both free floating and rock boring, and inhabit a wide range of latitudes spanning from polar oceans to warm seas.<sup>3</sup> Despite their immobility and nonaggression, sea urchin puncture wounds are common among divers, snorkelers, swimmers, surfers, and fishers who accidentally come into contact with their sharp spines. Although the epidemiology of sea urchin exposure and injury is difficult to assess, the American Association of Poison Control Centers’ most recent annual report in 2022 documents approximately 1426 annual aquatic bites and/or envenomations.<sup>4</sup></p> <h3>Sea Urchin Morphology and Toxicity</h3> <p>Echinoderms (a term of Greek origin meaning spiny skin) share a radially symmetric calcium carbonate skeleton (termed <i>stereom</i>) that is supported by collagenous ligaments.<sup>1</sup> Sea urchins possess spines composed of calcite crystals, which radiate from their body and play a role in locomotion and defense against predators—namely sea otters, starfish/sea stars, wolf eels, and triggerfish, among others (Figure).<sup>5</sup> These brittle spines can easily penetrate human skin and subsequently break off the sea urchin body. Most species of sea urchins possess solid spines, but a small percentage (80 of approximately 700 extant species) have hollow spines containing various toxic substances.<sup>6</sup> Penetration and systemic absorption of the toxins within these spines can generate severe systemic responses.</p> <p>The venomous flower urchin (<i>Toxopneustes pileolus</i>), found in the Indian and Pacific oceans, is one of the more common species known to produce a systemic reaction involving neuromuscular blockage.<sup>7-9</sup> The most common species harvested off the Pacific coast of the United States—<i>Strongylocentrotus purpuratus</i> (purple sea urchin) and <i>Strongylocentrotus franciscanus</i> (red sea urchins)—are not inherently venomous.<sup>8<br/><br/></sup>Both the sea urchin body and spines are covered in a unique epithelium thought to be responsible for the majority of their proinflammatory and pronociceptive properties. Epithelial compounds identified include serotonin, histamines, steroids, glycosides, hemolysins, proteases, and bradykininlike and cholinergic substances.<sup>5,7</sup> Additionally, certain sea urchin species possess 3-pronged pincerlike organs at the base of spines called pedicellariae, which are used in feeding.<sup>10</sup> Skin penetration by the pedicellariae is especially dangerous, as they tightly adhere to wounds and contain venom-producing organs that allow them to continue injecting toxins after their detachment from the sea urchin body.<sup>11</sup> </p> <h3>Presentation and Diagnosis of Sea Urchin Injuries</h3> <p>Sea urchin injuries have a wide range of manifestations depending on the number of spines involved, the presence of venom, the depth and location of spine penetration, the duration of spine retention in the skin, and the time before treatment initiation. The most common site of sea urchin injury unsurprisingly is the lower extremities and feet, often in the context of divers and swimmers walking across the sea floor. The hands are another frequently injured site, along with the legs, arms, back, scalp, and even oral mucosa.<sup>11</sup> </p> <p>Although clinical history and presentation frequently reveal the mechanism of aquatic injury, patients often are unsure of the agent to which they were exposed and may be unaware of retained foreign bodies. Dermoscopy can distinguish the distinct lines radiating from the core of sea urchin spines from other foreign bodies lodged within the skin.<sup>6</sup> It also can be used to locate spines for removal or for their analysis following punch biopsy.<sup>6,12 </sup>The radiopaque nature of sea urchin spines makes radiography and magnetic resonance imaging useful tools in assessment of periarticular soft-tissue damage and spine removal.<sup>8,11,13</sup> Ultrasonography can reveal spines that no longer appear on radiography due to absorption by human tissue.<sup>14</sup> </p> <h3>Immediate Dermatologic Effects</h3> <p>Sea urchin injuries can be broadly categorized into immediate and delayed reactions. Immediate manifestations of contact with sea urchin spines include localized pain, bleeding, erythema, myalgia, and edema at the site of injury that can last from a few hours to 1 week without proper wound care and spine removal.<sup>5</sup> Systemic symptoms ranging from dizziness, lightheadedness, paresthesia, aphonia, paralysis, coma, and death generally are only seen following injuries from venomous species, attachment of pedicellariae, injuries involving neurovascular structures, or penetration by more than 15 spines.<sup>7,11</sup> </p> <p>Initial treatment includes soaking the wound in hot water (113 <span class="body">°</span>F [45 <span class="body">°</span>C]) for 30 to 90 minutes and subsequently removing spines and pedicellariae to prevent development of delayed reactions.<sup>5,15,16</sup> The compounds in the sea urchin epithelium are heat labile and will be inactivated upon soaking in hot water.<sup>16</sup> Extraction of spines can be difficult, as they are brittle and easily break in the skin. Successful removal has been reported using forceps and a hypodermic needle as well as excision; both approaches may require local anesthesia.<sup>8,17</sup> Another technique involves freezing the localized area with liquid nitrogen to allow easier removal upon skin blistering.<sup>18</sup> Punch biopsy also has been utilized as an effective means of ensuring all spiny fragments are removed.<sup>9,19,20</sup> These spines often cause black or purple tattoolike staining at the puncture site, which can persist for a few days after spine extraction.<sup>8</sup> Ablation using the erbium-doped:YAG laser may be helpful for removal of associated pigment.<sup>21,22</sup></p> <h3>Delayed Dermatologic Effects</h3> <p>Delayed reactions to sea urchin injuries often are attributable to prolonged retention of spines in the skin. Granulomatous reactions typically manifest 2 weeks after injury as firm nonsuppurative nodules with central umbilication and a hyperkeratotic surface.<sup>7</sup> These nodules may or may not be painful. Histopathology most often reveals foreign body and sarcoidal-type granulomatous reactions. However, tuberculoid, necrobiotic, and suppurative granulomas also may develop.<sup>13</sup> Other microscopic features include inflammatory reactions, suppurative dermatitis, focal necrosis, and microabscesses.<sup>23</sup> Wounds with progression to granulomatous disease often require surgical debridement.</p> <p>Other more serious sequalae can result from involvement of joint capsules, especially in the hands and feet. Sea urchin injury involving joint spaces should be treated aggressively, as progression to inflammatory or infectious synovitis and tenosynovitis can cause irreversible loss of joint function. Inflammatory synovitis occurs 1 to 2 months on average after injury following a period of minimal symptoms and begins as a gradual increase in joint swelling and decrease in range of motion.<sup>8</sup> Infectious tenosynovitis manifests quite similarly. Although suppurative etiologies generally progress with a more acute onset, certain infectious organisms (eg, <i>Mycobacterium</i>) take on an indolent course and should not be overlooked as a cause of delayed symptoms.<sup>8</sup> The Kavanel cardinal signs are a sensitive tool used in the diagnosis of infectious flexor sheath tenosynovitis.<sup>8,24</sup> If suspicion for joint infection is high, emergency referral should be made for debridement and culture-guided antibiotic therapy. Left untreated, infectious tenosynovitis can result in tendon necrosis or rupture, digit necrosis, and systemic infection.<sup>24</sup> Patients with joint involvement should be referred to specialty care (eg, hand surgeon), as they often require synovectomy and surgical removal of foreign material.<sup>8<br/><br/></sup>From 1 month to 1 year after injury, prolonged granulomatous synovitis of the hand may eventually lead to joint destruction known as “sea urchin arthritis.” These patients present with decreased range of motion and numerous nodules on the hand with a hyperkeratotic surface. Radiography reveals joint space narrowing, osteolysis, subchondral sclerosis, and periosteal reaction. Synovectomy and debridement are necessary to prevent irreversible joint damage or the need for arthrodesis and bone grafting.<sup>24</sup> </p> <h3>Other Treatment Considerations</h3> <p>Other important considerations in the care of sea urchin spine injuries include assessment of tetanus immunization status and administration of necessary prophylaxis as soon as possible, even in delayed presentations (Table).<sup>16,25</sup> Cultures should be taken only if infection is suspected. Prophylactic antibiotics are not recommended unless the patient is immunocompromised or otherwise has impaired wound healing. If a patient presents with systemic symptoms, they should be referred to an emergency care facility for further management.</p> <h3>Final Thoughts</h3> <p>Sea urchin injuries can lead to serious complications if not diagnosed quickly and treated properly. Retention of sea urchin spines in the deep tissues and joint spaces may lead to granulomas, inflammatory and infectious tenosynovitis (including mycobacterial infection), and sea urchin arthritis requiring surgical debridement and possible irreversible joint damage, up to a year after initial injury. Patients should be educated on the possibility of developing these delayed reactions and instructed to seek immediate care. Joint deformities, range-of-motion deficits, and involvement of neurovascular structures should be considered emergent and referred for proper management. Shoes and diving gear offer some protection but are easily penetrable by sharp sea urchin spines. Preventive focus should be aimed at educating patients and providers on the importance of prompt spine removal upon injury. Although dermatologic and systemic manifestations vary widely, a thorough history, physical examination, and appropriate use of imaging modalities can facilitate accurate diagnosis and guide treatment. </p> <h2>References</h2> <p class="reference"> 1. Amemiya CT, Miyake T, Rast JP. Echinoderms. <i>Curr Biol.</i> 2005;15:R944-R946. doi:10.1016/j.cub.2005.11.026</p> <p class="reference"> 2. Koch NM, Coppard SE, Lessios HA, et al. A phylogenomic resolution of the sea urchin tree of life. <i>BMC Evol Biol.</i> 2018;18:189. doi:<span class="citation-doi">10.1186/s12862-018-1300-4<br/><br/></span> 3. Amir Y, Insler M, Giller A, et al. Senescence and longevity of sea urchins. <i>Genes (Basel).</i> 2020;11:573. doi:10.3390/genes11050573<br/><br/> 4. Gummin DD, Mowry JB, Beuhler MC, et al. 2022 Annual Report of the National Poison Data System<sup>®</sup> (NPDS) from America's Poison Centers<sup>®</sup>: 40th annual report. <i>Clin Toxicol (Phila).</i> 2023;61:717-939. doi:10.1080/15563650.2023.2268981 <br/><br/> 5. Gelman Y, Kong EL, Murphy-Lavoie HM. Sea urchin toxicity. In: <i>StatPearls</i> [Internet]. StatPearls Publishing; 2021.<br/><br/> 6. Suarez-Conde MF, Vallone MG, González VM, et al. Sea urchin skin lesions: a case report. <i>Dermatol Pract Concept.</i> 2021;11:E2021009. doi:10.5826/dpc.1102a09<br/><br/> 7. Al-Kathiri L, Al-Najjar T, Sulaiman I. Sea urchin granuloma of the hands: a case report. <i>Oman Med J.</i> 2019;34:350-353. doi:10.5001/omj.2019.68<br/><br/> 8. Dahl WJ, Jebson P, Louis DS. Sea urchin injuries to the hand: a case report and review of the literature. <i>Iowa Orthop J</i>. 2010;30:153-156.<br/><br/> 9. Hatakeyama T, Ichise A, Unno H, et al. Carbohydrate recognition by the rhamnose-binding lectin SUL-I with a novel three-domain structure isolated from the venom of globiferous pedicellariae of the flower sea urchin <i>Toxopneustes pileolus</i>. <i>Protein Sci</i>. 2017;26:1574-1583. doi:10.1002/pro.3185<br/><br/>10. Balhara KS, Stolbach A. Marine envenomations. <i>Emerg Med Clin North Am.</i> 2014;32:223-243. doi:10.1016/j.emc.2013.09.009<br/><br/>11. Schwartz Z, Cohen M, Lipner SR. Sea urchin injuries: a review and clinical approach algorithm. <i>J Dermatolog Treat.</i> 2021;32:150-156. doi:10.1080/09546634.2019.1638884<br/><br/>12. Park SJ, Park JW, Choi SY, et al. Use of dermoscopy after punch removal of a veiled sea urchin spine. <i>Dermatol Ther.</i> 2021;34:E14947. doi:10.1111/dth.14947 <br/><br/>13. Wada T, Soma T, Gaman K, et al. Sea urchin spine arthritis of the hand. <i>J Hand Surg Am.</i> 2008;33:398-401. doi:10.1016/j.jhsa.2007.11.016<br/><br/>14. Groleau S, Chhem RK, Younge D, et al. Ultrasonography of foreign-body tenosynovitis. <i>Can Assoc Radiol J. </i>1992;43:454-456. <br/><br/>15. Hornbeak KB, Auerbach PS. Marine envenomation. <i>Emerg Med Clin North Am.</i> 2017;35:321-337. doi:10.1016/j.emc.2016.12.004<br/><br/>16. Noonburg GE. Management of extremity trauma and related infections occurring in the aquatic environment. <i>J Am Acad Orthop Surg.</i> 2005;13:243-253. doi:10.5435/00124635-200507000-00004<br/><br/>17. Haddad Junior V. Observation of initial clinical manifestations and repercussions from the treatment of 314 human injuries caused by black sea urchins (<i>Echinometra lucunter</i>) on the southeastern Brazilian coast. <i>Rev Soc Bras Med Trop</i>. 2012;45:390-392. doi:10.1590/s0037-86822012000300021<br/><br/>18. Gargus MD, Morohashi DK. A sea-urchin spine chilling remedy. <i>N Engl J Med.</i> 2012;367:1867-1868. doi:10.1056/NEJMc1209382<br/><br/>19. Sjøberg T, de Weerd L. The usefulness of a skin biopsy punch to remove sea urchin spines. <i>ANZ J Surg</i>. 2010;80:383. doi:10.1111/j.1445-2197.2010.05296.x<br/><br/>20. Cardenas-de la Garza JA, Cuellar-Barboza A, Ancer-Arellano J, et al. Classic dermatological tools: foreign body removal with punch biopsy.<i>J Am Acad Dermatol.</i> 2019;81:E93-E94. doi:10.1016/j.jaad.2018.10.038<br/><br/>21. Gungor S, Tarikçi N, Gokdemir G. Removal of sea urchin spines using erbium-doped yttrium aluminum garnet ablation. <i>Dermatol Surg.</i> 2012;38:508-510. doi:10.1111/j.1524-4725.2011.02259.x<br/><br/>22. Böer A, Ochsendorf FR, Beier C, et al. Effective removal of sea-urchin spines by erbium:YAG laser ablation. <i>Br J Dermatol.</i> 2001;145:169-170. doi:10.1046/j.1365-2133.2001.04306.x<br/><br/>23. De La Torre C, Toribio J. Sea-urchin granuloma: histologic profile. a pathologic study of 50 biopsies. <i>J Cutan Pathol.</i> 2001;28:223-228. doi:10.1034/j.1600-0560.2001.028005223.x<br/><br/>24. Yi A, Kennedy C, Chia B, et al. Radiographic soft tissue thickness differentiating pyogenic flexor tenosynovitis from other finger infections. <i>J Hand Surg Am.</i> 2019;44:394-399. doi:10.1016/j.jhsa.2019.01.013<br/><br/>25. Callison C, Nguyen H. Tetanus prophylaxis. In: <i>StatPearls</i> [Internet]. StatPearls Publishing; 2022.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">From the Medical University of South Carolina, Charleston. Dr. Brailsford is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery. </p> <p class="disclosure">The authors report no conflict of interest.<br/><br/>Correspondence: Caroline J. Brailsford, MD, Medical University of South Carolina, 135 Rutledge Ave, 11th Floor, Charleston, SC 29425-5780 (cjbrailsford@gmail.com).<br/><br/><em>Cutis.</em> 2024 June;113(6):255-257. doi:10.12788/cutis.1034</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>Sea urchin spines easily become embedded in human skin upon contact and cause localized pain, edema, and black or purple pinpoint markings.</li> <li>Immediate treatment includes soaking in hot water (113 12<span class="body">°</span>F [45 12<span class="body">°</span>C]) for 30 to 90 minutes to inactivate proinflammatory compounds, followed by extraction of the spines.</li> <li>Successful methods of spine removal include the use of forceps and a hypodermic needle, as well as excision, liquid nitrogen, and punch biopsy. </li> <li>Prompt removal of the spines can reduce the incidence of delayed granulomatous reactions, synovitis, and sea urchin arthritis.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

 

Practice Points

  • Sea urchin spines easily become embedded in human skin upon contact and cause localized pain, edema, and black or purple pinpoint markings.
  • Immediate treatment includes soaking in hot water (113 12°F [45 12°C]) for 30 to 90 minutes to inactivate proinflammatory compounds, followed by extraction of the spines.
  • Successful methods of spine removal include the use of forceps and a hypodermic needle, as well as excision, liquid nitrogen, and punch biopsy.
  • Prompt removal of the spines can reduce the incidence of delayed granulomatous reactions, synovitis, and sea urchin arthritis.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Disable zoom
Off

Aquatic Antagonists: Seaweed Dermatitis (Lyngbya majuscula)

Article Type
Changed
Fri, 06/07/2024 - 11:04
Display Headline

Aquatic Antagonists: Seaweed Dermatitis (Lyngbya majuscula)

The filamentous cyanobacterium Lyngbya majuscula causes irritant contact dermatitis in beachgoers, fishers, and divers in tropical and subtropical marine environments worldwide.1 If fragments of L majuscula lodge in swimmers’ bathing suits, the toxins can become trapped against the skin and cause seaweed dermatitis.2 With climate change resulting in warmer oceans and more extreme storms, L majuscula blooms likely will become more frequent and widespread, thereby increasing the risk for human exposure.3,4 Herein, we describe the irritants that lead to dermatitis, clinical presentation, and prevention and management of seaweed dermatitis.

Identifying Features and Distribution of Plant

Lyngbya majuscula belongs to the family Oscillatoriaceae; these cyanobacteria grow as filaments and exhibit slow oscillating movements. Commonly referred to as blanketweed or mermaid’s hair due to its appearance, L majuscula grows fine hairlike clumps resembling a mass of olive-colored matted hair.1 Its thin filaments are 10- to 30-cm long and vary in color from red to white to brown.5 Microscopically, a rouleauxlike arrangement of discs provides the structure of each filament.6

First identified in Hawaii in 1912, L majuscula was not associated with seaweed dermatitis or dermatotoxicity by the medical community until the first outbreak occurred in Oahu in 1958, though fishermen and beachgoers previously had recognized a relationship between this particular seaweed and skin irritation.5,7 The first reporting included 125 confirmed cases, with many more mild unreported cases suspected.6 Now reported in about 100 locations worldwide, seaweed dermatitis outbreaks have occurred in Australia; Okinawa, Japan; Florida; and the Hawaiian and Marshall islands.1,2

Exposure to Seaweed

Lyngbya majuscula produces more than 70 biologically active compounds that irritate the skin, eyes, and respiratory system.2,8 It grows in marine and estuarine environments attached to seagrass, sand, and bedrock at depths of up to 30 m. Warm waters and maximal sunlight provide optimal growth conditions for L majuscula; therefore, the greatest risk for exposure occurs in the Northern and Southern hemispheres in the 1- to 2-month period following their summer solstices.5 Runoff during heavy rainfall, which is rich in soil extracts such as phosphorous, iron, and organic carbon, stimulates L majuscula growth and contributes to increased algal blooms.4

Dermatitis and Irritants

The dermatoxins Lyngbyatoxin A (LA) and debromoaplysiatoxin (DAT) cause the inflammatory and necrotic appearance of seaweed dermatitis.1,2,5,8 Lyngbyatoxin A is an indole alkaloid that is closely related to telocidin B, a poisonous compound associated with Streptomyces bacteria.9 Sampling of L majuscula and extraction of the dermatoxin, along with human and animal studies, confirmed DAT irritates the skin and induces dermatitis.5,6Stylocheilus longicauda (sea hare) feeds on L majuscula and contains isolates of DAT in its digestive tract.

Samples of L majuscula taken from several Hawaiian Islands where seaweed dermatitis outbreaks have occurred were examined for differences in toxicities via 6-hour patch tests on human skin.6 The samples obtained from the windward side of Oahu contained DAT and aplysiatoxin, while those obtained from the leeward side and Kahala Beach primarily contained LA. Although DAT and LA are vastly different in their molecular structures, testing elicited the same biologic response and induced the same level of skin irritation.6 Interestingly, not all strands of L majuscula produced LA and DAT and caused seaweed dermatitis; those that did lead to irritation were more red in color than nontoxic blooms.5,9

Cutaneous Manifestations

Seaweed dermatitis resembles chemical and thermal burns, ranging from a mild skin rash to severe contact dermatitis with itchy, swollen, ulcerated lesions.1,7 Patients typically develop a burning or itching sensation beneath their bathing suit or wetsuit that progresses to an erythematous papulovesicular eruption 2 to 24 hours after exposure.2,6 Within a week, vesicles and bullae desquamate, leaving behind tender erosions.1,2,6,8 Inframammary lesions are common in females and scrotal swelling in males.1,6 There is no known association between length of time spent in the water and severity of symptoms.5

Most reactions to L majuscula occur from exposure in the water; however, particles that become aerosolized during strong winds or storms can cause seaweed dermatitis on the face. Inhalation of L majuscula may lead to mucous membrane ulceration and pulmonary edema.1,5,6 Noncutaneous manifestations of seaweed dermatitis include headache, fatigue, and swelling of the eyes, nose, and throat (Figures 1 and 2).1,5

Prevention and Management

To prevent seaweed dermatitis, avoid swimming in ocean water during L majuscula blooms,10 which frequently occur following the summer solstices in the Northern and Southern hemispheres.5 The National Centers for Coastal Ocean Science Harmful Algae Bloom Monitoring System provides real-time access to algae bloom locations.11 Although this monitoring system is not specific to L majuscula, it may be helpful in determining where potential blooms are. Wearing protective clothing such as coveralls may benefit individuals who enter the water during blooms, but it does not guarantee protection.10

couagestemaclubacikofraswospetrauadraphibristochicreletocrecrodusispelithimathatethivodoswotivabegevofrushudrocovufrebrovepespiridedrawiwrephuladuphakohelaphukutrokekapre
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%20punch%20biopsy%20of%20an%20abdominal%20lesion%20in%20a%20patient%20with%20seaweed%20dermatitis%20(%3Cem%3ELyngbya%20majuscula%3C%2Fem%3E)%20showed%20an%20intraepidermal%20blister%20with%20superficial%20desquamation%20at%20the%20top%20(H%26amp%3BE%2C%20original%3Cbr%3Emagnification%20%C3%9740).%20Photograph%20courtesy%20of%20Scott%20Norton%2C%20MD%2C%20MPH%2C%20MSc%20(Washington%2C%20DC).%3C%2Fp%3E

theveslufrufrilitonosecihefroclufriuipephuwalustovigumespispewutradrophinunujepareprodushecrephohekiclah
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Classic%20erythematous%20papulovesicular%20rash%20on%20the%20abdomen%20of%20a%20patient%20with%20seaweed%20dermatitis%20(%3Cem%3ELyngbya%20majuscula%3C%2Fem%3E).%20Photograph%20courtesy%20of%20Scott%20Norton%2C%20MD%2C%20MPH%2C%20MSc%20(Washington%2C%20DC).%3C%2Fp%3E

Currently, there is no treatment for seaweed dermatitis, but symptom management may reduce discomfort and pain. Washing affected skin with soap and water within an hour of exposure may help reduce the severity of seaweed dermatitis, though studies have shown mixed results.6,7 Application of cool compresses and soothing ointments (eg, calamine) provide symptomatic relief and promote healing.7 The dermatitis typically self-resolves within 1 week.

References
  1. Werner K, Marquart L, Norton S. Lyngbya dermatitis (toxic seaweed dermatitis). Int J Dermatol. 2011;51:59-62. doi:10.1111/j.1365-4632.2011.05042.x
  2. Osborne N, Shaw G. Dermatitis associated with exposure to a marine cyanobacterium during recreational water exposure. BMC Dermatol. 2008;8:5. doi:10.1186/1471-5945-8-5
  3. Hays G, Richardson A, Robinson C. Climate change and marine plankton. Trends Ecol Evol. 2005;20:337-344. doi:10.1016/j.tree.2005.03.004
  4. Albert S, O’Neil J, Udy J, et al. Blooms of the cyanobacterium Lyngbya majuscula in costal Queensland, Australia: disparate sites, common factors. Mar Pollut Bull. 2004;51:428-437. doi:10.1016/j.marpolbul.2004.10.016
  5. Osborne N, Webb P, Shaw G. The toxins of Lyngbya majuscula and their human and ecological health effects. Environ Int. 2001;27:381-392. doi:10.1016/s0160-4120(01)00098-8
  6. Izumi A, Moore R. Seaweed ( Lyngbya majuscula ) dermatitis . Clin Dermatol . 1987;5:92-100. doi:10.1016/s0738-081x(87)80014-7
  7. Grauer F, Arnold H. Seaweed dermatitis: first report of a dermatitis-producing marine alga. Arch Dermatol. 1961; 84:720-732. doi:10.1001/archderm.1961.01580170014003
  8. Taylor M, Stahl-Timmins W, Redshaw C, et al. Toxic alkaloids in Lyngbya majuscula and related tropical marine cyanobacteria. Harmful Algae . 2014;31:1-8. doi:10.1016/j.hal.2013.09.003
  9. Cardellina J, Marner F, Moore R. Seaweed dermatitis: structure of lyngbyatoxin A. Science. 1979;204:193-195. doi:10.1126/science.107586
  10. Osborne N. Occupational dermatitis caused by Lyngbya majuscule in Australia. Int J Dermatol . 2012;5:122-123. doi:10.1111/j.1365-4632.2009.04455.x
  11. Harmful Algal Bloom Monitoring System. National Centers for Coastal Ocean Science. Accessed May 23, 2024. https://coastalscience.noaa.gov/research/stressor-impacts-mitigation/hab-monitoring-system/
Article PDF
Author and Disclosure Information

 

Dr. Hill is from the University of South Carolina School of Medicine, Greenville. Dr. Fulton is from Spartanburg Regional Medical Center, South Carolina. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.

The authors report no conflict of interest.

The images are in the public domain.

Correspondence: Kathleen L. Hill, MD, 607 Grove Rd, Greenville, SC 29605 (klhill@email.sc.edu).

Cutis. 2024 May;113(5):E38-E40. doi:10.12788/cutis.1032

Issue
Cutis - 113(5)
Publications
Topics
Page Number
E38-E40
Sections
Author and Disclosure Information

 

Dr. Hill is from the University of South Carolina School of Medicine, Greenville. Dr. Fulton is from Spartanburg Regional Medical Center, South Carolina. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.

The authors report no conflict of interest.

The images are in the public domain.

Correspondence: Kathleen L. Hill, MD, 607 Grove Rd, Greenville, SC 29605 (klhill@email.sc.edu).

Cutis. 2024 May;113(5):E38-E40. doi:10.12788/cutis.1032

Author and Disclosure Information

 

Dr. Hill is from the University of South Carolina School of Medicine, Greenville. Dr. Fulton is from Spartanburg Regional Medical Center, South Carolina. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.

The authors report no conflict of interest.

The images are in the public domain.

Correspondence: Kathleen L. Hill, MD, 607 Grove Rd, Greenville, SC 29605 (klhill@email.sc.edu).

Cutis. 2024 May;113(5):E38-E40. doi:10.12788/cutis.1032

Article PDF
Article PDF

The filamentous cyanobacterium Lyngbya majuscula causes irritant contact dermatitis in beachgoers, fishers, and divers in tropical and subtropical marine environments worldwide.1 If fragments of L majuscula lodge in swimmers’ bathing suits, the toxins can become trapped against the skin and cause seaweed dermatitis.2 With climate change resulting in warmer oceans and more extreme storms, L majuscula blooms likely will become more frequent and widespread, thereby increasing the risk for human exposure.3,4 Herein, we describe the irritants that lead to dermatitis, clinical presentation, and prevention and management of seaweed dermatitis.

Identifying Features and Distribution of Plant

Lyngbya majuscula belongs to the family Oscillatoriaceae; these cyanobacteria grow as filaments and exhibit slow oscillating movements. Commonly referred to as blanketweed or mermaid’s hair due to its appearance, L majuscula grows fine hairlike clumps resembling a mass of olive-colored matted hair.1 Its thin filaments are 10- to 30-cm long and vary in color from red to white to brown.5 Microscopically, a rouleauxlike arrangement of discs provides the structure of each filament.6

First identified in Hawaii in 1912, L majuscula was not associated with seaweed dermatitis or dermatotoxicity by the medical community until the first outbreak occurred in Oahu in 1958, though fishermen and beachgoers previously had recognized a relationship between this particular seaweed and skin irritation.5,7 The first reporting included 125 confirmed cases, with many more mild unreported cases suspected.6 Now reported in about 100 locations worldwide, seaweed dermatitis outbreaks have occurred in Australia; Okinawa, Japan; Florida; and the Hawaiian and Marshall islands.1,2

Exposure to Seaweed

Lyngbya majuscula produces more than 70 biologically active compounds that irritate the skin, eyes, and respiratory system.2,8 It grows in marine and estuarine environments attached to seagrass, sand, and bedrock at depths of up to 30 m. Warm waters and maximal sunlight provide optimal growth conditions for L majuscula; therefore, the greatest risk for exposure occurs in the Northern and Southern hemispheres in the 1- to 2-month period following their summer solstices.5 Runoff during heavy rainfall, which is rich in soil extracts such as phosphorous, iron, and organic carbon, stimulates L majuscula growth and contributes to increased algal blooms.4

Dermatitis and Irritants

The dermatoxins Lyngbyatoxin A (LA) and debromoaplysiatoxin (DAT) cause the inflammatory and necrotic appearance of seaweed dermatitis.1,2,5,8 Lyngbyatoxin A is an indole alkaloid that is closely related to telocidin B, a poisonous compound associated with Streptomyces bacteria.9 Sampling of L majuscula and extraction of the dermatoxin, along with human and animal studies, confirmed DAT irritates the skin and induces dermatitis.5,6Stylocheilus longicauda (sea hare) feeds on L majuscula and contains isolates of DAT in its digestive tract.

Samples of L majuscula taken from several Hawaiian Islands where seaweed dermatitis outbreaks have occurred were examined for differences in toxicities via 6-hour patch tests on human skin.6 The samples obtained from the windward side of Oahu contained DAT and aplysiatoxin, while those obtained from the leeward side and Kahala Beach primarily contained LA. Although DAT and LA are vastly different in their molecular structures, testing elicited the same biologic response and induced the same level of skin irritation.6 Interestingly, not all strands of L majuscula produced LA and DAT and caused seaweed dermatitis; those that did lead to irritation were more red in color than nontoxic blooms.5,9

Cutaneous Manifestations

Seaweed dermatitis resembles chemical and thermal burns, ranging from a mild skin rash to severe contact dermatitis with itchy, swollen, ulcerated lesions.1,7 Patients typically develop a burning or itching sensation beneath their bathing suit or wetsuit that progresses to an erythematous papulovesicular eruption 2 to 24 hours after exposure.2,6 Within a week, vesicles and bullae desquamate, leaving behind tender erosions.1,2,6,8 Inframammary lesions are common in females and scrotal swelling in males.1,6 There is no known association between length of time spent in the water and severity of symptoms.5

Most reactions to L majuscula occur from exposure in the water; however, particles that become aerosolized during strong winds or storms can cause seaweed dermatitis on the face. Inhalation of L majuscula may lead to mucous membrane ulceration and pulmonary edema.1,5,6 Noncutaneous manifestations of seaweed dermatitis include headache, fatigue, and swelling of the eyes, nose, and throat (Figures 1 and 2).1,5

Prevention and Management

To prevent seaweed dermatitis, avoid swimming in ocean water during L majuscula blooms,10 which frequently occur following the summer solstices in the Northern and Southern hemispheres.5 The National Centers for Coastal Ocean Science Harmful Algae Bloom Monitoring System provides real-time access to algae bloom locations.11 Although this monitoring system is not specific to L majuscula, it may be helpful in determining where potential blooms are. Wearing protective clothing such as coveralls may benefit individuals who enter the water during blooms, but it does not guarantee protection.10

couagestemaclubacikofraswospetrauadraphibristochicreletocrecrodusispelithimathatethivodoswotivabegevofrushudrocovufrebrovepespiridedrawiwrephuladuphakohelaphukutrokekapre
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%20punch%20biopsy%20of%20an%20abdominal%20lesion%20in%20a%20patient%20with%20seaweed%20dermatitis%20(%3Cem%3ELyngbya%20majuscula%3C%2Fem%3E)%20showed%20an%20intraepidermal%20blister%20with%20superficial%20desquamation%20at%20the%20top%20(H%26amp%3BE%2C%20original%3Cbr%3Emagnification%20%C3%9740).%20Photograph%20courtesy%20of%20Scott%20Norton%2C%20MD%2C%20MPH%2C%20MSc%20(Washington%2C%20DC).%3C%2Fp%3E

theveslufrufrilitonosecihefroclufriuipephuwalustovigumespispewutradrophinunujepareprodushecrephohekiclah
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Classic%20erythematous%20papulovesicular%20rash%20on%20the%20abdomen%20of%20a%20patient%20with%20seaweed%20dermatitis%20(%3Cem%3ELyngbya%20majuscula%3C%2Fem%3E).%20Photograph%20courtesy%20of%20Scott%20Norton%2C%20MD%2C%20MPH%2C%20MSc%20(Washington%2C%20DC).%3C%2Fp%3E

Currently, there is no treatment for seaweed dermatitis, but symptom management may reduce discomfort and pain. Washing affected skin with soap and water within an hour of exposure may help reduce the severity of seaweed dermatitis, though studies have shown mixed results.6,7 Application of cool compresses and soothing ointments (eg, calamine) provide symptomatic relief and promote healing.7 The dermatitis typically self-resolves within 1 week.

The filamentous cyanobacterium Lyngbya majuscula causes irritant contact dermatitis in beachgoers, fishers, and divers in tropical and subtropical marine environments worldwide.1 If fragments of L majuscula lodge in swimmers’ bathing suits, the toxins can become trapped against the skin and cause seaweed dermatitis.2 With climate change resulting in warmer oceans and more extreme storms, L majuscula blooms likely will become more frequent and widespread, thereby increasing the risk for human exposure.3,4 Herein, we describe the irritants that lead to dermatitis, clinical presentation, and prevention and management of seaweed dermatitis.

Identifying Features and Distribution of Plant

Lyngbya majuscula belongs to the family Oscillatoriaceae; these cyanobacteria grow as filaments and exhibit slow oscillating movements. Commonly referred to as blanketweed or mermaid’s hair due to its appearance, L majuscula grows fine hairlike clumps resembling a mass of olive-colored matted hair.1 Its thin filaments are 10- to 30-cm long and vary in color from red to white to brown.5 Microscopically, a rouleauxlike arrangement of discs provides the structure of each filament.6

First identified in Hawaii in 1912, L majuscula was not associated with seaweed dermatitis or dermatotoxicity by the medical community until the first outbreak occurred in Oahu in 1958, though fishermen and beachgoers previously had recognized a relationship between this particular seaweed and skin irritation.5,7 The first reporting included 125 confirmed cases, with many more mild unreported cases suspected.6 Now reported in about 100 locations worldwide, seaweed dermatitis outbreaks have occurred in Australia; Okinawa, Japan; Florida; and the Hawaiian and Marshall islands.1,2

Exposure to Seaweed

Lyngbya majuscula produces more than 70 biologically active compounds that irritate the skin, eyes, and respiratory system.2,8 It grows in marine and estuarine environments attached to seagrass, sand, and bedrock at depths of up to 30 m. Warm waters and maximal sunlight provide optimal growth conditions for L majuscula; therefore, the greatest risk for exposure occurs in the Northern and Southern hemispheres in the 1- to 2-month period following their summer solstices.5 Runoff during heavy rainfall, which is rich in soil extracts such as phosphorous, iron, and organic carbon, stimulates L majuscula growth and contributes to increased algal blooms.4

Dermatitis and Irritants

The dermatoxins Lyngbyatoxin A (LA) and debromoaplysiatoxin (DAT) cause the inflammatory and necrotic appearance of seaweed dermatitis.1,2,5,8 Lyngbyatoxin A is an indole alkaloid that is closely related to telocidin B, a poisonous compound associated with Streptomyces bacteria.9 Sampling of L majuscula and extraction of the dermatoxin, along with human and animal studies, confirmed DAT irritates the skin and induces dermatitis.5,6Stylocheilus longicauda (sea hare) feeds on L majuscula and contains isolates of DAT in its digestive tract.

Samples of L majuscula taken from several Hawaiian Islands where seaweed dermatitis outbreaks have occurred were examined for differences in toxicities via 6-hour patch tests on human skin.6 The samples obtained from the windward side of Oahu contained DAT and aplysiatoxin, while those obtained from the leeward side and Kahala Beach primarily contained LA. Although DAT and LA are vastly different in their molecular structures, testing elicited the same biologic response and induced the same level of skin irritation.6 Interestingly, not all strands of L majuscula produced LA and DAT and caused seaweed dermatitis; those that did lead to irritation were more red in color than nontoxic blooms.5,9

Cutaneous Manifestations

Seaweed dermatitis resembles chemical and thermal burns, ranging from a mild skin rash to severe contact dermatitis with itchy, swollen, ulcerated lesions.1,7 Patients typically develop a burning or itching sensation beneath their bathing suit or wetsuit that progresses to an erythematous papulovesicular eruption 2 to 24 hours after exposure.2,6 Within a week, vesicles and bullae desquamate, leaving behind tender erosions.1,2,6,8 Inframammary lesions are common in females and scrotal swelling in males.1,6 There is no known association between length of time spent in the water and severity of symptoms.5

Most reactions to L majuscula occur from exposure in the water; however, particles that become aerosolized during strong winds or storms can cause seaweed dermatitis on the face. Inhalation of L majuscula may lead to mucous membrane ulceration and pulmonary edema.1,5,6 Noncutaneous manifestations of seaweed dermatitis include headache, fatigue, and swelling of the eyes, nose, and throat (Figures 1 and 2).1,5

Prevention and Management

To prevent seaweed dermatitis, avoid swimming in ocean water during L majuscula blooms,10 which frequently occur following the summer solstices in the Northern and Southern hemispheres.5 The National Centers for Coastal Ocean Science Harmful Algae Bloom Monitoring System provides real-time access to algae bloom locations.11 Although this monitoring system is not specific to L majuscula, it may be helpful in determining where potential blooms are. Wearing protective clothing such as coveralls may benefit individuals who enter the water during blooms, but it does not guarantee protection.10

couagestemaclubacikofraswospetrauadraphibristochicreletocrecrodusispelithimathatethivodoswotivabegevofrushudrocovufrebrovepespiridedrawiwrephuladuphakohelaphukutrokekapre
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%20punch%20biopsy%20of%20an%20abdominal%20lesion%20in%20a%20patient%20with%20seaweed%20dermatitis%20(%3Cem%3ELyngbya%20majuscula%3C%2Fem%3E)%20showed%20an%20intraepidermal%20blister%20with%20superficial%20desquamation%20at%20the%20top%20(H%26amp%3BE%2C%20original%3Cbr%3Emagnification%20%C3%9740).%20Photograph%20courtesy%20of%20Scott%20Norton%2C%20MD%2C%20MPH%2C%20MSc%20(Washington%2C%20DC).%3C%2Fp%3E

theveslufrufrilitonosecihefroclufriuipephuwalustovigumespispewutradrophinunujepareprodushecrephohekiclah
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Classic%20erythematous%20papulovesicular%20rash%20on%20the%20abdomen%20of%20a%20patient%20with%20seaweed%20dermatitis%20(%3Cem%3ELyngbya%20majuscula%3C%2Fem%3E).%20Photograph%20courtesy%20of%20Scott%20Norton%2C%20MD%2C%20MPH%2C%20MSc%20(Washington%2C%20DC).%3C%2Fp%3E

Currently, there is no treatment for seaweed dermatitis, but symptom management may reduce discomfort and pain. Washing affected skin with soap and water within an hour of exposure may help reduce the severity of seaweed dermatitis, though studies have shown mixed results.6,7 Application of cool compresses and soothing ointments (eg, calamine) provide symptomatic relief and promote healing.7 The dermatitis typically self-resolves within 1 week.

References
  1. Werner K, Marquart L, Norton S. Lyngbya dermatitis (toxic seaweed dermatitis). Int J Dermatol. 2011;51:59-62. doi:10.1111/j.1365-4632.2011.05042.x
  2. Osborne N, Shaw G. Dermatitis associated with exposure to a marine cyanobacterium during recreational water exposure. BMC Dermatol. 2008;8:5. doi:10.1186/1471-5945-8-5
  3. Hays G, Richardson A, Robinson C. Climate change and marine plankton. Trends Ecol Evol. 2005;20:337-344. doi:10.1016/j.tree.2005.03.004
  4. Albert S, O’Neil J, Udy J, et al. Blooms of the cyanobacterium Lyngbya majuscula in costal Queensland, Australia: disparate sites, common factors. Mar Pollut Bull. 2004;51:428-437. doi:10.1016/j.marpolbul.2004.10.016
  5. Osborne N, Webb P, Shaw G. The toxins of Lyngbya majuscula and their human and ecological health effects. Environ Int. 2001;27:381-392. doi:10.1016/s0160-4120(01)00098-8
  6. Izumi A, Moore R. Seaweed ( Lyngbya majuscula ) dermatitis . Clin Dermatol . 1987;5:92-100. doi:10.1016/s0738-081x(87)80014-7
  7. Grauer F, Arnold H. Seaweed dermatitis: first report of a dermatitis-producing marine alga. Arch Dermatol. 1961; 84:720-732. doi:10.1001/archderm.1961.01580170014003
  8. Taylor M, Stahl-Timmins W, Redshaw C, et al. Toxic alkaloids in Lyngbya majuscula and related tropical marine cyanobacteria. Harmful Algae . 2014;31:1-8. doi:10.1016/j.hal.2013.09.003
  9. Cardellina J, Marner F, Moore R. Seaweed dermatitis: structure of lyngbyatoxin A. Science. 1979;204:193-195. doi:10.1126/science.107586
  10. Osborne N. Occupational dermatitis caused by Lyngbya majuscule in Australia. Int J Dermatol . 2012;5:122-123. doi:10.1111/j.1365-4632.2009.04455.x
  11. Harmful Algal Bloom Monitoring System. National Centers for Coastal Ocean Science. Accessed May 23, 2024. https://coastalscience.noaa.gov/research/stressor-impacts-mitigation/hab-monitoring-system/
References
  1. Werner K, Marquart L, Norton S. Lyngbya dermatitis (toxic seaweed dermatitis). Int J Dermatol. 2011;51:59-62. doi:10.1111/j.1365-4632.2011.05042.x
  2. Osborne N, Shaw G. Dermatitis associated with exposure to a marine cyanobacterium during recreational water exposure. BMC Dermatol. 2008;8:5. doi:10.1186/1471-5945-8-5
  3. Hays G, Richardson A, Robinson C. Climate change and marine plankton. Trends Ecol Evol. 2005;20:337-344. doi:10.1016/j.tree.2005.03.004
  4. Albert S, O’Neil J, Udy J, et al. Blooms of the cyanobacterium Lyngbya majuscula in costal Queensland, Australia: disparate sites, common factors. Mar Pollut Bull. 2004;51:428-437. doi:10.1016/j.marpolbul.2004.10.016
  5. Osborne N, Webb P, Shaw G. The toxins of Lyngbya majuscula and their human and ecological health effects. Environ Int. 2001;27:381-392. doi:10.1016/s0160-4120(01)00098-8
  6. Izumi A, Moore R. Seaweed ( Lyngbya majuscula ) dermatitis . Clin Dermatol . 1987;5:92-100. doi:10.1016/s0738-081x(87)80014-7
  7. Grauer F, Arnold H. Seaweed dermatitis: first report of a dermatitis-producing marine alga. Arch Dermatol. 1961; 84:720-732. doi:10.1001/archderm.1961.01580170014003
  8. Taylor M, Stahl-Timmins W, Redshaw C, et al. Toxic alkaloids in Lyngbya majuscula and related tropical marine cyanobacteria. Harmful Algae . 2014;31:1-8. doi:10.1016/j.hal.2013.09.003
  9. Cardellina J, Marner F, Moore R. Seaweed dermatitis: structure of lyngbyatoxin A. Science. 1979;204:193-195. doi:10.1126/science.107586
  10. Osborne N. Occupational dermatitis caused by Lyngbya majuscule in Australia. Int J Dermatol . 2012;5:122-123. doi:10.1111/j.1365-4632.2009.04455.x
  11. Harmful Algal Bloom Monitoring System. National Centers for Coastal Ocean Science. Accessed May 23, 2024. https://coastalscience.noaa.gov/research/stressor-impacts-mitigation/hab-monitoring-system/
Issue
Cutis - 113(5)
Issue
Cutis - 113(5)
Page Number
E38-E40
Page Number
E38-E40
Publications
Publications
Topics
Article Type
Display Headline

Aquatic Antagonists: Seaweed Dermatitis (Lyngbya majuscula)

Display Headline

Aquatic Antagonists: Seaweed Dermatitis (Lyngbya majuscula)

Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Hill Aquatic</fileName> <TBEID>0C02F80E.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F80E</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Hill Aquatic</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240604T155220</firstPublished> <LastPublished>20240604T155220</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240604T155219</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Kathleen L. Hill, MD</byline> <bylineText>Kathleen L. Hill, MD; Haley M. Fulton, MD; Thomas W. McGovern, MD</bylineText> <bylineFull>Kathleen L. Hill, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>E38-E40</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>The filamentous cyanobacterium Lyngbya majuscula causes irritant contact dermatitis in beachgoers, fishers, and divers in tropical and subtropical marine enviro</metaDescription> <articlePDF/> <teaserImage/> <title>Aquatic Antagonists: Seaweed Dermatitis (Lyngbya majuscula)</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>May</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>5</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2163</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>May 2024</pubIssueName> <pubArticleType>Online Exclusive | 2163</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">60</term> </sections> <topics> <term canonical="true">199</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Aquatic Antagonists: Seaweed Dermatitis (Lyngbya majuscula)</title> <deck/> </itemMeta> <itemContent> <p class="abstract">The cyanobacterium<em> Lyngbya majuscula</em> grows in marine and estuarine environments across the world and produces many biologically active compounds. Direct contact with <em>L majuscula </em>and its dermatoxins can cause seaweed dermatitis, which manifests as a papulovesicular eruption. As oceans warm, <em>L majuscula</em> will bloom more frequently; therefore, public awareness of <em>L majuscula</em> and seaweed dermatitis in oceanside communities can help promote precautions that can reduce the risk for exposure. This article describes the irritants that lead to dermatitis, clinical presentation, and prevention and management of seaweed dermatitis.</p> <p>The filamentous cyanobacterium <i>Lyngbya majuscula </i>causes irritant contact dermatitis in beachgoers, fishers, and divers in tropical and subtropical marine environments worldwide.<sup>1</sup> If fragments of <i>L majuscula </i>lodge in swimmers’ bathing suits, the toxins can become trapped against the skin and cause seaweed dermatitis.<sup>2</sup> With climate change resulting in warmer oceans and more extreme storms, <i>L majuscula</i> blooms likely will become more frequent and widespread, thereby increasing the risk for human exposure.<sup>3,4</sup> Herein, we describe the irritants that lead to dermatitis, clinical presentation, and prevention and management of seaweed dermatitis.</p> <h3>Identifying Features and Distribution of Plant</h3> <p><i>Lyngbya majuscula</i> belongs to the family Oscillatoriaceae; these cyanobacteria grow as filaments and exhibit slow oscillating movements. Commonly referred to as blanketweed or mermaid’s hair due to its appearance, <i>L majuscula</i> grows fine hairlike clumps resembling a mass of olive-colored matted hair.<sup>1</sup> Its thin filaments are 10- to 30-cm long and vary in color from red to white to brown.<sup>5</sup> Microscopically, a rouleauxlike arrangement of discs provides the structure of each filament.<sup>6</sup> </p> <p>First identified in Hawaii in 1912, <i>L majuscula</i> was not associated with seaweed dermatitis or dermatotoxicity by the medical community until the first outbreak occurred in Oahu in 1958, though fishermen and beachgoers previously had recognized a relationship between this particular seaweed and skin irritation.<sup>5,7</sup> The first reporting included 125 confirmed cases, with many more mild unreported cases suspected.<sup>6</sup> Now reported in about 100 locations worldwide, seaweed dermatitis outbreaks have occurred in Australia; Okinawa, Japan; Florida; and the Hawaiian and Marshall islands.<sup>1,2</sup> </p> <h3>Exposure to Seaweed</h3> <p><i>Lyngbya majuscula</i> produces more than 70 biologically active compounds that irritate the skin, eyes, and respiratory system.<sup>2,8</sup> It grows in marine and estuarine environments attached to seagrass, sand, and bedrock at depths of up to 30 m. Warm waters and maximal sunlight provide optimal growth conditions for <i>L majuscula</i>; therefore, the greatest risk for exposure occurs in the Northern and Southern hemispheres in the 1- to 2-month period following their summer solstices.<sup>5</sup> Runoff during heavy rainfall, which is rich in soil extracts such as phosphorous, iron, and organic carbon, stimulates <i>L majuscula</i> growth and contributes to increased algal blooms.<sup>4</sup></p> <h3>Dermatitis and Irritants</h3> <p>The dermatoxins Lyngbyatoxin A (LA) and debromoaplysiatoxin (DAT) cause the inflammatory and necrotic appearance of seaweed dermatitis.<sup>1,2,5,8</sup> Lyngbyatoxin A is an indole alkaloid that is closely related to telocidin B, a poisonous compound associated with <i>Streptomyces</i> bacteria.<sup>9</sup> Sampling of <i>L majuscula</i> and extraction of the dermatoxin, along with human and animal studies, confirmed DAT irritates the skin and induces dermatitis.<sup>5,6</sup> <i>Stylocheilus longicauda </i>(sea hare) feeds on <i>L majuscula </i>and contains isolates of DAT in its digestive tract<i>.</i></p> <p>Samples of <i>L majuscula</i> taken from several Hawaiian Islands where seaweed dermatitis outbreaks have occurred were examined for differences in toxicities via 6-hour patch tests on human skin.<sup>6</sup> The samples obtained from the windward side of Oahu contained DAT and aplysiatoxin, while those obtained from the leeward side and Kahala Beach primarily contained LA. Although DAT and LA are vastly different in their molecular structures, testing elicited the same biologic response and induced the same level of skin irritation.<sup>6</sup> Interestingly, not all strands of <i>L</i> <i>majuscula</i> produced LA and DAT and caused seaweed dermatitis; those that did lead to irritation were more red in color than nontoxic blooms.<sup>5,9</sup></p> <h3>Cutaneous Manifestations</h3> <p>Seaweed dermatitis resembles chemical and thermal burns, ranging from a mild skin rash to severe contact dermatitis with itchy, swollen, ulcerated lesions.<sup>1,7</sup> Patients typically develop a burning or itching sensation beneath their bathing suit or wetsuit that progresses to an erythematous papulovesicular eruption 2 to 24 hours after exposure.<sup>2,6</sup> Within a week, vesicles and bullae desquamate, leaving behind tender erosions.<sup>1,2,6,8</sup> Inframammary lesions are common in females and scrotal swelling in males.<sup>1,6</sup> There is no known association between length of time spent in the water and severity of symptoms.<sup>5</sup></p> <p>Most reactions to <i>L majuscula</i> occur from exposure in the water; however, particles that become aerosolized during strong winds or storms can cause seaweed dermatitis on the face. Inhalation of <i>L majuscula</i> may lead to mucous membrane ulceration and pulmonary edema.<sup>1,5,6</sup> Noncutaneous manifestations of seaweed dermatitis include headache, fatigue, and swelling of the eyes, nose, and throat (Figures 1 and 2).<sup>1,5</sup></p> <h3>Prevention and Management</h3> <p>To prevent seaweed dermatitis, avoid swimming in ocean water during <i>L majuscula</i> blooms,<sup>10</sup> which frequently occur following the summer solstices in the Northern and Southern hemispheres.<sup>5</sup> The National Centers for Coastal Ocean Science Harmful Algae Bloom Monitoring System provides real-time access to algae bloom locations.<sup>11</sup> Although this monitoring system is not specific to <i>L majuscula</i>, it may be helpful in determining where potential blooms are. Wearing protective clothing such as coveralls may benefit individuals who enter the water during blooms, but it does not guarantee protection.<sup>10</sup> </p> <p>Currently, there is no treatment for seaweed dermatitis, but symptom management may reduce discomfort and pain. Washing affected skin with soap and water within an hour of exposure may help reduce the severity of seaweed dermatitis, though studies have shown mixed results.<sup>6,7</sup> Application of cool compresses and soothing ointments (eg, calamine) provide symptomatic relief and promote healing.<sup>7</sup> The dermatitis typically self-resolves within 1 week.</p> <h2>References</h2> <p class="reference"> 1. Werner K, Marquart L, Norton S. <i>Lyngbya</i> dermatitis (toxic seaweed dermatitis). <i>Int J Dermatol</i>. 2011;51:59-62. doi:10.1111/j.1365-4632.2011.05042.x<br/><br/> 2. Osborne N, Shaw G. Dermatitis associated with exposure to a marine cyanobacterium during recreational water exposure. <i>BMC Dermatol</i>. 2008;8:5. doi:<a href="https://doi.org/10.1186/1471-5945-8-5">10.1186/1471-5945-8-5</a><br/><br/><span class="None"> 3. Hays G, Richardson A, Robinson C. Climate change and marine plankton. </span><span class="None"><i>Trends Ecol Evol.</i></span><span class="None"> 2005;20:337-344. doi:10.1016/j.tree.2005.03.004<br/><br/> 4. </span><span class="None">Albert S, O’Neil J, Udy J, et al. Blooms of the cyanobacterium </span><span class="None"><i>Lyngbya majuscula</i></span><span class="None"> in costal Queensland, Australia: disparate sites, common factors. </span><span class="None"><i>Mar Pollut Bull.</i></span><span class="None"> 2004;51:428-437. </span>doi:10.1016/j.marpolbul.2004.10.016<span class="None"><br/><br/> 5. Osborne N, Webb P, Shaw G. The toxins of </span><span class="None"><i>Lyngbya majuscula</i></span><span class="None"> and their human and ecological health effects. </span><span class="None"><i>Environ Int</i></span><span class="None">. 2001;27:381-392. </span>doi:10.1016/s0160-4120(01)00098-8</p> <p class="reference"> <span class="None"> 6. Izumi A, Moore R. Seaweed (</span> <span class="None"> <i>Lyngbya majuscula</i> </span> <span class="None">) dermatitis</span> <span class="None"> <i>. Clin Dermatol</i> </span> <span class="None">. 1987;5:92-100. doi:10.1016/s0738-081x(87)80014-7<br/><br/> 7. Grauer F, Arnold H. Seaweed dermatitis: first report of a dermatitis-producing marine alga. </span> <span class="None"> <i>Arch Dermatol</i> </span> <span class="None">. 1961; 84:720-732. doi:10.1001/archderm.1961.01580170014003<br/><br/> 8. Taylor M, Stahl-Timmins W, Redshaw C, et al. Toxic alkaloids in </span> <span class="None"> <i>Lyngbya majuscula</i> </span> <span class="None"> and related tropical marine cyanobacteria. </span> <span class="None"> <i>Harmful Algae</i> </span> <span class="None">. 2014;31:1-8. doi:10.1016/j.hal.2013.09.003<br/><br/> 9. Cardellina J, Marner F, Moore R. Seaweed dermatitis: structure of lyngbyatoxin A. </span> <span class="None"> <i>Science.</i> </span> <span class="None"> 1979;204:193-195. doi:10.1126/science.107586<br/><br/>10. Osborne N. Occupational dermatitis caused by </span> <span class="None"> <i>Lyngbya majuscule</i> </span> <span class="None"> in Australia. </span> <span class="None"> <i>Int J Dermatol</i> </span> <span class="None">. 2012;5:122-123. doi:10.1111/j.1365-4632.2009.04455.x<br/><br/>11. </span> <span class="None">Harmful Algal Bloom Monitoring System. National Centers for Coastal Ocean Science. Accessed May 23, 2024. https://coastalscience.noaa.gov/research/stressor-impacts-mitigation/hab-monitoring-system/</span> </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Dr. Hill is from the University of South Carolina School of Medicine, Greenville. Dr. Fulton is from Spartanburg Regional Medical Center, South Carolina. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.</p> <p class="disclosure">The authors report no conflict of interest.<br/><br/>The images are in the public domain. <br/><br/>Correspondence: Kathleen L. Hill, MD, 607 Grove Rd, Greenville, SC 29605 (klhill@email.sc.edu).<br/><br/><em>Cutis. </em>2024 May;113(5):E38-E40. doi:10.12788/cutis.1032</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">PRACTICE <strong>POINTS</strong></p> <ul class="insidebody"> <li><em>Lyngbya majuscula</em> causes seaweed dermatitis in swimmers and can be prevented by avoiding rough turbid waters in areas known to have<em> L majuscula</em> blooms.</li> <li>Seaweed dermatitis should be included in the differential diagnosis for erythematous papulovesicular rashes manifesting in patients who recently have spent time in the ocean.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

 

PRACTICE POINTS

  • Lyngbya majuscula causes seaweed dermatitis in swimmers and can be prevented by avoiding rough turbid waters in areas known to have L majuscula blooms.
  • Seaweed dermatitis should be included in the differential diagnosis for erythematous papulovesicular rashes manifesting in patients who recently have spent time in the ocean.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Disable zoom
Off

Botanical Briefs: Fig Phytophotodermatitis (Ficus carica)

Article Type
Changed
Wed, 04/10/2024 - 10:12
Display Headline
Botanical Briefs: Fig Phytophotodermatitis (Ficus carica)

Plant Parts and Nomenclature

Ficus carica (common fig) is a deciduous shrub or small tree with smooth gray bark that can grow up to 10 m in height (Figure 1). It is characterized by many spreading branches, but the trunk rarely grows beyond a diameter of 7 in. Its hairy leaves are coarse on the upper side and soft underneath with 3 to 7 deep lobes that can extend up to 25 cm in length or width; the leaves grow individually, alternating along the sides of the branches. Fig trees often can be seen adorning yards, gardens, and parks, especially in tropical and subtropical climates. Ficus carica should not be confused with Ficus benjamina (weeping fig), a common ornamental tree that also is used to provide shade in hot climates, though both can cause phototoxic skin eruptions.

Barker_0424_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%26nbsp%3B%26lt%3Bi%26gt%3BFicus%20carica%26lt%3B%2Fi%26gt%3B%20(common%20fig).%3C%2Fp%3E

The common fig tree originated in the Mediterranean and western Asia1 and has been cultivated by humans since the second and third millennia bc for its fruit, which commonly is used to sweeten cookies, cakes, and jams.2 Figs are the most commonly mentioned food plant in the Bible, with at least 56 references in the Old and New Testaments.3 The “fruit” technically is a syconium—a hollow fleshy receptacle with a small opening at the apex partly closed by small scales. It can be obovoid, turbinate, or pear shaped; can be 1 to 4 inches long; and can vary in color from yellowish green to coppery, bronze, or dark purple (Figure 2).

Barker_0424_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Immature%20fruit%20of%20the%20common%20fig%20tree.%3C%2Fp%3E

Ficus carica is a member of the Moraceae family (derived from the Latin name for the mulberry tree), which includes 53 genera and approximately 1400 species, of which about 850 belong to the genus Ficus (the Latin name for a fig tree). The term carica likely comes from the Latin word carricare (to load) to describe a tree loaded with figs. Family members include trees, shrubs, lianas, and herbs that usually contain laticifers with a milky latex.

Traditional Uses

For centuries, components of the fig tree have been used in herbal teas and pastes to treat ailments ranging from sore throats to diarrhea, though there is no evidence to support their efficacy.4 Ancient Indians and Egyptians used plants such as the common fig tree containing furocoumarins to induce hyperpigmentation in vitiligo.5

Phototoxic Components

The leaves and sap of the common fig tree contain psoralens, which are members of the furocoumarin group of chemical compounds and are the source of its phototoxicity. The fruit does not contain psoralens.6-9 The tree also produces proteolytic enzymes such as protease, amylase, ficin, triterpenoids, and lipodiastase that enhance its phototoxic effects.8 Exposure to UV light between 320 and 400 nm following contact with these phototoxic components triggers a reaction in the skin over the course of 1 to 3 days.5 The psoralens bind in epidermal cells, cross-link the DNA, and cause cell-membrane destruction, leading to edema and necrosis.10 The delay in symptoms may be attributed to the time needed to synthesize acute-phase reaction proteins such as tumor necrosis factor α and IL-1.11 In spring and summer months, an increased concentration of psoralens in the leaves and sap contribute to an increased incidence of phytophotodermatitis.9 Humidity and sweat also increase the percutaneous absorption of psoralens.12,13

Allergens

Fig trees produce a latex protein that can cause cross-reactive hypersensitivity reactions in those allergic to F benjamina latex and rubber latex.6 The latex proteins in fig trees can act as airborne respiratory allergens. Ingestion of figs can produce anaphylactic reactions in those sensitized to rubber latex and F benjamina latex.7 Other plant families associated with phototoxic reactions include Rutaceae (lemon, lime, bitter orange), Apiaceae (formerly Umbelliferae)(carrot, parsnip, parsley, dill, celery, hogweed), and Fabaceae (prairie turnip).

 

 

Cutaneous Manifestations

Most cases of fig phytophotodermatitis begin with burning, pain, and/or itching within hours of sunlight exposure in areas of the skin that encountered components of the fig tree, often in a linear pattern. The affected areas become erythematous and edematous with formation of bullae and unilocular vesicles over the course of 1 to 3 days.12,14,15 Lesions may extend beyond the region of contact with the fig tree as they spread across the skin due to sweat or friction, and pain may linger even after the lesions resolve.12,13,16 Adults who handle fig trees (eg, pruning) are susceptible to phototoxic reactions, especially those using chain saws or other mechanisms that result in spray exposure, as the photosensitizing sap permeates the wood and bark of the entire tree.17 Similarly, children who handle fig leaves or sap during outdoor play can develop bullous eruptions. Severe cases have resulted in hospital admission after prolonged exposure.16 Additionally, irritant dermatitis may arise from contact with the trichomes or “hairs” on various parts of the plant.

Barker_0424_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Leaves%20and%20milky%20sap%20of%20the%20common%20fig%20tree.%3C%2Fp%3E

Patients who use natural remedies containing components of the fig tree without the supervision of a medical provider put themselves at risk for unsafe or unwanted adverse effects, such as phytophotodermatitis.12,15,16,18 An entire family presented with burns after they applied fig leaf extract to the skin prior to tanning outside in the sun.19 A 42-year-old woman acquired a severe burn covering 81% of the body surface after topically applying fig leaf tea to the skin as a tanning agent.20 A subset of patients ingesting or applying fig tree components for conditions such as vitiligo, dermatitis, onychomycosis, and motor retardation developed similar cutaneous reactions.13,14,21,22 Lesions resembling finger marks can raise concerns for potential abuse or neglect in children.22

The differential diagnosis for fig phytophotodermatitis includes sunburn, chemical burns, drug-related photosensitivity, infectious lesions (eg, herpes simplex, bullous impetigo, Lyme disease, superficial lymphangitis), connective tissue disease (eg, systemic lupus erythematosus), contact dermatitis, and nonaccidental trauma.12,15,18 Compared to sunburn, phytophotodermatitis tends to increase in severity over days following exposure and heals with dramatic hyperpigmentation, which also prompts visits to dermatology.12

Treatment

Treatment of fig phytophotodermatitis chiefly is symptomatic, including analgesia, appropriate wound care, and infection prophylaxis. Topical and systemic corticosteroids may aid in the resolution of moderate to severe reactions.15,23,24 Even severe injuries over small areas or mild injuries to a high percentage of the total body surface area may require treatment in a burn unit. Patients should be encouraged to use mineral-based sunscreens on the affected areas to reduce the risk for hyperpigmentation. Individuals who regularly handle fig trees should use contact barriers including gloves and protective clothing (eg, long-sleeved shirts, long pants).

References
  1. Ikegami H, Nogata H, Hirashima K, et al. Analysis of genetic diversity among European and Asian fig varieties (Ficus carica L.) using ISSR, RAPD, and SSR markers. Genetic Resources and Crop Evolution. 2009;56:201-209.
  2. Zohary D, Spiegel-Roy P. Beginnings of fruit growing in the Old World. Science. 1975;187:319-327.
  3. Young R. Young’s Analytical Concordance. Thomas Nelson; 1982.
  4. Duke JA. Handbook of Medicinal Herbs. CRC Press; 2002.
  5. Pathak MA, Fitzpatrick TB. Bioassay of natural and synthetic furocoumarins (psoralens). J Invest Dermatol. 1959;32:509-518.
  6. Focke M, Hemmer W, Wöhrl S, et al. Cross-reactivity between Ficus benjamina latex and fig fruit in patients with clinical fig allergy. Clin Exp Allergy. 2003;33:971-977.
  7. Hemmer W, Focke M, Götz M, et al. Sensitization to Ficus benjamina: relationship to natural rubber latex allergy and identification of foods implicated in the Ficus-fruit syndrome. Clin Exp Allergy. 2004;34:1251-1258.
  8. Bonamonte D, Foti C, Lionetti N, et al. Photoallergic contact dermatitis to 8-methoxypsoralen in Ficus carica. Contact Dermatitis. 2010;62:343-348.
  9. Zaynoun ST, Aftimos BG, Abi Ali L, et al. Ficus carica; isolation and quantification of the photoactive components. Contact Dermatitis. 1984;11:21-25.
  10. Tessman JW, Isaacs ST, Hearst JE. Photochemistry of the furan-side 8-methoxypsoralen-thymidine monoadduct inside the DNA helix. conversion to diadduct and to pyrone-side monoadduct. Biochemistry. 1985;24:1669-1676.
  11. Geary P. Burns related to the use of psoralens as a tanning agent. Burns. 1996;22:636-637.
  12. Redgrave N, Solomon J. Severe phytophotodermatitis from fig sap: a little known phenomenon. BMJ Case Rep. 2021;14:E238745.
  13. Ozdamar E, Ozbek S, Akin S. An unusual cause of burn injury: fig leaf decoction used as a remedy for a dermatitis of unknown etiology. J Burn Care Rehabil. 2003;24:229-233; discussion 228.
  14. Berakha GJ, Lefkovits G. Psoralen phototherapy and phototoxicity. Ann Plast Surg. 1985;14:458-461.
  15. Papazoglou A, Mantadakis E. Fig tree leaves phytophotodermatitis. J Pediatr. 2021;239:244-245.
  16. Imen MS, Ahmadabadi A, Tavousi SH, et al. The curious cases of burn by fig tree leaves. Indian J Dermatol. 2019;64:71-73.
  17. Rouaiguia-Bouakkaz S, Amira-Guebailia H, Rivière C, et al. Identification and quantification of furanocoumarins in stem bark and wood of eight Algerian varieties of Ficus carica by RP-HPLC-DAD and RP-HPLC-DAD-MS. Nat Prod Commun. 2013;8:485-486.
  18. Oliveira AA, Morais J, Pires O, et al. Fig tree induced phytophotodermatitis. BMJ Case Rep. 2020;13:E233392.
  19. Bassioukas K, Stergiopoulou C, Hatzis J. Erythrodermic phytophotodermatitis after application of aqueous fig-leaf extract as an artificial suntan promoter and sunbathing. Contact Dermatitis. 2004;51:94-95.
  20. Sforza M, Andjelkov K, Zaccheddu R. Severe burn on 81% of body surface after sun tanning. Ulus Travma Acil Cerrahi Derg. 2013;19:383-384.
  21. Son JH, Jin H, You HS, et al. Five cases of phytophotodermatitis caused by fig leaves and relevant literature review. Ann Dermatol. 2017;29:86-90.
  22. Abali AE, Aka M, Aydogan C, et al. Burns or phytophotodermatitis, abuse or neglect: confusing aspects of skin lesions caused by the superstitious use of fig leaves. J Burn Care Res. 2012;33:E309-E312.
  23. Picard C, Morice C, Moreau A, et al. Phytophotodermatitis in children: a difficult diagnosis mimicking other dermatitis. 2017;5:1-3.
  24. Enjolras O, Soupre V, Picard A. Uncommon benign infantile vascular tumors. Adv Dermatol. 2008;24:105-124.
Article PDF
Author and Disclosure Information

Drs. Barker and Elston are from the Medical University of South Carolina, Charleston. Dr. Barker is from the Department of Internal Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.

The authors report no conflict of interest.

Correspondence: Catherine Shirer Barker, MD, 96 Jonathan Lucas St, Ste 807B, MSC 623, Charleston, SC 29425 (catherinesbarker@gmail.com).

Issue
Cutis - 113(4)
Publications
Topics
Page Number
167-169
Sections
Author and Disclosure Information

Drs. Barker and Elston are from the Medical University of South Carolina, Charleston. Dr. Barker is from the Department of Internal Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.

The authors report no conflict of interest.

Correspondence: Catherine Shirer Barker, MD, 96 Jonathan Lucas St, Ste 807B, MSC 623, Charleston, SC 29425 (catherinesbarker@gmail.com).

Author and Disclosure Information

Drs. Barker and Elston are from the Medical University of South Carolina, Charleston. Dr. Barker is from the Department of Internal Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.

The authors report no conflict of interest.

Correspondence: Catherine Shirer Barker, MD, 96 Jonathan Lucas St, Ste 807B, MSC 623, Charleston, SC 29425 (catherinesbarker@gmail.com).

Article PDF
Article PDF

Plant Parts and Nomenclature

Ficus carica (common fig) is a deciduous shrub or small tree with smooth gray bark that can grow up to 10 m in height (Figure 1). It is characterized by many spreading branches, but the trunk rarely grows beyond a diameter of 7 in. Its hairy leaves are coarse on the upper side and soft underneath with 3 to 7 deep lobes that can extend up to 25 cm in length or width; the leaves grow individually, alternating along the sides of the branches. Fig trees often can be seen adorning yards, gardens, and parks, especially in tropical and subtropical climates. Ficus carica should not be confused with Ficus benjamina (weeping fig), a common ornamental tree that also is used to provide shade in hot climates, though both can cause phototoxic skin eruptions.

Barker_0424_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%26nbsp%3B%26lt%3Bi%26gt%3BFicus%20carica%26lt%3B%2Fi%26gt%3B%20(common%20fig).%3C%2Fp%3E

The common fig tree originated in the Mediterranean and western Asia1 and has been cultivated by humans since the second and third millennia bc for its fruit, which commonly is used to sweeten cookies, cakes, and jams.2 Figs are the most commonly mentioned food plant in the Bible, with at least 56 references in the Old and New Testaments.3 The “fruit” technically is a syconium—a hollow fleshy receptacle with a small opening at the apex partly closed by small scales. It can be obovoid, turbinate, or pear shaped; can be 1 to 4 inches long; and can vary in color from yellowish green to coppery, bronze, or dark purple (Figure 2).

Barker_0424_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Immature%20fruit%20of%20the%20common%20fig%20tree.%3C%2Fp%3E

Ficus carica is a member of the Moraceae family (derived from the Latin name for the mulberry tree), which includes 53 genera and approximately 1400 species, of which about 850 belong to the genus Ficus (the Latin name for a fig tree). The term carica likely comes from the Latin word carricare (to load) to describe a tree loaded with figs. Family members include trees, shrubs, lianas, and herbs that usually contain laticifers with a milky latex.

Traditional Uses

For centuries, components of the fig tree have been used in herbal teas and pastes to treat ailments ranging from sore throats to diarrhea, though there is no evidence to support their efficacy.4 Ancient Indians and Egyptians used plants such as the common fig tree containing furocoumarins to induce hyperpigmentation in vitiligo.5

Phototoxic Components

The leaves and sap of the common fig tree contain psoralens, which are members of the furocoumarin group of chemical compounds and are the source of its phototoxicity. The fruit does not contain psoralens.6-9 The tree also produces proteolytic enzymes such as protease, amylase, ficin, triterpenoids, and lipodiastase that enhance its phototoxic effects.8 Exposure to UV light between 320 and 400 nm following contact with these phototoxic components triggers a reaction in the skin over the course of 1 to 3 days.5 The psoralens bind in epidermal cells, cross-link the DNA, and cause cell-membrane destruction, leading to edema and necrosis.10 The delay in symptoms may be attributed to the time needed to synthesize acute-phase reaction proteins such as tumor necrosis factor α and IL-1.11 In spring and summer months, an increased concentration of psoralens in the leaves and sap contribute to an increased incidence of phytophotodermatitis.9 Humidity and sweat also increase the percutaneous absorption of psoralens.12,13

Allergens

Fig trees produce a latex protein that can cause cross-reactive hypersensitivity reactions in those allergic to F benjamina latex and rubber latex.6 The latex proteins in fig trees can act as airborne respiratory allergens. Ingestion of figs can produce anaphylactic reactions in those sensitized to rubber latex and F benjamina latex.7 Other plant families associated with phototoxic reactions include Rutaceae (lemon, lime, bitter orange), Apiaceae (formerly Umbelliferae)(carrot, parsnip, parsley, dill, celery, hogweed), and Fabaceae (prairie turnip).

 

 

Cutaneous Manifestations

Most cases of fig phytophotodermatitis begin with burning, pain, and/or itching within hours of sunlight exposure in areas of the skin that encountered components of the fig tree, often in a linear pattern. The affected areas become erythematous and edematous with formation of bullae and unilocular vesicles over the course of 1 to 3 days.12,14,15 Lesions may extend beyond the region of contact with the fig tree as they spread across the skin due to sweat or friction, and pain may linger even after the lesions resolve.12,13,16 Adults who handle fig trees (eg, pruning) are susceptible to phototoxic reactions, especially those using chain saws or other mechanisms that result in spray exposure, as the photosensitizing sap permeates the wood and bark of the entire tree.17 Similarly, children who handle fig leaves or sap during outdoor play can develop bullous eruptions. Severe cases have resulted in hospital admission after prolonged exposure.16 Additionally, irritant dermatitis may arise from contact with the trichomes or “hairs” on various parts of the plant.

Barker_0424_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Leaves%20and%20milky%20sap%20of%20the%20common%20fig%20tree.%3C%2Fp%3E

Patients who use natural remedies containing components of the fig tree without the supervision of a medical provider put themselves at risk for unsafe or unwanted adverse effects, such as phytophotodermatitis.12,15,16,18 An entire family presented with burns after they applied fig leaf extract to the skin prior to tanning outside in the sun.19 A 42-year-old woman acquired a severe burn covering 81% of the body surface after topically applying fig leaf tea to the skin as a tanning agent.20 A subset of patients ingesting or applying fig tree components for conditions such as vitiligo, dermatitis, onychomycosis, and motor retardation developed similar cutaneous reactions.13,14,21,22 Lesions resembling finger marks can raise concerns for potential abuse or neglect in children.22

The differential diagnosis for fig phytophotodermatitis includes sunburn, chemical burns, drug-related photosensitivity, infectious lesions (eg, herpes simplex, bullous impetigo, Lyme disease, superficial lymphangitis), connective tissue disease (eg, systemic lupus erythematosus), contact dermatitis, and nonaccidental trauma.12,15,18 Compared to sunburn, phytophotodermatitis tends to increase in severity over days following exposure and heals with dramatic hyperpigmentation, which also prompts visits to dermatology.12

Treatment

Treatment of fig phytophotodermatitis chiefly is symptomatic, including analgesia, appropriate wound care, and infection prophylaxis. Topical and systemic corticosteroids may aid in the resolution of moderate to severe reactions.15,23,24 Even severe injuries over small areas or mild injuries to a high percentage of the total body surface area may require treatment in a burn unit. Patients should be encouraged to use mineral-based sunscreens on the affected areas to reduce the risk for hyperpigmentation. Individuals who regularly handle fig trees should use contact barriers including gloves and protective clothing (eg, long-sleeved shirts, long pants).

Plant Parts and Nomenclature

Ficus carica (common fig) is a deciduous shrub or small tree with smooth gray bark that can grow up to 10 m in height (Figure 1). It is characterized by many spreading branches, but the trunk rarely grows beyond a diameter of 7 in. Its hairy leaves are coarse on the upper side and soft underneath with 3 to 7 deep lobes that can extend up to 25 cm in length or width; the leaves grow individually, alternating along the sides of the branches. Fig trees often can be seen adorning yards, gardens, and parks, especially in tropical and subtropical climates. Ficus carica should not be confused with Ficus benjamina (weeping fig), a common ornamental tree that also is used to provide shade in hot climates, though both can cause phototoxic skin eruptions.

Barker_0424_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%26nbsp%3B%26lt%3Bi%26gt%3BFicus%20carica%26lt%3B%2Fi%26gt%3B%20(common%20fig).%3C%2Fp%3E

The common fig tree originated in the Mediterranean and western Asia1 and has been cultivated by humans since the second and third millennia bc for its fruit, which commonly is used to sweeten cookies, cakes, and jams.2 Figs are the most commonly mentioned food plant in the Bible, with at least 56 references in the Old and New Testaments.3 The “fruit” technically is a syconium—a hollow fleshy receptacle with a small opening at the apex partly closed by small scales. It can be obovoid, turbinate, or pear shaped; can be 1 to 4 inches long; and can vary in color from yellowish green to coppery, bronze, or dark purple (Figure 2).

Barker_0424_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Immature%20fruit%20of%20the%20common%20fig%20tree.%3C%2Fp%3E

Ficus carica is a member of the Moraceae family (derived from the Latin name for the mulberry tree), which includes 53 genera and approximately 1400 species, of which about 850 belong to the genus Ficus (the Latin name for a fig tree). The term carica likely comes from the Latin word carricare (to load) to describe a tree loaded with figs. Family members include trees, shrubs, lianas, and herbs that usually contain laticifers with a milky latex.

Traditional Uses

For centuries, components of the fig tree have been used in herbal teas and pastes to treat ailments ranging from sore throats to diarrhea, though there is no evidence to support their efficacy.4 Ancient Indians and Egyptians used plants such as the common fig tree containing furocoumarins to induce hyperpigmentation in vitiligo.5

Phototoxic Components

The leaves and sap of the common fig tree contain psoralens, which are members of the furocoumarin group of chemical compounds and are the source of its phototoxicity. The fruit does not contain psoralens.6-9 The tree also produces proteolytic enzymes such as protease, amylase, ficin, triterpenoids, and lipodiastase that enhance its phototoxic effects.8 Exposure to UV light between 320 and 400 nm following contact with these phototoxic components triggers a reaction in the skin over the course of 1 to 3 days.5 The psoralens bind in epidermal cells, cross-link the DNA, and cause cell-membrane destruction, leading to edema and necrosis.10 The delay in symptoms may be attributed to the time needed to synthesize acute-phase reaction proteins such as tumor necrosis factor α and IL-1.11 In spring and summer months, an increased concentration of psoralens in the leaves and sap contribute to an increased incidence of phytophotodermatitis.9 Humidity and sweat also increase the percutaneous absorption of psoralens.12,13

Allergens

Fig trees produce a latex protein that can cause cross-reactive hypersensitivity reactions in those allergic to F benjamina latex and rubber latex.6 The latex proteins in fig trees can act as airborne respiratory allergens. Ingestion of figs can produce anaphylactic reactions in those sensitized to rubber latex and F benjamina latex.7 Other plant families associated with phototoxic reactions include Rutaceae (lemon, lime, bitter orange), Apiaceae (formerly Umbelliferae)(carrot, parsnip, parsley, dill, celery, hogweed), and Fabaceae (prairie turnip).

 

 

Cutaneous Manifestations

Most cases of fig phytophotodermatitis begin with burning, pain, and/or itching within hours of sunlight exposure in areas of the skin that encountered components of the fig tree, often in a linear pattern. The affected areas become erythematous and edematous with formation of bullae and unilocular vesicles over the course of 1 to 3 days.12,14,15 Lesions may extend beyond the region of contact with the fig tree as they spread across the skin due to sweat or friction, and pain may linger even after the lesions resolve.12,13,16 Adults who handle fig trees (eg, pruning) are susceptible to phototoxic reactions, especially those using chain saws or other mechanisms that result in spray exposure, as the photosensitizing sap permeates the wood and bark of the entire tree.17 Similarly, children who handle fig leaves or sap during outdoor play can develop bullous eruptions. Severe cases have resulted in hospital admission after prolonged exposure.16 Additionally, irritant dermatitis may arise from contact with the trichomes or “hairs” on various parts of the plant.

Barker_0424_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Leaves%20and%20milky%20sap%20of%20the%20common%20fig%20tree.%3C%2Fp%3E

Patients who use natural remedies containing components of the fig tree without the supervision of a medical provider put themselves at risk for unsafe or unwanted adverse effects, such as phytophotodermatitis.12,15,16,18 An entire family presented with burns after they applied fig leaf extract to the skin prior to tanning outside in the sun.19 A 42-year-old woman acquired a severe burn covering 81% of the body surface after topically applying fig leaf tea to the skin as a tanning agent.20 A subset of patients ingesting or applying fig tree components for conditions such as vitiligo, dermatitis, onychomycosis, and motor retardation developed similar cutaneous reactions.13,14,21,22 Lesions resembling finger marks can raise concerns for potential abuse or neglect in children.22

The differential diagnosis for fig phytophotodermatitis includes sunburn, chemical burns, drug-related photosensitivity, infectious lesions (eg, herpes simplex, bullous impetigo, Lyme disease, superficial lymphangitis), connective tissue disease (eg, systemic lupus erythematosus), contact dermatitis, and nonaccidental trauma.12,15,18 Compared to sunburn, phytophotodermatitis tends to increase in severity over days following exposure and heals with dramatic hyperpigmentation, which also prompts visits to dermatology.12

Treatment

Treatment of fig phytophotodermatitis chiefly is symptomatic, including analgesia, appropriate wound care, and infection prophylaxis. Topical and systemic corticosteroids may aid in the resolution of moderate to severe reactions.15,23,24 Even severe injuries over small areas or mild injuries to a high percentage of the total body surface area may require treatment in a burn unit. Patients should be encouraged to use mineral-based sunscreens on the affected areas to reduce the risk for hyperpigmentation. Individuals who regularly handle fig trees should use contact barriers including gloves and protective clothing (eg, long-sleeved shirts, long pants).

References
  1. Ikegami H, Nogata H, Hirashima K, et al. Analysis of genetic diversity among European and Asian fig varieties (Ficus carica L.) using ISSR, RAPD, and SSR markers. Genetic Resources and Crop Evolution. 2009;56:201-209.
  2. Zohary D, Spiegel-Roy P. Beginnings of fruit growing in the Old World. Science. 1975;187:319-327.
  3. Young R. Young’s Analytical Concordance. Thomas Nelson; 1982.
  4. Duke JA. Handbook of Medicinal Herbs. CRC Press; 2002.
  5. Pathak MA, Fitzpatrick TB. Bioassay of natural and synthetic furocoumarins (psoralens). J Invest Dermatol. 1959;32:509-518.
  6. Focke M, Hemmer W, Wöhrl S, et al. Cross-reactivity between Ficus benjamina latex and fig fruit in patients with clinical fig allergy. Clin Exp Allergy. 2003;33:971-977.
  7. Hemmer W, Focke M, Götz M, et al. Sensitization to Ficus benjamina: relationship to natural rubber latex allergy and identification of foods implicated in the Ficus-fruit syndrome. Clin Exp Allergy. 2004;34:1251-1258.
  8. Bonamonte D, Foti C, Lionetti N, et al. Photoallergic contact dermatitis to 8-methoxypsoralen in Ficus carica. Contact Dermatitis. 2010;62:343-348.
  9. Zaynoun ST, Aftimos BG, Abi Ali L, et al. Ficus carica; isolation and quantification of the photoactive components. Contact Dermatitis. 1984;11:21-25.
  10. Tessman JW, Isaacs ST, Hearst JE. Photochemistry of the furan-side 8-methoxypsoralen-thymidine monoadduct inside the DNA helix. conversion to diadduct and to pyrone-side monoadduct. Biochemistry. 1985;24:1669-1676.
  11. Geary P. Burns related to the use of psoralens as a tanning agent. Burns. 1996;22:636-637.
  12. Redgrave N, Solomon J. Severe phytophotodermatitis from fig sap: a little known phenomenon. BMJ Case Rep. 2021;14:E238745.
  13. Ozdamar E, Ozbek S, Akin S. An unusual cause of burn injury: fig leaf decoction used as a remedy for a dermatitis of unknown etiology. J Burn Care Rehabil. 2003;24:229-233; discussion 228.
  14. Berakha GJ, Lefkovits G. Psoralen phototherapy and phototoxicity. Ann Plast Surg. 1985;14:458-461.
  15. Papazoglou A, Mantadakis E. Fig tree leaves phytophotodermatitis. J Pediatr. 2021;239:244-245.
  16. Imen MS, Ahmadabadi A, Tavousi SH, et al. The curious cases of burn by fig tree leaves. Indian J Dermatol. 2019;64:71-73.
  17. Rouaiguia-Bouakkaz S, Amira-Guebailia H, Rivière C, et al. Identification and quantification of furanocoumarins in stem bark and wood of eight Algerian varieties of Ficus carica by RP-HPLC-DAD and RP-HPLC-DAD-MS. Nat Prod Commun. 2013;8:485-486.
  18. Oliveira AA, Morais J, Pires O, et al. Fig tree induced phytophotodermatitis. BMJ Case Rep. 2020;13:E233392.
  19. Bassioukas K, Stergiopoulou C, Hatzis J. Erythrodermic phytophotodermatitis after application of aqueous fig-leaf extract as an artificial suntan promoter and sunbathing. Contact Dermatitis. 2004;51:94-95.
  20. Sforza M, Andjelkov K, Zaccheddu R. Severe burn on 81% of body surface after sun tanning. Ulus Travma Acil Cerrahi Derg. 2013;19:383-384.
  21. Son JH, Jin H, You HS, et al. Five cases of phytophotodermatitis caused by fig leaves and relevant literature review. Ann Dermatol. 2017;29:86-90.
  22. Abali AE, Aka M, Aydogan C, et al. Burns or phytophotodermatitis, abuse or neglect: confusing aspects of skin lesions caused by the superstitious use of fig leaves. J Burn Care Res. 2012;33:E309-E312.
  23. Picard C, Morice C, Moreau A, et al. Phytophotodermatitis in children: a difficult diagnosis mimicking other dermatitis. 2017;5:1-3.
  24. Enjolras O, Soupre V, Picard A. Uncommon benign infantile vascular tumors. Adv Dermatol. 2008;24:105-124.
References
  1. Ikegami H, Nogata H, Hirashima K, et al. Analysis of genetic diversity among European and Asian fig varieties (Ficus carica L.) using ISSR, RAPD, and SSR markers. Genetic Resources and Crop Evolution. 2009;56:201-209.
  2. Zohary D, Spiegel-Roy P. Beginnings of fruit growing in the Old World. Science. 1975;187:319-327.
  3. Young R. Young’s Analytical Concordance. Thomas Nelson; 1982.
  4. Duke JA. Handbook of Medicinal Herbs. CRC Press; 2002.
  5. Pathak MA, Fitzpatrick TB. Bioassay of natural and synthetic furocoumarins (psoralens). J Invest Dermatol. 1959;32:509-518.
  6. Focke M, Hemmer W, Wöhrl S, et al. Cross-reactivity between Ficus benjamina latex and fig fruit in patients with clinical fig allergy. Clin Exp Allergy. 2003;33:971-977.
  7. Hemmer W, Focke M, Götz M, et al. Sensitization to Ficus benjamina: relationship to natural rubber latex allergy and identification of foods implicated in the Ficus-fruit syndrome. Clin Exp Allergy. 2004;34:1251-1258.
  8. Bonamonte D, Foti C, Lionetti N, et al. Photoallergic contact dermatitis to 8-methoxypsoralen in Ficus carica. Contact Dermatitis. 2010;62:343-348.
  9. Zaynoun ST, Aftimos BG, Abi Ali L, et al. Ficus carica; isolation and quantification of the photoactive components. Contact Dermatitis. 1984;11:21-25.
  10. Tessman JW, Isaacs ST, Hearst JE. Photochemistry of the furan-side 8-methoxypsoralen-thymidine monoadduct inside the DNA helix. conversion to diadduct and to pyrone-side monoadduct. Biochemistry. 1985;24:1669-1676.
  11. Geary P. Burns related to the use of psoralens as a tanning agent. Burns. 1996;22:636-637.
  12. Redgrave N, Solomon J. Severe phytophotodermatitis from fig sap: a little known phenomenon. BMJ Case Rep. 2021;14:E238745.
  13. Ozdamar E, Ozbek S, Akin S. An unusual cause of burn injury: fig leaf decoction used as a remedy for a dermatitis of unknown etiology. J Burn Care Rehabil. 2003;24:229-233; discussion 228.
  14. Berakha GJ, Lefkovits G. Psoralen phototherapy and phototoxicity. Ann Plast Surg. 1985;14:458-461.
  15. Papazoglou A, Mantadakis E. Fig tree leaves phytophotodermatitis. J Pediatr. 2021;239:244-245.
  16. Imen MS, Ahmadabadi A, Tavousi SH, et al. The curious cases of burn by fig tree leaves. Indian J Dermatol. 2019;64:71-73.
  17. Rouaiguia-Bouakkaz S, Amira-Guebailia H, Rivière C, et al. Identification and quantification of furanocoumarins in stem bark and wood of eight Algerian varieties of Ficus carica by RP-HPLC-DAD and RP-HPLC-DAD-MS. Nat Prod Commun. 2013;8:485-486.
  18. Oliveira AA, Morais J, Pires O, et al. Fig tree induced phytophotodermatitis. BMJ Case Rep. 2020;13:E233392.
  19. Bassioukas K, Stergiopoulou C, Hatzis J. Erythrodermic phytophotodermatitis after application of aqueous fig-leaf extract as an artificial suntan promoter and sunbathing. Contact Dermatitis. 2004;51:94-95.
  20. Sforza M, Andjelkov K, Zaccheddu R. Severe burn on 81% of body surface after sun tanning. Ulus Travma Acil Cerrahi Derg. 2013;19:383-384.
  21. Son JH, Jin H, You HS, et al. Five cases of phytophotodermatitis caused by fig leaves and relevant literature review. Ann Dermatol. 2017;29:86-90.
  22. Abali AE, Aka M, Aydogan C, et al. Burns or phytophotodermatitis, abuse or neglect: confusing aspects of skin lesions caused by the superstitious use of fig leaves. J Burn Care Res. 2012;33:E309-E312.
  23. Picard C, Morice C, Moreau A, et al. Phytophotodermatitis in children: a difficult diagnosis mimicking other dermatitis. 2017;5:1-3.
  24. Enjolras O, Soupre V, Picard A. Uncommon benign infantile vascular tumors. Adv Dermatol. 2008;24:105-124.
Issue
Cutis - 113(4)
Issue
Cutis - 113(4)
Page Number
167-169
Page Number
167-169
Publications
Publications
Topics
Article Type
Display Headline
Botanical Briefs: Fig Phytophotodermatitis (Ficus carica)
Display Headline
Botanical Briefs: Fig Phytophotodermatitis (Ficus carica)
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Barker 0424</fileName> <TBEID>0C02F48C.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F48C</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Botanical Briefs:&#13;Fig Phytophot</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240408T094051</firstPublished> <LastPublished>20240408T094051</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240408T094051</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Catherine Shirer Barker, MD; Thomas W. McGovern, MD</byline> <bylineText/> <bylineFull>Catherine Shirer Barker, MD; Thomas W. McGovern, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType>(choose one)</newsDocType> <journalDocType>(choose one)</journalDocType> <linkLabel/> <pageRange>167-169</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Ficus carica (common fig) is a deciduous shrub or small tree with smooth gray bark that can grow up to 10 m in height (Figure 1). It is characterized by many sp</metaDescription> <articlePDF>300906</articlePDF> <teaserImage/> <title>Botanical Briefs: Fig Phytophotodermatitis (Ficus carica)Catherine Shirer Barker, MD; Thomas W. McGovern, MD; Dirk M. Elston, MD</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>April</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>4</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2165</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>April 2024</pubIssueName> <pubArticleType>Audio | 2165</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">60</term> </sections> <topics> <term canonical="true">199</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180026f8.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Botanical Briefs: Fig Phytophotodermatitis (Ficus carica)Catherine Shirer Barker, MD; Thomas W. McGovern, MD; Dirk M. Elston, MD</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Patients presenting with a linear, erythematous, blistering eruption may experience a sudden painful sunburn that seems to get worse rather than better with time. In warm climates, exposure to the common fig tree (<em>Ficus carica</em>) may be the culprit. Dermatologists should recognize fig phytophotodermatitis as a possible cause and help the patient connect their symptoms with the inciting agent as well as administer proper treatment. </p> <h3>Plant Parts and Nomenclature</h3> <p><i>Ficus</i> <i>carica </i>(common fig) is a deciduous shrub or small tree with smooth gray bark that can grow up to 10 m in height (Figure 1). It is characterized by many spreading branches, but the trunk rarely grows beyond a diameter of 7 in. Its hairy leaves are coarse on the upper side and soft underneath with 3 to 7 deep lobes that can extend up to 25 cm in length or width; the leaves grow individually, alternating along the sides of the branches. Fig trees often can be seen adorning yards, gardens, and parks, especially in tropical and subtropical climates. <i>Ficus carica </i>should not be confused with <i>Ficus benjamina</i> (weeping fig), a common ornamental tree that also is used to provide shade in hot climates, though both can cause phototoxic skin eruptions.</p> <p>The common fig tree originated in the Mediterranean and western Asia<sup>1</sup> and has been cultivated by humans since the second and third millennia <scaps>bc</scaps> for its fruit, which commonly is used to sweeten cookies, cakes, and jams.<sup>2</sup> Figs are the most commonly mentioned food plant in the Bible, with at least 56 references in the Old and New Testaments.<sup>3</sup> The “fruit” technically is a syconium—a hollow fleshy receptacle with a small opening at the apex partly closed by small scales. It can be obovoid, turbinate, or pear shaped; can be 1 to 4 inches long; and can vary in color from yellowish green to coppery, bronze, or dark purple (Figure 2).<i> <br/><br/>Ficus carica</i> is a member of the Moraceae family (derived from the Latin name for the mulberry tree), which includes 53 genera and approximately 1400 species, of which about 850 belong to the genus <i>Ficus</i> (the Latin name for a fig tree). The term <i>carica</i> likely comes from the Latin word<i> carricare</i> (to load) to describe a tree loaded with figs. Family members include trees, shrubs, lianas, and herbs that usually contain laticifers with a milky latex.</p> <h3>Traditional Uses</h3> <p>For centuries, components of the fig tree have been used in herbal teas and pastes to treat ailments ranging from sore throats to diarrhea, though there is no evidence to support their efficacy.<sup>4</sup> Ancient Indians and Egyptians used plants such as the common fig tree containing furocoumarins to induce hyperpigmentation in vitiligo.<sup>5</sup> </p> <h3>Phototoxic Components</h3> <p>The leaves and sap of the common fig tree contain psoralens, which are members of the furocoumarin group of chemical compounds and are the source of its phototoxicity. The fruit does not contain psoralens.<sup>6-9</sup> The tree also produces proteolytic enzymes such as protease, amylase, ficin, triterpenoids, and lipodiastase that enhance its phototoxic effects.<sup>8</sup> Exposure to UV light between 320 and 400 nm following contact with these phototoxic components triggers a reaction in the skin over the course of 1 to 3 days.<sup>5</sup> The psoralens bind in epidermal cells, cross-link the DNA, and cause cell-membrane destruction, leading to edema and necrosis.<sup>10</sup> The delay in symptoms may be attributed to the time needed to synthesize acute-phase reaction proteins such as tumor necrosis factor <span class="body">α</span> and IL-1.<sup>11</sup> In spring and summer months, an increased concentration of psoralens in the leaves and sap contribute to an increased incidence of phytophotodermatitis.<sup>9</sup> Humidity and sweat also increase the percutaneous absorption of psoralens.<sup>12,13</sup></p> <h3>Allergens</h3> <p>Fig trees produce a latex protein that can cause cross-reactive hypersensitivity reactions in those allergic to <i>F benjamina </i>latex and rubber latex.<sup>6</sup> The latex proteins in fig trees can act as airborne respiratory allergens. Ingestion of figs can produce anaphylactic reactions in those sensitized to rubber latex and <i>F benjamina </i>latex.<sup>7</sup> Other plant families associated with phototoxic reactions include Rutaceae (lemon, lime, bitter orange), Apiaceae (formerly Umbelliferae)(carrot, parsnip, parsley, dill, celery, hogweed), and Fabaceae (prairie turnip).</p> <h3>Cutaneous Manifestations</h3> <p>Most cases of fig phytophotodermatitis begin with burning, pain, and/or itching within hours of sunlight exposure in areas of the skin that encountered components of the fig tree, often in a linear pattern. The affected areas become erythematous and edematous with formation of bullae and unilocular vesicles over the course of 1 to 3 days.<sup>12,14,15</sup> Lesions may extend beyond the region of contact with the fig tree as they spread across the skin due to sweat or friction, and pain may linger even after the lesions resolve.<sup>12,13,16</sup> Adults who handle fig trees (eg, pruning) are susceptible to phototoxic reactions, especially those using chain saws or other mechanisms that result in spray exposure, as the photosensitizing sap permeates the wood and bark of the entire tree.<sup>17</sup> Similarly, children who handle fig leaves or sap during outdoor play can develop bullous eruptions. Severe cases have resulted in hospital admission after prolonged exposure.<sup>16</sup> Additionally, irritant dermatitis may arise from contact with the trichomes or “hairs” on various parts of the plant. </p> <p>Patients who use natural remedies containing components of the fig tree without the supervision of a medical provider put themselves at risk for unsafe or unwanted adverse effects, such as phytophotodermatitis.<sup>12,15,16,18</sup> An entire family presented with burns after they applied fig leaf extract to the skin prior to tanning outside in the sun.<sup>19</sup> A 42-year-old woman acquired a severe burn covering 81% of the body surface after topically applying fig leaf tea to the skin as a tanning agent.<sup>20</sup> A subset of patients ingesting or applying fig tree components for conditions such as vitiligo, dermatitis, onychomycosis, and motor retardation developed similar cutaneous reactions.<sup>13,14,21,22</sup> Lesions resembling finger marks can raise concerns for potential abuse or neglect in children.<sup>22</sup> <br/><br/>The differential diagnosis for fig phytophotodermatitis includes sunburn, chemical burns, drug-related photosensitivity, infectious lesions (eg, herpes simplex, bullous impetigo, Lyme disease, superficial lymphangitis), connective tissue disease (eg, systemic lupus erythematosus), contact dermatitis, and nonaccidental trauma.<sup>12,15,18</sup> Compared to sunburn, phytophotodermatitis tends to increase in severity over days following exposure and heals with dramatic hyperpigmentation, which also prompts visits to dermatology.<sup>12</sup></p> <h3>Treatment</h3> <p>Treatment of fig phytophotodermatitis chiefly is symptomatic, including analgesia, appropriate wound care, and infection prophylaxis. Topical and systemic corticosteroids may aid in the resolution of moderate to severe reactions.<sup>15,23,24</sup> Even severe injuries over small areas or mild injuries to a high percentage of the total body surface area may require treatment in a burn unit. Patients should be encouraged to use mineral-based sunscreens on the affected areas to reduce the risk for hyperpigmentation. Individuals who regularly handle fig trees should use contact barriers including gloves and protective clothing (eg, long-sleeved shirts, long pants).</p> <h2>References</h2> <p class="reference"> 1. Ikegami H, Nogata H, Hirashima K, et al. Analysis of genetic diversity among European and Asian fig varieties (<i>Ficus carica </i>L.) using ISSR, RAPD, and SSR markers. <i>Genetic Resources and Crop Evolution. </i>2009;56:201-209.</p> <p class="reference"> 2. Zohary D, Spiegel-Roy P. Beginnings of fruit growing in the Old World. <i>Science. </i>1975;187:319-327.<br/><br/> 3. Young R. <i>Young’s Analytical Concordance.</i> Thomas Nelson; 1982.<br/><br/> 4. Duke JA. <i>Handbook of Medicinal Herbs.</i> CRC Press; 2002.<br/><br/> 5. Pathak MA, Fitzpatrick TB. Bioassay of natural and synthetic furocoumarins (psoralens). <i>J Invest Dermatol. </i>1959;32:509-518.<br/><br/> 6. Focke M, Hemmer W, Wöhrl S, et al. Cross-reactivity between <i>Ficus benjamina</i> latex and fig fruit in patients with clinical fig allergy. <i>Clin Exp Allergy. </i>2003;33:971-977.<br/><br/> 7. Hemmer W, Focke M, Götz M, et al. Sensitization to <i>Ficus benjamina</i>: relationship to natural rubber latex allergy and identification of foods implicated in the <i>Ficus</i>-fruit syndrome. <i>Clin Exp Allergy. </i>2004;34:1251-1258.<br/><br/> 8. Bonamonte D, Foti C, Lionetti N, et al. Photoallergic contact dermatitis to 8-methoxypsoralen in <i>Ficus carica</i>. <i>Contact Dermatitis. </i>2010;62:343-348.<br/><br/> 9. Zaynoun ST, Aftimos BG, Abi Ali L, et al. <i>Ficus carica</i>; isolation and quantification of the photoactive components. <i>Contact Dermatitis. </i>1984;11:21-25.<br/><br/>10. Tessman JW, Isaacs ST, Hearst JE. Photochemistry of the furan-side 8-methoxypsoralen-thymidine monoadduct inside the DNA helix. conversion to diadduct and to pyrone-side monoadduct. <i>Biochemistry. </i>1985;24:1669-1676.<br/><br/>11. Geary P. Burns related to the use of psoralens as a tanning agent. <i>Burns. </i>1996;22:636-637.<br/><br/>12. Redgrave N, Solomon J. Severe phytophotodermatitis from fig sap: a little known phenomenon. <i>BMJ Case Rep. </i>2021;14:E238745.<br/><br/>13. Ozdamar E, Ozbek S, Akin S. An unusual cause of burn injury: fig leaf decoction used as a remedy for a dermatitis of unknown etiology. <i>J Burn Care Rehabil. </i>2003;24:229-233; discussion 228.<br/><br/>14. Berakha GJ, Lefkovits G. Psoralen phototherapy and phototoxicity. <i>Ann Plast Surg. </i>1985;14:458-461.<br/><br/>15. Papazoglou A, Mantadakis E. Fig tree leaves phytophotodermatitis. <i>J Pediatr. </i>2021;239:244-245.<br/><br/>16. Imen MS, Ahmadabadi A, Tavousi SH, et al. The curious cases of burn by fig tree leaves. <i>Indian J Dermatol. </i>2019;64:71-73.<br/><br/>17. Rouaiguia-Bouakkaz S, Amira-Guebailia H, Rivière C, et al. Identification and quantification of furanocoumarins in stem bark and wood of eight Algerian varieties of <i>Ficus carica </i>by RP-HPLC-DAD and RP-HPLC-DAD-MS. <i>Nat Prod Commun. </i>2013;8:485-486.<br/><br/>18. Oliveira AA, Morais J, Pires O, et al. Fig tree induced phytophotodermatitis. <i>BMJ Case Rep. </i>2020;13:<span class="cit">E233392</span>.<br/><br/>19. Bassioukas K, Stergiopoulou C, Hatzis J. Erythrodermic phytophotodermatitis after application of aqueous fig-leaf extract as an artificial suntan promoter and sunbathing. <i>Contact Dermatitis. </i>2004;51:94-95.<br/><br/>20. Sforza M, Andjelkov K, Zaccheddu R. Severe burn on 81% of body surface after sun tanning. <i>Ulus Travma Acil Cerrahi Derg. </i>2013;19:383-384.<br/><br/>21. Son JH, Jin H, You HS, et al. Five cases of phytophotodermatitis caused by fig leaves and relevant literature review. <i>Ann Dermatol. </i>2017;29:86-90.<br/><br/>22. Abali AE, Aka M, Aydogan C, et al. Burns or phytophotodermatitis, abuse or neglect: confusing aspects of skin lesions caused by the superstitious use of fig leaves. <i>J Burn Care Res. </i>2012;33:E309-E312.<br/><br/>23. Picard C, Morice C, Moreau A, et al. Phytophotodermatitis in children: a difficult diagnosis mimicking other dermatitis. 2017;5:1-3.<br/><br/>24. Enjolras O, Soupre V, Picard A. Uncommon benign infantile vascular tumors. <i>Adv Dermatol. </i>2008;24:105-124.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Drs. Barker and Elston are from the Medical University of South Carolina, Charleston. Dr. Barker is from the Department of Internal Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.</p> <p class="disclosure">The authors report no conflict of interest.<br/><br/>Correspondence: Catherine Shirer Barker, MD, 96 Jonathan Lucas St, Ste 807B, MSC 623, Charleston, SC 29425 (catherinesbarker@gmail.com).</p> <p class="disclosure"><em>Cutis. </em>2024 April;113(4):167-169. doi:10.12788/cutis.0990</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>Exposure to the components of the common fig tree <em>(Ficus carica) </em>can induce phytophotodermatitis. </li> <li>Notable postinflammatory hyperpigmentation typically occurs in the healing stage of fig phytophotodermatitis.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • Exposure to the components of the common fig tree (Ficus carica) can induce phytophotodermatitis.
  • Notable postinflammatory hyperpigmentation typically occurs in the healing stage of fig phytophotodermatitis.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
180026F8.SIG
Disable zoom
Off

What’s Eating You? Carpet Beetles (Dermestidae)

Article Type
Changed
Wed, 03/06/2024 - 16:32
Display Headline
What’s Eating You? Carpet Beetles (Dermestidae)

Carpet beetle larvae of the family Dermestidae have been documented to cause both acute and delayed hypersensitivity reactions in susceptible individuals. These larvae have specialized horizontal rows of spear-shaped hairs called hastisetae, which detach easily into the surrounding environment and are small enough to travel by air. Exposure to hastisetae has been tied to adverse effects ranging from dermatitis to rhinoconjunctivitis and acute asthma, with treatment being mostly empiric and symptom based. Due to the pervasiveness of carpet beetles in homes, improved awareness of dermestid-induced manifestations is valuable for clinicians.

Beetles in the Dermestidae family do not bite humans but have been reported to cause skin reactions in addition to other symptoms typical of an allergic reaction. Skin contact with larval hairs (hastisetae) of these insects—known as carpet, larder, or hide beetles may cause urticarial or edematous papules that are mistaken for papular urticaria or arthropod bites. 1 There are approximately 500 to 700 species of carpet beetles worldwide. Carpet beetles are a clinically underrecognized cause of allergic contact dermatitis given their frequent presence in homes across the world. 2 Carpet beetle larvae feed on shed skin, feathers, hair, wool, book bindings, felt, leather, wood, silk, and sometimes grains and thus can be found nearly anywhere. Most symptom-inducing exposures to Dermestidae beetles occur occupationally, such as in museum curators working hands-on with collection materials and workers handling infested materials such as wool. 3,4 In-home Dermestidae exposure may lead to symptoms, especially if regularly worn clothing and bedding materials are infested. The broad palate of dermestid members has resulted in substantial contamination of stored materials such as flour and fabric in addition to the destruction of museum collections. 5-7

The larvae of some dermestid species, most commonly of the genera Anthrenus and Dermestes, are 2 to 3 mm in length and have detachable hairlike hastisetae that shed into the surrounding environment throughout larval development (Figure 1).8 The hastisetae, located on the thoracic and abdominal segments (tergites), serve as a larval defense mechanism. When prodded, the round, hairy, wormlike larvae tense up and can raise their abdominal tergites while splaying the hastisetae out in a fanlike manner.9 Similar to porcupine quills, the hastisetae easily detach and can entrap the appendages of invertebrate predators. Hastisetae are not known to be sharp enough to puncture human skin, but friction and irritation from skin contact and superficial sticking of the hastisetae into mucous membranes and noncornified epithelium, such as in the bronchial airways, are thought to induce hypersensitivity reactions in susceptible individuals.

Johnson_carpet_beetles_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Dermestid%20larva.%20Horizontal%20rows%20of%20dark%20setae%20are%20visible%20on%20the%20larva.%20Thin%20lines%20are%20millimeter%20demarcations.%3C%2Fp%3E

Additionally, hastisetae and the exoskeletons of both adult and larval dermestid beetles are composed mostly of chitin, which is highly allergenic. Chitin has been found to play a proinflammatory role in ocular inflammation, asthma, and bronchial reactivity via T helper cell (TH2)–mediated cellular interactions.10-12 Larvae shed their exoskeletons, including hastisetae, multiple times over the course of their development, which contributes to their potential allergen burden (Figure 2). Reports of positive prick and/or patch testing to larval components indicate some cases of both acute type 1 and delayed type 4 hypersensitivity reactions.4,8,13

CT113003006_e_Fig2_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20A%20and%20B%2C%20Molted%20exoskeletons%20of%20dermestid%20larvae.%3C%2Fp%3E

Clinical Presentation and Diagnosis

Multiple erythematous urticarial papules, papulopustules, and papulovesicles are the typical manifestations of dermestid dermatitis.3,4,13-16 Figure 3 demonstrates several characteristic edematous papules with background erythema. Unlike the clusters seen with flea and bed bug bites, dermestid-induced lesions typically are single and scattered, with a propensity for exposed limbs and the face. Exposure to hastisetae commonly results in classic allergic symptoms including rhinitis, conjunctivitis, coughing, wheezing, sneezing, and intranasal and periocular pruritus, even in those with no personal history of atopy.17-19 Lymphadenopathy, vasculitis, and allergic alveolitis also have been reported.20 A large infestation in which many individual beetles as well as larvae can be found in 1 or more areas of the inhabited structure has been reported to cause more severe symptoms, including acute eczema, otitis externa, lymphocytic vasculitis, and allergic alveolitis, all of which resolved within 3 months of thorough deinfestation cleaning.21

CT113003006_e_Fig3_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20A%20and%20B%2C%20Edematous%20papules%20on%20the%20face%20with%20background%20erythema%20from%20dermestid%20larva%20contact.%3C%2Fp%3E

Skin-prick and/or patch testing is not necessary for this clinical diagnosis of dermestid-induced allergic contact dermatitis. This diagnosis is bolstered by (but does not require a history of) repeated symptom induction upon performing certain activities (eg, handling taxidermy specimens) and/or in certain environments (eg, only at home). Because of individual differences in hypersensitivity to dermestid parts, it is not typical for all members of a household to be affected.

When there are multiple potential suspected allergens or an unknown cause for symptoms despite a detailed history, allergy testing can be useful in confirming a diagnosis and directing management. Immediate-onset type 1 hypersensitivity reactions are evaluated using skin-prick testing or serum IgE levels, whereas delayed type 4 hypersensitivity reactions can be evaluated using patch testing. Type 1 reactions tend to present with classic allergy symptoms, especially where there are abundant mast cells to degranulate in the skin and mucosa of the gastrointestinal and respiratory tracts; these symptoms range from mild wheezing, urticaria, periorbital pruritus, and sneezing to outright asthma, diarrhea, rhinoconjunctivitis, and even anaphylaxis. With these reactions, initial exposure to an antigen such as chitin in the hastisetae leads to an asymptomatic sensitization against the antigen in which its introduction leads to a TH2-skewed cellular response, which promotes B-cell production of IgE antibodies. Upon subsequent exposure to this antigen, IgE antibodies bound to mast cells will lead them to degranulate with release of histamine and other proinflammatory molecules, resulting in clinical manifestations. The skin-prick test relies on introduction of potential antigens through the epidermis into the dermis with a sharp lancet to induce IgE antibody activation and then degranulation of the patient’s mast cells, resulting in a pruritic erythematous wheal. This IgE-mediated process has been shown to occur in response to dermestid larval parts among household dust, resulting in chronic coughing, sneezing, nasal pruritus, and asthma.15,17,22

 

 

Type 4 hypersensitivity reactions are T-cell mediated and also include a sensitization phase followed by symptom manifestation upon repeat exposure; however, these reactions usually are not immediate and can take up to 72 hours after exposure to manifest.23 This is because T cells specific to the antigen do not lead a process resulting in antibodies but instead recruit numerous other TH1-polarized mediators upon re-exposure to activate cytotoxic CD8+ T cells and macrophages to attempt to neutralize the antigen. Many type 4 reactions result in mostly cutaneous manifestations, such as contact dermatitis. Patch testing involves adhering potential allergens to the skin for a time with assessments at regular intervals to evaluate the level of reaction from weakly positive to severe. At minimum, most reports of dermestid-related manifestations include a rash such as erythematous papules, and several published cases involving patch testing have yielded positive results to various preparations of larval parts.3,14,21

Management and Treatment

Prevention of dermestid exposure is difficult given the myriad materials eaten by the larvae. An insect exterminator should verify and treat a carpet beetle infestation, while a dermatologist can treat symptomatic individuals. Treatment is driven by the severity of the patient’s discomfort and is aimed at both symptomatic relief and reducing dermestid exposure moving forward. Although in certain environments it will be nearly impossible to eradicate Dermestidae, cleaning thoroughly and regularly may go far to reduce exposure and associated symptoms.

Clothing and other materials such as bedding that will have direct skin contact should be washed to remove hastisetae and be stored in airtight containers in addition to items made with animal fibers, such as wool sweaters and down blankets. Mattresses, flooring, rugs, curtains, and other amenable areas should be vacuumed thoroughly, and the vacuum bag should be placed in the trash afterward. Protective pillow and mattress covers should be used. Stuffed animals in infested areas should be thrown away if not able to be completely washed and dried. Air conditioning systems may spread larval hairs away from the site of infestation and should be cleaned as much as possible. Surfaces where beetles and larvae also are commonly seen, such as windowsills, and hidden among closet and pantry items should also be wiped clean to remove both insects and potential substrate. In one case, scraping the wood flooring and applying a thick coat of varnish in addition to removing all stuffed animals from an affected individual’s home allowed for resolution of symptoms.17

Treatment for symptoms includes topical anti-inflammatory agents and/or oral antihistamines, with improvement in symptoms typically occurring within days and resolution dependent on level of exposure moving forward.

Final Thoughts

There is a broad overlap between dermestid habitats and human-occupied environments; thus, the opportunities for exposure and sensitization to allergenic dermestid parts are numerous. Dermatologists should be aware of the possible manifestations from dermestid exposure.

References
  1. Gumina ME, Yan AC. Carpet beetle dermatitis mimicking bullous impetigo. Pediatr Dermatol. 2021;38:329-331. doi:10.1111/pde.14453
  2. Bertone MA, Leong M, Bayless KM, et al. Arthropods of the great indoors: characterizing diversity inside urban and suburban homes. PeerJ. 2016;4:E1582. doi:10.7717/peerj.1582
  3. Siegel S, Lee N, Rohr A, et. al. Evaluation of dermestid sensitivity in museum personnel. J Allergy Clin Immunol. 1991;87:190. doi:10.1016/0091-6749(91)91488-F
  4. Brito FF, Mur P, Barber D, et al. Occupational rhinoconjunctivitis and asthma in a wool worker caused by Dermestidae spp. Allergy. 2002;57:1191-1194.
  5. Stengaard HL, Akerlund M, Grontoft T, et al. Future pest status of an insect pest in museums, Attagenus smirnovi: distribution and food consumption in relation to climate change. J Cult Herit. 2012;13:22l-227.
  6. Veer V, Negi BK, Rao KM. Dermestid beetles and some other insect pests associated with stored silkworm cocoons in India, including a world list of dermestid species found attacking this commodity. J Stored Products Research. 1996;32:69-89.
  7. Veer V, Prasad R, Rao KM. Taxonomic and biological notes on Attagenus and Anthrenus spp. (Coleoptera: Dermestidae) found damaging stored woolen fabrics in India. J Stored Products Research. 1991;27:189-198.
  8. Háva J. World Catalogue of Insects. Volume 13. Dermestidae (Coleoptera). Brill; 2015.
  9. Ruzzier E, Kadej M, Di Giulio A, et al. Entangling the enemy: ecological, systematic, and medical implications of dermestid beetle Hastisetae. Insects. 2021;12:436. doi:10.3390/insects12050436
  10. Arae K, Morita H, Unno H, et al. Chitin promotes antigen-specific Th2 cell-mediated murine asthma through induction of IL-33-mediated IL-1β production by DCs. Sci Rep. 2018;8:11721.
  11. Brinchmann BC, Bayat M, Brøgger T, et. al. A possible role of chitin in the pathogenesis of asthma and allergy. Ann Agric Environ Med. 2011;18:7-12.
  12. Bucolo C, Musumeci M, Musumeci S, et al. Acidic mammalian chitinase and the eye: implications for ocular inflammatory diseases. Front Pharmacol. 2011;2:1-4.
  13. Hoverson K, Wohltmann WE, Pollack RJ, et al. Dermestid dermatitis in a 2-year-old girl: case report and review of the literature. Pediatr Dermatol. 2015;32:E228-E233. doi:10.1111/pde.12641
  14. Simon L, Boukari F, Oumarou H, et al. Anthrenus sp. and an uncommon cluster of dermatitis. Emerg Infect Dis. 2021;27:1940-1943. doi:10.3201/eid2707.203245
  15. Ahmed R, Moy R, Barr R, et al. Carpet beetle dermatitis. J Am Acad Dermatol. 1981;5:428-432.
  16. MacArthur K, Richardson V, Novoa R, et al. Carpet beetle dermatitis: a possibly under-recognized entity. Int J Dermatol. 2016;55:577-579.
  17. Cuesta-Herranz J, de las Heras M, Sastre J, et al. Asthma caused by Dermestidae (black carpet beetle): a new allergen in house dust. J Allergy Clin Immunol. 1997;99(1 Pt 1):147-149.
  18. Bernstein J, Morgan M, Ghosh D, et al. Respiratory sensitization of a worker to the warehouse beetle Trogoderma variabile: an index case report. J Allergy Clin Immunol. 2009;123:1413-1416.
  19. Gorgojo IE, De Las Heras M, Pastor C, et al. Allergy to Dermestidae: a new indoor allergen? [abstract] J Allergy Clin Immunol. 2015;135:AB105.
  20. Ruzzier E, Kadej M, Battisti A. Occurrence, ecological function and medical importance of dermestid beetle hastisetae. PeerJ. 2020;8:E8340. doi:10.7717/peerj.8340
  21. Ramachandran J, Hern J, Almeyda J, et al. Contact dermatitis with cervical lymphadenopathy following exposure to the hide beetle, Dermestes peruvianus. Br J Dermatol. 1997;136:943-945.
  22. Horster S, Prinz J, Holm N, et al. Anthrenus-dermatitis. Hautarzt. 2002;53:328-331.
  23. Justiz Vaillant AA, Vashisht R, Zito PM. Immediate hypersensitivity reactions. In: StatPearls. StatPearls Publishing; 2023.
Article PDF
Author and Disclosure Information

From the Department of Dermatology, University Hospitals Cleveland Medical Center, Ohio.

The authors report no conflict of interest.

Correspondence: Amy G. Johnson, MD, Department of Dermatology, University Hospitals Cleveland Medical Center, 11000 Euclid Ave, Cleveland, OH 44106 (amy.johnson@uhhospitals.org).

Issue
Cutis - 113(3)
Publications
Topics
Page Number
E6-E9
Sections
Author and Disclosure Information

From the Department of Dermatology, University Hospitals Cleveland Medical Center, Ohio.

The authors report no conflict of interest.

Correspondence: Amy G. Johnson, MD, Department of Dermatology, University Hospitals Cleveland Medical Center, 11000 Euclid Ave, Cleveland, OH 44106 (amy.johnson@uhhospitals.org).

Author and Disclosure Information

From the Department of Dermatology, University Hospitals Cleveland Medical Center, Ohio.

The authors report no conflict of interest.

Correspondence: Amy G. Johnson, MD, Department of Dermatology, University Hospitals Cleveland Medical Center, 11000 Euclid Ave, Cleveland, OH 44106 (amy.johnson@uhhospitals.org).

Article PDF
Article PDF

Carpet beetle larvae of the family Dermestidae have been documented to cause both acute and delayed hypersensitivity reactions in susceptible individuals. These larvae have specialized horizontal rows of spear-shaped hairs called hastisetae, which detach easily into the surrounding environment and are small enough to travel by air. Exposure to hastisetae has been tied to adverse effects ranging from dermatitis to rhinoconjunctivitis and acute asthma, with treatment being mostly empiric and symptom based. Due to the pervasiveness of carpet beetles in homes, improved awareness of dermestid-induced manifestations is valuable for clinicians.

Beetles in the Dermestidae family do not bite humans but have been reported to cause skin reactions in addition to other symptoms typical of an allergic reaction. Skin contact with larval hairs (hastisetae) of these insects—known as carpet, larder, or hide beetles may cause urticarial or edematous papules that are mistaken for papular urticaria or arthropod bites. 1 There are approximately 500 to 700 species of carpet beetles worldwide. Carpet beetles are a clinically underrecognized cause of allergic contact dermatitis given their frequent presence in homes across the world. 2 Carpet beetle larvae feed on shed skin, feathers, hair, wool, book bindings, felt, leather, wood, silk, and sometimes grains and thus can be found nearly anywhere. Most symptom-inducing exposures to Dermestidae beetles occur occupationally, such as in museum curators working hands-on with collection materials and workers handling infested materials such as wool. 3,4 In-home Dermestidae exposure may lead to symptoms, especially if regularly worn clothing and bedding materials are infested. The broad palate of dermestid members has resulted in substantial contamination of stored materials such as flour and fabric in addition to the destruction of museum collections. 5-7

The larvae of some dermestid species, most commonly of the genera Anthrenus and Dermestes, are 2 to 3 mm in length and have detachable hairlike hastisetae that shed into the surrounding environment throughout larval development (Figure 1).8 The hastisetae, located on the thoracic and abdominal segments (tergites), serve as a larval defense mechanism. When prodded, the round, hairy, wormlike larvae tense up and can raise their abdominal tergites while splaying the hastisetae out in a fanlike manner.9 Similar to porcupine quills, the hastisetae easily detach and can entrap the appendages of invertebrate predators. Hastisetae are not known to be sharp enough to puncture human skin, but friction and irritation from skin contact and superficial sticking of the hastisetae into mucous membranes and noncornified epithelium, such as in the bronchial airways, are thought to induce hypersensitivity reactions in susceptible individuals.

Johnson_carpet_beetles_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Dermestid%20larva.%20Horizontal%20rows%20of%20dark%20setae%20are%20visible%20on%20the%20larva.%20Thin%20lines%20are%20millimeter%20demarcations.%3C%2Fp%3E

Additionally, hastisetae and the exoskeletons of both adult and larval dermestid beetles are composed mostly of chitin, which is highly allergenic. Chitin has been found to play a proinflammatory role in ocular inflammation, asthma, and bronchial reactivity via T helper cell (TH2)–mediated cellular interactions.10-12 Larvae shed their exoskeletons, including hastisetae, multiple times over the course of their development, which contributes to their potential allergen burden (Figure 2). Reports of positive prick and/or patch testing to larval components indicate some cases of both acute type 1 and delayed type 4 hypersensitivity reactions.4,8,13

CT113003006_e_Fig2_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20A%20and%20B%2C%20Molted%20exoskeletons%20of%20dermestid%20larvae.%3C%2Fp%3E

Clinical Presentation and Diagnosis

Multiple erythematous urticarial papules, papulopustules, and papulovesicles are the typical manifestations of dermestid dermatitis.3,4,13-16 Figure 3 demonstrates several characteristic edematous papules with background erythema. Unlike the clusters seen with flea and bed bug bites, dermestid-induced lesions typically are single and scattered, with a propensity for exposed limbs and the face. Exposure to hastisetae commonly results in classic allergic symptoms including rhinitis, conjunctivitis, coughing, wheezing, sneezing, and intranasal and periocular pruritus, even in those with no personal history of atopy.17-19 Lymphadenopathy, vasculitis, and allergic alveolitis also have been reported.20 A large infestation in which many individual beetles as well as larvae can be found in 1 or more areas of the inhabited structure has been reported to cause more severe symptoms, including acute eczema, otitis externa, lymphocytic vasculitis, and allergic alveolitis, all of which resolved within 3 months of thorough deinfestation cleaning.21

CT113003006_e_Fig3_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20A%20and%20B%2C%20Edematous%20papules%20on%20the%20face%20with%20background%20erythema%20from%20dermestid%20larva%20contact.%3C%2Fp%3E

Skin-prick and/or patch testing is not necessary for this clinical diagnosis of dermestid-induced allergic contact dermatitis. This diagnosis is bolstered by (but does not require a history of) repeated symptom induction upon performing certain activities (eg, handling taxidermy specimens) and/or in certain environments (eg, only at home). Because of individual differences in hypersensitivity to dermestid parts, it is not typical for all members of a household to be affected.

When there are multiple potential suspected allergens or an unknown cause for symptoms despite a detailed history, allergy testing can be useful in confirming a diagnosis and directing management. Immediate-onset type 1 hypersensitivity reactions are evaluated using skin-prick testing or serum IgE levels, whereas delayed type 4 hypersensitivity reactions can be evaluated using patch testing. Type 1 reactions tend to present with classic allergy symptoms, especially where there are abundant mast cells to degranulate in the skin and mucosa of the gastrointestinal and respiratory tracts; these symptoms range from mild wheezing, urticaria, periorbital pruritus, and sneezing to outright asthma, diarrhea, rhinoconjunctivitis, and even anaphylaxis. With these reactions, initial exposure to an antigen such as chitin in the hastisetae leads to an asymptomatic sensitization against the antigen in which its introduction leads to a TH2-skewed cellular response, which promotes B-cell production of IgE antibodies. Upon subsequent exposure to this antigen, IgE antibodies bound to mast cells will lead them to degranulate with release of histamine and other proinflammatory molecules, resulting in clinical manifestations. The skin-prick test relies on introduction of potential antigens through the epidermis into the dermis with a sharp lancet to induce IgE antibody activation and then degranulation of the patient’s mast cells, resulting in a pruritic erythematous wheal. This IgE-mediated process has been shown to occur in response to dermestid larval parts among household dust, resulting in chronic coughing, sneezing, nasal pruritus, and asthma.15,17,22

 

 

Type 4 hypersensitivity reactions are T-cell mediated and also include a sensitization phase followed by symptom manifestation upon repeat exposure; however, these reactions usually are not immediate and can take up to 72 hours after exposure to manifest.23 This is because T cells specific to the antigen do not lead a process resulting in antibodies but instead recruit numerous other TH1-polarized mediators upon re-exposure to activate cytotoxic CD8+ T cells and macrophages to attempt to neutralize the antigen. Many type 4 reactions result in mostly cutaneous manifestations, such as contact dermatitis. Patch testing involves adhering potential allergens to the skin for a time with assessments at regular intervals to evaluate the level of reaction from weakly positive to severe. At minimum, most reports of dermestid-related manifestations include a rash such as erythematous papules, and several published cases involving patch testing have yielded positive results to various preparations of larval parts.3,14,21

Management and Treatment

Prevention of dermestid exposure is difficult given the myriad materials eaten by the larvae. An insect exterminator should verify and treat a carpet beetle infestation, while a dermatologist can treat symptomatic individuals. Treatment is driven by the severity of the patient’s discomfort and is aimed at both symptomatic relief and reducing dermestid exposure moving forward. Although in certain environments it will be nearly impossible to eradicate Dermestidae, cleaning thoroughly and regularly may go far to reduce exposure and associated symptoms.

Clothing and other materials such as bedding that will have direct skin contact should be washed to remove hastisetae and be stored in airtight containers in addition to items made with animal fibers, such as wool sweaters and down blankets. Mattresses, flooring, rugs, curtains, and other amenable areas should be vacuumed thoroughly, and the vacuum bag should be placed in the trash afterward. Protective pillow and mattress covers should be used. Stuffed animals in infested areas should be thrown away if not able to be completely washed and dried. Air conditioning systems may spread larval hairs away from the site of infestation and should be cleaned as much as possible. Surfaces where beetles and larvae also are commonly seen, such as windowsills, and hidden among closet and pantry items should also be wiped clean to remove both insects and potential substrate. In one case, scraping the wood flooring and applying a thick coat of varnish in addition to removing all stuffed animals from an affected individual’s home allowed for resolution of symptoms.17

Treatment for symptoms includes topical anti-inflammatory agents and/or oral antihistamines, with improvement in symptoms typically occurring within days and resolution dependent on level of exposure moving forward.

Final Thoughts

There is a broad overlap between dermestid habitats and human-occupied environments; thus, the opportunities for exposure and sensitization to allergenic dermestid parts are numerous. Dermatologists should be aware of the possible manifestations from dermestid exposure.

Carpet beetle larvae of the family Dermestidae have been documented to cause both acute and delayed hypersensitivity reactions in susceptible individuals. These larvae have specialized horizontal rows of spear-shaped hairs called hastisetae, which detach easily into the surrounding environment and are small enough to travel by air. Exposure to hastisetae has been tied to adverse effects ranging from dermatitis to rhinoconjunctivitis and acute asthma, with treatment being mostly empiric and symptom based. Due to the pervasiveness of carpet beetles in homes, improved awareness of dermestid-induced manifestations is valuable for clinicians.

Beetles in the Dermestidae family do not bite humans but have been reported to cause skin reactions in addition to other symptoms typical of an allergic reaction. Skin contact with larval hairs (hastisetae) of these insects—known as carpet, larder, or hide beetles may cause urticarial or edematous papules that are mistaken for papular urticaria or arthropod bites. 1 There are approximately 500 to 700 species of carpet beetles worldwide. Carpet beetles are a clinically underrecognized cause of allergic contact dermatitis given their frequent presence in homes across the world. 2 Carpet beetle larvae feed on shed skin, feathers, hair, wool, book bindings, felt, leather, wood, silk, and sometimes grains and thus can be found nearly anywhere. Most symptom-inducing exposures to Dermestidae beetles occur occupationally, such as in museum curators working hands-on with collection materials and workers handling infested materials such as wool. 3,4 In-home Dermestidae exposure may lead to symptoms, especially if regularly worn clothing and bedding materials are infested. The broad palate of dermestid members has resulted in substantial contamination of stored materials such as flour and fabric in addition to the destruction of museum collections. 5-7

The larvae of some dermestid species, most commonly of the genera Anthrenus and Dermestes, are 2 to 3 mm in length and have detachable hairlike hastisetae that shed into the surrounding environment throughout larval development (Figure 1).8 The hastisetae, located on the thoracic and abdominal segments (tergites), serve as a larval defense mechanism. When prodded, the round, hairy, wormlike larvae tense up and can raise their abdominal tergites while splaying the hastisetae out in a fanlike manner.9 Similar to porcupine quills, the hastisetae easily detach and can entrap the appendages of invertebrate predators. Hastisetae are not known to be sharp enough to puncture human skin, but friction and irritation from skin contact and superficial sticking of the hastisetae into mucous membranes and noncornified epithelium, such as in the bronchial airways, are thought to induce hypersensitivity reactions in susceptible individuals.

Johnson_carpet_beetles_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Dermestid%20larva.%20Horizontal%20rows%20of%20dark%20setae%20are%20visible%20on%20the%20larva.%20Thin%20lines%20are%20millimeter%20demarcations.%3C%2Fp%3E

Additionally, hastisetae and the exoskeletons of both adult and larval dermestid beetles are composed mostly of chitin, which is highly allergenic. Chitin has been found to play a proinflammatory role in ocular inflammation, asthma, and bronchial reactivity via T helper cell (TH2)–mediated cellular interactions.10-12 Larvae shed their exoskeletons, including hastisetae, multiple times over the course of their development, which contributes to their potential allergen burden (Figure 2). Reports of positive prick and/or patch testing to larval components indicate some cases of both acute type 1 and delayed type 4 hypersensitivity reactions.4,8,13

CT113003006_e_Fig2_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20A%20and%20B%2C%20Molted%20exoskeletons%20of%20dermestid%20larvae.%3C%2Fp%3E

Clinical Presentation and Diagnosis

Multiple erythematous urticarial papules, papulopustules, and papulovesicles are the typical manifestations of dermestid dermatitis.3,4,13-16 Figure 3 demonstrates several characteristic edematous papules with background erythema. Unlike the clusters seen with flea and bed bug bites, dermestid-induced lesions typically are single and scattered, with a propensity for exposed limbs and the face. Exposure to hastisetae commonly results in classic allergic symptoms including rhinitis, conjunctivitis, coughing, wheezing, sneezing, and intranasal and periocular pruritus, even in those with no personal history of atopy.17-19 Lymphadenopathy, vasculitis, and allergic alveolitis also have been reported.20 A large infestation in which many individual beetles as well as larvae can be found in 1 or more areas of the inhabited structure has been reported to cause more severe symptoms, including acute eczema, otitis externa, lymphocytic vasculitis, and allergic alveolitis, all of which resolved within 3 months of thorough deinfestation cleaning.21

CT113003006_e_Fig3_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20A%20and%20B%2C%20Edematous%20papules%20on%20the%20face%20with%20background%20erythema%20from%20dermestid%20larva%20contact.%3C%2Fp%3E

Skin-prick and/or patch testing is not necessary for this clinical diagnosis of dermestid-induced allergic contact dermatitis. This diagnosis is bolstered by (but does not require a history of) repeated symptom induction upon performing certain activities (eg, handling taxidermy specimens) and/or in certain environments (eg, only at home). Because of individual differences in hypersensitivity to dermestid parts, it is not typical for all members of a household to be affected.

When there are multiple potential suspected allergens or an unknown cause for symptoms despite a detailed history, allergy testing can be useful in confirming a diagnosis and directing management. Immediate-onset type 1 hypersensitivity reactions are evaluated using skin-prick testing or serum IgE levels, whereas delayed type 4 hypersensitivity reactions can be evaluated using patch testing. Type 1 reactions tend to present with classic allergy symptoms, especially where there are abundant mast cells to degranulate in the skin and mucosa of the gastrointestinal and respiratory tracts; these symptoms range from mild wheezing, urticaria, periorbital pruritus, and sneezing to outright asthma, diarrhea, rhinoconjunctivitis, and even anaphylaxis. With these reactions, initial exposure to an antigen such as chitin in the hastisetae leads to an asymptomatic sensitization against the antigen in which its introduction leads to a TH2-skewed cellular response, which promotes B-cell production of IgE antibodies. Upon subsequent exposure to this antigen, IgE antibodies bound to mast cells will lead them to degranulate with release of histamine and other proinflammatory molecules, resulting in clinical manifestations. The skin-prick test relies on introduction of potential antigens through the epidermis into the dermis with a sharp lancet to induce IgE antibody activation and then degranulation of the patient’s mast cells, resulting in a pruritic erythematous wheal. This IgE-mediated process has been shown to occur in response to dermestid larval parts among household dust, resulting in chronic coughing, sneezing, nasal pruritus, and asthma.15,17,22

 

 

Type 4 hypersensitivity reactions are T-cell mediated and also include a sensitization phase followed by symptom manifestation upon repeat exposure; however, these reactions usually are not immediate and can take up to 72 hours after exposure to manifest.23 This is because T cells specific to the antigen do not lead a process resulting in antibodies but instead recruit numerous other TH1-polarized mediators upon re-exposure to activate cytotoxic CD8+ T cells and macrophages to attempt to neutralize the antigen. Many type 4 reactions result in mostly cutaneous manifestations, such as contact dermatitis. Patch testing involves adhering potential allergens to the skin for a time with assessments at regular intervals to evaluate the level of reaction from weakly positive to severe. At minimum, most reports of dermestid-related manifestations include a rash such as erythematous papules, and several published cases involving patch testing have yielded positive results to various preparations of larval parts.3,14,21

Management and Treatment

Prevention of dermestid exposure is difficult given the myriad materials eaten by the larvae. An insect exterminator should verify and treat a carpet beetle infestation, while a dermatologist can treat symptomatic individuals. Treatment is driven by the severity of the patient’s discomfort and is aimed at both symptomatic relief and reducing dermestid exposure moving forward. Although in certain environments it will be nearly impossible to eradicate Dermestidae, cleaning thoroughly and regularly may go far to reduce exposure and associated symptoms.

Clothing and other materials such as bedding that will have direct skin contact should be washed to remove hastisetae and be stored in airtight containers in addition to items made with animal fibers, such as wool sweaters and down blankets. Mattresses, flooring, rugs, curtains, and other amenable areas should be vacuumed thoroughly, and the vacuum bag should be placed in the trash afterward. Protective pillow and mattress covers should be used. Stuffed animals in infested areas should be thrown away if not able to be completely washed and dried. Air conditioning systems may spread larval hairs away from the site of infestation and should be cleaned as much as possible. Surfaces where beetles and larvae also are commonly seen, such as windowsills, and hidden among closet and pantry items should also be wiped clean to remove both insects and potential substrate. In one case, scraping the wood flooring and applying a thick coat of varnish in addition to removing all stuffed animals from an affected individual’s home allowed for resolution of symptoms.17

Treatment for symptoms includes topical anti-inflammatory agents and/or oral antihistamines, with improvement in symptoms typically occurring within days and resolution dependent on level of exposure moving forward.

Final Thoughts

There is a broad overlap between dermestid habitats and human-occupied environments; thus, the opportunities for exposure and sensitization to allergenic dermestid parts are numerous. Dermatologists should be aware of the possible manifestations from dermestid exposure.

References
  1. Gumina ME, Yan AC. Carpet beetle dermatitis mimicking bullous impetigo. Pediatr Dermatol. 2021;38:329-331. doi:10.1111/pde.14453
  2. Bertone MA, Leong M, Bayless KM, et al. Arthropods of the great indoors: characterizing diversity inside urban and suburban homes. PeerJ. 2016;4:E1582. doi:10.7717/peerj.1582
  3. Siegel S, Lee N, Rohr A, et. al. Evaluation of dermestid sensitivity in museum personnel. J Allergy Clin Immunol. 1991;87:190. doi:10.1016/0091-6749(91)91488-F
  4. Brito FF, Mur P, Barber D, et al. Occupational rhinoconjunctivitis and asthma in a wool worker caused by Dermestidae spp. Allergy. 2002;57:1191-1194.
  5. Stengaard HL, Akerlund M, Grontoft T, et al. Future pest status of an insect pest in museums, Attagenus smirnovi: distribution and food consumption in relation to climate change. J Cult Herit. 2012;13:22l-227.
  6. Veer V, Negi BK, Rao KM. Dermestid beetles and some other insect pests associated with stored silkworm cocoons in India, including a world list of dermestid species found attacking this commodity. J Stored Products Research. 1996;32:69-89.
  7. Veer V, Prasad R, Rao KM. Taxonomic and biological notes on Attagenus and Anthrenus spp. (Coleoptera: Dermestidae) found damaging stored woolen fabrics in India. J Stored Products Research. 1991;27:189-198.
  8. Háva J. World Catalogue of Insects. Volume 13. Dermestidae (Coleoptera). Brill; 2015.
  9. Ruzzier E, Kadej M, Di Giulio A, et al. Entangling the enemy: ecological, systematic, and medical implications of dermestid beetle Hastisetae. Insects. 2021;12:436. doi:10.3390/insects12050436
  10. Arae K, Morita H, Unno H, et al. Chitin promotes antigen-specific Th2 cell-mediated murine asthma through induction of IL-33-mediated IL-1β production by DCs. Sci Rep. 2018;8:11721.
  11. Brinchmann BC, Bayat M, Brøgger T, et. al. A possible role of chitin in the pathogenesis of asthma and allergy. Ann Agric Environ Med. 2011;18:7-12.
  12. Bucolo C, Musumeci M, Musumeci S, et al. Acidic mammalian chitinase and the eye: implications for ocular inflammatory diseases. Front Pharmacol. 2011;2:1-4.
  13. Hoverson K, Wohltmann WE, Pollack RJ, et al. Dermestid dermatitis in a 2-year-old girl: case report and review of the literature. Pediatr Dermatol. 2015;32:E228-E233. doi:10.1111/pde.12641
  14. Simon L, Boukari F, Oumarou H, et al. Anthrenus sp. and an uncommon cluster of dermatitis. Emerg Infect Dis. 2021;27:1940-1943. doi:10.3201/eid2707.203245
  15. Ahmed R, Moy R, Barr R, et al. Carpet beetle dermatitis. J Am Acad Dermatol. 1981;5:428-432.
  16. MacArthur K, Richardson V, Novoa R, et al. Carpet beetle dermatitis: a possibly under-recognized entity. Int J Dermatol. 2016;55:577-579.
  17. Cuesta-Herranz J, de las Heras M, Sastre J, et al. Asthma caused by Dermestidae (black carpet beetle): a new allergen in house dust. J Allergy Clin Immunol. 1997;99(1 Pt 1):147-149.
  18. Bernstein J, Morgan M, Ghosh D, et al. Respiratory sensitization of a worker to the warehouse beetle Trogoderma variabile: an index case report. J Allergy Clin Immunol. 2009;123:1413-1416.
  19. Gorgojo IE, De Las Heras M, Pastor C, et al. Allergy to Dermestidae: a new indoor allergen? [abstract] J Allergy Clin Immunol. 2015;135:AB105.
  20. Ruzzier E, Kadej M, Battisti A. Occurrence, ecological function and medical importance of dermestid beetle hastisetae. PeerJ. 2020;8:E8340. doi:10.7717/peerj.8340
  21. Ramachandran J, Hern J, Almeyda J, et al. Contact dermatitis with cervical lymphadenopathy following exposure to the hide beetle, Dermestes peruvianus. Br J Dermatol. 1997;136:943-945.
  22. Horster S, Prinz J, Holm N, et al. Anthrenus-dermatitis. Hautarzt. 2002;53:328-331.
  23. Justiz Vaillant AA, Vashisht R, Zito PM. Immediate hypersensitivity reactions. In: StatPearls. StatPearls Publishing; 2023.
References
  1. Gumina ME, Yan AC. Carpet beetle dermatitis mimicking bullous impetigo. Pediatr Dermatol. 2021;38:329-331. doi:10.1111/pde.14453
  2. Bertone MA, Leong M, Bayless KM, et al. Arthropods of the great indoors: characterizing diversity inside urban and suburban homes. PeerJ. 2016;4:E1582. doi:10.7717/peerj.1582
  3. Siegel S, Lee N, Rohr A, et. al. Evaluation of dermestid sensitivity in museum personnel. J Allergy Clin Immunol. 1991;87:190. doi:10.1016/0091-6749(91)91488-F
  4. Brito FF, Mur P, Barber D, et al. Occupational rhinoconjunctivitis and asthma in a wool worker caused by Dermestidae spp. Allergy. 2002;57:1191-1194.
  5. Stengaard HL, Akerlund M, Grontoft T, et al. Future pest status of an insect pest in museums, Attagenus smirnovi: distribution and food consumption in relation to climate change. J Cult Herit. 2012;13:22l-227.
  6. Veer V, Negi BK, Rao KM. Dermestid beetles and some other insect pests associated with stored silkworm cocoons in India, including a world list of dermestid species found attacking this commodity. J Stored Products Research. 1996;32:69-89.
  7. Veer V, Prasad R, Rao KM. Taxonomic and biological notes on Attagenus and Anthrenus spp. (Coleoptera: Dermestidae) found damaging stored woolen fabrics in India. J Stored Products Research. 1991;27:189-198.
  8. Háva J. World Catalogue of Insects. Volume 13. Dermestidae (Coleoptera). Brill; 2015.
  9. Ruzzier E, Kadej M, Di Giulio A, et al. Entangling the enemy: ecological, systematic, and medical implications of dermestid beetle Hastisetae. Insects. 2021;12:436. doi:10.3390/insects12050436
  10. Arae K, Morita H, Unno H, et al. Chitin promotes antigen-specific Th2 cell-mediated murine asthma through induction of IL-33-mediated IL-1β production by DCs. Sci Rep. 2018;8:11721.
  11. Brinchmann BC, Bayat M, Brøgger T, et. al. A possible role of chitin in the pathogenesis of asthma and allergy. Ann Agric Environ Med. 2011;18:7-12.
  12. Bucolo C, Musumeci M, Musumeci S, et al. Acidic mammalian chitinase and the eye: implications for ocular inflammatory diseases. Front Pharmacol. 2011;2:1-4.
  13. Hoverson K, Wohltmann WE, Pollack RJ, et al. Dermestid dermatitis in a 2-year-old girl: case report and review of the literature. Pediatr Dermatol. 2015;32:E228-E233. doi:10.1111/pde.12641
  14. Simon L, Boukari F, Oumarou H, et al. Anthrenus sp. and an uncommon cluster of dermatitis. Emerg Infect Dis. 2021;27:1940-1943. doi:10.3201/eid2707.203245
  15. Ahmed R, Moy R, Barr R, et al. Carpet beetle dermatitis. J Am Acad Dermatol. 1981;5:428-432.
  16. MacArthur K, Richardson V, Novoa R, et al. Carpet beetle dermatitis: a possibly under-recognized entity. Int J Dermatol. 2016;55:577-579.
  17. Cuesta-Herranz J, de las Heras M, Sastre J, et al. Asthma caused by Dermestidae (black carpet beetle): a new allergen in house dust. J Allergy Clin Immunol. 1997;99(1 Pt 1):147-149.
  18. Bernstein J, Morgan M, Ghosh D, et al. Respiratory sensitization of a worker to the warehouse beetle Trogoderma variabile: an index case report. J Allergy Clin Immunol. 2009;123:1413-1416.
  19. Gorgojo IE, De Las Heras M, Pastor C, et al. Allergy to Dermestidae: a new indoor allergen? [abstract] J Allergy Clin Immunol. 2015;135:AB105.
  20. Ruzzier E, Kadej M, Battisti A. Occurrence, ecological function and medical importance of dermestid beetle hastisetae. PeerJ. 2020;8:E8340. doi:10.7717/peerj.8340
  21. Ramachandran J, Hern J, Almeyda J, et al. Contact dermatitis with cervical lymphadenopathy following exposure to the hide beetle, Dermestes peruvianus. Br J Dermatol. 1997;136:943-945.
  22. Horster S, Prinz J, Holm N, et al. Anthrenus-dermatitis. Hautarzt. 2002;53:328-331.
  23. Justiz Vaillant AA, Vashisht R, Zito PM. Immediate hypersensitivity reactions. In: StatPearls. StatPearls Publishing; 2023.
Issue
Cutis - 113(3)
Issue
Cutis - 113(3)
Page Number
E6-E9
Page Number
E6-E9
Publications
Publications
Topics
Article Type
Display Headline
What’s Eating You? Carpet Beetles (Dermestidae)
Display Headline
What’s Eating You? Carpet Beetles (Dermestidae)
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Johnson carpet beetles</fileName> <TBEID>0C02F394.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F394</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Johnson carpet beetles</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240306T150748</firstPublished> <LastPublished>20240306T150748</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240306T150748</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Amy G. Johnson, MD; Bethany R. Rohr, MD</byline> <bylineText>Amy G. Johnson, MD; Bethany R. Rohr, MD</bylineText> <bylineFull>Amy G. Johnson, MD; Bethany R. Rohr, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>E6-E9</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>B eetles in the Dermestidae family do not bite humans but have been reported to cause skin reactions in addition to other symptoms typical of an allergic reacti</metaDescription> <articlePDF>300513</articlePDF> <teaserImage/> <title>What’s Eating You? Carpet Beetles (Dermestidae)</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>March</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>3</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2159</CMSID> </CMSIDs> <keywords> <keyword>contact dermatitis</keyword> <keyword> carpet beetle</keyword> <keyword> dermestidae</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>March 2024</pubIssueName> <pubArticleType>Departments | 2159</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">60</term> </sections> <topics> <term canonical="true">199</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180026e9.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>What’s Eating You? Carpet Beetles (Dermestidae)</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Carpet beetle larvae of the family Dermestidae have been documented to cause both acute and delayed hypersensitivity reactions in susceptible individuals. These larvae have specialized horizontal rows of spear-shaped hairs called hastisetae, which detach easily into the surrounding environment and are small enough to travel by air. Exposure to hastisetae has been tied to adverse effects ranging from dermatitis to rhinoconjunctivitis and acute asthma, with treatment being mostly empiric and symptom based. Due to the pervasiveness of carpet beetles in homes, improved awareness of dermestid-induced manifestations is valuable for clinicians.</p> <p> <span class="body">B</span> eetles in the Dermestidae family do not bite humans but have been reported to cause skin reactions in addition to other symptoms typical of an allergic reaction. Skin contact with larval hairs (hastisetae) of these insects—known as carpet, larder, or hide beetles <i>—</i> may cause urticarial or edematous papules that are mistaken for papular urticaria or arthropod bites. <sup>1</sup> There are approximately 500 to 700 species of carpet beetles worldwide. Carpet beetles are a clinically underrecognized cause of allergic contact dermatitis given their frequent presence in homes across the world. <sup>2</sup> Carpet beetle larvae feed on shed skin, feathers, hair, wool, book bindings, felt, leather, wood, silk, and sometimes grains and thus can be found nearly anywhere. Most symptom-inducing exposures to Dermestidae beetles occur occupationally, such as in museum curators working hands-on with collection materials and workers handling infested materials such as wool. <sup>3,4</sup> In-home Dermestidae exposure may lead to symptoms, especially if regularly worn clothing and bedding materials are infested. The broad palate of dermestid members has resulted in substantial contamination of stored materials such as flour and fabric in addition to the destruction of museum collections. <sup>5-7</sup> </p> <p>The larvae of some dermestid species, most commonly of the genera <i>Anthrenus</i> and <i>Dermestes</i>, are 2 to 3 mm in length and have detachable hairlike hastisetae that shed into the surrounding environment throughout larval development (Figure 1).<sup>8</sup> The hastisetae, located on the thoracic and abdominal segments (tergites), serve as a larval defense mechanism. When prodded, the round, hairy, wormlike larvae tense up and can raise their abdominal tergites while splaying the hastisetae out in a fanlike manner.<sup>9</sup> Similar to porcupine quills, the hastisetae easily detach and can entrap the appendages of invertebrate predators. Hastisetae are not known to be sharp enough to puncture human skin, but friction and irritation from skin contact and superficial sticking of the hastisetae into mucous membranes and noncornified epithelium, such as in the bronchial airways, are thought to induce hypersensitivity reactions in susceptible individuals.<br/><br/>Additionally, hastisetae and the exoskeletons of both adult and larval dermestid beetles are composed mostly of chitin, which is highly allergenic. Chitin has been found to play a proinflammatory role in ocular inflammation, asthma, and bronchial reactivity via T helper cell (T<sub>H</sub>2)–mediated cellular interactions.<sup>10-12</sup> Larvae shed their exoskeletons, including hastisetae, multiple times over the course of their development, which contributes to their potential allergen burden (Figure 2). Reports of positive prick and/or patch testing to larval components indicate some cases of both acute type 1 and delayed type 4 hypersensitivity reactions.<sup>4,8,13</sup></p> <h3>Clinical Presentation and Diagnosis</h3> <p>Multiple erythematous urticarial papules, papulopustules, and papulovesicles are the typical manifestations of dermestid dermatitis.<sup>3,4,13-16</sup> Figure 3 demonstrates several characteristic edematous papules with background erythema. Unlike the clusters seen with flea and bed bug bites, dermestid-induced lesions typically are single and scattered, with a propensity for exposed limbs and the face. Exposure to hastisetae commonly results in classic allergic symptoms including rhinitis, conjunctivitis, coughing, wheezing, sneezing, and intranasal and periocular pruritus, even in those with no personal history of atopy.<sup>17-19</sup> Lymphadenopathy, vasculitis, and allergic alveolitis also have been reported.<sup>20</sup> A large infestation in which many individual beetles as well as larvae can be found in 1 or more areas of the inhabited structure has been reported to cause more severe symptoms, including acute eczema, otitis externa, lymphocytic vasculitis, and allergic alveolitis, all of which resolved within 3 months of thorough deinfestation cleaning.<sup>21</sup></p> <p>Skin-prick and/or patch testing is not necessary for this clinical diagnosis of dermestid-induced allergic contact dermatitis. This diagnosis is bolstered by (but does not require a history of) repeated symptom induction upon performing certain activities (eg, handling taxidermy specimens) and/or in certain environments (eg, only at home). Because of individual differences in hypersensitivity to dermestid parts, it is not typical for all members of a household to be affected.<br/><br/>When there are multiple potential suspected allergens or an unknown cause for symptoms despite a detailed history, allergy testing can be useful in confirming a diagnosis and directing management. Immediate-onset type 1 hypersensitivity reactions are evaluated using skin-prick testing or serum IgE levels, whereas delayed type 4 hypersensitivity reactions can be evaluated using patch testing. Type 1 reactions tend to present with classic allergy symptoms, especially where there are abundant mast cells to degranulate in the skin and mucosa of the gastrointestinal and respiratory tracts; these symptoms range from mild wheezing, urticaria, periorbital pruritus, and sneezing to outright asthma, diarrhea, rhinoconjunctivitis, and even anaphylaxis. With these reactions, initial exposure to an antigen such as chitin in the hastisetae leads to an asymptomatic sensitization against the antigen in which its introduction leads to a T<sub>H</sub>2-skewed cellular response, which promotes B-cell production of IgE antibodies. Upon subsequent exposure to this antigen, IgE antibodies bound to mast cells will lead them to degranulate with release of histamine and other proinflammatory molecules, resulting in clinical manifestations. The skin-prick test relies on introduction of potential antigens through the epidermis into the dermis with a sharp lancet to induce IgE antibody activation and then degranulation of the patient’s mast cells, resulting in a pruritic erythematous wheal. This IgE-mediated process has been shown to occur in response to dermestid larval parts among household dust, resulting in chronic coughing, sneezing, nasal pruritus, and asthma.<sup>15,17,22<br/><br/></sup>Type 4 hypersensitivity reactions are T-cell mediated and also include a sensitization phase followed by symptom manifestation upon repeat exposure; however, these reactions usually are not immediate and can take up to 72 hours after exposure to manifest.<sup>23</sup> This is because T cells specific to the antigen do not lead a process resulting in antibodies but instead recruit numerous other T<sub>H</sub>1-polarized mediators upon re-exposure to activate cytotoxic CD8<span class="body"><sup>+</sup></span> T cells and macrophages to attempt to neutralize the antigen. Many type 4 reactions result in mostly cutaneous manifestations, such as contact dermatitis. Patch testing involves adhering potential allergens to the skin for a time with assessments at regular intervals to evaluate the level of reaction from weakly positive to severe. At minimum, most reports of dermestid-related manifestations include a rash such as erythematous papules, and several published cases involving patch testing have yielded positive results to various preparations of larval parts.<sup>3,14,21</sup></p> <h3>Management and Treatment</h3> <p>Prevention of dermestid exposure is difficult given the myriad materials eaten by the larvae. An insect exterminator should verify and treat a carpet beetle infestation, while a dermatologist can treat symptomatic individuals. Treatment is driven by the severity of the patient’s discomfort and is aimed at both symptomatic relief and reducing dermestid exposure moving forward. Although in certain environments it will be nearly impossible to eradicate Dermestidae, cleaning thoroughly and regularly may go far to reduce exposure and associated symptoms.</p> <p>Clothing and other materials such as bedding that will have direct skin contact should be washed to remove hastisetae and be stored in airtight containers in addition to items made with animal fibers, such as wool sweaters and down blankets. Mattresses, flooring, rugs, curtains, and other amenable areas should be vacuumed thoroughly, and the vacuum bag should be placed in the trash afterward. Protective pillow and mattress covers should be used. Stuffed animals in infested areas should be thrown away if not able to be completely washed and dried. Air conditioning systems may spread larval hairs away from the site of infestation and should be cleaned as much as possible. Surfaces where beetles and larvae also are commonly seen, such as windowsills, and hidden among closet and pantry items should also be wiped clean to remove both insects and potential substrate. In one case, scraping the wood flooring and applying a thick coat of varnish in addition to removing all stuffed animals from an affected individual’s home allowed for resolution of symptoms.<sup>17<br/><br/></sup>Treatment for symptoms includes topical anti-inflammatory agents and/or oral antihistamines, with improvement in symptoms typically occurring within days and resolution dependent on level of exposure moving forward.</p> <h3>Final Thoughts</h3> <p><hl name="3"/>There is a broad overlap between dermestid habitats and human-occupied environments; thus, the opportunities for exposure and sensitization to allergenic dermestid parts are numerous. Dermatologists should be aware of the possible manifestations from dermestid exposure. </p> <h2>References</h2> <p class="reference"> 1. Gumina ME, Yan AC. Carpet beetle dermatitis mimicking bullous impetigo. <i>Pediatr Dermatol</i>. 2021;38:329-331. doi:10.1111/pde.14453<br/><br/> 2. Bertone MA, Leong M, Bayless KM, et al. Arthropods of the great indoors: characterizing diversity inside urban and suburban homes. <i>PeerJ. </i>2016;4:E1582. doi:10.7717/peerj.1582<br/><br/> 3. Siegel S, Lee N, Rohr A, et. al. Evaluation of dermestid sensitivity in museum personnel. <i>J Allergy Clin Immunol.</i> 1991;87:190. doi:10.1016/0091-6749(91)91488-F<br/><br/> 4. Brito FF, Mur P, Barber D, et al. Occupational rhinoconjunctivitis and asthma in a wool worker caused by Dermestidae spp. <i>Allergy</i>. 2002;57:1191-1194.<br/><br/> 5. Stengaard HL, Akerlund M, Grontoft T, et al. Future pest status of an insect pest in museums, <i>Attagenus smirnovi</i>: distribution and food consumption in relation to climate change. <i>J Cult Herit</i>. 2012;13:22l-227.<br/><br/> 6. Veer V, Negi BK, Rao KM. Dermestid beetles and some other insect pests associated with stored silkworm cocoons in India, including a world list of dermestid species found attacking this commodity. <i>J Stored Products Research</i>. 1996;32:69-89. <br/><br/> 7. Veer V, Prasad R, Rao KM. Taxonomic and biological notes on <i>Attagenus</i> and <i>Anthrenus</i> spp. (Coleoptera: Dermestidae) found damaging stored woolen fabrics in India. <i>J Stored Products Research</i>. 1991;27:189-198.<br/><br/> 8. Háva J. <i>World Catalogue of Insects. </i>Volume 13.<i> Dermestidae (Coleoptera)</i>. Brill; 2015.<br/><br/> 9. Ruzzier E, Kadej M, Di Giulio A, et al. Entangling the enemy: ecological, systematic, and medical implications of dermestid beetle Hastisetae. <i>Insects</i>. 2021;12:436. doi:10.3390/insects12050436<br/><br/>10. Arae K, Morita H, Unno H, et al. Chitin promotes antigen-specific Th2 cell-mediated murine asthma through induction of IL-33-mediated IL-1<span class="body">β</span> production by DCs. <i>Sci Rep</i>. 2018;8:11721.<br/><br/>11. Brinchmann BC, Bayat M, Brøgger T, et. al. A possible role of chitin in the pathogenesis of asthma and allergy. <i>Ann Agric Environ Med</i>. 2011;18:7-12.<br/><br/>12. Bucolo C, Musumeci M, Musumeci S, et al. Acidic mammalian chitinase and the eye: implications for ocular inflammatory diseases. <i>Front Pharmacol</i>. 2011;2:1-4.<br/><br/>13. Hoverson K, Wohltmann WE, Pollack RJ, et al. Dermestid dermatitis in a 2-year-old girl: case report and review of the literature. <i>Pediatr Dermatol</i>. 2015;32:E228-E233. doi:10.1111/pde.12641<br/><br/>14. Simon L, Boukari F, Oumarou H, et al. Anthrenus sp. and an uncommon cluster of dermatitis. <i>Emerg Infect Dis</i>. 2021;27:1940-1943. doi:10.3201/eid2707.203245<br/><br/>15. Ahmed R, Moy R, Barr R, et al. Carpet beetle dermatitis. <i>J Am Acad Dermatol</i>. 1981;5:428-432.<br/><br/>16. MacArthur K, Richardson V, Novoa R, et al. Carpet beetle dermatitis: a possibly under-recognized entity. <i>Int J Dermatol</i>. 2016;55:577-579.<br/><br/>17. Cuesta-Herranz J, de las Heras M, Sastre J, et al. Asthma caused by Dermestidae (black carpet beetle): a new allergen in house dust. <i>J Allergy Clin Immunol</i>. 1997;99(1 Pt 1):147-149.<br/><br/>18. Bernstein J, Morgan M, Ghosh D, et al. Respiratory sensitization of a worker to the warehouse beetle <i>Trogoderma variabile</i>: an index case report. <i>J Allergy Clin Immunol</i>. 2009;123:1413-1416.<br/><br/>19. Gorgojo IE, De Las Heras M, Pastor C, et al. Allergy to Dermestidae: a new indoor allergen? [abstract] <i>J Allergy Clin Immunol</i>. 2015;135:AB105.<br/><br/>20. Ruzzier E, Kadej M, Battisti A. Occurrence, ecological function and medical importance of dermestid beetle hastisetae. <i>PeerJ.</i> 2020;8:E8340. doi:10.7717/peerj.8340<br/><br/>21. Ramachandran J, Hern J, Almeyda J, et al. Contact dermatitis with cervical lymphadenopathy following exposure to the hide beetle, <i>Dermestes peruvianus</i>. <i>Br J Dermatol</i>. 1997;136:943-945.<br/><br/>22. Horster S, Prinz J, Holm N, et al. Anthrenus-dermatitis. <i>Hautarzt</i>. 2002;53:328-331.<br/><br/>23. Justiz Vaillant AA, Vashisht R, Zito PM. Immediate hypersensitivity reactions. In: <i>StatPearls</i>. StatPearls Publishing; 2023.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">From the Department of Dermatology, University Hospitals Cleveland Medical Center, Ohio.</p> <p class="disclosure">The authors report no conflict of interest.<br/><br/>Correspondence: Amy G. Johnson, MD, Department of Dermatology, University Hospitals Cleveland Medical Center, 11000 Euclid Ave, Cleveland, OH 44106 (amy.johnson@uhhospitals.org).<br/><br/><i>Cutis.</i> 2024 March;113(3):E6-E9. doi:10.12788/cutis.0979</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>Given their ubiquity, dermatologists should be aware of the potential for hypersensitivity reactions to carpet beetles (Dermestidae). </li> <li>Pruritic erythematous papules, pustules, and vesicles are the most common manifestations of exposure to larval hairs. </li> <li>Treatment is symptom based, and future exposure can be greatly diminished with thorough cleaning of the patient’s environment.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • Given their ubiquity, dermatologists should be aware of the potential for hypersensitivity reactions to carpet beetles (Dermestidae).
  • Pruritic erythematous papules, pustules, and vesicles are the most common manifestations of exposure to larval hairs.
  • Treatment is symptom based, and future exposure can be greatly diminished with thorough cleaning of the patient’s environment.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
180026E9.SIG
Disable zoom
Off

Aquatic Antagonists: Scorpionfish Envenomation

Article Type
Changed
Wed, 03/06/2024 - 11:47
Display Headline
Aquatic Antagonists: Scorpionfish Envenomation

With the growing popularity of water sports and a proliferation of invasive species, human injuries from marine animal envenomation continue to rise.1-3 Members of the scorpionfish family Scorpaenidae are second only to stingrays as the leading cause of the 40,000 to 50,000 injuries annually from marine life worldwide.4 Because scorpionfish represent a growing threat and competition with native species, it has been suggested that they could replace endangered species on restaurant menus.5-8 Scorpionfish have been introduced by humans from tropical to temperate seas and are now common off the coast of California and the eastern coast from New York to Florida, as well as in the Caribbean, the Bahamas, and off the southern coast of Brazil. Victims of scorpionfish stings experience considerable pain and may require days to weeks to fully recover, highlighting the socioeconomic costs and burden of scorpionfish envenomation.9,10 Fishers, divers, swimmers, and aquarium owners are most often affected.

Family

The common term scorpionfish refers to both the family Scorpaenidae and the genus Scorpaena. Members of this family possess similar dorsal, anal, and pelvic fins, though they vary between genera in their size and the potency of the venom they insulate. Other familiar members include the genus Pterois (lionfish) and Synanceja (stonefish). Synanceja are the most venomous within the group, but scorpionfish stings more commonly arise from Pterois and Scorpaena.8 Because of the rare shapes and vibrant colors of scorpionfish, some traders and aquarium owners will seek and pay high prices for these fish, providing further opportunity for envenomation.11,12

Characteristics

Scorpionfish have with a high variation in color, ranging from lighter grays to intense reds depending on their geographic location and habitat. Synanceja are bland in coloration, blending in with rocks and gravel, but the more dramatic-appearing Scorpaena exhibit a large cranium and wide range of multicolored patterns (Figure 1).13Pterois serve as the most conspicuous member of the group with brightly colored red and white stripes (Figure 2). Scorpionfish commonly grow up to 19 inches long and boast 12 dorsal, 2 pelvic, and 3 anal spines housing 5 to 10 mg of venom.14 An integumentary sheath encapsulates each spine housing the glandular tissue that produces the potent venom.

Afvari_scorpionfish_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Red%20scorpionfish%20(%3Cem%3EScorpaena%20scrofa%3C%2Fem%3E).%3C%2Fp%3E

Toxin Properties

Unlike Pterois and Synanceja, Scorpaena do not have venom ducts around their glands, complicating the work of marine biologists aiming to extract and study the venomous toxins. Several studies have managed to isolate scorpionfish venom and overcome its unstable heat-labile nature to investigate its biologic properties.15-20 Several high-molecular-weight proteins (50–800 kDa) comprise the venom, including hyaluronidase, integrin-inhibiting factors, capillary permeability factor, proteases, and some less-understood cytolytic toxins. These factors provoke the inflammatory, proteolytic, hemorrhagic, cardiovascular, and hemolytic biologic activities at both the local and systemic levels, directing damage to wounded tissues and inducing vascular and tissue permeability to reach cellular processes far and wide. Mediators of inflammation include tumor necrosis factor, IL-6, and monocyte chemoattractant protein 1, followed by neutrophils and other mononuclear cells, initiating the immune response at the wound site. Toxin potency remains for up to 2 days after fish death.1

Afvari_scorpionfish_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Lionfish%20(%3Cem%3EPterois%20volitans%3C%2Fem%3E).%3C%2Fp%3E

Clinical Manifestation

Physicians may be guided by clinical symptoms in identifying scorpionfish stings, as the patient may not know the identity of their marine assailant. Initially, individuals punctured by scorpionfish spikes will experience an acute pain and burning sensation at the puncture site that may be accompanied by systemic symptoms such as nausea, vomiting, diarrhea, tachycardia, hypotension, loss of consciousness, difficulty breathing, and delirium.9,21-23 The pain will intensify and radiate distal to the site of envenomation, and the wound may exhibit vesiculation, erythema, bruising, pallor, and notable edema.4,24 Pain intensity peaks at 30 to 90 minutes after envenomation, and other systemic symptoms generally last for 24 to 48 hours.25 If patients do not seek prompt treatment, secondary infection may ensue, and the lingering venom in the blister may cause dermal necrosis, paresthesia, and anesthesia. Chronic sequelae may include joint contractures, compartment syndrome, necrotic ulcers, and chronic neuropathy.1

Management

Treatment of scorpionfish stings primarily is palliative and aimed at symptom reduction. Patients should immediately treat wounds with hot but not scalding water immersion.26,27 Given the thermolabile components of scorpionfish venom, the most effective treatment is to soak the affected limb in water 42 °C to 45 °C for 30 to 90 minutes. Any higher temperature may pose risk for scalding burns. Children should be monitored throughout treatment.28 If hot water immersion does not provide relief, oral analgesics may be considered. Stonefish antivenom is available and may be used for any scorpionfish sting given the shared biologic properties between genera. Providers evaluating stings could use sterile irrigation to clean wounds and search for foreign bodies including spine fragments; probing should be accomplished by instruments rather than a gloved finger. Providers should consider culturing wounds and prescribing antibiotics for suspected secondary infections. A tetanus toxoid history also should be elicited, and patients may have a booster administered, as indicated.29

References
  1. Rensch G, Murphy-Lavoie HM. Lionfish, scorpionfish, and stonefish toxicity. StatPearls. StatPearls Publishing; May 10, 2022.
  2. Cearnal L. Red lionfish and ciguatoxin: menace spreading through western hemisphere. Ann Emerg Med. 2012;60:21A-22A. doi:10.1016/j.annemergmed.2012.05.022
  3. Côté IM, Green SJ. Potential effects of climate change on a marine invasion: the importance of current context. Curr Zool. 2012;58:1-8. doi:10.1093/czoolo/58.1.1
  4. Venomology of scorpionfishes. In: Santhanam R. Biology and Ecology of Venomous Marine Scorpionfishes. Academic Press; 2019:263-278.
  5. Ferri J, Staglicˇic´ N, Matić-Skoko S. The black scorpionfish, Scorpaena porcus (Scorpaenidae): could it serve as reliable indicator of Mediterranean coastal communities’ health? Ecol Indicators. 2012;18:25-30. doi:10.1016/j.ecolind.2011.11.004
  6. Santhanam R. Biology and Ecology of Venomous Marine Scorpionfishes. Academic Press; 2019.
  7. Morris JA, Akins JL. Feeding ecology of invasive lionfish (Pterois volitans) in the Bahamian Archipelago. Environ Biol Fishes. 2009;86:389-398. doi:10.1007/s10641-009-9538-8 
  8. Albins MA, Hixon MA. Worst case scenario: potential long-term effects of invasive predatory lionfish (Pterois volitans) on Atlantic and Caribbean coral-reef communities. Environ Biol Fishes. 2013;96:1151–1157. doi:10.1007/s10641-011-9795-1
  9. Haddad V Jr, Martins IA, Makyama HM. Injuries caused by scorpionfishes (Scorpaena plumieri Bloch, 1789 and Scorpaena brasiliensis Cuvier, 1829) in the Southwestern Atlantic Ocean (Brazilian coast): epidemiologic, clinic and therapeutic aspects of 23 stings in humans. Toxicon. 2003;42:79-83. doi:10.1016/s0041-0101(03)00103-x
  10. Campos FV, Menezes TN, Malacarne PF, et al. A review on the Scorpaena plumieri fish venom and its bioactive compounds. J Venom Anim Toxins Incl Trop Dis. 2016;22:35. doi:10.1186/s40409-016-0090-7
  11. Needleman RK, Neylan IP, Erickson TB. Environmental and ecological effects of climate change on venomous marine and amphibious species in the wilderness. Wilderness Environ Med. 2018;29:343-356. doi:10.1016/j.wem.2018.04.003
  12. Aldred B, Erickson T, Lipscomb J. Lionfish envenomations in an urban wilderness. Wilderness Environ Med. 1996;7:291-296. doi:10.1580/1080-6032(1996)007[0291:leiauw]2.3.co;2
  13. Stewart J, Hughes JM. Life-history traits of the southern hemisphere eastern red scorpionfish, Scorpaena cardinalis (Scorpaenidae: Scorpaeninae). Mar Freshw Res. 2010;61:1290-1297. doi:10.1071/MF10040
  14. Auerbach PS. Marine envenomations. N Engl J Med. 1991;325:486-493. doi:10.1056/NEJM199108153250707
  15. Andrich F, Carnielli JB, Cassoli JS, et al. A potent vasoactive cytolysin isolated from Scorpaena plumieri scorpionfish venom. Toxicon. 2010;56:487-496. doi:10.1016/j.toxicon.2010.05.003
  16. Gomes HL, Andrich F, Mauad H, et al. Cardiovascular effects of scorpionfish (Scorpaena plumieri) venom. Toxicon. 2010;55(2-3):580-589. doi:10.1016/j.toxicon.2009.10.012
  17. Menezes TN, Carnielli JB, Gomes HL, et al. Local inflammatory response induced by scorpionfish Scorpaena plumieri venom in mice. Toxicon. 2012;60:4-11. doi:10.1016/j.toxicon.2012.03.008
  18. Schaeffer RC Jr, Carlson RW, Russell FE. Some chemical properties of the venom of the scorpionfish Scorpaena guttata. Toxicon. 1971;9:69-78. doi:10.1016/0041-0101(71)90045-6
  19. Khalil AM, Wahsha MA, Abu Khadra KM, et al. Biochemical and histopathological effects of the stonefish (Synanceia verrucosa) venom in rats. Toxicon. 2018;142:45-51. doi:10.1016/j.toxicon.2017.12.052
  20. Mouchbahani-Constance S, Lesperance LS, Petitjean H, et al. Lionfish venom elicits pain predominantly through the activation of nonpeptidergic nociceptors. Pain. 2018;159:2255-2266. doi:10.1097/j.pain.0000000000001326
  21. Ottuso P. Aquatic dermatology: encounters with the denizens of the deep (and not so deep)—a review. part II: the vertebrates, single-celled organisms, and aquatic biotoxins. Int J Dermatol. 2013;52:268-278. doi:10.1111/j.1365-4632.2011.05426.x
  22. Bayley HH. Injuries caused by scorpion fish. Trans R Soc Trop Med Hyg. 1940;34:227-230. doi:10.1016/s0035-9203(40)90072-4
  23. González D. Epidemiological and clinical aspects of certain venomous animals of Spain. Toxicon. 1982;20:925-928. doi:10.1016/0041-0101(82)90080-0
  24. Halstead BW. Injurious effects from the sting of the scorpionfish, Scorpaena guttata. with report of a case. Calif Med. 1951;74:395-396.
  25. Vasievich MP, Villarreal JD, Tomecki KJ. Got the travel bug? a review of common infections, infestations, bites, and stings among returning travelers. Am J Clin Dermatol. 2016;17:451-462. doi:10.1007/s40257-016-0203-7
  26. Barnett S, Saggiomo S, Smout M, et al. Heat deactivation of the stonefish Synanceia horrida venom—implications for first-aid management. Diving Hyperb Med. 2017;47:155-158. doi:10.28920/dhm47.3.155-158
  27. Russell FE. Weever fish sting: the last word. Br Med J (Clin Res Ed). 1983;287:981-982. doi:10.1136/bmj.287.6397.981-c
  28. Tomlinson H, Elston DM. Aquatic antagonists: lionfish (Pterois volitans). Cutis. 2018;102:232-234.
  29. Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337. doi:10.1016/j.emc.2016.12.004
Article PDF
Author and Disclosure Information

Shawn Afvari is from the New York Medical College School of Medicine, Valhalla. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Correspondence: Shawn Afvari, BS (safvari@student.nymc.edu).

Issue
Cutis - 113(3)
Publications
Topics
Page Number
133-134,136
Sections
Author and Disclosure Information

Shawn Afvari is from the New York Medical College School of Medicine, Valhalla. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Correspondence: Shawn Afvari, BS (safvari@student.nymc.edu).

Author and Disclosure Information

Shawn Afvari is from the New York Medical College School of Medicine, Valhalla. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Correspondence: Shawn Afvari, BS (safvari@student.nymc.edu).

Article PDF
Article PDF

With the growing popularity of water sports and a proliferation of invasive species, human injuries from marine animal envenomation continue to rise.1-3 Members of the scorpionfish family Scorpaenidae are second only to stingrays as the leading cause of the 40,000 to 50,000 injuries annually from marine life worldwide.4 Because scorpionfish represent a growing threat and competition with native species, it has been suggested that they could replace endangered species on restaurant menus.5-8 Scorpionfish have been introduced by humans from tropical to temperate seas and are now common off the coast of California and the eastern coast from New York to Florida, as well as in the Caribbean, the Bahamas, and off the southern coast of Brazil. Victims of scorpionfish stings experience considerable pain and may require days to weeks to fully recover, highlighting the socioeconomic costs and burden of scorpionfish envenomation.9,10 Fishers, divers, swimmers, and aquarium owners are most often affected.

Family

The common term scorpionfish refers to both the family Scorpaenidae and the genus Scorpaena. Members of this family possess similar dorsal, anal, and pelvic fins, though they vary between genera in their size and the potency of the venom they insulate. Other familiar members include the genus Pterois (lionfish) and Synanceja (stonefish). Synanceja are the most venomous within the group, but scorpionfish stings more commonly arise from Pterois and Scorpaena.8 Because of the rare shapes and vibrant colors of scorpionfish, some traders and aquarium owners will seek and pay high prices for these fish, providing further opportunity for envenomation.11,12

Characteristics

Scorpionfish have with a high variation in color, ranging from lighter grays to intense reds depending on their geographic location and habitat. Synanceja are bland in coloration, blending in with rocks and gravel, but the more dramatic-appearing Scorpaena exhibit a large cranium and wide range of multicolored patterns (Figure 1).13Pterois serve as the most conspicuous member of the group with brightly colored red and white stripes (Figure 2). Scorpionfish commonly grow up to 19 inches long and boast 12 dorsal, 2 pelvic, and 3 anal spines housing 5 to 10 mg of venom.14 An integumentary sheath encapsulates each spine housing the glandular tissue that produces the potent venom.

Afvari_scorpionfish_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Red%20scorpionfish%20(%3Cem%3EScorpaena%20scrofa%3C%2Fem%3E).%3C%2Fp%3E

Toxin Properties

Unlike Pterois and Synanceja, Scorpaena do not have venom ducts around their glands, complicating the work of marine biologists aiming to extract and study the venomous toxins. Several studies have managed to isolate scorpionfish venom and overcome its unstable heat-labile nature to investigate its biologic properties.15-20 Several high-molecular-weight proteins (50–800 kDa) comprise the venom, including hyaluronidase, integrin-inhibiting factors, capillary permeability factor, proteases, and some less-understood cytolytic toxins. These factors provoke the inflammatory, proteolytic, hemorrhagic, cardiovascular, and hemolytic biologic activities at both the local and systemic levels, directing damage to wounded tissues and inducing vascular and tissue permeability to reach cellular processes far and wide. Mediators of inflammation include tumor necrosis factor, IL-6, and monocyte chemoattractant protein 1, followed by neutrophils and other mononuclear cells, initiating the immune response at the wound site. Toxin potency remains for up to 2 days after fish death.1

Afvari_scorpionfish_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Lionfish%20(%3Cem%3EPterois%20volitans%3C%2Fem%3E).%3C%2Fp%3E

Clinical Manifestation

Physicians may be guided by clinical symptoms in identifying scorpionfish stings, as the patient may not know the identity of their marine assailant. Initially, individuals punctured by scorpionfish spikes will experience an acute pain and burning sensation at the puncture site that may be accompanied by systemic symptoms such as nausea, vomiting, diarrhea, tachycardia, hypotension, loss of consciousness, difficulty breathing, and delirium.9,21-23 The pain will intensify and radiate distal to the site of envenomation, and the wound may exhibit vesiculation, erythema, bruising, pallor, and notable edema.4,24 Pain intensity peaks at 30 to 90 minutes after envenomation, and other systemic symptoms generally last for 24 to 48 hours.25 If patients do not seek prompt treatment, secondary infection may ensue, and the lingering venom in the blister may cause dermal necrosis, paresthesia, and anesthesia. Chronic sequelae may include joint contractures, compartment syndrome, necrotic ulcers, and chronic neuropathy.1

Management

Treatment of scorpionfish stings primarily is palliative and aimed at symptom reduction. Patients should immediately treat wounds with hot but not scalding water immersion.26,27 Given the thermolabile components of scorpionfish venom, the most effective treatment is to soak the affected limb in water 42 °C to 45 °C for 30 to 90 minutes. Any higher temperature may pose risk for scalding burns. Children should be monitored throughout treatment.28 If hot water immersion does not provide relief, oral analgesics may be considered. Stonefish antivenom is available and may be used for any scorpionfish sting given the shared biologic properties between genera. Providers evaluating stings could use sterile irrigation to clean wounds and search for foreign bodies including spine fragments; probing should be accomplished by instruments rather than a gloved finger. Providers should consider culturing wounds and prescribing antibiotics for suspected secondary infections. A tetanus toxoid history also should be elicited, and patients may have a booster administered, as indicated.29

With the growing popularity of water sports and a proliferation of invasive species, human injuries from marine animal envenomation continue to rise.1-3 Members of the scorpionfish family Scorpaenidae are second only to stingrays as the leading cause of the 40,000 to 50,000 injuries annually from marine life worldwide.4 Because scorpionfish represent a growing threat and competition with native species, it has been suggested that they could replace endangered species on restaurant menus.5-8 Scorpionfish have been introduced by humans from tropical to temperate seas and are now common off the coast of California and the eastern coast from New York to Florida, as well as in the Caribbean, the Bahamas, and off the southern coast of Brazil. Victims of scorpionfish stings experience considerable pain and may require days to weeks to fully recover, highlighting the socioeconomic costs and burden of scorpionfish envenomation.9,10 Fishers, divers, swimmers, and aquarium owners are most often affected.

Family

The common term scorpionfish refers to both the family Scorpaenidae and the genus Scorpaena. Members of this family possess similar dorsal, anal, and pelvic fins, though they vary between genera in their size and the potency of the venom they insulate. Other familiar members include the genus Pterois (lionfish) and Synanceja (stonefish). Synanceja are the most venomous within the group, but scorpionfish stings more commonly arise from Pterois and Scorpaena.8 Because of the rare shapes and vibrant colors of scorpionfish, some traders and aquarium owners will seek and pay high prices for these fish, providing further opportunity for envenomation.11,12

Characteristics

Scorpionfish have with a high variation in color, ranging from lighter grays to intense reds depending on their geographic location and habitat. Synanceja are bland in coloration, blending in with rocks and gravel, but the more dramatic-appearing Scorpaena exhibit a large cranium and wide range of multicolored patterns (Figure 1).13Pterois serve as the most conspicuous member of the group with brightly colored red and white stripes (Figure 2). Scorpionfish commonly grow up to 19 inches long and boast 12 dorsal, 2 pelvic, and 3 anal spines housing 5 to 10 mg of venom.14 An integumentary sheath encapsulates each spine housing the glandular tissue that produces the potent venom.

Afvari_scorpionfish_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Red%20scorpionfish%20(%3Cem%3EScorpaena%20scrofa%3C%2Fem%3E).%3C%2Fp%3E

Toxin Properties

Unlike Pterois and Synanceja, Scorpaena do not have venom ducts around their glands, complicating the work of marine biologists aiming to extract and study the venomous toxins. Several studies have managed to isolate scorpionfish venom and overcome its unstable heat-labile nature to investigate its biologic properties.15-20 Several high-molecular-weight proteins (50–800 kDa) comprise the venom, including hyaluronidase, integrin-inhibiting factors, capillary permeability factor, proteases, and some less-understood cytolytic toxins. These factors provoke the inflammatory, proteolytic, hemorrhagic, cardiovascular, and hemolytic biologic activities at both the local and systemic levels, directing damage to wounded tissues and inducing vascular and tissue permeability to reach cellular processes far and wide. Mediators of inflammation include tumor necrosis factor, IL-6, and monocyte chemoattractant protein 1, followed by neutrophils and other mononuclear cells, initiating the immune response at the wound site. Toxin potency remains for up to 2 days after fish death.1

Afvari_scorpionfish_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Lionfish%20(%3Cem%3EPterois%20volitans%3C%2Fem%3E).%3C%2Fp%3E

Clinical Manifestation

Physicians may be guided by clinical symptoms in identifying scorpionfish stings, as the patient may not know the identity of their marine assailant. Initially, individuals punctured by scorpionfish spikes will experience an acute pain and burning sensation at the puncture site that may be accompanied by systemic symptoms such as nausea, vomiting, diarrhea, tachycardia, hypotension, loss of consciousness, difficulty breathing, and delirium.9,21-23 The pain will intensify and radiate distal to the site of envenomation, and the wound may exhibit vesiculation, erythema, bruising, pallor, and notable edema.4,24 Pain intensity peaks at 30 to 90 minutes after envenomation, and other systemic symptoms generally last for 24 to 48 hours.25 If patients do not seek prompt treatment, secondary infection may ensue, and the lingering venom in the blister may cause dermal necrosis, paresthesia, and anesthesia. Chronic sequelae may include joint contractures, compartment syndrome, necrotic ulcers, and chronic neuropathy.1

Management

Treatment of scorpionfish stings primarily is palliative and aimed at symptom reduction. Patients should immediately treat wounds with hot but not scalding water immersion.26,27 Given the thermolabile components of scorpionfish venom, the most effective treatment is to soak the affected limb in water 42 °C to 45 °C for 30 to 90 minutes. Any higher temperature may pose risk for scalding burns. Children should be monitored throughout treatment.28 If hot water immersion does not provide relief, oral analgesics may be considered. Stonefish antivenom is available and may be used for any scorpionfish sting given the shared biologic properties between genera. Providers evaluating stings could use sterile irrigation to clean wounds and search for foreign bodies including spine fragments; probing should be accomplished by instruments rather than a gloved finger. Providers should consider culturing wounds and prescribing antibiotics for suspected secondary infections. A tetanus toxoid history also should be elicited, and patients may have a booster administered, as indicated.29

References
  1. Rensch G, Murphy-Lavoie HM. Lionfish, scorpionfish, and stonefish toxicity. StatPearls. StatPearls Publishing; May 10, 2022.
  2. Cearnal L. Red lionfish and ciguatoxin: menace spreading through western hemisphere. Ann Emerg Med. 2012;60:21A-22A. doi:10.1016/j.annemergmed.2012.05.022
  3. Côté IM, Green SJ. Potential effects of climate change on a marine invasion: the importance of current context. Curr Zool. 2012;58:1-8. doi:10.1093/czoolo/58.1.1
  4. Venomology of scorpionfishes. In: Santhanam R. Biology and Ecology of Venomous Marine Scorpionfishes. Academic Press; 2019:263-278.
  5. Ferri J, Staglicˇic´ N, Matić-Skoko S. The black scorpionfish, Scorpaena porcus (Scorpaenidae): could it serve as reliable indicator of Mediterranean coastal communities’ health? Ecol Indicators. 2012;18:25-30. doi:10.1016/j.ecolind.2011.11.004
  6. Santhanam R. Biology and Ecology of Venomous Marine Scorpionfishes. Academic Press; 2019.
  7. Morris JA, Akins JL. Feeding ecology of invasive lionfish (Pterois volitans) in the Bahamian Archipelago. Environ Biol Fishes. 2009;86:389-398. doi:10.1007/s10641-009-9538-8 
  8. Albins MA, Hixon MA. Worst case scenario: potential long-term effects of invasive predatory lionfish (Pterois volitans) on Atlantic and Caribbean coral-reef communities. Environ Biol Fishes. 2013;96:1151–1157. doi:10.1007/s10641-011-9795-1
  9. Haddad V Jr, Martins IA, Makyama HM. Injuries caused by scorpionfishes (Scorpaena plumieri Bloch, 1789 and Scorpaena brasiliensis Cuvier, 1829) in the Southwestern Atlantic Ocean (Brazilian coast): epidemiologic, clinic and therapeutic aspects of 23 stings in humans. Toxicon. 2003;42:79-83. doi:10.1016/s0041-0101(03)00103-x
  10. Campos FV, Menezes TN, Malacarne PF, et al. A review on the Scorpaena plumieri fish venom and its bioactive compounds. J Venom Anim Toxins Incl Trop Dis. 2016;22:35. doi:10.1186/s40409-016-0090-7
  11. Needleman RK, Neylan IP, Erickson TB. Environmental and ecological effects of climate change on venomous marine and amphibious species in the wilderness. Wilderness Environ Med. 2018;29:343-356. doi:10.1016/j.wem.2018.04.003
  12. Aldred B, Erickson T, Lipscomb J. Lionfish envenomations in an urban wilderness. Wilderness Environ Med. 1996;7:291-296. doi:10.1580/1080-6032(1996)007[0291:leiauw]2.3.co;2
  13. Stewart J, Hughes JM. Life-history traits of the southern hemisphere eastern red scorpionfish, Scorpaena cardinalis (Scorpaenidae: Scorpaeninae). Mar Freshw Res. 2010;61:1290-1297. doi:10.1071/MF10040
  14. Auerbach PS. Marine envenomations. N Engl J Med. 1991;325:486-493. doi:10.1056/NEJM199108153250707
  15. Andrich F, Carnielli JB, Cassoli JS, et al. A potent vasoactive cytolysin isolated from Scorpaena plumieri scorpionfish venom. Toxicon. 2010;56:487-496. doi:10.1016/j.toxicon.2010.05.003
  16. Gomes HL, Andrich F, Mauad H, et al. Cardiovascular effects of scorpionfish (Scorpaena plumieri) venom. Toxicon. 2010;55(2-3):580-589. doi:10.1016/j.toxicon.2009.10.012
  17. Menezes TN, Carnielli JB, Gomes HL, et al. Local inflammatory response induced by scorpionfish Scorpaena plumieri venom in mice. Toxicon. 2012;60:4-11. doi:10.1016/j.toxicon.2012.03.008
  18. Schaeffer RC Jr, Carlson RW, Russell FE. Some chemical properties of the venom of the scorpionfish Scorpaena guttata. Toxicon. 1971;9:69-78. doi:10.1016/0041-0101(71)90045-6
  19. Khalil AM, Wahsha MA, Abu Khadra KM, et al. Biochemical and histopathological effects of the stonefish (Synanceia verrucosa) venom in rats. Toxicon. 2018;142:45-51. doi:10.1016/j.toxicon.2017.12.052
  20. Mouchbahani-Constance S, Lesperance LS, Petitjean H, et al. Lionfish venom elicits pain predominantly through the activation of nonpeptidergic nociceptors. Pain. 2018;159:2255-2266. doi:10.1097/j.pain.0000000000001326
  21. Ottuso P. Aquatic dermatology: encounters with the denizens of the deep (and not so deep)—a review. part II: the vertebrates, single-celled organisms, and aquatic biotoxins. Int J Dermatol. 2013;52:268-278. doi:10.1111/j.1365-4632.2011.05426.x
  22. Bayley HH. Injuries caused by scorpion fish. Trans R Soc Trop Med Hyg. 1940;34:227-230. doi:10.1016/s0035-9203(40)90072-4
  23. González D. Epidemiological and clinical aspects of certain venomous animals of Spain. Toxicon. 1982;20:925-928. doi:10.1016/0041-0101(82)90080-0
  24. Halstead BW. Injurious effects from the sting of the scorpionfish, Scorpaena guttata. with report of a case. Calif Med. 1951;74:395-396.
  25. Vasievich MP, Villarreal JD, Tomecki KJ. Got the travel bug? a review of common infections, infestations, bites, and stings among returning travelers. Am J Clin Dermatol. 2016;17:451-462. doi:10.1007/s40257-016-0203-7
  26. Barnett S, Saggiomo S, Smout M, et al. Heat deactivation of the stonefish Synanceia horrida venom—implications for first-aid management. Diving Hyperb Med. 2017;47:155-158. doi:10.28920/dhm47.3.155-158
  27. Russell FE. Weever fish sting: the last word. Br Med J (Clin Res Ed). 1983;287:981-982. doi:10.1136/bmj.287.6397.981-c
  28. Tomlinson H, Elston DM. Aquatic antagonists: lionfish (Pterois volitans). Cutis. 2018;102:232-234.
  29. Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337. doi:10.1016/j.emc.2016.12.004
References
  1. Rensch G, Murphy-Lavoie HM. Lionfish, scorpionfish, and stonefish toxicity. StatPearls. StatPearls Publishing; May 10, 2022.
  2. Cearnal L. Red lionfish and ciguatoxin: menace spreading through western hemisphere. Ann Emerg Med. 2012;60:21A-22A. doi:10.1016/j.annemergmed.2012.05.022
  3. Côté IM, Green SJ. Potential effects of climate change on a marine invasion: the importance of current context. Curr Zool. 2012;58:1-8. doi:10.1093/czoolo/58.1.1
  4. Venomology of scorpionfishes. In: Santhanam R. Biology and Ecology of Venomous Marine Scorpionfishes. Academic Press; 2019:263-278.
  5. Ferri J, Staglicˇic´ N, Matić-Skoko S. The black scorpionfish, Scorpaena porcus (Scorpaenidae): could it serve as reliable indicator of Mediterranean coastal communities’ health? Ecol Indicators. 2012;18:25-30. doi:10.1016/j.ecolind.2011.11.004
  6. Santhanam R. Biology and Ecology of Venomous Marine Scorpionfishes. Academic Press; 2019.
  7. Morris JA, Akins JL. Feeding ecology of invasive lionfish (Pterois volitans) in the Bahamian Archipelago. Environ Biol Fishes. 2009;86:389-398. doi:10.1007/s10641-009-9538-8 
  8. Albins MA, Hixon MA. Worst case scenario: potential long-term effects of invasive predatory lionfish (Pterois volitans) on Atlantic and Caribbean coral-reef communities. Environ Biol Fishes. 2013;96:1151–1157. doi:10.1007/s10641-011-9795-1
  9. Haddad V Jr, Martins IA, Makyama HM. Injuries caused by scorpionfishes (Scorpaena plumieri Bloch, 1789 and Scorpaena brasiliensis Cuvier, 1829) in the Southwestern Atlantic Ocean (Brazilian coast): epidemiologic, clinic and therapeutic aspects of 23 stings in humans. Toxicon. 2003;42:79-83. doi:10.1016/s0041-0101(03)00103-x
  10. Campos FV, Menezes TN, Malacarne PF, et al. A review on the Scorpaena plumieri fish venom and its bioactive compounds. J Venom Anim Toxins Incl Trop Dis. 2016;22:35. doi:10.1186/s40409-016-0090-7
  11. Needleman RK, Neylan IP, Erickson TB. Environmental and ecological effects of climate change on venomous marine and amphibious species in the wilderness. Wilderness Environ Med. 2018;29:343-356. doi:10.1016/j.wem.2018.04.003
  12. Aldred B, Erickson T, Lipscomb J. Lionfish envenomations in an urban wilderness. Wilderness Environ Med. 1996;7:291-296. doi:10.1580/1080-6032(1996)007[0291:leiauw]2.3.co;2
  13. Stewart J, Hughes JM. Life-history traits of the southern hemisphere eastern red scorpionfish, Scorpaena cardinalis (Scorpaenidae: Scorpaeninae). Mar Freshw Res. 2010;61:1290-1297. doi:10.1071/MF10040
  14. Auerbach PS. Marine envenomations. N Engl J Med. 1991;325:486-493. doi:10.1056/NEJM199108153250707
  15. Andrich F, Carnielli JB, Cassoli JS, et al. A potent vasoactive cytolysin isolated from Scorpaena plumieri scorpionfish venom. Toxicon. 2010;56:487-496. doi:10.1016/j.toxicon.2010.05.003
  16. Gomes HL, Andrich F, Mauad H, et al. Cardiovascular effects of scorpionfish (Scorpaena plumieri) venom. Toxicon. 2010;55(2-3):580-589. doi:10.1016/j.toxicon.2009.10.012
  17. Menezes TN, Carnielli JB, Gomes HL, et al. Local inflammatory response induced by scorpionfish Scorpaena plumieri venom in mice. Toxicon. 2012;60:4-11. doi:10.1016/j.toxicon.2012.03.008
  18. Schaeffer RC Jr, Carlson RW, Russell FE. Some chemical properties of the venom of the scorpionfish Scorpaena guttata. Toxicon. 1971;9:69-78. doi:10.1016/0041-0101(71)90045-6
  19. Khalil AM, Wahsha MA, Abu Khadra KM, et al. Biochemical and histopathological effects of the stonefish (Synanceia verrucosa) venom in rats. Toxicon. 2018;142:45-51. doi:10.1016/j.toxicon.2017.12.052
  20. Mouchbahani-Constance S, Lesperance LS, Petitjean H, et al. Lionfish venom elicits pain predominantly through the activation of nonpeptidergic nociceptors. Pain. 2018;159:2255-2266. doi:10.1097/j.pain.0000000000001326
  21. Ottuso P. Aquatic dermatology: encounters with the denizens of the deep (and not so deep)—a review. part II: the vertebrates, single-celled organisms, and aquatic biotoxins. Int J Dermatol. 2013;52:268-278. doi:10.1111/j.1365-4632.2011.05426.x
  22. Bayley HH. Injuries caused by scorpion fish. Trans R Soc Trop Med Hyg. 1940;34:227-230. doi:10.1016/s0035-9203(40)90072-4
  23. González D. Epidemiological and clinical aspects of certain venomous animals of Spain. Toxicon. 1982;20:925-928. doi:10.1016/0041-0101(82)90080-0
  24. Halstead BW. Injurious effects from the sting of the scorpionfish, Scorpaena guttata. with report of a case. Calif Med. 1951;74:395-396.
  25. Vasievich MP, Villarreal JD, Tomecki KJ. Got the travel bug? a review of common infections, infestations, bites, and stings among returning travelers. Am J Clin Dermatol. 2016;17:451-462. doi:10.1007/s40257-016-0203-7
  26. Barnett S, Saggiomo S, Smout M, et al. Heat deactivation of the stonefish Synanceia horrida venom—implications for first-aid management. Diving Hyperb Med. 2017;47:155-158. doi:10.28920/dhm47.3.155-158
  27. Russell FE. Weever fish sting: the last word. Br Med J (Clin Res Ed). 1983;287:981-982. doi:10.1136/bmj.287.6397.981-c
  28. Tomlinson H, Elston DM. Aquatic antagonists: lionfish (Pterois volitans). Cutis. 2018;102:232-234.
  29. Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337. doi:10.1016/j.emc.2016.12.004
Issue
Cutis - 113(3)
Issue
Cutis - 113(3)
Page Number
133-134,136
Page Number
133-134,136
Publications
Publications
Topics
Article Type
Display Headline
Aquatic Antagonists: Scorpionfish Envenomation
Display Headline
Aquatic Antagonists: Scorpionfish Envenomation
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Afvari scorpionfish</fileName> <TBEID>0C02F309.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F309</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Afvari scorpionfish</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240305T083640</firstPublished> <LastPublished>20240305T083640</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240305T083639</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Shawn Afvari, BS; Dirk M. Elston, MD</byline> <bylineText>Shawn Afvari, BS; Dirk M. Elston, MD</bylineText> <bylineFull>Shawn Afvari, BS; Dirk M. Elston, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>133-134,136</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>With the growing popularity of water sports and a proliferation of invasive species, human injuries from marine animal envenomation continue to rise.1-3 Members</metaDescription> <articlePDF>300457</articlePDF> <teaserImage/> <title>Aquatic Antagonists: Scorpionfish Envenomation</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>March</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>3</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2159</CMSID> </CMSIDs> <keywords> <keyword>wounds</keyword> <keyword> scorpionfish envenomation</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>March 2024</pubIssueName> <pubArticleType>Departments | 2159</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">60</term> </sections> <topics> <term canonical="true">313</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180026e1.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Aquatic Antagonists: Scorpionfish Envenomation</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Scorpionfish are among the most venomous creatures found in American and Caribbean seas. Their envenomation is responsible for considerable morbidity and socioeconomic burden associated with marine animal injuries. Avoiding physical contact with scorpionfish through proper identification prevails as the chief prevention method for stings. This article discusses common features of scorpionfish as well as the clinical presentation and treatment options following exposure to its toxins.</p> <p> <em><em>Cutis.</em> 2024;113:133-134, 136.</em> </p> <p>With the growing popularity of water sports and a proliferation of invasive species, human injuries from marine animal envenomation continue to rise.<sup>1-3</sup> Members of the scorpionfish family Scorpaenidae are second only to stingrays as the leading cause of the 40,000 to 50,000 injuries annually from marine life worldwide.<sup>4</sup> Because scorpionfish represent a growing threat and competition with native species, it has been suggested that they could replace endangered species on restaurant menus.<sup>5-8</sup> Scorpionfish have been introduced by humans from tropical to temperate seas and are now common off the coast of California and the eastern coast from New York to Florida, as well as in the Caribbean, the Bahamas, and off the southern coast of Brazil. Victims of scorpionfish stings experience considerable pain and may require days to weeks to fully recover, highlighting the socioeconomic costs and burden of scorpionfish envenomation.<sup>9,10</sup> Fishers, divers, swimmers, and aquarium owners are most often affected. </p> <h3>Family</h3> <p>The common term <i>scorpionfish</i> refers to both the family Scorpaenidae and the genus <i>Scorpaena</i>. Members of this family possess similar dorsal, anal, and pelvic fins, though they vary between genera in their size and the potency of the venom they insulate. Other familiar members include the genus <i>Pterois</i> (lionfish) and <i>Synanceja</i> (stonefish). <i>Synanceja</i> are the most venomous within the group, but scorpionfish stings more commonly arise from <i>Pterois </i>and <i>Scorpaena.</i><sup>8</sup><i> </i>Because of the rare shapes and vibrant colors of scorpionfish, some traders and aquarium owners will seek and pay high prices for these fish, providing further opportunity for envenomation.<sup>11,12</sup> </p> <h3>Characteristics</h3> <p>Scorpionfish have with a high variation in color, ranging from lighter grays to intense reds depending on their geographic location and habitat. <i>Synanceja</i> are bland in coloration, blending in with rocks and gravel, but the more dramatic-appearing <i>Scorpaena </i>exhibit a large cranium and wide range of multicolored patterns (Figure 1).<sup>13</sup> <i>Pterois</i> serve as the most conspicuous member of the group with brightly colored red and white stripes (Figure 2). Scorpionfish commonly grow up to 19 inches long and boast 12 dorsal, 2 pelvic, and 3 anal spines housing 5 to 10 mg of venom.<sup>14</sup> An integumentary sheath encapsulates each spine housing the glandular tissue that produces the potent venom. </p> <h3>Toxin Properties </h3> <p>Unlike <i>Pterois </i>and<i> Synanceja, Scorpaena </i>do not have venom ducts around their glands, complicating the work of marine biologists aiming to extract and study the venomous toxins. Several studies have managed to isolate scorpionfish venom and overcome its unstable heat-labile nature to investigate its biologic properties.<sup>15-20</sup> Several high-molecular-weight proteins (50–800 kDa) comprise the venom, including hyaluronidase, integrin-inhibiting factors, capillary permeability factor, proteases, and some less-understood cytolytic toxins. These factors provoke the inflammatory, proteolytic, hemorrhagic, cardiovascular, and hemolytic biologic activities at both the local and systemic levels, directing damage to wounded tissues and inducing vascular and tissue permeability to reach cellular processes far and wide. Mediators of inflammation include tumor necrosis factor, IL-6, and monocyte chemoattractant protein 1, followed by neutrophils and other mononuclear cells, initiating the immune response at the wound site. Toxin potency remains for up to 2 days after fish death.<sup>1</sup> </p> <h3>Clinical Manifestation</h3> <p>Physicians may be guided by clinical symptoms in identifying scorpionfish stings, as the patient may not know the identity of their marine assailant. Initially, individuals punctured by scorpionfish spikes will experience an acute pain and burning sensation at the puncture site that may be accompanied by systemic symptoms such as nausea, vomiting, diarrhea, tachycardia, hypotension, loss of consciousness, difficulty breathing, and delirium.<sup>9,21-23</sup> The pain will intensify and radiate distal to the site of envenomation, and the wound may exhibit vesiculation, erythema, bruising, pallor, and notable edema.<sup>4,24</sup> Pain intensity peaks at 30 to 90 minutes after envenomation, and other systemic symptoms generally last for 24 to 48 hours.<sup>25</sup> If patients do not seek prompt treatment, secondary infection may ensue, and the lingering venom in the blister may cause dermal necrosis, paresthesia, and anesthesia. Chronic sequelae may include joint contractures, compartment syndrome, necrotic ulcers, and chronic neuropathy.<sup>1</sup></p> <h3>Management </h3> <p>Treatment of scorpionfish stings primarily is palliative and aimed at symptom reduction. Patients should immediately treat wounds with hot but not scalding water immersion.<sup>26,27</sup> Given the thermolabile components of scorpionfish venom, the most effective treatment is to soak the affected limb in water 42 <span class="body">°</span>C to 45 <span class="body">°</span>C for 30 to 90 minutes. Any higher temperature may pose risk for scalding burns. Children should be monitored throughout treatment.<sup>28</sup> If hot water immersion does not provide relief, oral analgesics may be considered. Stonefish antivenom is available and may be used for any scorpionfish sting given the shared biologic properties between genera. Providers evaluating stings could use sterile irrigation to clean wounds and search for foreign bodies including spine fragments; probing should be accomplished by instruments rather than a gloved finger. Providers should consider culturing wounds and prescribing antibiotics for suspected secondary infections. A tetanus toxoid history also should be elicited, and patients may have a booster administered, as indicated.<sup>29</sup></p> <h2>References</h2> <p class="reference"> 1. Rensch G, Murphy-Lavoie HM. Lionfish, scorpionfish, and stonefish toxicity. <i>StatPearls</i>. StatPearls Publishing; May 10, 2022.<br/><br/> 2. Cearnal L. Red lionfish and ciguatoxin: menace spreading through western hemisphere. <i>Ann Emerg Med</i>. 2012;60:21A-22A. doi:10.1016/j.annemergmed.2012.05.022<br/><br/> 3. Côté IM, Green SJ. Potential effects of climate change on a marine invasion: the importance of current context. <i>Curr Zool.</i> 2012;58:1-8. doi:10.1093/czoolo/58.1.1<br/><br/> 4. Venomology of scorpionfishes. In: Santhanam R. <i>Biology and Ecology of Venomous Marine Scorpionfishes</i>. Academic Press; 2019:263-278.<br/><br/> 5. Ferri J, Staglicˇic´ N, Matić-Skoko S. The black scorpionfish, <i>Scorpaena porcus</i> (Scorpaenidae): could it serve as reliable indicator of Mediterranean coastal communities’ health? <i>Ecol Indicators</i>. 2012;18:25-30. doi:10.1016/j.ecolind.2011.11.004 <br/><br/> 6. Santhanam R.<i> Biology and Ecology of Venomous Marine Scorpionfishes</i>. Academic Press; 2019.<br/><br/> 7. Morris JA, Akins JL. Feeding ecology of invasive lionfish (<i>Pterois volitans</i>) in the Bahamian Archipelago. <i>Environ Biol Fishes</i>. 2009;86:389-398. doi:10.1007/s10641-009-9538-8 <br/><br/> <br/><br/> 8. Albins MA, Hixon MA. Worst case scenario: potential long-term effects of invasive predatory lionfish (<i>Pterois volitans</i>) on Atlantic and Caribbean coral-reef communities. <i>Environ Biol Fishes. </i>2013;96:1151–1157. doi:10.1007/s10641-011-9795-1</p> <p class="reference"> 9. Haddad V Jr, Martins IA, Makyama HM. Injuries caused by scorpionfishes (<i>Scorpaena plumieri</i> Bloch, 1789 and <i>Scorpaena brasiliensis</i> Cuvier, 1829) in the Southwestern Atlantic Ocean (Brazilian coast): epidemiologic, clinic and therapeutic aspects of 23 stings in humans. <i>Toxicon</i>. 2003;42:79-83. doi:10.1016/s0041-0101(03)00103-x<br/><br/>10. Campos FV, Menezes TN, Malacarne PF, et al. A review on the <i>Scorpaena plumieri</i> fish venom and its bioactive compounds. <i>J Venom Anim Toxins Incl Trop Dis</i>. 2016;22:35. doi:10.1186/s40409-016-0090-7<br/><br/>11. Needleman RK, Neylan IP, Erickson TB. Environmental and ecological effects of climate change on venomous marine and amphibious species in the wilderness. <i>Wilderness Environ Med</i>. 2018;29:343-356. doi:10.1016/j.wem.2018.04.003<br/><br/>12. Aldred B, Erickson T, Lipscomb J. Lionfish envenomations in an urban wilderness. <i>Wilderness Environ Med</i>. 1996;7:291-296. doi:10.1580/1080-6032(1996)007[0291:leiauw]2.3.co;2<br/><br/>13. Stewart J, Hughes JM. Life-history traits of the southern hemisphere eastern red scorpionfish, <i>Scorpaena cardinalis</i> (Scorpaenidae: Scorpaeninae). <i>Mar Freshw Res. </i>2010;61:1290-1297. doi:10.1071/MF10040<br/><br/>14. Auerbach PS. Marine envenomations. <i>N Engl J Med</i>. 1991;325:486-493. doi:10.1056/NEJM199108153250707<br/><br/>15. Andrich F, Carnielli JB, Cassoli JS, et al. A potent vasoactive cytolysin isolated from <i>Scorpaena plumieri</i> scorpionfish venom. <i>Toxicon</i>. 2010;56:487-496. doi:10.1016/j.toxicon.2010.05.003<br/><br/>16. Gomes HL, Andrich F, Mauad H, et al. Cardiovascular effects of scorpionfish (<i>Scorpaena plumieri</i>) venom. <i>Toxicon</i>. 2010;55(2-3):580-589. doi:10.1016/j.toxicon.2009.10.012<br/><br/>17. Menezes TN, Carnielli JB, Gomes HL, et al. Local inflammatory response induced by scorpionfish <i>Scorpaena plumieri</i> venom in mice. <i>Toxicon</i>. 2012;60:4-11. doi:10.1016/j.toxicon.2012.03.008<br/><br/>18. Schaeffer RC Jr, Carlson RW, Russell FE. Some chemical properties of the venom of the scorpionfish <i>Scorpaena guttata</i>. <i>Toxicon</i>. 1971;9:69-78. doi:10.1016/0041-0101(71)90045-6<br/><br/>19. Khalil AM, Wahsha MA, Abu Khadra KM, et al. Biochemical and histopathological effects of the stonefish (<i>Synanceia verrucosa</i>) venom in rats. <i>Toxicon</i>. 2018;142:45-51. doi:10.1016/j.toxicon.2017.12.052<br/><br/>20. Mouchbahani-Constance S, Lesperance LS, Petitjean H, et al. Lionfish venom elicits pain predominantly through the activation of nonpeptidergic nociceptors. <i>Pain</i>. 2018;159:2255-2266. doi:10.1097/j.pain.0000000000001326<br/><br/>21. Ottuso P. Aquatic dermatology: encounters with the denizens of the deep (and not so deep)—a review. part II: the vertebrates, single-celled organisms, and aquatic biotoxins. <i>Int J Dermatol</i>. 2013;52:268-278. doi:10.1111/j.1365-4632.2011.05426.x<br/><br/>22. Bayley HH. Injuries caused by scorpion fish. <i>Trans R Soc Trop Med Hyg.</i> 1940;34:227-230. doi:10.1016/s0035-9203(40)90072-4 <br/><br/>23. González D. Epidemiological and clinical aspects of certain venomous animals of Spain. <i>Toxicon</i>. 1982;20:925-928. doi:10.1016/0041-0101(82)90080-0<br/><br/>24. Halstead BW. Injurious effects from the sting of the scorpionfish, <i>Scorpaena guttata</i>. with report of a case. <i>Calif Med</i>. 1951;74:395-396.<br/><br/>25. Vasievich MP, Villarreal JD, Tomecki KJ. Got the travel bug? a review of common infections, infestations, bites, and stings among returning travelers. <i>Am J Clin Dermatol</i>. 2016;17:451-462. doi:10.1007/s40257-016-0203-7<br/><br/>26. Barnett S, Saggiomo S, Smout M, et al. Heat deactivation of the stonefish <i>Synanceia horrida</i> venom—implications for first-aid management. <i>Diving Hyperb Med</i>. 2017;47:155-158. doi:10.28920/dhm47.3.155-158<br/><br/>27. Russell FE. Weever fish sting: the last word. <i>Br Med J (Clin Res Ed)</i>. 1983;287:981-982. doi:10.1136/bmj.287.6397.981-c<br/><br/>28. Tomlinson H, Elston DM. Aquatic antagonists: lionfish (<i>Pterois volitans</i>). <i>Cutis</i>. 2018;102:232-234.<br/><br/>29. Hornbeak KB, Auerbach PS. Marine envenomation. <i>Emerg Med Clin North Am</i>. 2017;35:321-337. doi:10.1016/j.emc.2016.12.004 </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Shawn Afvari is from the New York Medical College School of Medicine, Valhalla. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.</p> <p class="disclosure">The authors report no conflict of interest.<br/><br/>Correspondence: Shawn Afvari, BS (safvari@student.nymc.edu).doi:10.12788/cutis.0973</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>As some species of scorpionfish proliferate, providers may see an increase in envenomation cases. </li> <li>Physicians should suspect scorpionfish stings based on clinical symptoms and physical examination. </li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • As some species of scorpionfish proliferate, providers may see an increase in envenomation cases.
  • Physicians should suspect scorpionfish stings based on clinical symptoms and physical examination.
  • Scorpionfish toxins are thermolabile, and patients can find symptom relief by immediately immersing the affected area in hot water (42 °C–45 °C) for 30 to 90 minutes.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
180026E1.SIG
Disable zoom
Off

What’s Eating You? Rhipicephalus Ticks Revisited

Article Type
Changed
Wed, 01/31/2024 - 16:13
Display Headline
What’s Eating You? Rhipicephalus Ticks Revisited

Characteristics

Rhipicephalus ticks belong to the Ixodidae family of hard-bodied ticks. They are large and teardrop shaped with an inornate scutum (hard dorsal plate) and relatively short mouthparts attached at a hexagonal basis capitulum (base of the head to which mouthparts are attached)(Figure).1 Widely spaced eyes and festoons also are present. The first pair of coxae—attachment base for the first pair of legs—are characteristically bifid; males have a pair of sclerotized adanal plates on the ventral surface adjacent to the anus as well as accessory adanal shields.2Rhipicephalus (formerly Boophilus) microplus (the so-called cattle tick) is a newly added species; it lacks posterior festoons, and the anal groove is absent.3

Brantley_Figure.jpg
%3Cp%3E%3Cem%3ERhipicephalus%3C%2Fem%3E%20ticks%20are%20brown%20and%20teardrop%20shaped%20with%20an%20inornate%20scutum.%20The%20hexagonal%20basis%20capitulum%20is%20a%20defining%20characteristic.%20The%20image%20is%20in%20the%20public%20domain.%3C%2Fp%3E

Almost all Rhipicephalus ticks, except for R microplus, are 3-host ticks in which a single blood meal is consumed from a vertebrate host at each active life stage—larva, nymph, and adult—to complete development.4,5 In contrast to most ixodid ticks, which are exophilic (living outside of human habitation), the Rhipicephalus sanguineus sensu lato species (the brown dog tick) is highly endophilic (adapted to indoor living) and often can be found hidden in cracks and crevices of walls in homes and peridomestic structures.6 It is predominately monotropic (all developmental stages feed on the same host species) and has a strong host preference for dogs, though it occasionally feeds on other hosts (eg, humans).7 Although most common in tropical and subtropical climates, they can be found anywhere there are dogs due to their ability to colonize indoor dwellings.8 In contrast, R microplus ticks have a predilection for cattle and livestock rather than humans, posing a notable concern to livestock worldwide. Infestation results in transmission of disease-causing pathogens, such as Babesia and Anaplasma species, which costs the cattle industry billions of dollars annually.9

Clinical Manifestations and Treatment

Tick bites usually manifest as intensely pruritic, erythematous papules at the site of tick attachment due to a local type IV hypersensitivity reaction to antigens in the tick’s saliva. This reaction can be long-lasting. In addition to pruritic papules following a bite, an attached tick can be mistaken for a skin neoplasm or nevus. Given that ticks are small, especially during the larval stage, dermoscopy may be helpful in making a diagnosis.10 Symptomatic relief usually can be achieved with topical antipruritics or oral antihistamines.

Of public health concern, brown dog ticks are important vectors of Rickettsia rickettsii (the causative organism of Rocky Mountain spotted fever [RMSF]) in the Western hemisphere, and Rickettsia conorii (the causative organism of Mediterranean spotted fever [MSF][also known as Boutonneuse fever]) in the Eastern hemisphere.11 Bites by ticks carrying rickettsial disease classically manifest with early symptoms of fever, headache, and myalgia, followed by a rash or by a localized eschar or tache noire (a black, necrotic, scabbed lesion) that represents direct endothelial invasion and vascular damage by Rickettsia.12 Rocky Mountain spotted fever and MSF are more prevalent during summer, likely due, in part, to the combination of increased outdoor activity and a higher rate of tick-questing (host-seeking) behavior in warmer climates.4,7

Rocky Mountain Spotted FeverDermacentor variabilis is the primary vector of RMSF in the southeastern United States; Dermacentor andersoni is the major vector of RMSF in Rocky Mountain states. Rhipicephalus sanguineus sensu lato is an important vector of RMSF in the southwestern United States, Mexico, and Central America.11,13

Early symptoms of RMSF are nonspecific and can include fever, headache, arthralgia, myalgia, and malaise. Gastrointestinal tract symptoms (eg, nausea, vomiting, anorexia) may occur; notable abdominal pain occurs in some patients, particularly children. A characteristic petechial rash occurs in as many as 90% of patients, typically at the third to fifth day of illness, and classically begins on the wrists and ankles, with progression to the palms and soles before spreading centripetally to the arms, legs, and trunk.14 An eschar at the inoculation site is uncommon in RMSF; when present, it is more suggestive of MSF.15

The classic triad of fever, headache, and rash is present in 3% of patients during the first 3 days after a tick bite and in 60% to 70% within 2 weeks.16 A rash often is absent when patients first seek medical attention and may not develop (absent in 9% to 12% of cases; so-called spotless RMSF). Therefore, absence of rash should not be a reason to withhold treatment.16 Empiric treatment with doxycycline should be started promptly for all suspected cases of RMSF because of the rapid progression of disease and an increased risk for morbidity and mortality with delayed diagnosis.

 

 

Patients do not become antibody positive until 7 to 10 days after symptoms begin; therefore, treatment should not be delayed while awaiting serologic test results. The case fatality rate in the United States is estimated to be 5% to 10% overall and as high as 40% to 50% among patients who are not treated until day 8 or 9 of illness.17

Cutaneous complications include skin necrosis and gangrene due to continuous tissue damage in severe cases.16 Severe infection also may manifest with signs of multiorgan system damage, including altered mental status, cerebral edema, meningismus, transient deafness, myocarditis, pulmonary hemorrhage and edema, conjunctivitis, retinal abnormalities, and acute renal failure.14,16 Risk factors for more severe illness include delayed treatment, age 40 years or older or younger than 10 years, and underlying medical conditions such as alcoholic liver disease and glucose-6-phosphate dehydrogenase deficiency. However, even some healthy young patients die of this disease.17

Mediterranean Spotted FeverRhipicephalus sanguineus sensu lato is the primary vector of MSF, which is prevalent in areas adjacent to the Mediterranean Sea, including southern Europe, Africa, and Central Asia; Sicily is the most highly affected region.18 Findings with MSF are nearly identical to those of RMSF, except that tache noire is more common, present in as many as 70% of cases at the site of the inoculating tick bite, and MSF typically follows a less severe clinical course.12 Similar to other rickettsial diseases, the pathogenesis of MSF involves direct injury to vascular endothelial cells, causing a vasculitis that is responsible for the clinical abnormalities observed.

Patients with severe MSF experience complications similar to severe RMSF, including neurologic manifestations and multiorgan damage.18 Risk factors include advanced age, immunocompromised state, cardiac disease, chronic alcoholism, diabetes mellitus, glucose-6-phosphate dehydrogenase deficiency, respiratory insufficiency, and delayed treatment.18

Treatment—For all spotted fever group rickettsial infections, doxycycline is the treatment of choice for all patients, including children and pregnant women. Treatment should be started without delay; recommended dosages are 100 mg twice daily for children weighing more than 45 kg and adults, and 2.2 mg/kg twice daily for children weighing 45 kg or less.12

Rhipicephalus tick bites rarely can result in paralysis; however, Dermacentor ticks are responsible for most cases of tick-related paralysis in North America. Other pathogens proven or reputed to be transmitted by Rhipicephalus sanguineus sensu lato with zoonotic potential include but are not limited to Rickettsia massiliae, Coxiella burnetti, Anaplasma platys, Leishmania infantum, and Crimean-Congo hemorrhagic fever virus (Nairovirus).19

Environmental Treatment and Prevention

The most effective way to prevent tick-borne illness is avoidance of tick bites. Primary prevention methods include vector control, use of repellents (eg, N,N-diethyl-meta-toluamide [DEET]), picaridin, permethrin), avoidance of areas with a high tick burden, use of protective clothing, and detection and removal of ticks as soon as possible.

 

 

Environmental and veterinary controls also are important methods of tick-bite prevention. A veterinarian can recommend a variety of agents for dogs and cats that prevent attachment of ticks. Environmental controls include synthetic or natural product-based chemical acaricides and nonchemical methods, such as landscape management (eg, sealing cracks and crevices in homes and controlling tall grasses, weeds, and leaf debris) to minimize potential tick habitat.20 Secondary prevention includes antibiotics for prophylaxis or for treatment of tick-borne disease, when indicated.

Numerous tick repellents are available commercially; others are being studied. DEET, the most widely used topical repellent, has a broad spectrum of activity against many tick species.21 In addition, DEET has a well-known safety and toxicity profile, with rare adverse effects, and is safe for use in pregnant women and children older than 2 years. Alternative repellents, such as those containing picaridin, ethyl butylacetylaminopropionate (IR3535 [Merck]), oil of lemon eucalyptus, and 2-undecanone can be effective; some show efficacy comparable to that of DEET.22 Permethrin, a synthetic pyrethroid, is a highly efficacious tick repellent and insecticide, especially when used in conjunction with a topical repellent such as DEET. Unlike topically applied repellents, permethrin spray is applied to fabric (eg, clothing, shoes, bed nets, camping gear), not to skin.

Indiscriminate use of acaricides worldwide has led to increasing selection of acaricide resistance in Rhipicephalus tick species, which is especially true with the use of acaricides in controlling R microplus livestock infestations; several tick populations now show resistance to all major classes of these compounds.23-25 For that reason, there has been an increasing effort to develop new chemical and nonchemical approaches to tick control that are more environmentally sustainable and strategies to minimize development and progression of resistance such as rotation of acaricides; reducing the frequency of their application; use of pesticide mixtures, synergists, or both; and increasing use of nonacaricidal methods of control.26

Prompt removal of ticks is important for preventing the transmission of tick-borne disease. Proper removal involves rubbing the tick in a circular motion with a moist gauze pad or using fine-tipped tweezers to grasp the tick as close to the skin surface as possible and pulling upward with a steady pressure.17,27 It is important not to jerk, twist, squeeze, smash, or burn the tick, as this can result in insufficient removal of mouthparts or spread contaminated tick fluids to mucous membranes, increasing the risk for infection. Application of petroleum jelly or nail polish to aid in tick removal have not been shown to be effective and are not recommended.16,28

References
  1. Dantas-Torres F. The brown dog tick, Rhipicephalus sanguineus (Latreille, 1806) (Acari: Ixodidae): from taxonomy to control. Vet Parasitol. 2008;152:173-185. doi:10.1016/j.vetpar.2007.12.030
  2. Madder M, Fourie JJ, Schetters TPM. Arachnida, Metastigmata, Ixodidae (except Ixodes holocyclus). In: Marchiondo AA, Cruthers LR, Fourie JJ, eds. Parasiticide Screening: In Vitro and In Vivo Tests With Relevant Parasite Rearing and Host Infection/Infestation Methods. Volume 1. Elsevier Academic Press; 2019:19-20.
  3. Burger TD, Shao R, Barker SC. Phylogenetic analysis of mitochondrial genome sequences indicates that the cattle tick, Rhipicephalus (Boophilus) microplus, contains a cryptic species. Mol Phylogenet Evol. 2014;76:241-253. doi:10.1016/j.ympev.2014.03.017
  4. Gray J, Dantas-Torres F, Estrada-Peña A, et al. Systematics and ecology of the brown dog tick, Rhipicephalus sanguineus. Ticks Tick Borne Dis. 2013;4:171-180. doi:10.1016/j.ttbdis.2012.12.003
  5. Tian Y, Lord CC, Kaufman PE. Brown dog tick, Rhipicephalus Sanguineus Latrielle (Arachnida: Acari: Ixodidae): EENY-221/IN378. EDIS. March 26, 2020. Accessed January 3, 2024. https://doi.org/10.32473/edis-in378-2020
  6. Saleh MN, Allen KE, Lineberry MW, et al. Ticks infesting dogs and cats in North America: biology, geographic distribution, and pathogen transmission. Vet Parasitol. 2021;294:109392. doi:10.1016/j.vetpar.2021.109392
  7. Dantas-Torres F. Biology and ecology of the brown dog tick, Rhipicephalus sanguineus. Parasit Vectors. 2010;3:26. doi:10.1186/1756-3305-3-26
  8. Dryden MW, Payne PA. Biology and control of ticks infesting dogs and cats in North America. Vet Ther. 2004;5:139-154.
  9. Nyangiwe N, Yawa M, Muchenje V. Driving forces for changes in geographic range of cattle ticks (Acari: Ixodidae) in Africa: a Review. S Afr J Anim Sci. 2018;48:829. doi:10.4314/sajas.v48i5.4
  10. Ramot Y, Zlotogorski A, Mumcuoglu KY. Brown dog tick (Rhipicephalus sanguineus) infestation of the penis detected by dermoscopy. Int J Dermatol. 2012;51:1402-1403. doi:10.1111/j.1365-4632.2010.04756.x
  11. Tucker NSG, Weeks ENI, Beati L, et al. Prevalence and distribution of pathogen infection and permethrin resistance in tropical and temperate populations of Rhipicephalus sanguineus s.l. collected worldwide. Med Vet Entomol. 2021;35:147-157. doi:10.1111/mve.12479
  12. McClain MT, Sexton DJ, Hall KK, eds. Other spotted fever group rickettsial infections. UpToDate. Updated October 10, 2022. Accessed January 3, 2024. https://www.uptodate.com/contents/other-spotted-fever-group-rickettsial-infections
  13. Ribeiro CM, Carvalho JLB, Bastos PAS, et al. Prevalence of Rickettsia rickettsii in ticks: systematic review and meta-analysis. Vector Borne Zoonotic Dis. 2021;21:557-565. doi:10.1089/vbz.2021.0004
  14. Pace EJ, O’Reilly M. Tickborne diseases: diagnosis and management. Am Fam Physician. 2020;101:530-540.
  15. Patterson JW. Weedon’s Skin Pathology. 5th ed. Elsevier; 2020.
  16. Dantas-Torres F. Rocky Mountain spotted fever. Lancet Infect Dis. 2007;7:724-732. doi:10.1016/S1473-3099(07)70261-X
  17. Biggs HM, Behravesh CB, Bradley KK, et al. Diagnosis and management of tickborne rickettsial diseases: Rocky Mountain spotted fever and other spotted fever group rickettsioses, ehrlichioses, and anaplasmosis—United States. MMWR Recomm Rep. 2016;65:1-44. doi:10.15585/mmwr.rr6502a1
  18. Rossio R, Conalbi V, Castagna V, et al. Mediterranean spotted fever and hearing impairment: a rare complication. Int J Infect Dis. 2015;35:34-36. doi:10.1016/j.ijid.2015.04.005
  19. Dantas-Torres F, Otranto D. Further thoughts on the taxonomy and vector role of Rhipicephalus sanguineus group ticks. Vet Parasitol. 2015;208:9-13. doi:10.1016/j.vetpar.2014.12.014
  20. Eisen RJ, Kugeler KJ, Eisen L, et al. Tick-borne zoonoses in the United States: persistent and emerging threats to human health. ILAR J. 2017;58:319-335. doi:10.1093/ilar/ilx005
  21. Nguyen QD, Vu MN, Hebert AA. Insect repellents: an updated review for the clinician. J Am Acad Dermatol. 2018;88:123-130. doi:10.1016/j.jaad.2018.10.053
  22. Pages F, Dautel H, Duvallet G, et al. Tick repellents for human use: prevention of tick bites and tick-borne diseases. Vector Borne Zoonotic Dis. 2014;14:85-93. doi:10.1089/vbz.2013.1410
  23. Rodriguez-Vivas RI, Alonso-Díaz MA, et al. Prevalence and potential risk factors for organophosphate and pyrethroid resistance in Boophilus microplus ticks on cattle ranches from the State of Yucatan, Mexico. Vet Parasitol. 2006;136:335-342. doi:10.1016/j.vetpar.2005.05.069
  24. Rodríguez-Vivas RI, Rodríguez-Arevalo F, Alonso-Díaz MA, et al. Prevalence and potential risk factors for amitraz resistance in Boophilus microplus ticks in cattle farms in the State of Yucatan, Mexico. Prev Vet Med. 2006;75:280-286. doi:10.1016/j.prevetmed.2006.04.001
  25. Perez-Cogollo LC, Rodriguez-Vivas RI, Ramirez-Cruz GT, et al. First report of the cattle tick Rhipicephalus microplus resistant to ivermectin in Mexico. Vet Parasitol. 2010;168:165-169. doi:10.1016/j.vetpar.2009.10.021
  26. Rodriguez-Vivas RI, Jonsson NN, Bhushan C. Strategies for the control of Rhipicephalus microplus ticks in a world of conventional acaricide and macrocyclic lactone resistance. Parasitol Res.2018;117:3-29. doi:10.1007/s00436-017-5677-6
  27. Centers for Disease Control and Prevention. Tick removal. Updated May 13, 2022. Accessed January 3, 2024. https://www.cdc.gov/ticks/removing_a_tick.html
  28. Diaz JH. Chemical and plant-based insect repellents: efficacy, safety, and toxicity. Wilderness Environ Med. 2016;27:153-163. doi:10.1016/j.wem.2015.11.007
Article PDF
Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Rebecca A. Brantley is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Dirk M. Elston, MD (elstond@musc.edu).

Issue
Cutis - 113(1)
Publications
Topics
Page Number
E44-E47
Sections
Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Rebecca A. Brantley is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Dirk M. Elston, MD (elstond@musc.edu).

Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Rebecca A. Brantley is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Dirk M. Elston, MD (elstond@musc.edu).

Article PDF
Article PDF

Characteristics

Rhipicephalus ticks belong to the Ixodidae family of hard-bodied ticks. They are large and teardrop shaped with an inornate scutum (hard dorsal plate) and relatively short mouthparts attached at a hexagonal basis capitulum (base of the head to which mouthparts are attached)(Figure).1 Widely spaced eyes and festoons also are present. The first pair of coxae—attachment base for the first pair of legs—are characteristically bifid; males have a pair of sclerotized adanal plates on the ventral surface adjacent to the anus as well as accessory adanal shields.2Rhipicephalus (formerly Boophilus) microplus (the so-called cattle tick) is a newly added species; it lacks posterior festoons, and the anal groove is absent.3

Brantley_Figure.jpg
%3Cp%3E%3Cem%3ERhipicephalus%3C%2Fem%3E%20ticks%20are%20brown%20and%20teardrop%20shaped%20with%20an%20inornate%20scutum.%20The%20hexagonal%20basis%20capitulum%20is%20a%20defining%20characteristic.%20The%20image%20is%20in%20the%20public%20domain.%3C%2Fp%3E

Almost all Rhipicephalus ticks, except for R microplus, are 3-host ticks in which a single blood meal is consumed from a vertebrate host at each active life stage—larva, nymph, and adult—to complete development.4,5 In contrast to most ixodid ticks, which are exophilic (living outside of human habitation), the Rhipicephalus sanguineus sensu lato species (the brown dog tick) is highly endophilic (adapted to indoor living) and often can be found hidden in cracks and crevices of walls in homes and peridomestic structures.6 It is predominately monotropic (all developmental stages feed on the same host species) and has a strong host preference for dogs, though it occasionally feeds on other hosts (eg, humans).7 Although most common in tropical and subtropical climates, they can be found anywhere there are dogs due to their ability to colonize indoor dwellings.8 In contrast, R microplus ticks have a predilection for cattle and livestock rather than humans, posing a notable concern to livestock worldwide. Infestation results in transmission of disease-causing pathogens, such as Babesia and Anaplasma species, which costs the cattle industry billions of dollars annually.9

Clinical Manifestations and Treatment

Tick bites usually manifest as intensely pruritic, erythematous papules at the site of tick attachment due to a local type IV hypersensitivity reaction to antigens in the tick’s saliva. This reaction can be long-lasting. In addition to pruritic papules following a bite, an attached tick can be mistaken for a skin neoplasm or nevus. Given that ticks are small, especially during the larval stage, dermoscopy may be helpful in making a diagnosis.10 Symptomatic relief usually can be achieved with topical antipruritics or oral antihistamines.

Of public health concern, brown dog ticks are important vectors of Rickettsia rickettsii (the causative organism of Rocky Mountain spotted fever [RMSF]) in the Western hemisphere, and Rickettsia conorii (the causative organism of Mediterranean spotted fever [MSF][also known as Boutonneuse fever]) in the Eastern hemisphere.11 Bites by ticks carrying rickettsial disease classically manifest with early symptoms of fever, headache, and myalgia, followed by a rash or by a localized eschar or tache noire (a black, necrotic, scabbed lesion) that represents direct endothelial invasion and vascular damage by Rickettsia.12 Rocky Mountain spotted fever and MSF are more prevalent during summer, likely due, in part, to the combination of increased outdoor activity and a higher rate of tick-questing (host-seeking) behavior in warmer climates.4,7

Rocky Mountain Spotted FeverDermacentor variabilis is the primary vector of RMSF in the southeastern United States; Dermacentor andersoni is the major vector of RMSF in Rocky Mountain states. Rhipicephalus sanguineus sensu lato is an important vector of RMSF in the southwestern United States, Mexico, and Central America.11,13

Early symptoms of RMSF are nonspecific and can include fever, headache, arthralgia, myalgia, and malaise. Gastrointestinal tract symptoms (eg, nausea, vomiting, anorexia) may occur; notable abdominal pain occurs in some patients, particularly children. A characteristic petechial rash occurs in as many as 90% of patients, typically at the third to fifth day of illness, and classically begins on the wrists and ankles, with progression to the palms and soles before spreading centripetally to the arms, legs, and trunk.14 An eschar at the inoculation site is uncommon in RMSF; when present, it is more suggestive of MSF.15

The classic triad of fever, headache, and rash is present in 3% of patients during the first 3 days after a tick bite and in 60% to 70% within 2 weeks.16 A rash often is absent when patients first seek medical attention and may not develop (absent in 9% to 12% of cases; so-called spotless RMSF). Therefore, absence of rash should not be a reason to withhold treatment.16 Empiric treatment with doxycycline should be started promptly for all suspected cases of RMSF because of the rapid progression of disease and an increased risk for morbidity and mortality with delayed diagnosis.

 

 

Patients do not become antibody positive until 7 to 10 days after symptoms begin; therefore, treatment should not be delayed while awaiting serologic test results. The case fatality rate in the United States is estimated to be 5% to 10% overall and as high as 40% to 50% among patients who are not treated until day 8 or 9 of illness.17

Cutaneous complications include skin necrosis and gangrene due to continuous tissue damage in severe cases.16 Severe infection also may manifest with signs of multiorgan system damage, including altered mental status, cerebral edema, meningismus, transient deafness, myocarditis, pulmonary hemorrhage and edema, conjunctivitis, retinal abnormalities, and acute renal failure.14,16 Risk factors for more severe illness include delayed treatment, age 40 years or older or younger than 10 years, and underlying medical conditions such as alcoholic liver disease and glucose-6-phosphate dehydrogenase deficiency. However, even some healthy young patients die of this disease.17

Mediterranean Spotted FeverRhipicephalus sanguineus sensu lato is the primary vector of MSF, which is prevalent in areas adjacent to the Mediterranean Sea, including southern Europe, Africa, and Central Asia; Sicily is the most highly affected region.18 Findings with MSF are nearly identical to those of RMSF, except that tache noire is more common, present in as many as 70% of cases at the site of the inoculating tick bite, and MSF typically follows a less severe clinical course.12 Similar to other rickettsial diseases, the pathogenesis of MSF involves direct injury to vascular endothelial cells, causing a vasculitis that is responsible for the clinical abnormalities observed.

Patients with severe MSF experience complications similar to severe RMSF, including neurologic manifestations and multiorgan damage.18 Risk factors include advanced age, immunocompromised state, cardiac disease, chronic alcoholism, diabetes mellitus, glucose-6-phosphate dehydrogenase deficiency, respiratory insufficiency, and delayed treatment.18

Treatment—For all spotted fever group rickettsial infections, doxycycline is the treatment of choice for all patients, including children and pregnant women. Treatment should be started without delay; recommended dosages are 100 mg twice daily for children weighing more than 45 kg and adults, and 2.2 mg/kg twice daily for children weighing 45 kg or less.12

Rhipicephalus tick bites rarely can result in paralysis; however, Dermacentor ticks are responsible for most cases of tick-related paralysis in North America. Other pathogens proven or reputed to be transmitted by Rhipicephalus sanguineus sensu lato with zoonotic potential include but are not limited to Rickettsia massiliae, Coxiella burnetti, Anaplasma platys, Leishmania infantum, and Crimean-Congo hemorrhagic fever virus (Nairovirus).19

Environmental Treatment and Prevention

The most effective way to prevent tick-borne illness is avoidance of tick bites. Primary prevention methods include vector control, use of repellents (eg, N,N-diethyl-meta-toluamide [DEET]), picaridin, permethrin), avoidance of areas with a high tick burden, use of protective clothing, and detection and removal of ticks as soon as possible.

 

 

Environmental and veterinary controls also are important methods of tick-bite prevention. A veterinarian can recommend a variety of agents for dogs and cats that prevent attachment of ticks. Environmental controls include synthetic or natural product-based chemical acaricides and nonchemical methods, such as landscape management (eg, sealing cracks and crevices in homes and controlling tall grasses, weeds, and leaf debris) to minimize potential tick habitat.20 Secondary prevention includes antibiotics for prophylaxis or for treatment of tick-borne disease, when indicated.

Numerous tick repellents are available commercially; others are being studied. DEET, the most widely used topical repellent, has a broad spectrum of activity against many tick species.21 In addition, DEET has a well-known safety and toxicity profile, with rare adverse effects, and is safe for use in pregnant women and children older than 2 years. Alternative repellents, such as those containing picaridin, ethyl butylacetylaminopropionate (IR3535 [Merck]), oil of lemon eucalyptus, and 2-undecanone can be effective; some show efficacy comparable to that of DEET.22 Permethrin, a synthetic pyrethroid, is a highly efficacious tick repellent and insecticide, especially when used in conjunction with a topical repellent such as DEET. Unlike topically applied repellents, permethrin spray is applied to fabric (eg, clothing, shoes, bed nets, camping gear), not to skin.

Indiscriminate use of acaricides worldwide has led to increasing selection of acaricide resistance in Rhipicephalus tick species, which is especially true with the use of acaricides in controlling R microplus livestock infestations; several tick populations now show resistance to all major classes of these compounds.23-25 For that reason, there has been an increasing effort to develop new chemical and nonchemical approaches to tick control that are more environmentally sustainable and strategies to minimize development and progression of resistance such as rotation of acaricides; reducing the frequency of their application; use of pesticide mixtures, synergists, or both; and increasing use of nonacaricidal methods of control.26

Prompt removal of ticks is important for preventing the transmission of tick-borne disease. Proper removal involves rubbing the tick in a circular motion with a moist gauze pad or using fine-tipped tweezers to grasp the tick as close to the skin surface as possible and pulling upward with a steady pressure.17,27 It is important not to jerk, twist, squeeze, smash, or burn the tick, as this can result in insufficient removal of mouthparts or spread contaminated tick fluids to mucous membranes, increasing the risk for infection. Application of petroleum jelly or nail polish to aid in tick removal have not been shown to be effective and are not recommended.16,28

Characteristics

Rhipicephalus ticks belong to the Ixodidae family of hard-bodied ticks. They are large and teardrop shaped with an inornate scutum (hard dorsal plate) and relatively short mouthparts attached at a hexagonal basis capitulum (base of the head to which mouthparts are attached)(Figure).1 Widely spaced eyes and festoons also are present. The first pair of coxae—attachment base for the first pair of legs—are characteristically bifid; males have a pair of sclerotized adanal plates on the ventral surface adjacent to the anus as well as accessory adanal shields.2Rhipicephalus (formerly Boophilus) microplus (the so-called cattle tick) is a newly added species; it lacks posterior festoons, and the anal groove is absent.3

Brantley_Figure.jpg
%3Cp%3E%3Cem%3ERhipicephalus%3C%2Fem%3E%20ticks%20are%20brown%20and%20teardrop%20shaped%20with%20an%20inornate%20scutum.%20The%20hexagonal%20basis%20capitulum%20is%20a%20defining%20characteristic.%20The%20image%20is%20in%20the%20public%20domain.%3C%2Fp%3E

Almost all Rhipicephalus ticks, except for R microplus, are 3-host ticks in which a single blood meal is consumed from a vertebrate host at each active life stage—larva, nymph, and adult—to complete development.4,5 In contrast to most ixodid ticks, which are exophilic (living outside of human habitation), the Rhipicephalus sanguineus sensu lato species (the brown dog tick) is highly endophilic (adapted to indoor living) and often can be found hidden in cracks and crevices of walls in homes and peridomestic structures.6 It is predominately monotropic (all developmental stages feed on the same host species) and has a strong host preference for dogs, though it occasionally feeds on other hosts (eg, humans).7 Although most common in tropical and subtropical climates, they can be found anywhere there are dogs due to their ability to colonize indoor dwellings.8 In contrast, R microplus ticks have a predilection for cattle and livestock rather than humans, posing a notable concern to livestock worldwide. Infestation results in transmission of disease-causing pathogens, such as Babesia and Anaplasma species, which costs the cattle industry billions of dollars annually.9

Clinical Manifestations and Treatment

Tick bites usually manifest as intensely pruritic, erythematous papules at the site of tick attachment due to a local type IV hypersensitivity reaction to antigens in the tick’s saliva. This reaction can be long-lasting. In addition to pruritic papules following a bite, an attached tick can be mistaken for a skin neoplasm or nevus. Given that ticks are small, especially during the larval stage, dermoscopy may be helpful in making a diagnosis.10 Symptomatic relief usually can be achieved with topical antipruritics or oral antihistamines.

Of public health concern, brown dog ticks are important vectors of Rickettsia rickettsii (the causative organism of Rocky Mountain spotted fever [RMSF]) in the Western hemisphere, and Rickettsia conorii (the causative organism of Mediterranean spotted fever [MSF][also known as Boutonneuse fever]) in the Eastern hemisphere.11 Bites by ticks carrying rickettsial disease classically manifest with early symptoms of fever, headache, and myalgia, followed by a rash or by a localized eschar or tache noire (a black, necrotic, scabbed lesion) that represents direct endothelial invasion and vascular damage by Rickettsia.12 Rocky Mountain spotted fever and MSF are more prevalent during summer, likely due, in part, to the combination of increased outdoor activity and a higher rate of tick-questing (host-seeking) behavior in warmer climates.4,7

Rocky Mountain Spotted FeverDermacentor variabilis is the primary vector of RMSF in the southeastern United States; Dermacentor andersoni is the major vector of RMSF in Rocky Mountain states. Rhipicephalus sanguineus sensu lato is an important vector of RMSF in the southwestern United States, Mexico, and Central America.11,13

Early symptoms of RMSF are nonspecific and can include fever, headache, arthralgia, myalgia, and malaise. Gastrointestinal tract symptoms (eg, nausea, vomiting, anorexia) may occur; notable abdominal pain occurs in some patients, particularly children. A characteristic petechial rash occurs in as many as 90% of patients, typically at the third to fifth day of illness, and classically begins on the wrists and ankles, with progression to the palms and soles before spreading centripetally to the arms, legs, and trunk.14 An eschar at the inoculation site is uncommon in RMSF; when present, it is more suggestive of MSF.15

The classic triad of fever, headache, and rash is present in 3% of patients during the first 3 days after a tick bite and in 60% to 70% within 2 weeks.16 A rash often is absent when patients first seek medical attention and may not develop (absent in 9% to 12% of cases; so-called spotless RMSF). Therefore, absence of rash should not be a reason to withhold treatment.16 Empiric treatment with doxycycline should be started promptly for all suspected cases of RMSF because of the rapid progression of disease and an increased risk for morbidity and mortality with delayed diagnosis.

 

 

Patients do not become antibody positive until 7 to 10 days after symptoms begin; therefore, treatment should not be delayed while awaiting serologic test results. The case fatality rate in the United States is estimated to be 5% to 10% overall and as high as 40% to 50% among patients who are not treated until day 8 or 9 of illness.17

Cutaneous complications include skin necrosis and gangrene due to continuous tissue damage in severe cases.16 Severe infection also may manifest with signs of multiorgan system damage, including altered mental status, cerebral edema, meningismus, transient deafness, myocarditis, pulmonary hemorrhage and edema, conjunctivitis, retinal abnormalities, and acute renal failure.14,16 Risk factors for more severe illness include delayed treatment, age 40 years or older or younger than 10 years, and underlying medical conditions such as alcoholic liver disease and glucose-6-phosphate dehydrogenase deficiency. However, even some healthy young patients die of this disease.17

Mediterranean Spotted FeverRhipicephalus sanguineus sensu lato is the primary vector of MSF, which is prevalent in areas adjacent to the Mediterranean Sea, including southern Europe, Africa, and Central Asia; Sicily is the most highly affected region.18 Findings with MSF are nearly identical to those of RMSF, except that tache noire is more common, present in as many as 70% of cases at the site of the inoculating tick bite, and MSF typically follows a less severe clinical course.12 Similar to other rickettsial diseases, the pathogenesis of MSF involves direct injury to vascular endothelial cells, causing a vasculitis that is responsible for the clinical abnormalities observed.

Patients with severe MSF experience complications similar to severe RMSF, including neurologic manifestations and multiorgan damage.18 Risk factors include advanced age, immunocompromised state, cardiac disease, chronic alcoholism, diabetes mellitus, glucose-6-phosphate dehydrogenase deficiency, respiratory insufficiency, and delayed treatment.18

Treatment—For all spotted fever group rickettsial infections, doxycycline is the treatment of choice for all patients, including children and pregnant women. Treatment should be started without delay; recommended dosages are 100 mg twice daily for children weighing more than 45 kg and adults, and 2.2 mg/kg twice daily for children weighing 45 kg or less.12

Rhipicephalus tick bites rarely can result in paralysis; however, Dermacentor ticks are responsible for most cases of tick-related paralysis in North America. Other pathogens proven or reputed to be transmitted by Rhipicephalus sanguineus sensu lato with zoonotic potential include but are not limited to Rickettsia massiliae, Coxiella burnetti, Anaplasma platys, Leishmania infantum, and Crimean-Congo hemorrhagic fever virus (Nairovirus).19

Environmental Treatment and Prevention

The most effective way to prevent tick-borne illness is avoidance of tick bites. Primary prevention methods include vector control, use of repellents (eg, N,N-diethyl-meta-toluamide [DEET]), picaridin, permethrin), avoidance of areas with a high tick burden, use of protective clothing, and detection and removal of ticks as soon as possible.

 

 

Environmental and veterinary controls also are important methods of tick-bite prevention. A veterinarian can recommend a variety of agents for dogs and cats that prevent attachment of ticks. Environmental controls include synthetic or natural product-based chemical acaricides and nonchemical methods, such as landscape management (eg, sealing cracks and crevices in homes and controlling tall grasses, weeds, and leaf debris) to minimize potential tick habitat.20 Secondary prevention includes antibiotics for prophylaxis or for treatment of tick-borne disease, when indicated.

Numerous tick repellents are available commercially; others are being studied. DEET, the most widely used topical repellent, has a broad spectrum of activity against many tick species.21 In addition, DEET has a well-known safety and toxicity profile, with rare adverse effects, and is safe for use in pregnant women and children older than 2 years. Alternative repellents, such as those containing picaridin, ethyl butylacetylaminopropionate (IR3535 [Merck]), oil of lemon eucalyptus, and 2-undecanone can be effective; some show efficacy comparable to that of DEET.22 Permethrin, a synthetic pyrethroid, is a highly efficacious tick repellent and insecticide, especially when used in conjunction with a topical repellent such as DEET. Unlike topically applied repellents, permethrin spray is applied to fabric (eg, clothing, shoes, bed nets, camping gear), not to skin.

Indiscriminate use of acaricides worldwide has led to increasing selection of acaricide resistance in Rhipicephalus tick species, which is especially true with the use of acaricides in controlling R microplus livestock infestations; several tick populations now show resistance to all major classes of these compounds.23-25 For that reason, there has been an increasing effort to develop new chemical and nonchemical approaches to tick control that are more environmentally sustainable and strategies to minimize development and progression of resistance such as rotation of acaricides; reducing the frequency of their application; use of pesticide mixtures, synergists, or both; and increasing use of nonacaricidal methods of control.26

Prompt removal of ticks is important for preventing the transmission of tick-borne disease. Proper removal involves rubbing the tick in a circular motion with a moist gauze pad or using fine-tipped tweezers to grasp the tick as close to the skin surface as possible and pulling upward with a steady pressure.17,27 It is important not to jerk, twist, squeeze, smash, or burn the tick, as this can result in insufficient removal of mouthparts or spread contaminated tick fluids to mucous membranes, increasing the risk for infection. Application of petroleum jelly or nail polish to aid in tick removal have not been shown to be effective and are not recommended.16,28

References
  1. Dantas-Torres F. The brown dog tick, Rhipicephalus sanguineus (Latreille, 1806) (Acari: Ixodidae): from taxonomy to control. Vet Parasitol. 2008;152:173-185. doi:10.1016/j.vetpar.2007.12.030
  2. Madder M, Fourie JJ, Schetters TPM. Arachnida, Metastigmata, Ixodidae (except Ixodes holocyclus). In: Marchiondo AA, Cruthers LR, Fourie JJ, eds. Parasiticide Screening: In Vitro and In Vivo Tests With Relevant Parasite Rearing and Host Infection/Infestation Methods. Volume 1. Elsevier Academic Press; 2019:19-20.
  3. Burger TD, Shao R, Barker SC. Phylogenetic analysis of mitochondrial genome sequences indicates that the cattle tick, Rhipicephalus (Boophilus) microplus, contains a cryptic species. Mol Phylogenet Evol. 2014;76:241-253. doi:10.1016/j.ympev.2014.03.017
  4. Gray J, Dantas-Torres F, Estrada-Peña A, et al. Systematics and ecology of the brown dog tick, Rhipicephalus sanguineus. Ticks Tick Borne Dis. 2013;4:171-180. doi:10.1016/j.ttbdis.2012.12.003
  5. Tian Y, Lord CC, Kaufman PE. Brown dog tick, Rhipicephalus Sanguineus Latrielle (Arachnida: Acari: Ixodidae): EENY-221/IN378. EDIS. March 26, 2020. Accessed January 3, 2024. https://doi.org/10.32473/edis-in378-2020
  6. Saleh MN, Allen KE, Lineberry MW, et al. Ticks infesting dogs and cats in North America: biology, geographic distribution, and pathogen transmission. Vet Parasitol. 2021;294:109392. doi:10.1016/j.vetpar.2021.109392
  7. Dantas-Torres F. Biology and ecology of the brown dog tick, Rhipicephalus sanguineus. Parasit Vectors. 2010;3:26. doi:10.1186/1756-3305-3-26
  8. Dryden MW, Payne PA. Biology and control of ticks infesting dogs and cats in North America. Vet Ther. 2004;5:139-154.
  9. Nyangiwe N, Yawa M, Muchenje V. Driving forces for changes in geographic range of cattle ticks (Acari: Ixodidae) in Africa: a Review. S Afr J Anim Sci. 2018;48:829. doi:10.4314/sajas.v48i5.4
  10. Ramot Y, Zlotogorski A, Mumcuoglu KY. Brown dog tick (Rhipicephalus sanguineus) infestation of the penis detected by dermoscopy. Int J Dermatol. 2012;51:1402-1403. doi:10.1111/j.1365-4632.2010.04756.x
  11. Tucker NSG, Weeks ENI, Beati L, et al. Prevalence and distribution of pathogen infection and permethrin resistance in tropical and temperate populations of Rhipicephalus sanguineus s.l. collected worldwide. Med Vet Entomol. 2021;35:147-157. doi:10.1111/mve.12479
  12. McClain MT, Sexton DJ, Hall KK, eds. Other spotted fever group rickettsial infections. UpToDate. Updated October 10, 2022. Accessed January 3, 2024. https://www.uptodate.com/contents/other-spotted-fever-group-rickettsial-infections
  13. Ribeiro CM, Carvalho JLB, Bastos PAS, et al. Prevalence of Rickettsia rickettsii in ticks: systematic review and meta-analysis. Vector Borne Zoonotic Dis. 2021;21:557-565. doi:10.1089/vbz.2021.0004
  14. Pace EJ, O’Reilly M. Tickborne diseases: diagnosis and management. Am Fam Physician. 2020;101:530-540.
  15. Patterson JW. Weedon’s Skin Pathology. 5th ed. Elsevier; 2020.
  16. Dantas-Torres F. Rocky Mountain spotted fever. Lancet Infect Dis. 2007;7:724-732. doi:10.1016/S1473-3099(07)70261-X
  17. Biggs HM, Behravesh CB, Bradley KK, et al. Diagnosis and management of tickborne rickettsial diseases: Rocky Mountain spotted fever and other spotted fever group rickettsioses, ehrlichioses, and anaplasmosis—United States. MMWR Recomm Rep. 2016;65:1-44. doi:10.15585/mmwr.rr6502a1
  18. Rossio R, Conalbi V, Castagna V, et al. Mediterranean spotted fever and hearing impairment: a rare complication. Int J Infect Dis. 2015;35:34-36. doi:10.1016/j.ijid.2015.04.005
  19. Dantas-Torres F, Otranto D. Further thoughts on the taxonomy and vector role of Rhipicephalus sanguineus group ticks. Vet Parasitol. 2015;208:9-13. doi:10.1016/j.vetpar.2014.12.014
  20. Eisen RJ, Kugeler KJ, Eisen L, et al. Tick-borne zoonoses in the United States: persistent and emerging threats to human health. ILAR J. 2017;58:319-335. doi:10.1093/ilar/ilx005
  21. Nguyen QD, Vu MN, Hebert AA. Insect repellents: an updated review for the clinician. J Am Acad Dermatol. 2018;88:123-130. doi:10.1016/j.jaad.2018.10.053
  22. Pages F, Dautel H, Duvallet G, et al. Tick repellents for human use: prevention of tick bites and tick-borne diseases. Vector Borne Zoonotic Dis. 2014;14:85-93. doi:10.1089/vbz.2013.1410
  23. Rodriguez-Vivas RI, Alonso-Díaz MA, et al. Prevalence and potential risk factors for organophosphate and pyrethroid resistance in Boophilus microplus ticks on cattle ranches from the State of Yucatan, Mexico. Vet Parasitol. 2006;136:335-342. doi:10.1016/j.vetpar.2005.05.069
  24. Rodríguez-Vivas RI, Rodríguez-Arevalo F, Alonso-Díaz MA, et al. Prevalence and potential risk factors for amitraz resistance in Boophilus microplus ticks in cattle farms in the State of Yucatan, Mexico. Prev Vet Med. 2006;75:280-286. doi:10.1016/j.prevetmed.2006.04.001
  25. Perez-Cogollo LC, Rodriguez-Vivas RI, Ramirez-Cruz GT, et al. First report of the cattle tick Rhipicephalus microplus resistant to ivermectin in Mexico. Vet Parasitol. 2010;168:165-169. doi:10.1016/j.vetpar.2009.10.021
  26. Rodriguez-Vivas RI, Jonsson NN, Bhushan C. Strategies for the control of Rhipicephalus microplus ticks in a world of conventional acaricide and macrocyclic lactone resistance. Parasitol Res.2018;117:3-29. doi:10.1007/s00436-017-5677-6
  27. Centers for Disease Control and Prevention. Tick removal. Updated May 13, 2022. Accessed January 3, 2024. https://www.cdc.gov/ticks/removing_a_tick.html
  28. Diaz JH. Chemical and plant-based insect repellents: efficacy, safety, and toxicity. Wilderness Environ Med. 2016;27:153-163. doi:10.1016/j.wem.2015.11.007
References
  1. Dantas-Torres F. The brown dog tick, Rhipicephalus sanguineus (Latreille, 1806) (Acari: Ixodidae): from taxonomy to control. Vet Parasitol. 2008;152:173-185. doi:10.1016/j.vetpar.2007.12.030
  2. Madder M, Fourie JJ, Schetters TPM. Arachnida, Metastigmata, Ixodidae (except Ixodes holocyclus). In: Marchiondo AA, Cruthers LR, Fourie JJ, eds. Parasiticide Screening: In Vitro and In Vivo Tests With Relevant Parasite Rearing and Host Infection/Infestation Methods. Volume 1. Elsevier Academic Press; 2019:19-20.
  3. Burger TD, Shao R, Barker SC. Phylogenetic analysis of mitochondrial genome sequences indicates that the cattle tick, Rhipicephalus (Boophilus) microplus, contains a cryptic species. Mol Phylogenet Evol. 2014;76:241-253. doi:10.1016/j.ympev.2014.03.017
  4. Gray J, Dantas-Torres F, Estrada-Peña A, et al. Systematics and ecology of the brown dog tick, Rhipicephalus sanguineus. Ticks Tick Borne Dis. 2013;4:171-180. doi:10.1016/j.ttbdis.2012.12.003
  5. Tian Y, Lord CC, Kaufman PE. Brown dog tick, Rhipicephalus Sanguineus Latrielle (Arachnida: Acari: Ixodidae): EENY-221/IN378. EDIS. March 26, 2020. Accessed January 3, 2024. https://doi.org/10.32473/edis-in378-2020
  6. Saleh MN, Allen KE, Lineberry MW, et al. Ticks infesting dogs and cats in North America: biology, geographic distribution, and pathogen transmission. Vet Parasitol. 2021;294:109392. doi:10.1016/j.vetpar.2021.109392
  7. Dantas-Torres F. Biology and ecology of the brown dog tick, Rhipicephalus sanguineus. Parasit Vectors. 2010;3:26. doi:10.1186/1756-3305-3-26
  8. Dryden MW, Payne PA. Biology and control of ticks infesting dogs and cats in North America. Vet Ther. 2004;5:139-154.
  9. Nyangiwe N, Yawa M, Muchenje V. Driving forces for changes in geographic range of cattle ticks (Acari: Ixodidae) in Africa: a Review. S Afr J Anim Sci. 2018;48:829. doi:10.4314/sajas.v48i5.4
  10. Ramot Y, Zlotogorski A, Mumcuoglu KY. Brown dog tick (Rhipicephalus sanguineus) infestation of the penis detected by dermoscopy. Int J Dermatol. 2012;51:1402-1403. doi:10.1111/j.1365-4632.2010.04756.x
  11. Tucker NSG, Weeks ENI, Beati L, et al. Prevalence and distribution of pathogen infection and permethrin resistance in tropical and temperate populations of Rhipicephalus sanguineus s.l. collected worldwide. Med Vet Entomol. 2021;35:147-157. doi:10.1111/mve.12479
  12. McClain MT, Sexton DJ, Hall KK, eds. Other spotted fever group rickettsial infections. UpToDate. Updated October 10, 2022. Accessed January 3, 2024. https://www.uptodate.com/contents/other-spotted-fever-group-rickettsial-infections
  13. Ribeiro CM, Carvalho JLB, Bastos PAS, et al. Prevalence of Rickettsia rickettsii in ticks: systematic review and meta-analysis. Vector Borne Zoonotic Dis. 2021;21:557-565. doi:10.1089/vbz.2021.0004
  14. Pace EJ, O’Reilly M. Tickborne diseases: diagnosis and management. Am Fam Physician. 2020;101:530-540.
  15. Patterson JW. Weedon’s Skin Pathology. 5th ed. Elsevier; 2020.
  16. Dantas-Torres F. Rocky Mountain spotted fever. Lancet Infect Dis. 2007;7:724-732. doi:10.1016/S1473-3099(07)70261-X
  17. Biggs HM, Behravesh CB, Bradley KK, et al. Diagnosis and management of tickborne rickettsial diseases: Rocky Mountain spotted fever and other spotted fever group rickettsioses, ehrlichioses, and anaplasmosis—United States. MMWR Recomm Rep. 2016;65:1-44. doi:10.15585/mmwr.rr6502a1
  18. Rossio R, Conalbi V, Castagna V, et al. Mediterranean spotted fever and hearing impairment: a rare complication. Int J Infect Dis. 2015;35:34-36. doi:10.1016/j.ijid.2015.04.005
  19. Dantas-Torres F, Otranto D. Further thoughts on the taxonomy and vector role of Rhipicephalus sanguineus group ticks. Vet Parasitol. 2015;208:9-13. doi:10.1016/j.vetpar.2014.12.014
  20. Eisen RJ, Kugeler KJ, Eisen L, et al. Tick-borne zoonoses in the United States: persistent and emerging threats to human health. ILAR J. 2017;58:319-335. doi:10.1093/ilar/ilx005
  21. Nguyen QD, Vu MN, Hebert AA. Insect repellents: an updated review for the clinician. J Am Acad Dermatol. 2018;88:123-130. doi:10.1016/j.jaad.2018.10.053
  22. Pages F, Dautel H, Duvallet G, et al. Tick repellents for human use: prevention of tick bites and tick-borne diseases. Vector Borne Zoonotic Dis. 2014;14:85-93. doi:10.1089/vbz.2013.1410
  23. Rodriguez-Vivas RI, Alonso-Díaz MA, et al. Prevalence and potential risk factors for organophosphate and pyrethroid resistance in Boophilus microplus ticks on cattle ranches from the State of Yucatan, Mexico. Vet Parasitol. 2006;136:335-342. doi:10.1016/j.vetpar.2005.05.069
  24. Rodríguez-Vivas RI, Rodríguez-Arevalo F, Alonso-Díaz MA, et al. Prevalence and potential risk factors for amitraz resistance in Boophilus microplus ticks in cattle farms in the State of Yucatan, Mexico. Prev Vet Med. 2006;75:280-286. doi:10.1016/j.prevetmed.2006.04.001
  25. Perez-Cogollo LC, Rodriguez-Vivas RI, Ramirez-Cruz GT, et al. First report of the cattle tick Rhipicephalus microplus resistant to ivermectin in Mexico. Vet Parasitol. 2010;168:165-169. doi:10.1016/j.vetpar.2009.10.021
  26. Rodriguez-Vivas RI, Jonsson NN, Bhushan C. Strategies for the control of Rhipicephalus microplus ticks in a world of conventional acaricide and macrocyclic lactone resistance. Parasitol Res.2018;117:3-29. doi:10.1007/s00436-017-5677-6
  27. Centers for Disease Control and Prevention. Tick removal. Updated May 13, 2022. Accessed January 3, 2024. https://www.cdc.gov/ticks/removing_a_tick.html
  28. Diaz JH. Chemical and plant-based insect repellents: efficacy, safety, and toxicity. Wilderness Environ Med. 2016;27:153-163. doi:10.1016/j.wem.2015.11.007
Issue
Cutis - 113(1)
Issue
Cutis - 113(1)
Page Number
E44-E47
Page Number
E44-E47
Publications
Publications
Topics
Article Type
Display Headline
What’s Eating You? Rhipicephalus Ticks Revisited
Display Headline
What’s Eating You? Rhipicephalus Ticks Revisited
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Brantley</fileName> <TBEID>0C02F1CB.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F1CB</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Brantley</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240131T134800</firstPublished> <LastPublished>20240131T134800</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240131T134759</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Rebecca A. Brantley, BS; Dirk M. Elston, MD</byline> <bylineText>Rebecca A. Brantley, BS; Dirk M. Elston, MD</bylineText> <bylineFull>Rebecca A. Brantley, BS; Dirk M. Elston, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>E44-E47</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Rhipicephalus ticks belong to the Ixodidae family of hard-bodied ticks. They are large and teardrop shaped with an inornate scutum (hard dorsal plate) and relat</metaDescription> <articlePDF>300123</articlePDF> <teaserImage/> <title>What’s Eating You? Rhipicephalus Ticks Revisited</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>January</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>1</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2159</CMSID> </CMSIDs> <keywords> <keyword>infectious disease</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>January 2024</pubIssueName> <pubArticleType>Departments | 2159</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">60</term> </sections> <topics> <term canonical="true">234</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180026b8.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>What’s Eating You? Rhipicephalus Ticks Revisited</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><em>Rhipicephalus </em>ticks are vectors of disease in humans and animals. <em>Rhipicephalus sanguineus </em>sensu lato (the brown dog tick) is one of the most geographically widespread tick species worldwide, likely due to its ability to colonize human and canine dwellings over a range of habitats. They transmit a variety of diseases to dogs and humans, including canine babesiosis, canine monocytic ehrlichiosis, hepatozoonosis, Mediterranean spotted fever, and Rocky Mountain spotted fever. Tick bites manifest as intensely pruritic, erythematous papules at the site of tick attachment; symptomatic relief usually can be achieved with topical antipruritics. Prevention of tick bites is best achieved through a combination of veterinary and environmental control; protective clothing; repellents, such as N,N-diethyl-meta-toluamide (DEET) and permethrin; and prompt identification and removal of ticks. </p> <p> <em><em>Cutis.</em> 2024;113:E44-E47.</em> </p> <h3>Characteristics </h3> <p><i>Rhipicephalus </i>ticks belong to the Ixodidae family of hard-bodied ticks. They are large and teardrop shaped with an inornate scutum (hard dorsal plate) and relatively short mouthparts attached at a hexagonal basis capitulum (base of the head to which mouthparts are attached)(Figure).<sup>1</sup> Widely spaced eyes and festoons also are present. The first pair of coxae—attachment base for the first pair of legs—are characteristically bifid; males have a pair of sclerotized adanal plates on the ventral surface adjacent to the anus as well as accessory adanal shields.<sup>2</sup> <i>Rhipicephalus </i>(formerly <i>Boophilus</i>) <i>microplus</i> (the so-called cattle tick) is a newly added species; it lacks posterior festoons, and the anal groove is absent.<sup>3</sup> </p> <p>Almost all <i>Rhipicephalus</i> ticks, except for <i>R microplus</i>, are 3-host ticks in which a single blood meal is consumed from a vertebrate host at each active life stage—larva, nymph, and adult—to complete development.<sup>4,5</sup> In contrast to most ixodid ticks, which are exophilic (living outside of human habitation), the <i>Rhipicephalus sanguineus </i>sensu lato species (the brown dog tick) is highly endophilic (adapted to indoor living) and often can be found hidden in cracks and crevices of walls in homes and peridomestic structures.<sup>6</sup> It is predominately monotropic (all developmental stages feed on the same host species) and has a strong host preference for dogs, though it occasionally feeds on other hosts (eg, humans).<sup>7</sup> Although most common in tropical and subtropical climates, they can be found anywhere there are dogs due to their ability to colonize indoor dwellings.<sup>8</sup> In contrast, <i>R microplus</i> ticks have a predilection for cattle and livestock rather than humans, posing a notable concern to livestock worldwide. Infestation results in transmission of disease-causing pathogens, such as <i>Babesia</i> and <i>Anaplasma </i>species, which costs the cattle industry billions of dollars annually.<sup>9</sup></p> <h3>Clinical Manifestations and Treatment</h3> <p>Tick bites usually manifest as intensely pruritic, erythematous papules at the site of tick attachment due to a local type IV hypersensitivity reaction to antigens in the tick’s saliva. This reaction can be long-lasting. In addition to pruritic papules following a bite, an attached tick can be mistaken for a skin neoplasm or nevus. Given that ticks are small, especially during the larval stage, dermoscopy may be helpful in making a diagnosis.<sup>10</sup> Symptomatic relief usually can be achieved with topical antipruritics or oral antihistamines. </p> <p>Of public health concern, brown dog ticks are important vectors of <i>Rickettsia rickettsii</i> (the causative organism of Rocky Mountain spotted fever [RMSF]) in the Western hemisphere, and <i>Rickettsia conorii</i> (the causative organism of Mediterranean spotted fever [MSF][also known as Boutonneuse fever]) in the Eastern hemisphere.<sup>11</sup> Bites by<span class="apple-converted-space"> ticks carrying rickettsial disease classically manifest with early symptoms of fever, headache, and myalgia, followed by a rash or by a localized eschar or tache noire (a black, necrotic, scabbed lesion) that represents direct endothelial invasion and vascular damage by </span><span class="apple-converted-space"><i>Rickettsia</i></span><span class="apple-converted-space">.</span><span class="apple-converted-space"><sup>12</sup></span><span class="apple-converted-space"> </span>Rocky Mountain spotted fever<span class="apple-converted-space"> and MSF </span>are more prevalent during summer, likely due, in part, to the combination of increased outdoor activity and a higher rate of tick-questing (host-seeking) behavior in warmer climates.<sup>4,7<br/><br/></sup><i>Rocky Mountain Spotted Fever</i>—<i>Dermacentor variabilis</i> is the primary vector of RMSF in the southeastern United States; <i>Dermacentor andersoni</i> is the major vector of RMSF in Rocky Mountain states. <i>Rhipicephalus sanguineus </i>sensu lato is an important vector of RMSF in the southwestern United States, Mexico, and Central America.<sup>11,13<br/><br/></sup>Early symptoms of RMSF are nonspecific and can include fever, headache, arthralgia, myalgia, and malaise. Gastrointestinal tract symptoms (eg, nausea, vomiting, anorexia) may occur; notable abdominal pain occurs in some patients, particularly children. A characteristic petechial rash occurs in as many as 90% of patients, typically at the third to fifth day of illness, and classically begins on the wrists and ankles, with progression to the palms and soles before spreading centripetally to the arms, legs, and trunk.<span class="apple-converted-space"><sup>14</sup></span> An es<span class="apple-converted-space">char at the inoculation site is uncommon in RMSF; when present, it is more suggestive of MSF.</span><span class="apple-converted-space"><sup>15</sup></span><span class="apple-converted-space"> <br/><br/>T</span>he classic triad of fever, headache, and rash is present in 3% of patients during the first 3 days after a tick bite and in 60% to 70% within 2 weeks.<sup>16</sup> A r<span class="apple-converted-space">ash often is absent when patients first seek medical attention and may not develop (absent in 9% to 12% of cases; so-called spotless RMSF). Therefore, absence of rash should not be a reason to withhold treatment.</span><span class="apple-converted-space"><sup>16</sup></span><span class="apple-converted-space"> Empiric treatment with doxycycline should be started promptly for all suspected cases of RMSF because of the rapid progression of disease and an increased risk for morbidity and mortality with delayed diagnosis. <br/><br/></span>Patients do not become antibody positive until 7 to 10 days after symptoms begin; therefore, treatment should not be delayed while awaiting serologic test results. <span class="apple-converted-space">The case fatality rate in the United States is estimated to be 5% to 10% overall and as high as 40% to 50% among patients who are not treated until day 8 or 9 of illness.</span><span class="apple-converted-space"><sup>17 <br/><br/></sup></span><span class="apple-converted-space">Cutaneous complications include skin necrosis and gangrene due to continuous tissue damage in severe cases.</span><span class="apple-converted-space"><sup>16</sup></span><span class="apple-converted-space"> Severe infection also may manifest with signs of multiorgan system damage, including</span> altered mental status, cerebral edema, meningismus, transient deafness, myocarditis, pulmonary hemorrhage and edema, conjunctivitis, retinal abnormalities, and acute renal failure.<sup>14,16</sup> Risk factors for more severe illness include delayed treatment, age 40 years or older or younger than 10 years, and underlying medical conditions such as alcoholic liver disease and glucose-6-phosphate dehydrogenase deficiency. However, even some healthy young patients die of this disease.<sup>17<br/><br/></sup><i>Mediterranean Spotted Fever</i>—<i>Rhipicephalus sanguineus </i>sensu lato is the primary vector of MSF, which is prevalent in areas adjacent to the Mediterranean Sea, including southern Europe, Africa, and Central Asia; Sicily is the most highly affected region.<sup>18</sup> Findings with MSF are nearly identical to those of RMSF, except that tache noire is more common, present in as many as 70% of cases at the site of the inoculating tick bite, and MSF typically follows a less severe clinical course.<sup>12</sup> Similar to other rickettsial diseases, the pathogenesis of MSF involves direct injury to vascular endothelial cells, causing a vasculitis that is responsible for the clinical abnormalities observed. <br/><br/>Patients with severe MSF experience complications similar to severe RMSF, including neurologic manifestations and multiorgan damage.<sup>18</sup> Risk factors include advanced age, immunocompromised state, cardiac disease, chronic alcoholism, diabetes mellitus, glucose-6-phosphate dehydrogenase deficiency, respiratory insufficiency, and delayed treatment.<sup>18<br/><br/></sup><i>Treatment—</i>For all spotted fever group rickettsial infections, doxycycline is the treatment of choice for all patients, including children and pregnant women. Treatment should be started without delay; recommended dosages are 100 mg twice daily for children weighing more than 45 kg and adults, and 2.2 mg/kg twice daily for children weighing 45 kg or less.<sup>12<br/><br/></sup><i>Rhipicephalus </i>tick bites rarely can result in paralysis; however, <i>Dermacentor</i> ticks are responsible for most cases of tick-related paralysis in North America. Other pathogens proven or reputed to be transmitted by <i>Rhipicephalus sanguineus </i>sensu lato with zoonotic potential include but are not limited to <i>Rickettsia massiliae</i>, <i>Coxiella burnetti</i>, <i>Anaplasma platys</i>, <i>Leishmania infantum</i>, and Crimean-Congo hemorrhagic fever virus (Nairovirus).<sup>19</sup> </p> <h3>Environmental Treatment and Prevention </h3> <p>The most effective way to prevent tick-borne illness is avoidance of tick bites. Primary prevention methods include vector control, use of repellents (eg, N,N-diethyl-meta-toluamide [DEET]), picaridin, permethrin), avoidance of areas with a high tick burden, use of protective clothing, and detection and removal of ticks as soon as possible. </p> <p>Environmental and veterinary controls also are important methods of tick-bite prevention. A veterinarian can recommend a variety of agents for dogs and cats that prevent attachment of ticks. Environmental controls include synthetic or natural product-based chemical acaricides and nonchemical methods, such as landscape management (eg, sealing cracks and crevices in homes and controlling tall grasses, weeds, and leaf debris) to minimize potential tick habitat.<sup>20</sup> Secondary prevention includes antibiotics for prophylaxis or for treatment of tick-borne disease, when indicated.<br/><br/>Numerous tick repellents are available commercially; others are being studied. DEET, the most widely used topical repellent, has a broad spectrum of activity against many tick species.<sup>21</sup> In addition, DEET has a well-known safety and toxicity profile, with rare adverse effects, and is safe for use in pregnant women and children older than 2 years. Alternative repellents, such as those containing picaridin, ethyl butylacetylaminopropionate (IR3535 [Merck]), oil of lemon eucalyptus, and 2-undecanone can be effective; some show efficacy comparable to that of DEET.<sup>22</sup> Permethrin, a synthetic pyrethroid, is a highly efficacious tick repellent and insecticide, especially when used in conjunction with a topical repellent such as DEET. Unlike topically applied repellents, permethrin spray is applied to fabric (eg, clothing, shoes, bed nets, camping gear), not to skin. <br/><br/>Indiscriminate use of acaricides worldwide has led to increasing selection of acaricide resistance in <i>Rhipicephalus</i> tick species, which is especially true with the use of acaricides in controlling <i>R microplus</i> livestock infestations; several tick populations now show resistance to all major classes of these compounds.<sup>23-25</sup> For that reason, there has been an increasing effort to develop new chemical and nonchemical approaches to tick control that are more environmentally sustainable and strategies to minimize development and progression of resistance such as rotation of acaricides; reducing the frequency of their application; use of pesticide mixtures, synergists, or both; and increasing use of nonacaricidal methods of control.<sup>26 <br/><br/></sup>Prompt removal of ticks is important for preventing the transmission of tick-borne disease. Proper removal involves rubbing the tick in a circular motion with a moist gauze pad or using fine-tipped tweezers to grasp the tick as close to the skin surface as possible and pulling upward with a steady pressure.<sup>17,27</sup> It is important not to jerk, twist, squeeze, smash, or burn the tick, as this can result in insufficient removal of mouthparts or spread contaminated tick fluids to mucous membranes, increasing the risk for infection. Application of petroleum jelly or nail polish to aid in tick removal have not been shown to be effective and are not recommended.<sup>16,28</sup></p> <h2>REFERENCES</h2> <p class="reference"> 1. Dantas-Torres F. The brown dog tick, <i>Rhipicephalus sanguineus</i> (Latreille, 1806) (Acari: Ixodidae): from taxonomy to control. <i>Vet Parasitol</i>. 2008;152:173-185. doi:10.1016/j.vetpar.2007.12.030 </p> <p class="reference"> 2. Madder M, Fourie JJ, Schetters TPM. Arachnida, Metastigmata, Ixodidae (except <i>Ixodes holocyclus</i>). In:<span class="apple-converted-space"> </span>Marchiondo AA, Cruthers LR, Fourie JJ, eds. <i>Parasiticide Screening: In Vitro and In Vivo Tests With Relevant Parasite Rearing and Host Infection/Infestation Methods</i><span class="apple-converted-space">. </span>Volume 1. Elsevier Academic Press; 2019:19-20.<br/><br/> 3. Burger TD, Shao R, Barker SC. Phylogenetic analysis of mitochondrial genome sequences indicates that the cattle tick, <i>Rhipicephalus (Boophilus) microplus</i>, contains a cryptic species. <i>Mol Phylogenet Evol</i>. 2014;76:241-253. doi:10.1016/j.ympev.2014.03.017 <br/><br/> 4. Gray J, Dantas-Torres F, Estrada-Peña A, et al. Systematics and ecology of the brown dog tick, <i>Rhipicephalus sanguineus</i>. <i>Ticks Tick Borne Dis</i>. 2013;4:171-180. doi:10.1016/j.ttbdis.2012.12.003<br/><br/> 5. Tian Y, Lord CC, Kaufman PE. Brown dog tick, <i>Rhipicephalus Sanguineus</i> Latrielle (Arachnida: Acari: Ixodidae): EENY-221/IN378. <i>EDIS</i>. March 26, 2020. Accessed January 3, 2024. https://doi.org/10.32473/edis-in378-2020<br/><br/> 6. Saleh MN, Allen KE, Lineberry MW, et al. Ticks infesting dogs and cats in North America: biology, geographic distribution, and pathogen transmission. <i>Vet Parasitol</i>. 2021;294:109392. doi:10.1016/j.vetpar.2021.109392<br/><br/> 7. Dantas-Torres F. Biology and ecology of the brown dog tick, <i>Rhipicephalus sanguineus</i>. <i>Parasit Vectors</i>. 2010;3:26. doi:10.1186/1756-3305-3-26<br/><br/> 8. Dryden MW, Payne PA. Biology and control of ticks infesting dogs and cats in North America. <i>Vet Ther</i>. 2004;5:139-154.<br/><br/> 9. Nyangiwe N, Yawa M, Muchenje V. Driving forces for changes in geographic range of cattle ticks (Acari: Ixodidae) in Africa: a Review. <i>S Afr J Anim Sci</i>. 2018;48:829. doi:10.4314/sajas.v48i5.4<br/><br/>10. Ramot Y, Zlotogorski A, Mumcuoglu KY. Brown dog tick (<i>Rhipicephalus</i><i> sanguineus</i>) infestation of the penis detected by dermoscopy. <i>Int J Dermatol</i>. 2012;51:1402-1403. doi:10.1111/j.1365-4632.2010.04756.x<br/><br/>11. Tucker NSG, Weeks ENI, Beati L, et al. Prevalence and distribution of pathogen infection and permethrin resistance in tropical and temperate populations of <i>Rhipicephalus sanguineus</i> s.l. collected worldwide. <i>Med Vet Entomol</i>. 2021;35:147-157. doi:10.1111/mve.12479<br/><br/>12. McClain MT, Sexton DJ, Hall KK, eds. Other spotted fever group rickettsial infections. <i>UpToDate</i>. Updated October 10, 2022. Accessed January 3, 2024. https://www.uptodate.com/contents/other-spotted-fever-group-rickettsial-infections</p> <p class="reference">13. Ribeiro CM, Carvalho JLB, Bastos PAS, et al. Prevalence of<span class="apple-converted-space"> </span><i>Rickettsia rickettsii</i><span class="apple-converted-space"> </span>in ticks: systematic review and meta-analysis. <i>Vector Borne Zoonotic Dis</i>. 2021;21:557-565. doi:10.1089/vbz.2021.0004<br/><br/>14. Pace EJ, O’Reilly M. Tickborne diseases: diagnosis and management. <i>Am Fam Phy</i>s<i>ician</i>. 2020;101:530-540.<br/><br/>15. Patterson JW. <i>Weedon’s Skin Pathology. </i>5th ed. Elsevier; 2020.<br/><br/>16. Dantas-Torres F. Rocky Mountain spotted fever. <i>Lancet Infect Dis</i>. 2007;7:724-732. doi:10.1016/S1473-3099(07)70261-X<br/><br/>17. Biggs HM, Behravesh CB, Bradley KK, et al. Diagnosis and management of tickborne rickettsial diseases: Rocky Mountain spotted fever and other spotted fever group rickettsioses, ehrlichioses, and anaplasmosis—United States. <i>MMWR Recomm Rep</i>. 2016;65:1-44. doi:10.15585/mmwr.rr6502a1<br/><br/>18. Rossio R, Conalbi V, Castagna V, et al. Mediterranean spotted fever and hearing impairment: a rare complication. <i>Int J Infect Dis</i>. 2015;35:34-36. doi:10.1016/j.ijid.2015.04.005<br/><br/>19. Dantas-Torres F, Otranto D. Further thoughts on the taxonomy and vector role of <i>Rhipicephalus sanguineus</i> group ticks. <i>Vet Parasitol</i>. 2015;208:9-13. doi:10.1016/j.vetpar.2014.12.014<br/><br/>20. Eisen RJ, Kugeler KJ, Eisen L, et al. Tick-borne zoonoses in the United States: persistent and emerging threats to human health. <i>ILAR J</i>. 2017;58:319-335. doi:10.1093/ilar/ilx005<br/><br/>21. Nguyen QD, Vu MN, Hebert AA. Insect repellents: an updated review for the clinician. <i>J Am Acad Dermatol</i>. 2018;88:123-130. doi:10.1016/j.jaad.2018.10.053<br/><br/>22. Pages F, Dautel H, Duvallet G, et al. Tick repellents for human use: prevention of tick bites and tick-borne diseases. <i>Vector Borne Zoonotic Dis</i>. 2014;14:85-93. doi:10.1089/vbz.2013.1410<br/><br/>23. Rodriguez-Vivas RI, Alonso-Díaz MA, et al. Prevalence and potential risk factors for organophosphate and pyrethroid resistance in <i>Boophilus microplus </i>ticks on cattle ranches from the State of Yucatan, Mexico. <i>Vet Parasitol</i>. 2006;136:335-342. <span class="citation-doi">doi:10.1016/j.vetpar.2005.05.069<br/><br/></span>24. Rodríguez-Vivas RI, Rodríguez-Arevalo F, Alonso-Díaz MA, et al. Prevalence and potential risk factors for amitraz resistance in <i>Boophilus microplus</i> ticks in cattle farms in the State of Yucatan, Mexico. <i>Prev Vet Med</i>. 2006;75:280-286. <span class="citation-doi">doi:10.1016/j.prevetmed.2006.04.001<br/><br/></span>25. Perez-Cogollo LC, Rodriguez-Vivas RI, Ramirez-Cruz GT, et al. First report of the cattle tick <i>Rhipicephalus microplus</i> resistant to ivermectin in Mexico. <i>Vet Parasitol</i>. 2010;168:165-169. doi:10.1016/j.vetpar.2009.10.021<br/><br/>26. Rodriguez-Vivas RI, Jonsson NN, Bhushan C. Strategies for the control of <i>Rhipicephalus microplus</i> ticks in a world of conventional acaricide and macrocyclic lactone resistance<i>. Parasitol Res</i>.2018;117:3-29. <span class="citation-doi">doi:10.1007/s00436-017-5677-6<br/><br/></span>27. Centers for Disease Control and Prevention. Tick removal. Updated May 13, 2022. Accessed January 3, 2024. https://www.cdc.gov/ticks/removing_a_tick.html<br/><br/>28. Diaz JH. Chemical and plant-based insect repellents: efficacy, safety, and toxicity. <i>Wilderness Environ Med</i>. 2016;27:153-163. doi:10.1016/j.wem.2015.11.007</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">From the Medical University of South Carolina, Charleston. Rebecca A. Brantley is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.</p> <p class="disclosure">The authors report no conflict of interest.<br/><br/>Correspondence: Dirk M. Elston, MD (elstond@musc.edu).doi:10.12788/cutis.0955</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>points</strong></p> <ul class="insidebody"> <li><em>Rhipicephalus </em>ticks are vectors of a variety of diseases, including the rickettsial diseases Rocky Mountain spotted fever and Mediterranean spotted fever. </li> <li>Presenting symptoms of a tick bite include intensely pruritic, erythematous papules and nodules at the site of tick attachment. </li> <li>If rickettsial disease is suspected, treatment with doxycycline should be initiated immediately; do not delay treatment to await results of confirmatory tests or because of the absence of a rash.</li> <li>Primary methods of prevention of tick-borne disease include repellents, protective clothing, vector control, and prompt removal of the tick. </li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

PRACTICE POINTS

  • Rhipicephalus ticks are vectors of a variety of diseases, including the rickettsial diseases Rocky Mountain spotted fever and Mediterranean spotted fever.
  • Presenting symptoms of a tick bite include intensely pruritic, erythematous papules and nodules at the site of tick attachment.
  • If rickettsial disease is suspected, treatment with doxycycline should be initiated immediately; do not delay treatment to await results of confirmatory tests or because of the absence of a rash.
  • Primary methods of prevention of tick-borne disease include repellents, protective clothing, vector control, and prompt removal of the tick.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
180026B8.SIG
Disable zoom
Off

Botanical Briefs: Neem Oil (Azadirachta indica)

Article Type
Changed
Tue, 01/09/2024 - 12:56
Display Headline
Botanical Briefs: Neem Oil (Azadirachta indica)

Commonly known as neem or nimba, Azadirachta indica traditionally has been used as an oil or poultice to lighten skin pigment and reduce joint inflammation. Neem is a drought-resistant evergreen tree with thin serrated leaves, white fragrant flowers, and olivelike fruit (Figure 1). This plant is indigenous to India but also is readily found within tropical and semitropical environments throughout the Middle East, Southeast Asia, North Africa, and Australia.

Patel_neem_1.jpg
%3Cp%3EFIGURE%201.%20Leaves%20of%20a%20neem%20plant%20(%3Cem%3EAzadirachta%20indica%3C%2Fem%3E).%3C%2Fp%3E

Traditional Uses

For more than 4000 years, neem leaves, bark, fruit, and seeds have been used in food, insecticide, and herbal medicine cross-culturally in Indian Ayurvedic medicine and across Southeast Asia, particularly in Cambodia, Laos, Thailand, Myanmar, and Vietnam.1-3 Because of its many essential nutrients—oleic acid, palmitic acid, stearic acid, linoleic acid, behenic acid, arachidic acid, and palmitoleic acid—and readily available nature, some ethnic groups include neem in their diet.4 Neem commonly is used as a seasoning in soups and rice, eaten as a cooked vegetable, infused into teas and tonics, and pickled with other spices.5

All parts of the neem tree—both externally and internally—have been utilized in traditional medicine for the treatment of various diseases and ailments. The flowers have been used to treat eye diseases and dyspepsia, the fruit has been employed as an anthelmintic, the seeds and leaves have been used for malaria treatment and insecticide, the stem bark has been used for the treatment of diarrhea, and the root bark has been used for skin diseases and inflammation.6 Neem oil is a yellow-brown bitter substance that often is utilized to treat skin diseases such as psoriasis, eczema, fungal infections, and abscesses.

Case Report—A 77-year-old man presented with a diffuse rash across the lower back. He reported that he had been using topical neem oil to alleviate lower back pain and arthritis for the last 6 months with noted relief and improvement of back pain. After roughly 3 to 4 months of using neem oil, he noted a rash on the lower back, bilateral flanks, and buttocks (Figure 2). The rash was asymptomatic, and he denied any pruritus, scaling, pain, or burning. The patient was referred to dermatology and received a diagnosis of chemical leukoderma secondary to contact with A indica. The patient was advised to stop using the topical neem oil, and the rash was simply monitored, as it was asymptomatic.

Patel_neem_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Hypopigmentation%20on%20the%20lower%20back%2C%20bilateral%20flanks%2C%20and%20buttocks%20due%20to%20neem%20oil%E2%80%93induced%20chemical%20leukoderma.%3C%2Fp%3E

Bioactivity

Research has elucidated multiple bioactivity mechanisms of neem, including melanogenesis-inhibitory activity, toxicity against pests, antimalarial activity, and antioxidant activity.1,7-9 Literature on the diverse phytochemical components of A indica indicate high levels of limonoids, flavonoids, and triterpenoids that are responsible for much of its antioxidant, anti-inflammatory, and insecticide properties.1,10

Melanogenesis-Inhibitory Activity—To date, neem has been added to a number of cosmetic products used in Ayurvedic medicine. One study of isolated compounds in A indica showed superior inhibitory activities against melanogenesis with minimal toxicity to cells (86.5%–105.1% cell viability). Western blot analysis of samples extracted and isolated from neem root and bark showed melanogenesis-inhibitory activities in B16 melanoma cells through the inhibition of microphthalmia-associated transcription factor expression and decreased expression of tyrosinase, as well as tyrosinase-related proteins 1 and 2, which are largely responsible for melanin synthesis.11 In another study, A indica flowers and their extracted constituents—6-deacetylnimbin and kaempferide—suggest melanogenesis-inhibitory activities in B16 melanoma cells with little to no toxicity to the cells (81.0%–111.7% cell viability).1 In an evaluationof A indica seed extracts, some of the isolated limonoids and diterpenoids exhibited a marked melanogenesis-inhibitory effect (74%–91% reduction of melanin content) with no toxicity to the cell.5 All of these studies indicate that active compounds in neem root, bark, flowers, and seeds may be potential skin-lightening agents.

Toxicity Against PestsNeem seeds have phytochemicals that convey some insecticidal properties. The seeds often are ground into a powder, combined with water, and sprayed onto crops to act as an insecticide. As a natural method of nonpesticidal management, A indica acts as an antifeedant, insect repellent, and egg-laying deterrent that protects crops from damage. Studies of A indica have noted effective nonpesticidal management against arthropod pests such as armyworm, termites, and the oriental fruit fly.7,12,13

 

 

Antimalarial Activity—One study indicated that nimbolide, a limonoid from the neem plant, demonstrated antimalarial activity against Plasmodium falciparum. In separate cultures of asexual parasites and mature gametocytes, parasite numbers were less than 50% of the number in control cultures (8.0% vs 8.5% parasitemia, respectively).14 Thus, the lower parasite numbers indicated by this study highlight the antimalarial utility of nimbolide and neem oil.

Antioxidant and Anti-inflammatory Activity—Neem bark has been reported to have considerable antioxidant activity due to its high phenolic content.1,15 One study showed that azadirachtin and nimbolide in neem exhibited concentration-dependent antiradical scavenging activity and antioxidant properties.16

The anti-inflammatory potential for neem may occur via the inhibition of the nuclear factor-κB signaling pathway, which is linked to cancer, inflammation, and apoptosis.17 It also has been observed that nimbidin within neem extracts—such as leaves, bark, and seed extract—suppresses the function of macrophages and neutrophils relevant to inflammation.16 Another study indicated neem’s anti-inflammatory activity due to the regulation of proinflammatory enzymes such as cyclooxygenase and lipoxygenase.18

Safety, Toxicity, and Risks

Ingestion—Although neem is safe to use in the general population, neem oil poisoning has been reported, particularly in young children. Ingesting large quantities of neem has resulted in vomiting, hepatic toxicity, metabolic acidosis, late neurologic sequelae, and encephalopathy in young children.19 The diagnosis of neem oil poisoning is based on patient history, clinical examination, and imaging findings. Poisoning can manifest as drowsiness, tachypnea, and generalized seizures.20

Topical Application—Topical use of neem appears to be safe if the substance is diluted with other ingredients. However, direct application to the skin is not advised, as it may cause leukoderma and could induce allergic contact dermatitis and other allergic reactions.4

Final Thoughts

The use of neem extract for disease prevention and treatment has been prevalent around the world since ancient times. Neem has been documented to possess melanogenesis-inhibitory activity, toxicity against pests, antimalarial activity, and antioxidant activity by means of tyrosinase inhibition, phytochemical production, limonoid expression, and nuclear factor-κB regulation, respectively. However, topical use of neem may trigger a cutaneous response, highlighting the importance of considering a diagnosis of neem oil–induced chemical leukoderma when patients present with a hypopigmented rash and relevant history.

References
  1. Kitdamrongtham W, Ishii K, Ebina K, et al. Limonoids and flavonoids from the flowers of Azadirachta indica var. siamensis, and their melanogenesis-inhibitory and cytotoxic activities. Chem Biodivers. 2014;11:73-84. doi:10.1002/cbdv.201300266
  2. Singh A, Srivastava PS, Lakshmikumaran M. Comparison of AFLP and SAMPL markers for assessment of intra-population genetic variation in Azadirachta indica A. Juss. Plant Sci. 2002;162:17-25. doi:10.1016/S0168-9452(01)00503-9
  3. Pandey G, Verma K, Singh M. Evaluation of phytochemical, antibacterial and free radical scavenging properties of Azadirachta Indica (neem) leaves. Int J Pharm Pharmaceut Sci. 2014;6:444-447.
  4. Romita P, Calogiuri G, Bellino M, et al. Allergic contact dermatitis caused by neem oil: an underrated allergen. Contact Dermatitis. 2019;81:133-134. doi:10.1111/cod. 13256
  5. Akihisa T, Noto T, Takahashi A, et al. Melanogenesis inhibitory, anti-inflammatory, and chemopreventive effects of limonoids from the seeds of Azadirachta indica A. Juss. (neem). J Oleo Sci. 2009;58:581-594.
  6. Subapriya R, Nagini S. Medicinal properties of neem leaves: a review. Curr Med Chem Anticancer Agents. 2005;5:149-156. doi:10.2174/1568011053174828
  7. Areekul S, Sinchaisri P, Tigvatananon S. Effect of Thai plant extracts on the Oriental fruit fly. I: toxicity test. Agriculture and Natural Resources. 1987;21:395-407.
  8. Rochanakij S, Thebtaranonth Y, Yenjai C, et al. Nimbolide, a constituent of Azadirachta indica, inhibits Plasmodium falciparum in culture. Southeast Asian J Trop Med Public Health. 1985;16:66-72.
  9. Sithisarn P, Supabphol R, Gritsanapan W. Antioxidant activity of Siamese neem tree (VP1209). J Ethnopharmacol. 2005;99:109-112. doi:10.1016/j.jep.2005.02.008
  10. Yin F, Lei XX, Cheng L, et al. Isolation and structure identification of the compounds from the seeds and leaves of Azadirachta indica A. Juss. J China Pharmaceut University. 2005;36:10-12.
  11. Su S, Cheng J, Zhang C, et al. Melanogenesis-inhibitory activities of limonoids and tricyclic diterpenoids from Azadirachta indica. Bioorganic Chemistry. 2020;100:103941. doi:j.bioorg.2020.103941
  12. Tulashie SK, Adjei F, Abraham J, et al. Potential of neem extracts as natural insecticide against fall armyworm (Spodoptera frugiperda (JE Smith)(Lepidoptera: Noctuidae). Case Stud Chem Environ Eng. 2021;4:100130. doi:10.1016/j.cscee.2021.100130
  13. Yashroy RC, Gupta PK. Neem-seed oil inhibits growth of termite surface-tunnels. Indian J Toxicol. 2000;7:49-50.
  14. Udeinya JI, Shu EN, Quakyi I, et al. An antimalarial neem leaf extract has both schizonticidal and gametocytocidal activities. Am J Therapeutics. 2008;15:108-110. doi:10.1097/MJT.0b013e31804c6d1d
  15. Bindurani R, Kumar K. Evaluation of antioxidant activity of hydro distilled extracts of leaf, heart wood and flower of Azadirachta indica. Int J Pharm Sci Rev Res. 2013;20:222.
  16. Alzohairy MA. Therapeutics role of Azadirachta indica (Neem) and their active constituents in diseases prevention and treatment [published online March 1, 2016]. Evid Based Complement Alternat Med. doi:10.1155/2016/7382506 
  17. Schumacher M, Cerella C, Reuter S, et al. Anti-inflammatory, pro-apoptotic, and anti-proliferative effects of a methanolic neem (Azadirachta indica) leaf extract are mediated via modulation of the nuclear factor-κB pathway. Genes Nutr. 2011;6:149-160. doi:10.1007/s12263-010-0194-6
  18. Kaur G, Sarwar Alam M, Athar M. Nimbidin suppresses functions of macrophages and neutrophils: relevance to its anti-inflammatory mechanisms. Phytotherapy Res. 2004;18:419-424. doi:10.1002/ptr.1474
  19. Dhongade RK, Kavade SG, Damle RS. Neem oil poisoning. Indian Pediatr. 2008;45:56-57.
  20. Bhaskar MV, Pramod SJ, Jeevika MU, et al. MR imaging findings of neem oil poisoning. Am J Neuroradiol. 2010;31:E60-E61. doi:10.3174/ajnr.A2146
Article PDF
Author and Disclosure Information

Nina Patel is from the Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois. Drs. Knabel and Speiser and from the Loyola University Medical Center, Maywood. Dr. Knabel is from the Division of Dermatology, and Dr. Speiser is from the Department of Pathology.

The authors report no conflict of interest.

Correspondence: Jodi Speiser, MD, Department of Pathology, Loyola University Medical Center, 2160 S First Ave, Maywood, IL 60153 (jspeiser@lumc.edu).

Issue
Cutis - 113(1)
Publications
Topics
Page Number
22-24
Sections
Author and Disclosure Information

Nina Patel is from the Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois. Drs. Knabel and Speiser and from the Loyola University Medical Center, Maywood. Dr. Knabel is from the Division of Dermatology, and Dr. Speiser is from the Department of Pathology.

The authors report no conflict of interest.

Correspondence: Jodi Speiser, MD, Department of Pathology, Loyola University Medical Center, 2160 S First Ave, Maywood, IL 60153 (jspeiser@lumc.edu).

Author and Disclosure Information

Nina Patel is from the Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois. Drs. Knabel and Speiser and from the Loyola University Medical Center, Maywood. Dr. Knabel is from the Division of Dermatology, and Dr. Speiser is from the Department of Pathology.

The authors report no conflict of interest.

Correspondence: Jodi Speiser, MD, Department of Pathology, Loyola University Medical Center, 2160 S First Ave, Maywood, IL 60153 (jspeiser@lumc.edu).

Article PDF
Article PDF

Commonly known as neem or nimba, Azadirachta indica traditionally has been used as an oil or poultice to lighten skin pigment and reduce joint inflammation. Neem is a drought-resistant evergreen tree with thin serrated leaves, white fragrant flowers, and olivelike fruit (Figure 1). This plant is indigenous to India but also is readily found within tropical and semitropical environments throughout the Middle East, Southeast Asia, North Africa, and Australia.

Patel_neem_1.jpg
%3Cp%3EFIGURE%201.%20Leaves%20of%20a%20neem%20plant%20(%3Cem%3EAzadirachta%20indica%3C%2Fem%3E).%3C%2Fp%3E

Traditional Uses

For more than 4000 years, neem leaves, bark, fruit, and seeds have been used in food, insecticide, and herbal medicine cross-culturally in Indian Ayurvedic medicine and across Southeast Asia, particularly in Cambodia, Laos, Thailand, Myanmar, and Vietnam.1-3 Because of its many essential nutrients—oleic acid, palmitic acid, stearic acid, linoleic acid, behenic acid, arachidic acid, and palmitoleic acid—and readily available nature, some ethnic groups include neem in their diet.4 Neem commonly is used as a seasoning in soups and rice, eaten as a cooked vegetable, infused into teas and tonics, and pickled with other spices.5

All parts of the neem tree—both externally and internally—have been utilized in traditional medicine for the treatment of various diseases and ailments. The flowers have been used to treat eye diseases and dyspepsia, the fruit has been employed as an anthelmintic, the seeds and leaves have been used for malaria treatment and insecticide, the stem bark has been used for the treatment of diarrhea, and the root bark has been used for skin diseases and inflammation.6 Neem oil is a yellow-brown bitter substance that often is utilized to treat skin diseases such as psoriasis, eczema, fungal infections, and abscesses.

Case Report—A 77-year-old man presented with a diffuse rash across the lower back. He reported that he had been using topical neem oil to alleviate lower back pain and arthritis for the last 6 months with noted relief and improvement of back pain. After roughly 3 to 4 months of using neem oil, he noted a rash on the lower back, bilateral flanks, and buttocks (Figure 2). The rash was asymptomatic, and he denied any pruritus, scaling, pain, or burning. The patient was referred to dermatology and received a diagnosis of chemical leukoderma secondary to contact with A indica. The patient was advised to stop using the topical neem oil, and the rash was simply monitored, as it was asymptomatic.

Patel_neem_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Hypopigmentation%20on%20the%20lower%20back%2C%20bilateral%20flanks%2C%20and%20buttocks%20due%20to%20neem%20oil%E2%80%93induced%20chemical%20leukoderma.%3C%2Fp%3E

Bioactivity

Research has elucidated multiple bioactivity mechanisms of neem, including melanogenesis-inhibitory activity, toxicity against pests, antimalarial activity, and antioxidant activity.1,7-9 Literature on the diverse phytochemical components of A indica indicate high levels of limonoids, flavonoids, and triterpenoids that are responsible for much of its antioxidant, anti-inflammatory, and insecticide properties.1,10

Melanogenesis-Inhibitory Activity—To date, neem has been added to a number of cosmetic products used in Ayurvedic medicine. One study of isolated compounds in A indica showed superior inhibitory activities against melanogenesis with minimal toxicity to cells (86.5%–105.1% cell viability). Western blot analysis of samples extracted and isolated from neem root and bark showed melanogenesis-inhibitory activities in B16 melanoma cells through the inhibition of microphthalmia-associated transcription factor expression and decreased expression of tyrosinase, as well as tyrosinase-related proteins 1 and 2, which are largely responsible for melanin synthesis.11 In another study, A indica flowers and their extracted constituents—6-deacetylnimbin and kaempferide—suggest melanogenesis-inhibitory activities in B16 melanoma cells with little to no toxicity to the cells (81.0%–111.7% cell viability).1 In an evaluationof A indica seed extracts, some of the isolated limonoids and diterpenoids exhibited a marked melanogenesis-inhibitory effect (74%–91% reduction of melanin content) with no toxicity to the cell.5 All of these studies indicate that active compounds in neem root, bark, flowers, and seeds may be potential skin-lightening agents.

Toxicity Against PestsNeem seeds have phytochemicals that convey some insecticidal properties. The seeds often are ground into a powder, combined with water, and sprayed onto crops to act as an insecticide. As a natural method of nonpesticidal management, A indica acts as an antifeedant, insect repellent, and egg-laying deterrent that protects crops from damage. Studies of A indica have noted effective nonpesticidal management against arthropod pests such as armyworm, termites, and the oriental fruit fly.7,12,13

 

 

Antimalarial Activity—One study indicated that nimbolide, a limonoid from the neem plant, demonstrated antimalarial activity against Plasmodium falciparum. In separate cultures of asexual parasites and mature gametocytes, parasite numbers were less than 50% of the number in control cultures (8.0% vs 8.5% parasitemia, respectively).14 Thus, the lower parasite numbers indicated by this study highlight the antimalarial utility of nimbolide and neem oil.

Antioxidant and Anti-inflammatory Activity—Neem bark has been reported to have considerable antioxidant activity due to its high phenolic content.1,15 One study showed that azadirachtin and nimbolide in neem exhibited concentration-dependent antiradical scavenging activity and antioxidant properties.16

The anti-inflammatory potential for neem may occur via the inhibition of the nuclear factor-κB signaling pathway, which is linked to cancer, inflammation, and apoptosis.17 It also has been observed that nimbidin within neem extracts—such as leaves, bark, and seed extract—suppresses the function of macrophages and neutrophils relevant to inflammation.16 Another study indicated neem’s anti-inflammatory activity due to the regulation of proinflammatory enzymes such as cyclooxygenase and lipoxygenase.18

Safety, Toxicity, and Risks

Ingestion—Although neem is safe to use in the general population, neem oil poisoning has been reported, particularly in young children. Ingesting large quantities of neem has resulted in vomiting, hepatic toxicity, metabolic acidosis, late neurologic sequelae, and encephalopathy in young children.19 The diagnosis of neem oil poisoning is based on patient history, clinical examination, and imaging findings. Poisoning can manifest as drowsiness, tachypnea, and generalized seizures.20

Topical Application—Topical use of neem appears to be safe if the substance is diluted with other ingredients. However, direct application to the skin is not advised, as it may cause leukoderma and could induce allergic contact dermatitis and other allergic reactions.4

Final Thoughts

The use of neem extract for disease prevention and treatment has been prevalent around the world since ancient times. Neem has been documented to possess melanogenesis-inhibitory activity, toxicity against pests, antimalarial activity, and antioxidant activity by means of tyrosinase inhibition, phytochemical production, limonoid expression, and nuclear factor-κB regulation, respectively. However, topical use of neem may trigger a cutaneous response, highlighting the importance of considering a diagnosis of neem oil–induced chemical leukoderma when patients present with a hypopigmented rash and relevant history.

Commonly known as neem or nimba, Azadirachta indica traditionally has been used as an oil or poultice to lighten skin pigment and reduce joint inflammation. Neem is a drought-resistant evergreen tree with thin serrated leaves, white fragrant flowers, and olivelike fruit (Figure 1). This plant is indigenous to India but also is readily found within tropical and semitropical environments throughout the Middle East, Southeast Asia, North Africa, and Australia.

Patel_neem_1.jpg
%3Cp%3EFIGURE%201.%20Leaves%20of%20a%20neem%20plant%20(%3Cem%3EAzadirachta%20indica%3C%2Fem%3E).%3C%2Fp%3E

Traditional Uses

For more than 4000 years, neem leaves, bark, fruit, and seeds have been used in food, insecticide, and herbal medicine cross-culturally in Indian Ayurvedic medicine and across Southeast Asia, particularly in Cambodia, Laos, Thailand, Myanmar, and Vietnam.1-3 Because of its many essential nutrients—oleic acid, palmitic acid, stearic acid, linoleic acid, behenic acid, arachidic acid, and palmitoleic acid—and readily available nature, some ethnic groups include neem in their diet.4 Neem commonly is used as a seasoning in soups and rice, eaten as a cooked vegetable, infused into teas and tonics, and pickled with other spices.5

All parts of the neem tree—both externally and internally—have been utilized in traditional medicine for the treatment of various diseases and ailments. The flowers have been used to treat eye diseases and dyspepsia, the fruit has been employed as an anthelmintic, the seeds and leaves have been used for malaria treatment and insecticide, the stem bark has been used for the treatment of diarrhea, and the root bark has been used for skin diseases and inflammation.6 Neem oil is a yellow-brown bitter substance that often is utilized to treat skin diseases such as psoriasis, eczema, fungal infections, and abscesses.

Case Report—A 77-year-old man presented with a diffuse rash across the lower back. He reported that he had been using topical neem oil to alleviate lower back pain and arthritis for the last 6 months with noted relief and improvement of back pain. After roughly 3 to 4 months of using neem oil, he noted a rash on the lower back, bilateral flanks, and buttocks (Figure 2). The rash was asymptomatic, and he denied any pruritus, scaling, pain, or burning. The patient was referred to dermatology and received a diagnosis of chemical leukoderma secondary to contact with A indica. The patient was advised to stop using the topical neem oil, and the rash was simply monitored, as it was asymptomatic.

Patel_neem_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Hypopigmentation%20on%20the%20lower%20back%2C%20bilateral%20flanks%2C%20and%20buttocks%20due%20to%20neem%20oil%E2%80%93induced%20chemical%20leukoderma.%3C%2Fp%3E

Bioactivity

Research has elucidated multiple bioactivity mechanisms of neem, including melanogenesis-inhibitory activity, toxicity against pests, antimalarial activity, and antioxidant activity.1,7-9 Literature on the diverse phytochemical components of A indica indicate high levels of limonoids, flavonoids, and triterpenoids that are responsible for much of its antioxidant, anti-inflammatory, and insecticide properties.1,10

Melanogenesis-Inhibitory Activity—To date, neem has been added to a number of cosmetic products used in Ayurvedic medicine. One study of isolated compounds in A indica showed superior inhibitory activities against melanogenesis with minimal toxicity to cells (86.5%–105.1% cell viability). Western blot analysis of samples extracted and isolated from neem root and bark showed melanogenesis-inhibitory activities in B16 melanoma cells through the inhibition of microphthalmia-associated transcription factor expression and decreased expression of tyrosinase, as well as tyrosinase-related proteins 1 and 2, which are largely responsible for melanin synthesis.11 In another study, A indica flowers and their extracted constituents—6-deacetylnimbin and kaempferide—suggest melanogenesis-inhibitory activities in B16 melanoma cells with little to no toxicity to the cells (81.0%–111.7% cell viability).1 In an evaluationof A indica seed extracts, some of the isolated limonoids and diterpenoids exhibited a marked melanogenesis-inhibitory effect (74%–91% reduction of melanin content) with no toxicity to the cell.5 All of these studies indicate that active compounds in neem root, bark, flowers, and seeds may be potential skin-lightening agents.

Toxicity Against PestsNeem seeds have phytochemicals that convey some insecticidal properties. The seeds often are ground into a powder, combined with water, and sprayed onto crops to act as an insecticide. As a natural method of nonpesticidal management, A indica acts as an antifeedant, insect repellent, and egg-laying deterrent that protects crops from damage. Studies of A indica have noted effective nonpesticidal management against arthropod pests such as armyworm, termites, and the oriental fruit fly.7,12,13

 

 

Antimalarial Activity—One study indicated that nimbolide, a limonoid from the neem plant, demonstrated antimalarial activity against Plasmodium falciparum. In separate cultures of asexual parasites and mature gametocytes, parasite numbers were less than 50% of the number in control cultures (8.0% vs 8.5% parasitemia, respectively).14 Thus, the lower parasite numbers indicated by this study highlight the antimalarial utility of nimbolide and neem oil.

Antioxidant and Anti-inflammatory Activity—Neem bark has been reported to have considerable antioxidant activity due to its high phenolic content.1,15 One study showed that azadirachtin and nimbolide in neem exhibited concentration-dependent antiradical scavenging activity and antioxidant properties.16

The anti-inflammatory potential for neem may occur via the inhibition of the nuclear factor-κB signaling pathway, which is linked to cancer, inflammation, and apoptosis.17 It also has been observed that nimbidin within neem extracts—such as leaves, bark, and seed extract—suppresses the function of macrophages and neutrophils relevant to inflammation.16 Another study indicated neem’s anti-inflammatory activity due to the regulation of proinflammatory enzymes such as cyclooxygenase and lipoxygenase.18

Safety, Toxicity, and Risks

Ingestion—Although neem is safe to use in the general population, neem oil poisoning has been reported, particularly in young children. Ingesting large quantities of neem has resulted in vomiting, hepatic toxicity, metabolic acidosis, late neurologic sequelae, and encephalopathy in young children.19 The diagnosis of neem oil poisoning is based on patient history, clinical examination, and imaging findings. Poisoning can manifest as drowsiness, tachypnea, and generalized seizures.20

Topical Application—Topical use of neem appears to be safe if the substance is diluted with other ingredients. However, direct application to the skin is not advised, as it may cause leukoderma and could induce allergic contact dermatitis and other allergic reactions.4

Final Thoughts

The use of neem extract for disease prevention and treatment has been prevalent around the world since ancient times. Neem has been documented to possess melanogenesis-inhibitory activity, toxicity against pests, antimalarial activity, and antioxidant activity by means of tyrosinase inhibition, phytochemical production, limonoid expression, and nuclear factor-κB regulation, respectively. However, topical use of neem may trigger a cutaneous response, highlighting the importance of considering a diagnosis of neem oil–induced chemical leukoderma when patients present with a hypopigmented rash and relevant history.

References
  1. Kitdamrongtham W, Ishii K, Ebina K, et al. Limonoids and flavonoids from the flowers of Azadirachta indica var. siamensis, and their melanogenesis-inhibitory and cytotoxic activities. Chem Biodivers. 2014;11:73-84. doi:10.1002/cbdv.201300266
  2. Singh A, Srivastava PS, Lakshmikumaran M. Comparison of AFLP and SAMPL markers for assessment of intra-population genetic variation in Azadirachta indica A. Juss. Plant Sci. 2002;162:17-25. doi:10.1016/S0168-9452(01)00503-9
  3. Pandey G, Verma K, Singh M. Evaluation of phytochemical, antibacterial and free radical scavenging properties of Azadirachta Indica (neem) leaves. Int J Pharm Pharmaceut Sci. 2014;6:444-447.
  4. Romita P, Calogiuri G, Bellino M, et al. Allergic contact dermatitis caused by neem oil: an underrated allergen. Contact Dermatitis. 2019;81:133-134. doi:10.1111/cod. 13256
  5. Akihisa T, Noto T, Takahashi A, et al. Melanogenesis inhibitory, anti-inflammatory, and chemopreventive effects of limonoids from the seeds of Azadirachta indica A. Juss. (neem). J Oleo Sci. 2009;58:581-594.
  6. Subapriya R, Nagini S. Medicinal properties of neem leaves: a review. Curr Med Chem Anticancer Agents. 2005;5:149-156. doi:10.2174/1568011053174828
  7. Areekul S, Sinchaisri P, Tigvatananon S. Effect of Thai plant extracts on the Oriental fruit fly. I: toxicity test. Agriculture and Natural Resources. 1987;21:395-407.
  8. Rochanakij S, Thebtaranonth Y, Yenjai C, et al. Nimbolide, a constituent of Azadirachta indica, inhibits Plasmodium falciparum in culture. Southeast Asian J Trop Med Public Health. 1985;16:66-72.
  9. Sithisarn P, Supabphol R, Gritsanapan W. Antioxidant activity of Siamese neem tree (VP1209). J Ethnopharmacol. 2005;99:109-112. doi:10.1016/j.jep.2005.02.008
  10. Yin F, Lei XX, Cheng L, et al. Isolation and structure identification of the compounds from the seeds and leaves of Azadirachta indica A. Juss. J China Pharmaceut University. 2005;36:10-12.
  11. Su S, Cheng J, Zhang C, et al. Melanogenesis-inhibitory activities of limonoids and tricyclic diterpenoids from Azadirachta indica. Bioorganic Chemistry. 2020;100:103941. doi:j.bioorg.2020.103941
  12. Tulashie SK, Adjei F, Abraham J, et al. Potential of neem extracts as natural insecticide against fall armyworm (Spodoptera frugiperda (JE Smith)(Lepidoptera: Noctuidae). Case Stud Chem Environ Eng. 2021;4:100130. doi:10.1016/j.cscee.2021.100130
  13. Yashroy RC, Gupta PK. Neem-seed oil inhibits growth of termite surface-tunnels. Indian J Toxicol. 2000;7:49-50.
  14. Udeinya JI, Shu EN, Quakyi I, et al. An antimalarial neem leaf extract has both schizonticidal and gametocytocidal activities. Am J Therapeutics. 2008;15:108-110. doi:10.1097/MJT.0b013e31804c6d1d
  15. Bindurani R, Kumar K. Evaluation of antioxidant activity of hydro distilled extracts of leaf, heart wood and flower of Azadirachta indica. Int J Pharm Sci Rev Res. 2013;20:222.
  16. Alzohairy MA. Therapeutics role of Azadirachta indica (Neem) and their active constituents in diseases prevention and treatment [published online March 1, 2016]. Evid Based Complement Alternat Med. doi:10.1155/2016/7382506 
  17. Schumacher M, Cerella C, Reuter S, et al. Anti-inflammatory, pro-apoptotic, and anti-proliferative effects of a methanolic neem (Azadirachta indica) leaf extract are mediated via modulation of the nuclear factor-κB pathway. Genes Nutr. 2011;6:149-160. doi:10.1007/s12263-010-0194-6
  18. Kaur G, Sarwar Alam M, Athar M. Nimbidin suppresses functions of macrophages and neutrophils: relevance to its anti-inflammatory mechanisms. Phytotherapy Res. 2004;18:419-424. doi:10.1002/ptr.1474
  19. Dhongade RK, Kavade SG, Damle RS. Neem oil poisoning. Indian Pediatr. 2008;45:56-57.
  20. Bhaskar MV, Pramod SJ, Jeevika MU, et al. MR imaging findings of neem oil poisoning. Am J Neuroradiol. 2010;31:E60-E61. doi:10.3174/ajnr.A2146
References
  1. Kitdamrongtham W, Ishii K, Ebina K, et al. Limonoids and flavonoids from the flowers of Azadirachta indica var. siamensis, and their melanogenesis-inhibitory and cytotoxic activities. Chem Biodivers. 2014;11:73-84. doi:10.1002/cbdv.201300266
  2. Singh A, Srivastava PS, Lakshmikumaran M. Comparison of AFLP and SAMPL markers for assessment of intra-population genetic variation in Azadirachta indica A. Juss. Plant Sci. 2002;162:17-25. doi:10.1016/S0168-9452(01)00503-9
  3. Pandey G, Verma K, Singh M. Evaluation of phytochemical, antibacterial and free radical scavenging properties of Azadirachta Indica (neem) leaves. Int J Pharm Pharmaceut Sci. 2014;6:444-447.
  4. Romita P, Calogiuri G, Bellino M, et al. Allergic contact dermatitis caused by neem oil: an underrated allergen. Contact Dermatitis. 2019;81:133-134. doi:10.1111/cod. 13256
  5. Akihisa T, Noto T, Takahashi A, et al. Melanogenesis inhibitory, anti-inflammatory, and chemopreventive effects of limonoids from the seeds of Azadirachta indica A. Juss. (neem). J Oleo Sci. 2009;58:581-594.
  6. Subapriya R, Nagini S. Medicinal properties of neem leaves: a review. Curr Med Chem Anticancer Agents. 2005;5:149-156. doi:10.2174/1568011053174828
  7. Areekul S, Sinchaisri P, Tigvatananon S. Effect of Thai plant extracts on the Oriental fruit fly. I: toxicity test. Agriculture and Natural Resources. 1987;21:395-407.
  8. Rochanakij S, Thebtaranonth Y, Yenjai C, et al. Nimbolide, a constituent of Azadirachta indica, inhibits Plasmodium falciparum in culture. Southeast Asian J Trop Med Public Health. 1985;16:66-72.
  9. Sithisarn P, Supabphol R, Gritsanapan W. Antioxidant activity of Siamese neem tree (VP1209). J Ethnopharmacol. 2005;99:109-112. doi:10.1016/j.jep.2005.02.008
  10. Yin F, Lei XX, Cheng L, et al. Isolation and structure identification of the compounds from the seeds and leaves of Azadirachta indica A. Juss. J China Pharmaceut University. 2005;36:10-12.
  11. Su S, Cheng J, Zhang C, et al. Melanogenesis-inhibitory activities of limonoids and tricyclic diterpenoids from Azadirachta indica. Bioorganic Chemistry. 2020;100:103941. doi:j.bioorg.2020.103941
  12. Tulashie SK, Adjei F, Abraham J, et al. Potential of neem extracts as natural insecticide against fall armyworm (Spodoptera frugiperda (JE Smith)(Lepidoptera: Noctuidae). Case Stud Chem Environ Eng. 2021;4:100130. doi:10.1016/j.cscee.2021.100130
  13. Yashroy RC, Gupta PK. Neem-seed oil inhibits growth of termite surface-tunnels. Indian J Toxicol. 2000;7:49-50.
  14. Udeinya JI, Shu EN, Quakyi I, et al. An antimalarial neem leaf extract has both schizonticidal and gametocytocidal activities. Am J Therapeutics. 2008;15:108-110. doi:10.1097/MJT.0b013e31804c6d1d
  15. Bindurani R, Kumar K. Evaluation of antioxidant activity of hydro distilled extracts of leaf, heart wood and flower of Azadirachta indica. Int J Pharm Sci Rev Res. 2013;20:222.
  16. Alzohairy MA. Therapeutics role of Azadirachta indica (Neem) and their active constituents in diseases prevention and treatment [published online March 1, 2016]. Evid Based Complement Alternat Med. doi:10.1155/2016/7382506 
  17. Schumacher M, Cerella C, Reuter S, et al. Anti-inflammatory, pro-apoptotic, and anti-proliferative effects of a methanolic neem (Azadirachta indica) leaf extract are mediated via modulation of the nuclear factor-κB pathway. Genes Nutr. 2011;6:149-160. doi:10.1007/s12263-010-0194-6
  18. Kaur G, Sarwar Alam M, Athar M. Nimbidin suppresses functions of macrophages and neutrophils: relevance to its anti-inflammatory mechanisms. Phytotherapy Res. 2004;18:419-424. doi:10.1002/ptr.1474
  19. Dhongade RK, Kavade SG, Damle RS. Neem oil poisoning. Indian Pediatr. 2008;45:56-57.
  20. Bhaskar MV, Pramod SJ, Jeevika MU, et al. MR imaging findings of neem oil poisoning. Am J Neuroradiol. 2010;31:E60-E61. doi:10.3174/ajnr.A2146
Issue
Cutis - 113(1)
Issue
Cutis - 113(1)
Page Number
22-24
Page Number
22-24
Publications
Publications
Topics
Article Type
Display Headline
Botanical Briefs: Neem Oil (Azadirachta indica)
Display Headline
Botanical Briefs: Neem Oil (Azadirachta indica)
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Patel neem</fileName> <TBEID>0C02EFD2.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02EFD2</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Close Encounters</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240108T111552</firstPublished> <LastPublished>20240108T111552</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240108T111551</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Patel</byline> <bylineText>Nina Patel, MS; Michael Knabel, MD; Jodi Speiser, MD</bylineText> <bylineFull>Patel</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>22-24</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Commonly known as neem or nimba, Azadirachta indica traditionally has been used as an oil or poultice to lighten skin pigment and reduce joint inflammation. Nee</metaDescription> <articlePDF>299907</articlePDF> <teaserImage/> <title>Botanical Briefs: Neem Oil (Azadirachta indica)</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>January</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>1</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2271</CMSID> <CMSID>2159</CMSID> </CMSIDs> <keywords> <keyword>pigmentation disorder</keyword> <keyword> neem oil</keyword> <keyword> azadirachta indica</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>January 2024</pubIssueName> <pubArticleType>Departments | 2159</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Close Encounters with the Environment | 2271<pubSubsection/></pubSection> </pubSections> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">60</term> </sections> <topics> <term canonical="true">276</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/1800269e.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Botanical Briefs: Neem Oil (Azadirachta indica)</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><i>Azadirachta indica,</i> commonly known as neem, has many uses as a natural remedy. We review and discuss the pharmacologic, biologic, and medicinal properties of neem in disease management. We also report a rare clinical case of a 77-year-old man who presented with a hypopigmented rash on the lower back, bilateral flanks, and buttocks after 6 months of repeated application of neem oil to treat persistent arthritis and lower back pain.</p> <p> <em><i>Cutis</i>. 2024;113:22-24.</em> </p> <p>Commonly known as neem or nimba, <i>Azadirachta indica</i> traditionally has been used as an oil or poultice to lighten skin pigment and reduce joint inflammation. Neem is a drought-resistant evergreen tree with thin serrated leaves, white fragrant flowers, and olivelike fruit (Figure 1). This plant is indigenous to India but also is readily found within tropical and semitropical environments throughout the Middle East, Southeast Asia, North Africa, and Australia.</p> <h3>Traditional Uses</h3> <p>For more than 4000 years, neem leaves, bark, fruit, and seeds have been used in food, insecticide, and herbal medicine cross-culturally in Indian Ayurvedic medicine and across Southeast Asia, particularly in Cambodia, Laos, Thailand, Myanmar, and Vietnam.<sup>1-3</sup> Because of its many essential nutrients—oleic acid, palmitic acid, stearic acid, linoleic acid, behenic acid, arachidic acid, and palmitoleic acid—and readily available nature, some ethnic groups include neem in their diet.<sup>4</sup> Neem commonly is used as a seasoning in soups and rice, eaten as a cooked vegetable, infused into teas and tonics, and pickled with other spices.<sup>5</sup> </p> <p>All parts of the neem tree—both externally and internally—have been utilized in traditional medicine for the treatment of various diseases and ailments. The flowers have been used to treat eye diseases and dyspepsia, the fruit has been employed as an anthelmintic, the seeds and leaves have been used for malaria treatment and insecticide, the stem bark has been used for the treatment of diarrhea, and the root bark has been used for skin diseases and inflammation.<sup>6</sup> Neem oil is a yellow-brown bitter substance that often is utilized to treat skin diseases such as psoriasis, eczema, fungal infections, and abscesses. <br/><br/><span class="sub3">Case Report—</span>A 77-year-old man presented with a diffuse rash across the lower back. He reported that he had been using topical neem oil to alleviate lower back pain and arthritis for the last 6 months with noted relief and improvement of back pain. After roughly 3 to 4 months of using neem oil, he noted a rash on the lower back, bilateral flanks, and buttocks (Figure 2). The rash was asymptomatic, and he denied any pruritus, scaling, pain, or burning. The patient was referred to dermatology and received a diagnosis of chemical leukoderma secondary to contact with <i>A indica</i>. The patient was advised to stop using the topical neem oil, and the rash was simply monitored, as it was asymptomatic. </p> <h3>Bioactivity</h3> <p>Research has elucidated multiple bioactivity mechanisms of neem, including melanogenesis-inhibitory activity, toxicity against pests, antimalarial activity, and antioxidant activity.<sup>1,7-9</sup> Literature on the diverse phytochemical components of <i>A indica </i>indicate high levels of limonoids, flavonoids, and triterpenoids that are responsible for much of its antioxidant, anti-inflammatory, and insecticide properties.<sup>1,10</sup> </p> <p><span class="sub3">Melanogenesis-Inhibitory Activity</span>—To date, neem has been added to a number of cosmetic products used in Ayurvedic medicine. One study of isolated compounds in <i>A indica </i>showed superior inhibitory activities against melanogenesis with minimal toxicity to cells (86.5%–105.1% cell viability). Western blot analysis of samples extracted and isolated from neem root and bark showed melanogenesis-inhibitory activities in B16 melanoma cells through the inhibition of microphthalmia-associated transcription factor expression and decreased expression of tyrosinase, as well as tyrosinase-related proteins 1 and 2, which are largely responsible for melanin synthesis.<sup>11</sup> In another study, <i>A indica</i> flowers and their extracted constituents—6-deacetylnimbin and kaempferide—suggest melanogenesis-inhibitory activities in B16 melanoma cells with little to no toxicity to the cells (81.0%–111.7% cell viability).<sup>1</sup> In an evaluationof <i>A indica</i> seed extracts, some of the isolated limonoids and diterpenoids exhibited a marked melanogenesis-inhibitory effect (74%–91% reduction of melanin content) with no toxicity to the cell.<sup>5</sup> All of these studies indicate that active compounds in neem root, bark, flowers, and seeds may be potential skin-lightening agents. <br/><br/><span class="sub3">Toxicity Against Pests</span><i>—</i>Neem seeds have phytochemicals that convey some insecticidal properties. The seeds often are ground into a powder, combined with water, and sprayed onto crops to act as an insecticide. As a natural method of nonpesticidal management, <i>A indica</i> acts as an antifeedant, insect repellent, and egg-laying deterrent that protects crops from damage. Studies of <i>A indica </i>have noted effective nonpesticidal management against arthropod pests such as armyworm, termites, and the oriental fruit fly.<sup>7,12,13</sup> <br/><br/><span class="sub3">Antimalarial Activity</span>—One study indicated that nimbolide, a limonoid from the neem plant, demonstrated antimalarial activity against <i>Plasmodium falciparum</i>. In separate cultures of asexual parasites and mature gametocytes, parasite numbers were less than 50% of the number in control cultures (8.0% vs 8.5% parasitemia, respectively).<sup>14</sup> Thus, the lower parasite numbers indicated by this study highlight the antimalarial utility of nimbolide and neem oil. <br/><br/><span class="sub3">Antioxidant and Anti-inflammatory Activity</span>—Neem bark has been reported to have considerable antioxidant activity due to its high phenolic content.<sup>1,15</sup> One study showed that azadirachtin and nimbolide in neem exhibited concentration-dependent antiradical scavenging activity and antioxidant properties.<sup>16</sup> <br/><br/>The anti-inflammatory potential for neem may occur via the inhibition of the nuclear factor-κB signaling pathway, which is linked to cancer, inflammation, and apoptosis.<sup>17</sup> It also has been observed that nimbidin within neem extracts—such as leaves, bark, and seed extract—suppresses the function of macrophages and neutrophils relevant to inflammation.<sup>16</sup> Another study indicated neem’s anti-inflammatory activity due to the regulation of proinflammatory enzymes such as cyclooxygenase and lipoxygenase.<sup>18</sup></p> <h3>Safety, Toxicity, and Risks</h3> <p><span class="sub3">Ingestion</span>—Although neem is safe to use in the general population, neem oil poisoning has been reported, particularly in young children. Ingesting large quantities of neem has resulted in vomiting, hepatic toxicity, metabolic acidosis, late neurologic sequelae, and encephalopathy in young children.<sup>19</sup> The diagnosis of neem oil poisoning is based on patient history, clinical examination, and imaging findings. Poisoning can manifest as drowsiness, tachypnea, and generalized seizures.<sup>20</sup> </p> <p><span class="sub3">Topical Application</span>—Topical use of neem appears to be safe if the substance is diluted with other ingredients. However, direct application to the skin is not advised, as it may cause leukoderma and could induce allergic contact dermatitis and other allergic reactions.<sup>4</sup></p> <h3>Final Thoughts</h3> <p>The use of neem extract for disease prevention and treatment has been prevalent around the world since ancient times. Neem has been documented to possess melanogenesis-inhibitory activity, toxicity against pests, antimalarial activity, and antioxidant activity by means of tyrosinase inhibition, phytochemical production, limonoid expression, and nuclear factor-κB regulation, respectively. However, topical use of neem may trigger a cutaneous response, highlighting the importance of considering a diagnosis of neem oil–induced chemical leukoderma when patients present with a hypopigmented rash and relevant history.</p> <h2>References</h2> <p class="reference"> 1. Kitdamrongtham W, Ishii K, Ebina K, et al. Limonoids and flavonoids from the flowers of <i>Azadirachta indica</i> var. siamensis, and their melanogenesis-inhibitory and cytotoxic activities. <i>Chem Biodivers.</i> 2014;11:73-84. doi:10.1002/cbdv.201300266<br/><br/> 2. Singh A, Srivastava PS, Lakshmikumaran M. Comparison of AFLP and SAMPL markers for assessment of intra-population genetic variation in <i>Azadirachta indica</i> A. Juss.<i> Plant Sci.</i> 2002;162:17-25. doi:10.1016/S0168-9452(01)00503-9<br/><br/> 3. Pandey G, Verma K, Singh M. Evaluation of phytochemical, antibacterial and free radical scavenging properties of <i>Azadirachta Indica</i> (neem) leaves. <i>Int J Pharm Pharmaceut Sci.</i> 2014;6:444-447.<br/><br/> 4. Romita P, Calogiuri G, Bellino M, et al. Allergic contact dermatitis caused by neem oil: an underrated allergen. <i>Contact Dermatitis</i>. 2019;81:133-134. doi:10.1111/cod. 13256<br/><br/> 5. Akihisa T, Noto T, Takahashi A, et al. Melanogenesis inhibitory, anti-inflammatory, and chemopreventive effects of limonoids from the seeds of <i>Azadirachta indica</i> A. Juss. (neem). <i>J Oleo Sci</i>. 2009;58:581-594.<br/><br/> 6. Subapriya R, Nagini S. Medicinal properties of neem leaves: a review. <i>Curr Med Chem Anticancer Agents</i>. 2005;5:149-156. doi:10.2174/1568011053174828<br/><br/> 7. Areekul S, Sinchaisri P, Tigvatananon S. Effect of Thai plant extracts on the Oriental fruit fly. I: toxicity test. <i>Agriculture and Natural Resources</i>. 1987;21:395-407.<br/><br/> 8. Rochanakij S, Thebtaranonth Y, Yenjai C, et al. Nimbolide, a constituent of <i>Azadirachta indica</i>, inhibits <em>Plasmodium falciparum</em> in culture. <i>Southeast Asian J Trop Med Public Health</i>. 1985;16:66-72.<br/><br/> 9. Sithisarn P, Supabphol R, Gritsanapan W. Antioxidant activity of Siamese neem tree (VP1209). <i>J Ethnopharmacol</i>. 2005;99:109-112. doi:10.1016/j.jep.2005.02.008<br/><br/>10. Yin F, Lei XX, Cheng L, et al. Isolation and structure identification of the compounds from the seeds and leaves of <i>Azadirachta indica</i> A. Juss. <i>J China Pharmaceut University</i>. 2005;36:10-12.<br/><br/>11. Su S, Cheng J, Zhang C, et al. Melanogenesis-inhibitory activities of limonoids and tricyclic diterpenoids from <i>Azadirachta indica</i>. <i>Bioorganic Chemistry</i>. 2020;100:103941. doi:j.bioorg.2020.103941<br/><br/>12. Tulashie SK, Adjei F, Abraham J, et al. Potential of neem extracts as natural insecticide against fall armyworm (<i>Spodoptera frugiperda</i> (JE Smith)(Lepidoptera: Noctuidae). <i>Case Stud Chem Environ Eng</i>. 2021;4:100130. doi:10.1016/j.cscee.2021.100130<br/><br/>13. Yashroy RC, Gupta PK. Neem-seed oil inhibits growth of termite surface-tunnels. <i>Indian J Toxicol</i>. 2000;7:49-50.<br/><br/>14. Udeinya JI, Shu EN, Quakyi I, et al. An antimalarial neem leaf extract has both schizonticidal and gametocytocidal activities. <i>Am J Therapeutics</i>. 2008;15:108-110. doi:10.1097/MJT.0b013e31804c6d1d<br/><br/>15. Bindurani R, Kumar K. Evaluation of antioxidant activity of hydro distilled extracts of leaf, heart wood and flower of <i>Azadirachta indica</i>. <i>Int J Pharm Sci Rev Res.</i> 2013;20:222.<br/><br/>16. Alzohairy MA. Therapeutics role of <i>Azadirachta indica</i> (Neem) and their active constituents in diseases prevention and treatment [published online March 1, 2016]. <i>Evid Based Complement Alternat Med</i>. doi:10.1155/2016/7382506 <br/><br/>17. Schumacher M, Cerella C, Reuter S, et al. Anti-inflammatory, pro-apoptotic, and anti-proliferative effects of a methanolic neem (<i>Azadirachta indica</i>) leaf extract are mediated via modulation of the nuclear factor-κB pathway. <i>Genes Nutr</i>. 2011;6:149-160. doi:10.1007/s12263-010-0194-6<br/><br/>18. Kaur G, Sarwar Alam M, Athar M. Nimbidin suppresses functions of macrophages and neutrophils: relevance to its anti-inflammatory mechanisms. <i>Phytotherapy Res. </i>2004;18:419-424. doi:10.1002/ptr.1474<br/><br/>19. Dhongade RK, Kavade SG, Damle RS. Neem oil poisoning. <i>Indian Pediatr. </i>2008;45:56-57.<br/><br/>20. Bhaskar MV, Pramod SJ, Jeevika MU, et al. MR imaging findings of neem oil poisoning. <i>Am J Neuroradiol</i>. 2010;31:E60-E61. doi:10.3174/ajnr.A2146</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Nina Patel is from the Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois. Drs. Knabel and Speiser and from the Loyola University Medical Center, Maywood. Dr. Knabel is from the Division of Dermatology, and Dr. Speiser is from the Department of Pathology. </p> <p class="disclosure">The authors report no conflict of interest. <br/><br/>Correspondence: Jodi Speiser, MD, Department of Pathology, Loyola University Medical Center, 2160 S First Ave, Maywood, IL 60153 (jspeiser@lumc.edu).<br/><br/>doi:10.12788/cutis.0928</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li> Neem is a traditional herb with various bioactivities, such as melanogenesis-inhibitory activity, toxicity against pests, antimalarial activity, and antioxidant activity.</li> <li> Neem should be used with caution as a remedy because of its skin-lightening properties, which are attributed to melanogenesis-inhibitory activity via tyrosinase inhibition.</li> <li> Chemical leukoderma should be included in the differential diagnosis when a patient presents with a hypopigmented rash after topical use of neem products.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • Neem is a traditional herb with various bioactivities, such as melanogenesis-inhibitory activity, toxicity against pests, antimalarial activity, and antioxidant activity.
  • Neem should be used with caution as a remedy because of its skin-lightening properties, which are attributed to melanogenesis-inhibitory activity via tyrosinase inhibition.
  • Chemical leukoderma should be included in the differential diagnosis when a patient presents with a hypopigmented rash after topical use of neem products.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
1800269E.SIG
Disable zoom
Off

Botanical Briefs: Contact Dermatitis Induced by Western Poison Ivy (Toxicodendron rydbergii)

Article Type
Changed
Thu, 01/04/2024 - 14:49
Display Headline
Botanical Briefs: Contact Dermatitis Induced by Western Poison Ivy (Toxicodendron rydbergii)

Clinical Importance

Western poison ivy (Toxicodendron rydbergii) is responsible for many of the cases of Toxicodendron contact dermatitis (TCD) reported in the western and northern United States. Toxicodendron plants cause more cases of allergic contact dermatitis (ACD) in North America than any other allergen1; 9 million Americans present to physician offices and 1.6 million present to emergency departments annually for ACD, emphasizing the notable medical burden of this condition.2,3 Exposure to urushiol, a plant resin containing potent allergens, precipitates this form of ACD.

An estimated 50% to 75% of adults in the United States demonstrate clinical sensitivity and exhibit ACD following contact with T rydbergii.4 Campers, hikers, firefighters, and forest workers often risk increased exposure through physical contact or aerosolized allergens in smoke. According to the Centers for Disease Control and Prevention, the incidence of visits to US emergency departments for TCD nearly doubled from 2002 to 2012,5 which may be explained by atmospheric CO2 levels that both promote increased growth of Toxicodendron species and augment their toxicity.6

Cutaneous Manifestations

The clinical presentation of T rydbergii contact dermatitis is similar to other allergenic members of the Toxicodendron genus. Patients sensitive to urushiol typically develop a pruritic erythematous rash within 1 to 2 days of exposure (range, 5 hours to 15 days).7 Erythematous and edematous streaks initially manifest on the extremities and often progress to bullae and oozing papulovesicles. In early disease, patients also may display black lesions on or near the rash8 (so-called black-dot dermatitis) caused by oxidized urushiol deposited on the skin—an uncommon yet classic presentation of TCD. Generally, symptoms resolve without complications and with few sequalae, though hyperpigmentation or a secondary infection can develop on or near affected areas.9,10

Taxonomy

The Toxicodendron genus belongs to the Anacardiaceae family, which includes pistachios, mangos, and cashews, and causes more cases of ACD than every other plant combined.4 (Shelled pistachios and cashews do not possess cross-reacting allergens and should not worry consumers; mango skin does contain urushiol.)

Toxicodendron (formerly part of the Rhus genus) includes several species of poison oak, poison ivy, and poison sumac and can be found in shrubs (T rydbergii and Toxicodendron diversilobum), vines (Toxicodendron radicans and Toxicodendron pubescens), and trees (Toxicodendron vernix). In addition, Toxicodendron taxa can hybridize with other taxa in close geographic proximity to form morphologic intermediates. Some individual plants have features of multiple species.11

Etymology

The common name of T rydbergii—western poison ivy—misleads the public; the plant contains no poison that can cause death and does not grow as ivy by wrapping around trees, as T radicans and English ivy (Hedera helix) do. Its formal genus, Toxicodendron, means “poison tree” in Greek and was given its generic name by the English botanist Phillip Miller in 1768,12 which caused the renaming of Rhus rydbergii as T rydbergii. The species name honors Per Axel Rydberg, a 19th and 20th century Swedish-American botanist.

Distribution

Toxicodendron rydbergii grows in California and other states in the western half of the United States as well as the states bordering Canada and Mexico. In Canada, it reigns as the most dominant form of poison ivy.13 Hikers and campers find T rydbergii in a variety of areas, including roadsides, river bottoms, sandy shores, talus slopes, precipices, and floodplains.11 This taxon grows under a variety of conditions and in distinct regions, and it thrives in both full sun or shade.

 

 

Identifying Features

Toxicodendron rydbergii turns red earlier than most plants; early red summer leaves should serve as a warning sign to hikers from a distance (Figure 1). It displays trifoliate ovate leaves (ie, each leaf contains 3 leaflets) on a dwarf nonclimbing shrub (Figure 2). Although the plant shares common features with its cousin T radicans (eastern poison ivy), T rydbergii is easily distinguished by its thicker stems, absence of aerial rootlets (abundant in T radicans), and short (approximately 1 meter) height.4

Afvari_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Hiker%E2%80%99s%20view%20of%20red%20leaves%20on%20a%20western%20poison%20ivy%20shrub%20(%3Cem%3EToxicodendron%20rydbergii%3C%2Fem%3E)(photographed%20from%20a%20distance%20of%203%20meters)%20in%20Spearfish%20Canyon%2C%20South%20Dakota.%3C%2Fp%3E

Curly hairs occupy the underside of T rydbergii leaflets and along the midrib; leaflet margins appear lobed or rounded. Lenticels appear as small holes in the bark that turn gray in the cold and become brighter come spring.13

Afvari_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Five%20characteristic%20features%20for%20identifying%20western%20poison%20ivy%20(%3Cem%3EToxicodendron%20rydbergii%3C%2Fem%3E)%3A%20(1)%20leaves%20with%203%20leaflets%3B%20(2)%20a%20low-growing%2C%20nonclimbing%20habitat%3B%20(3)%20early%20autumn%20colors%20starting%20in%20summer%3B%20(4)%20lack%20of%20deposits%20of%20oxidized%20urushiol%3B%20and%20(5)%20drupes%2C%20or%20fruit%20(arrows)%2C%20where%20the%20petiole%20meets%20the%20branch%20or%20root%20(Spearfish%20Canyon%2C%20South%20Dakota).%3C%2Fp%3E

The plant bears glabrous long petioles (leaf stems) and densely grouped clusters of yellow flowers. In autumn, the globose fruit—formed in clusters between each twig and leaf petiole (known as an axillary position)—change from yellow-green to tan (Figure 3). When urushiol exudes from damaged leaflets or other plant parts, it oxidizes on exposure to air and creates hardened black deposits on the plant. Even when grown in garden pots, T rydbergii maintains its distinguishing features.11

Afvari_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Mature%20fruit%20of%20%3Cem%3EToxicodendron%20rydbergii%3C%2Fem%3E%20in%20winter.%3C%2Fp%3E

Dermatitis-Inducing Plant Parts

All parts of T rydbergii including leaves, stems, roots, and fruit contain the allergenic sap throughout the year.14 A person must damage or bruise the plant for urushiol to be released and produce its allergenic effects; softly brushing against undamaged plants typically does not induce dermatitis.4

Pathophysiology of Urushiol

Urushiol, a pale yellow, oily mixture of organic compounds conserved throughout all Toxicodendron species, contains highly allergenic alkyl catechols. These catechols possess hydroxyl groups at positions 1 and 2 on a benzene ring; the hydrocarbon side chain of poison ivies (typically 15–carbon atoms long) attaches at position 3.15 The catechols and the aliphatic side chain contribute to the plant’s antigenic and dermatitis-inducing properties.16

The high lipophilicity of urushiol allows for rapid and unforgiving absorption into the skin, notwithstanding attempts to wash it off. Upon direct contact, catechols of urushiol penetrate the epidermis and become oxidized to quinone intermediates that bind to antigen-presenting cells in the epidermis and dermis. Epidermal Langerhans cells and dermal macrophages internalize and present the antigen to CD4+ T cells in nearby lymph nodes. This sequence results in production of inflammatory mediators, clonal expansion of T-effector and T-memory cells specific to the allergenic catechols, and an ensuing cytotoxic response against epidermal cells and the dermal vasculature. Keratinocytes and monocytes mediate the inflammatory response by releasing other cytokines.4,17

Sensitization to urushiol generally occurs at 8 to 14 years of age; therefore, infants have lower susceptibility to dermatitis upon contact with T rydbergii.18 Most animals do not experience sensitization upon contact; in fact, birds and forest animals consume the urushiol-rich fruit of T rydbergii without harm.3

 

 

Prevention and Treatment

Toxicodendron dermatitis typically lasts 1 to 3 weeks but can remain for as long as 6 weeks without treatment.19 Recognition and physical avoidance of the plant provides the most promising preventive strategy. Immediate rinsing with soap and water can prevent TCD by breaking down urushiol and its allergenic components; however, this is an option for only a short time, as the skin absorbs 50% of urushiol within 10 minutes after contact.20 Nevertheless, patients must seize the earliest opportunity to wash off the affected area and remove any residual urushiol. Patients must be cautious when removing and washing clothing to prevent further contact.

Most health care providers treat TCD with a corticosteroid to reduce inflammation and intense pruritus. A high-potency topical corticosteroid (eg, clobetasol) may prove effective in providing early therapeutic relief in mild disease.21 A short course of a systemic steroid quickly and effectively quenches intense itching and should not be limited to what the clinician considers severe disease. Do not underestimate the patient’s symptoms with this eruption.

Prednisone dosing begins at 1 mg/kg daily and is then tapered slowly over 2 weeks (no shorter a time) for an optimal treatment course of 15 days.22 Prescribing an inadequate dosage and course of a corticosteroid leaves the patient susceptible to rebound dermatitis—and loss of trust in their provider.

Intramuscular injection of the long-acting corticosteroid triamcinolone acetonide with rapid-onset betamethasone provides rapid relief and fewer adverse effects than an oral corticosteroid.22 Despite the long-standing use of sedating oral antihistamines by clinicians, these drugs provide no benefit for pruritus or sleep because the histamine does not cause the itching of TCD, and antihistamines disrupt normal sleep architecture.23-25

Patients can consider several over-the-counter products that have varying degrees of efficacy.4,26 The few products for which prospective studies support their use include Tecnu (Tec Laboraties Inc), Zanfel (RhusTox), and the well-known soaps Dial (Henkel Corporation) and Goop (Critzas Industries, Inc).27,28

Aside from treating the direct effects of TCD, clinicians also must take note of any look for signs of secondary infection and occasionally should consider supplementing treatment with an antibiotic.

References
  1. Lofgran T, Mahabal GD. Toxicodendron toxicity. StatPearls [Internet]. Updated May 16, 2023. Accessed December 23, 2023. https://www.ncbi.nlm.nih.gov/books/NBK557866/
  2. The Lewin Group. The Burden of Skin Diseases 2005. Society for Investigative Dermatology and American Academy of Dermatology Association; 2005:37-40. Accessed December 26, 2023. https://www.lewin.com/content/dam/Lewin/Resources/Site_Sections/Publications/april2005skindisease.pdf
  3. Monroe J. Toxicodendron contact dermatitis: a case report and brief review. J Clin Aesthet Dermatol. 2020;13(9 Suppl 1):S29-S34.
  4. Gladman AC. Toxicodendron dermatitis: poison ivy, oak, and sumac. Wilderness Environ Med. 2006;17:120-128. doi:10.1580/pr31-05.1
  5. Fretwell S. Poison ivy cases on the rise. The State. Updated May 15,2017. Accessed December 26, 2023. https://www.thestate.com/news/local/article150403932.html
  6. Mohan JE, Ziska LH, Schlesinger WH, et al. Biomass and toxicity responses of poison ivy (Toxicodendron radicans) to elevated atmospheric CO2Proc Natl Acad Sci U S A. 2006;103:9086-9089. doi:10.1073/pnas.0602392103
  7. Williams JV, Light J, Marks JG Jr. Individual variations in allergic contact dermatitis from urushiol. Arch Dermatol. 1999;135:1002-1003. doi:10.1001/archderm.135.8.1002
  8. Kurlan JG, Lucky AW. Black spot poison ivy: a report of 5 cases and a review of the literature. J Am Acad Dermatol. 2001;45:246-249. doi:10.1067/mjd.2001.114295
  9. Fisher AA. Poison ivy/oak/sumac. part II: specific features. Cutis. 1996;58:22-24.
  10. Brook I, Frazier EH, Yeager JK. Microbiology of infected poison ivy dermatitis. Br J Dermatol. 2000;142:943-946. doi:10.1046/j.1365-2133.2000.03475.x
  11. Gillis WT. The systematics and ecology of poison-ivy and the poison-oaks (Toxicodendron, Anacardiaceae). Rhodora. 1971;73:370-443.
  12. Reveal JL. Typification of six Philip Miller names of temperate North American Toxicodendron (Anacardiaceae) with proposals (999-1000) to reject T. crenatum and T. volubileTAXON. 1991;40:333-335. doi:10.2307/1222994 
  13. Guin JD, Gillis WT, Beaman JH. Recognizing the Toxicodendrons (poison ivy, poison oak, and poison sumac). J Am Acad Dermatol. 1981;4:99-114. doi:10.1016/s0190-9622(81)70014-8
  14. Lee NP, Arriola ER. Poison ivy, oak, and sumac dermatitis. West J Med. 1999;171:354-355.
  15. Marks JG Jr, Anderson BE, DeLeo VA, eds. Contact and Occupational Dermatology. Jaypee Brothers Medical Publishers Ltd; 2016.
  16. Dawson CR. The chemistry of poison ivy. Trans N Y Acad Sci. 1956;18:427-443. doi:10.1111/j.2164-0947.1956.tb00465.x
  17. Kalish RS. Recent developments in the pathogenesis of allergic contact dermatitis. Arch Dermatol. 1991;127:1558-1563.
  18. Fisher AA, Mitchell J. Toxicodendron plants and spices. In: Rietschel RL, Fowler JF Jr. Fisher’s Contact Dermatitis. 4th ed. Williams & Wilkins; 1995:461-523.
  19. Labib A, Yosipovitch G. Itchy Toxicodendron plant dermatitis. Allergies. 2022;2:16-22. doi:10.3390/allergies2010002 
  20. Fisher AA. Poison ivy/oak dermatitis part I: prevention—soap and water, topical barriers, hyposensitization. Cutis. 1996;57:384-386.
  21. Kim Y, Flamm A, ElSohly MA, et al. Poison ivy, oak, and sumac dermatitis: what is known and what is new? 2019;30:183-190. doi:10.1097/DER.0000000000000472
  22. Prok L, McGovern T. Poison ivy (Toxicodendron) dermatitis. UpToDate. Updated October 16, 2023. Accessed December 26, 2023. https://www.uptodate.com/contents/poison-ivy-toxicodendron-dermatitis
  23. Klein PA, Clark RA. An evidence-based review of the efficacy of antihistamines in relieving pruritus in atopic dermatitis. Arch Dermatol. 1999;135:1522-1525. doi:10.1001/archderm.135.12.1522
  24. He A, Feldman SR, Fleischer AB Jr. An assessment of the use of antihistamines in the management of atopic dermatitis. J Am Acad Dermatol. 2018;79:92-96. doi:10.1016/j.jaad.2017.12.077
  25. van Zuuren EJ, Apfelbacher CJ, Fedorowicz Z, et al. No high level evidence to support the use of oral H1 antihistamines as monotherapy for eczema: a summary of a Cochrane systematic review. Syst Rev. 2014;3:25. doi:10.1186/2046-4053-3-25
  26. Neill BC, Neill JA, Brauker J, et al. Postexposure prevention of Toxicodendron dermatitis by early forceful unidirectional washing with liquid dishwashing soap. J Am Acad Dermatol. 2019;81:E25. doi:10.1016/j.jaad.2017.12.081
  27. Stibich AS, Yagan M, Sharma V, et al. Cost-effective post-exposure prevention of poison ivy dermatitis. Int J Dermatol. 2000;39:515-518. doi:10.1046/j.1365-4362.2000.00003.x
  28. Davila A, Laurora M, Fulton J, et al. A new topical agent, Zanfel, ameliorates urushiol-induced Toxicodendron allergic contact dermatitis [abstract]. Ann Emerg Med. 2003;42:S98.
Article PDF
Author and Disclosure Information

Shawn Afvari is from New York Medical College School of Medicine, Valhalla. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.

The authors report no conflict of interest.

Correspondence: Shawn Afvari, BS (safvari@student.nymc.edu).

Issue
Cutis - 113(1)
Publications
Topics
Page Number
E11-E14
Sections
Author and Disclosure Information

Shawn Afvari is from New York Medical College School of Medicine, Valhalla. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.

The authors report no conflict of interest.

Correspondence: Shawn Afvari, BS (safvari@student.nymc.edu).

Author and Disclosure Information

Shawn Afvari is from New York Medical College School of Medicine, Valhalla. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.

The authors report no conflict of interest.

Correspondence: Shawn Afvari, BS (safvari@student.nymc.edu).

Article PDF
Article PDF

Clinical Importance

Western poison ivy (Toxicodendron rydbergii) is responsible for many of the cases of Toxicodendron contact dermatitis (TCD) reported in the western and northern United States. Toxicodendron plants cause more cases of allergic contact dermatitis (ACD) in North America than any other allergen1; 9 million Americans present to physician offices and 1.6 million present to emergency departments annually for ACD, emphasizing the notable medical burden of this condition.2,3 Exposure to urushiol, a plant resin containing potent allergens, precipitates this form of ACD.

An estimated 50% to 75% of adults in the United States demonstrate clinical sensitivity and exhibit ACD following contact with T rydbergii.4 Campers, hikers, firefighters, and forest workers often risk increased exposure through physical contact or aerosolized allergens in smoke. According to the Centers for Disease Control and Prevention, the incidence of visits to US emergency departments for TCD nearly doubled from 2002 to 2012,5 which may be explained by atmospheric CO2 levels that both promote increased growth of Toxicodendron species and augment their toxicity.6

Cutaneous Manifestations

The clinical presentation of T rydbergii contact dermatitis is similar to other allergenic members of the Toxicodendron genus. Patients sensitive to urushiol typically develop a pruritic erythematous rash within 1 to 2 days of exposure (range, 5 hours to 15 days).7 Erythematous and edematous streaks initially manifest on the extremities and often progress to bullae and oozing papulovesicles. In early disease, patients also may display black lesions on or near the rash8 (so-called black-dot dermatitis) caused by oxidized urushiol deposited on the skin—an uncommon yet classic presentation of TCD. Generally, symptoms resolve without complications and with few sequalae, though hyperpigmentation or a secondary infection can develop on or near affected areas.9,10

Taxonomy

The Toxicodendron genus belongs to the Anacardiaceae family, which includes pistachios, mangos, and cashews, and causes more cases of ACD than every other plant combined.4 (Shelled pistachios and cashews do not possess cross-reacting allergens and should not worry consumers; mango skin does contain urushiol.)

Toxicodendron (formerly part of the Rhus genus) includes several species of poison oak, poison ivy, and poison sumac and can be found in shrubs (T rydbergii and Toxicodendron diversilobum), vines (Toxicodendron radicans and Toxicodendron pubescens), and trees (Toxicodendron vernix). In addition, Toxicodendron taxa can hybridize with other taxa in close geographic proximity to form morphologic intermediates. Some individual plants have features of multiple species.11

Etymology

The common name of T rydbergii—western poison ivy—misleads the public; the plant contains no poison that can cause death and does not grow as ivy by wrapping around trees, as T radicans and English ivy (Hedera helix) do. Its formal genus, Toxicodendron, means “poison tree” in Greek and was given its generic name by the English botanist Phillip Miller in 1768,12 which caused the renaming of Rhus rydbergii as T rydbergii. The species name honors Per Axel Rydberg, a 19th and 20th century Swedish-American botanist.

Distribution

Toxicodendron rydbergii grows in California and other states in the western half of the United States as well as the states bordering Canada and Mexico. In Canada, it reigns as the most dominant form of poison ivy.13 Hikers and campers find T rydbergii in a variety of areas, including roadsides, river bottoms, sandy shores, talus slopes, precipices, and floodplains.11 This taxon grows under a variety of conditions and in distinct regions, and it thrives in both full sun or shade.

 

 

Identifying Features

Toxicodendron rydbergii turns red earlier than most plants; early red summer leaves should serve as a warning sign to hikers from a distance (Figure 1). It displays trifoliate ovate leaves (ie, each leaf contains 3 leaflets) on a dwarf nonclimbing shrub (Figure 2). Although the plant shares common features with its cousin T radicans (eastern poison ivy), T rydbergii is easily distinguished by its thicker stems, absence of aerial rootlets (abundant in T radicans), and short (approximately 1 meter) height.4

Afvari_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Hiker%E2%80%99s%20view%20of%20red%20leaves%20on%20a%20western%20poison%20ivy%20shrub%20(%3Cem%3EToxicodendron%20rydbergii%3C%2Fem%3E)(photographed%20from%20a%20distance%20of%203%20meters)%20in%20Spearfish%20Canyon%2C%20South%20Dakota.%3C%2Fp%3E

Curly hairs occupy the underside of T rydbergii leaflets and along the midrib; leaflet margins appear lobed or rounded. Lenticels appear as small holes in the bark that turn gray in the cold and become brighter come spring.13

Afvari_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Five%20characteristic%20features%20for%20identifying%20western%20poison%20ivy%20(%3Cem%3EToxicodendron%20rydbergii%3C%2Fem%3E)%3A%20(1)%20leaves%20with%203%20leaflets%3B%20(2)%20a%20low-growing%2C%20nonclimbing%20habitat%3B%20(3)%20early%20autumn%20colors%20starting%20in%20summer%3B%20(4)%20lack%20of%20deposits%20of%20oxidized%20urushiol%3B%20and%20(5)%20drupes%2C%20or%20fruit%20(arrows)%2C%20where%20the%20petiole%20meets%20the%20branch%20or%20root%20(Spearfish%20Canyon%2C%20South%20Dakota).%3C%2Fp%3E

The plant bears glabrous long petioles (leaf stems) and densely grouped clusters of yellow flowers. In autumn, the globose fruit—formed in clusters between each twig and leaf petiole (known as an axillary position)—change from yellow-green to tan (Figure 3). When urushiol exudes from damaged leaflets or other plant parts, it oxidizes on exposure to air and creates hardened black deposits on the plant. Even when grown in garden pots, T rydbergii maintains its distinguishing features.11

Afvari_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Mature%20fruit%20of%20%3Cem%3EToxicodendron%20rydbergii%3C%2Fem%3E%20in%20winter.%3C%2Fp%3E

Dermatitis-Inducing Plant Parts

All parts of T rydbergii including leaves, stems, roots, and fruit contain the allergenic sap throughout the year.14 A person must damage or bruise the plant for urushiol to be released and produce its allergenic effects; softly brushing against undamaged plants typically does not induce dermatitis.4

Pathophysiology of Urushiol

Urushiol, a pale yellow, oily mixture of organic compounds conserved throughout all Toxicodendron species, contains highly allergenic alkyl catechols. These catechols possess hydroxyl groups at positions 1 and 2 on a benzene ring; the hydrocarbon side chain of poison ivies (typically 15–carbon atoms long) attaches at position 3.15 The catechols and the aliphatic side chain contribute to the plant’s antigenic and dermatitis-inducing properties.16

The high lipophilicity of urushiol allows for rapid and unforgiving absorption into the skin, notwithstanding attempts to wash it off. Upon direct contact, catechols of urushiol penetrate the epidermis and become oxidized to quinone intermediates that bind to antigen-presenting cells in the epidermis and dermis. Epidermal Langerhans cells and dermal macrophages internalize and present the antigen to CD4+ T cells in nearby lymph nodes. This sequence results in production of inflammatory mediators, clonal expansion of T-effector and T-memory cells specific to the allergenic catechols, and an ensuing cytotoxic response against epidermal cells and the dermal vasculature. Keratinocytes and monocytes mediate the inflammatory response by releasing other cytokines.4,17

Sensitization to urushiol generally occurs at 8 to 14 years of age; therefore, infants have lower susceptibility to dermatitis upon contact with T rydbergii.18 Most animals do not experience sensitization upon contact; in fact, birds and forest animals consume the urushiol-rich fruit of T rydbergii without harm.3

 

 

Prevention and Treatment

Toxicodendron dermatitis typically lasts 1 to 3 weeks but can remain for as long as 6 weeks without treatment.19 Recognition and physical avoidance of the plant provides the most promising preventive strategy. Immediate rinsing with soap and water can prevent TCD by breaking down urushiol and its allergenic components; however, this is an option for only a short time, as the skin absorbs 50% of urushiol within 10 minutes after contact.20 Nevertheless, patients must seize the earliest opportunity to wash off the affected area and remove any residual urushiol. Patients must be cautious when removing and washing clothing to prevent further contact.

Most health care providers treat TCD with a corticosteroid to reduce inflammation and intense pruritus. A high-potency topical corticosteroid (eg, clobetasol) may prove effective in providing early therapeutic relief in mild disease.21 A short course of a systemic steroid quickly and effectively quenches intense itching and should not be limited to what the clinician considers severe disease. Do not underestimate the patient’s symptoms with this eruption.

Prednisone dosing begins at 1 mg/kg daily and is then tapered slowly over 2 weeks (no shorter a time) for an optimal treatment course of 15 days.22 Prescribing an inadequate dosage and course of a corticosteroid leaves the patient susceptible to rebound dermatitis—and loss of trust in their provider.

Intramuscular injection of the long-acting corticosteroid triamcinolone acetonide with rapid-onset betamethasone provides rapid relief and fewer adverse effects than an oral corticosteroid.22 Despite the long-standing use of sedating oral antihistamines by clinicians, these drugs provide no benefit for pruritus or sleep because the histamine does not cause the itching of TCD, and antihistamines disrupt normal sleep architecture.23-25

Patients can consider several over-the-counter products that have varying degrees of efficacy.4,26 The few products for which prospective studies support their use include Tecnu (Tec Laboraties Inc), Zanfel (RhusTox), and the well-known soaps Dial (Henkel Corporation) and Goop (Critzas Industries, Inc).27,28

Aside from treating the direct effects of TCD, clinicians also must take note of any look for signs of secondary infection and occasionally should consider supplementing treatment with an antibiotic.

Clinical Importance

Western poison ivy (Toxicodendron rydbergii) is responsible for many of the cases of Toxicodendron contact dermatitis (TCD) reported in the western and northern United States. Toxicodendron plants cause more cases of allergic contact dermatitis (ACD) in North America than any other allergen1; 9 million Americans present to physician offices and 1.6 million present to emergency departments annually for ACD, emphasizing the notable medical burden of this condition.2,3 Exposure to urushiol, a plant resin containing potent allergens, precipitates this form of ACD.

An estimated 50% to 75% of adults in the United States demonstrate clinical sensitivity and exhibit ACD following contact with T rydbergii.4 Campers, hikers, firefighters, and forest workers often risk increased exposure through physical contact or aerosolized allergens in smoke. According to the Centers for Disease Control and Prevention, the incidence of visits to US emergency departments for TCD nearly doubled from 2002 to 2012,5 which may be explained by atmospheric CO2 levels that both promote increased growth of Toxicodendron species and augment their toxicity.6

Cutaneous Manifestations

The clinical presentation of T rydbergii contact dermatitis is similar to other allergenic members of the Toxicodendron genus. Patients sensitive to urushiol typically develop a pruritic erythematous rash within 1 to 2 days of exposure (range, 5 hours to 15 days).7 Erythematous and edematous streaks initially manifest on the extremities and often progress to bullae and oozing papulovesicles. In early disease, patients also may display black lesions on or near the rash8 (so-called black-dot dermatitis) caused by oxidized urushiol deposited on the skin—an uncommon yet classic presentation of TCD. Generally, symptoms resolve without complications and with few sequalae, though hyperpigmentation or a secondary infection can develop on or near affected areas.9,10

Taxonomy

The Toxicodendron genus belongs to the Anacardiaceae family, which includes pistachios, mangos, and cashews, and causes more cases of ACD than every other plant combined.4 (Shelled pistachios and cashews do not possess cross-reacting allergens and should not worry consumers; mango skin does contain urushiol.)

Toxicodendron (formerly part of the Rhus genus) includes several species of poison oak, poison ivy, and poison sumac and can be found in shrubs (T rydbergii and Toxicodendron diversilobum), vines (Toxicodendron radicans and Toxicodendron pubescens), and trees (Toxicodendron vernix). In addition, Toxicodendron taxa can hybridize with other taxa in close geographic proximity to form morphologic intermediates. Some individual plants have features of multiple species.11

Etymology

The common name of T rydbergii—western poison ivy—misleads the public; the plant contains no poison that can cause death and does not grow as ivy by wrapping around trees, as T radicans and English ivy (Hedera helix) do. Its formal genus, Toxicodendron, means “poison tree” in Greek and was given its generic name by the English botanist Phillip Miller in 1768,12 which caused the renaming of Rhus rydbergii as T rydbergii. The species name honors Per Axel Rydberg, a 19th and 20th century Swedish-American botanist.

Distribution

Toxicodendron rydbergii grows in California and other states in the western half of the United States as well as the states bordering Canada and Mexico. In Canada, it reigns as the most dominant form of poison ivy.13 Hikers and campers find T rydbergii in a variety of areas, including roadsides, river bottoms, sandy shores, talus slopes, precipices, and floodplains.11 This taxon grows under a variety of conditions and in distinct regions, and it thrives in both full sun or shade.

 

 

Identifying Features

Toxicodendron rydbergii turns red earlier than most plants; early red summer leaves should serve as a warning sign to hikers from a distance (Figure 1). It displays trifoliate ovate leaves (ie, each leaf contains 3 leaflets) on a dwarf nonclimbing shrub (Figure 2). Although the plant shares common features with its cousin T radicans (eastern poison ivy), T rydbergii is easily distinguished by its thicker stems, absence of aerial rootlets (abundant in T radicans), and short (approximately 1 meter) height.4

Afvari_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Hiker%E2%80%99s%20view%20of%20red%20leaves%20on%20a%20western%20poison%20ivy%20shrub%20(%3Cem%3EToxicodendron%20rydbergii%3C%2Fem%3E)(photographed%20from%20a%20distance%20of%203%20meters)%20in%20Spearfish%20Canyon%2C%20South%20Dakota.%3C%2Fp%3E

Curly hairs occupy the underside of T rydbergii leaflets and along the midrib; leaflet margins appear lobed or rounded. Lenticels appear as small holes in the bark that turn gray in the cold and become brighter come spring.13

Afvari_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Five%20characteristic%20features%20for%20identifying%20western%20poison%20ivy%20(%3Cem%3EToxicodendron%20rydbergii%3C%2Fem%3E)%3A%20(1)%20leaves%20with%203%20leaflets%3B%20(2)%20a%20low-growing%2C%20nonclimbing%20habitat%3B%20(3)%20early%20autumn%20colors%20starting%20in%20summer%3B%20(4)%20lack%20of%20deposits%20of%20oxidized%20urushiol%3B%20and%20(5)%20drupes%2C%20or%20fruit%20(arrows)%2C%20where%20the%20petiole%20meets%20the%20branch%20or%20root%20(Spearfish%20Canyon%2C%20South%20Dakota).%3C%2Fp%3E

The plant bears glabrous long petioles (leaf stems) and densely grouped clusters of yellow flowers. In autumn, the globose fruit—formed in clusters between each twig and leaf petiole (known as an axillary position)—change from yellow-green to tan (Figure 3). When urushiol exudes from damaged leaflets or other plant parts, it oxidizes on exposure to air and creates hardened black deposits on the plant. Even when grown in garden pots, T rydbergii maintains its distinguishing features.11

Afvari_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Mature%20fruit%20of%20%3Cem%3EToxicodendron%20rydbergii%3C%2Fem%3E%20in%20winter.%3C%2Fp%3E

Dermatitis-Inducing Plant Parts

All parts of T rydbergii including leaves, stems, roots, and fruit contain the allergenic sap throughout the year.14 A person must damage or bruise the plant for urushiol to be released and produce its allergenic effects; softly brushing against undamaged plants typically does not induce dermatitis.4

Pathophysiology of Urushiol

Urushiol, a pale yellow, oily mixture of organic compounds conserved throughout all Toxicodendron species, contains highly allergenic alkyl catechols. These catechols possess hydroxyl groups at positions 1 and 2 on a benzene ring; the hydrocarbon side chain of poison ivies (typically 15–carbon atoms long) attaches at position 3.15 The catechols and the aliphatic side chain contribute to the plant’s antigenic and dermatitis-inducing properties.16

The high lipophilicity of urushiol allows for rapid and unforgiving absorption into the skin, notwithstanding attempts to wash it off. Upon direct contact, catechols of urushiol penetrate the epidermis and become oxidized to quinone intermediates that bind to antigen-presenting cells in the epidermis and dermis. Epidermal Langerhans cells and dermal macrophages internalize and present the antigen to CD4+ T cells in nearby lymph nodes. This sequence results in production of inflammatory mediators, clonal expansion of T-effector and T-memory cells specific to the allergenic catechols, and an ensuing cytotoxic response against epidermal cells and the dermal vasculature. Keratinocytes and monocytes mediate the inflammatory response by releasing other cytokines.4,17

Sensitization to urushiol generally occurs at 8 to 14 years of age; therefore, infants have lower susceptibility to dermatitis upon contact with T rydbergii.18 Most animals do not experience sensitization upon contact; in fact, birds and forest animals consume the urushiol-rich fruit of T rydbergii without harm.3

 

 

Prevention and Treatment

Toxicodendron dermatitis typically lasts 1 to 3 weeks but can remain for as long as 6 weeks without treatment.19 Recognition and physical avoidance of the plant provides the most promising preventive strategy. Immediate rinsing with soap and water can prevent TCD by breaking down urushiol and its allergenic components; however, this is an option for only a short time, as the skin absorbs 50% of urushiol within 10 minutes after contact.20 Nevertheless, patients must seize the earliest opportunity to wash off the affected area and remove any residual urushiol. Patients must be cautious when removing and washing clothing to prevent further contact.

Most health care providers treat TCD with a corticosteroid to reduce inflammation and intense pruritus. A high-potency topical corticosteroid (eg, clobetasol) may prove effective in providing early therapeutic relief in mild disease.21 A short course of a systemic steroid quickly and effectively quenches intense itching and should not be limited to what the clinician considers severe disease. Do not underestimate the patient’s symptoms with this eruption.

Prednisone dosing begins at 1 mg/kg daily and is then tapered slowly over 2 weeks (no shorter a time) for an optimal treatment course of 15 days.22 Prescribing an inadequate dosage and course of a corticosteroid leaves the patient susceptible to rebound dermatitis—and loss of trust in their provider.

Intramuscular injection of the long-acting corticosteroid triamcinolone acetonide with rapid-onset betamethasone provides rapid relief and fewer adverse effects than an oral corticosteroid.22 Despite the long-standing use of sedating oral antihistamines by clinicians, these drugs provide no benefit for pruritus or sleep because the histamine does not cause the itching of TCD, and antihistamines disrupt normal sleep architecture.23-25

Patients can consider several over-the-counter products that have varying degrees of efficacy.4,26 The few products for which prospective studies support their use include Tecnu (Tec Laboraties Inc), Zanfel (RhusTox), and the well-known soaps Dial (Henkel Corporation) and Goop (Critzas Industries, Inc).27,28

Aside from treating the direct effects of TCD, clinicians also must take note of any look for signs of secondary infection and occasionally should consider supplementing treatment with an antibiotic.

References
  1. Lofgran T, Mahabal GD. Toxicodendron toxicity. StatPearls [Internet]. Updated May 16, 2023. Accessed December 23, 2023. https://www.ncbi.nlm.nih.gov/books/NBK557866/
  2. The Lewin Group. The Burden of Skin Diseases 2005. Society for Investigative Dermatology and American Academy of Dermatology Association; 2005:37-40. Accessed December 26, 2023. https://www.lewin.com/content/dam/Lewin/Resources/Site_Sections/Publications/april2005skindisease.pdf
  3. Monroe J. Toxicodendron contact dermatitis: a case report and brief review. J Clin Aesthet Dermatol. 2020;13(9 Suppl 1):S29-S34.
  4. Gladman AC. Toxicodendron dermatitis: poison ivy, oak, and sumac. Wilderness Environ Med. 2006;17:120-128. doi:10.1580/pr31-05.1
  5. Fretwell S. Poison ivy cases on the rise. The State. Updated May 15,2017. Accessed December 26, 2023. https://www.thestate.com/news/local/article150403932.html
  6. Mohan JE, Ziska LH, Schlesinger WH, et al. Biomass and toxicity responses of poison ivy (Toxicodendron radicans) to elevated atmospheric CO2Proc Natl Acad Sci U S A. 2006;103:9086-9089. doi:10.1073/pnas.0602392103
  7. Williams JV, Light J, Marks JG Jr. Individual variations in allergic contact dermatitis from urushiol. Arch Dermatol. 1999;135:1002-1003. doi:10.1001/archderm.135.8.1002
  8. Kurlan JG, Lucky AW. Black spot poison ivy: a report of 5 cases and a review of the literature. J Am Acad Dermatol. 2001;45:246-249. doi:10.1067/mjd.2001.114295
  9. Fisher AA. Poison ivy/oak/sumac. part II: specific features. Cutis. 1996;58:22-24.
  10. Brook I, Frazier EH, Yeager JK. Microbiology of infected poison ivy dermatitis. Br J Dermatol. 2000;142:943-946. doi:10.1046/j.1365-2133.2000.03475.x
  11. Gillis WT. The systematics and ecology of poison-ivy and the poison-oaks (Toxicodendron, Anacardiaceae). Rhodora. 1971;73:370-443.
  12. Reveal JL. Typification of six Philip Miller names of temperate North American Toxicodendron (Anacardiaceae) with proposals (999-1000) to reject T. crenatum and T. volubileTAXON. 1991;40:333-335. doi:10.2307/1222994 
  13. Guin JD, Gillis WT, Beaman JH. Recognizing the Toxicodendrons (poison ivy, poison oak, and poison sumac). J Am Acad Dermatol. 1981;4:99-114. doi:10.1016/s0190-9622(81)70014-8
  14. Lee NP, Arriola ER. Poison ivy, oak, and sumac dermatitis. West J Med. 1999;171:354-355.
  15. Marks JG Jr, Anderson BE, DeLeo VA, eds. Contact and Occupational Dermatology. Jaypee Brothers Medical Publishers Ltd; 2016.
  16. Dawson CR. The chemistry of poison ivy. Trans N Y Acad Sci. 1956;18:427-443. doi:10.1111/j.2164-0947.1956.tb00465.x
  17. Kalish RS. Recent developments in the pathogenesis of allergic contact dermatitis. Arch Dermatol. 1991;127:1558-1563.
  18. Fisher AA, Mitchell J. Toxicodendron plants and spices. In: Rietschel RL, Fowler JF Jr. Fisher’s Contact Dermatitis. 4th ed. Williams & Wilkins; 1995:461-523.
  19. Labib A, Yosipovitch G. Itchy Toxicodendron plant dermatitis. Allergies. 2022;2:16-22. doi:10.3390/allergies2010002 
  20. Fisher AA. Poison ivy/oak dermatitis part I: prevention—soap and water, topical barriers, hyposensitization. Cutis. 1996;57:384-386.
  21. Kim Y, Flamm A, ElSohly MA, et al. Poison ivy, oak, and sumac dermatitis: what is known and what is new? 2019;30:183-190. doi:10.1097/DER.0000000000000472
  22. Prok L, McGovern T. Poison ivy (Toxicodendron) dermatitis. UpToDate. Updated October 16, 2023. Accessed December 26, 2023. https://www.uptodate.com/contents/poison-ivy-toxicodendron-dermatitis
  23. Klein PA, Clark RA. An evidence-based review of the efficacy of antihistamines in relieving pruritus in atopic dermatitis. Arch Dermatol. 1999;135:1522-1525. doi:10.1001/archderm.135.12.1522
  24. He A, Feldman SR, Fleischer AB Jr. An assessment of the use of antihistamines in the management of atopic dermatitis. J Am Acad Dermatol. 2018;79:92-96. doi:10.1016/j.jaad.2017.12.077
  25. van Zuuren EJ, Apfelbacher CJ, Fedorowicz Z, et al. No high level evidence to support the use of oral H1 antihistamines as monotherapy for eczema: a summary of a Cochrane systematic review. Syst Rev. 2014;3:25. doi:10.1186/2046-4053-3-25
  26. Neill BC, Neill JA, Brauker J, et al. Postexposure prevention of Toxicodendron dermatitis by early forceful unidirectional washing with liquid dishwashing soap. J Am Acad Dermatol. 2019;81:E25. doi:10.1016/j.jaad.2017.12.081
  27. Stibich AS, Yagan M, Sharma V, et al. Cost-effective post-exposure prevention of poison ivy dermatitis. Int J Dermatol. 2000;39:515-518. doi:10.1046/j.1365-4362.2000.00003.x
  28. Davila A, Laurora M, Fulton J, et al. A new topical agent, Zanfel, ameliorates urushiol-induced Toxicodendron allergic contact dermatitis [abstract]. Ann Emerg Med. 2003;42:S98.
References
  1. Lofgran T, Mahabal GD. Toxicodendron toxicity. StatPearls [Internet]. Updated May 16, 2023. Accessed December 23, 2023. https://www.ncbi.nlm.nih.gov/books/NBK557866/
  2. The Lewin Group. The Burden of Skin Diseases 2005. Society for Investigative Dermatology and American Academy of Dermatology Association; 2005:37-40. Accessed December 26, 2023. https://www.lewin.com/content/dam/Lewin/Resources/Site_Sections/Publications/april2005skindisease.pdf
  3. Monroe J. Toxicodendron contact dermatitis: a case report and brief review. J Clin Aesthet Dermatol. 2020;13(9 Suppl 1):S29-S34.
  4. Gladman AC. Toxicodendron dermatitis: poison ivy, oak, and sumac. Wilderness Environ Med. 2006;17:120-128. doi:10.1580/pr31-05.1
  5. Fretwell S. Poison ivy cases on the rise. The State. Updated May 15,2017. Accessed December 26, 2023. https://www.thestate.com/news/local/article150403932.html
  6. Mohan JE, Ziska LH, Schlesinger WH, et al. Biomass and toxicity responses of poison ivy (Toxicodendron radicans) to elevated atmospheric CO2Proc Natl Acad Sci U S A. 2006;103:9086-9089. doi:10.1073/pnas.0602392103
  7. Williams JV, Light J, Marks JG Jr. Individual variations in allergic contact dermatitis from urushiol. Arch Dermatol. 1999;135:1002-1003. doi:10.1001/archderm.135.8.1002
  8. Kurlan JG, Lucky AW. Black spot poison ivy: a report of 5 cases and a review of the literature. J Am Acad Dermatol. 2001;45:246-249. doi:10.1067/mjd.2001.114295
  9. Fisher AA. Poison ivy/oak/sumac. part II: specific features. Cutis. 1996;58:22-24.
  10. Brook I, Frazier EH, Yeager JK. Microbiology of infected poison ivy dermatitis. Br J Dermatol. 2000;142:943-946. doi:10.1046/j.1365-2133.2000.03475.x
  11. Gillis WT. The systematics and ecology of poison-ivy and the poison-oaks (Toxicodendron, Anacardiaceae). Rhodora. 1971;73:370-443.
  12. Reveal JL. Typification of six Philip Miller names of temperate North American Toxicodendron (Anacardiaceae) with proposals (999-1000) to reject T. crenatum and T. volubileTAXON. 1991;40:333-335. doi:10.2307/1222994 
  13. Guin JD, Gillis WT, Beaman JH. Recognizing the Toxicodendrons (poison ivy, poison oak, and poison sumac). J Am Acad Dermatol. 1981;4:99-114. doi:10.1016/s0190-9622(81)70014-8
  14. Lee NP, Arriola ER. Poison ivy, oak, and sumac dermatitis. West J Med. 1999;171:354-355.
  15. Marks JG Jr, Anderson BE, DeLeo VA, eds. Contact and Occupational Dermatology. Jaypee Brothers Medical Publishers Ltd; 2016.
  16. Dawson CR. The chemistry of poison ivy. Trans N Y Acad Sci. 1956;18:427-443. doi:10.1111/j.2164-0947.1956.tb00465.x
  17. Kalish RS. Recent developments in the pathogenesis of allergic contact dermatitis. Arch Dermatol. 1991;127:1558-1563.
  18. Fisher AA, Mitchell J. Toxicodendron plants and spices. In: Rietschel RL, Fowler JF Jr. Fisher’s Contact Dermatitis. 4th ed. Williams & Wilkins; 1995:461-523.
  19. Labib A, Yosipovitch G. Itchy Toxicodendron plant dermatitis. Allergies. 2022;2:16-22. doi:10.3390/allergies2010002 
  20. Fisher AA. Poison ivy/oak dermatitis part I: prevention—soap and water, topical barriers, hyposensitization. Cutis. 1996;57:384-386.
  21. Kim Y, Flamm A, ElSohly MA, et al. Poison ivy, oak, and sumac dermatitis: what is known and what is new? 2019;30:183-190. doi:10.1097/DER.0000000000000472
  22. Prok L, McGovern T. Poison ivy (Toxicodendron) dermatitis. UpToDate. Updated October 16, 2023. Accessed December 26, 2023. https://www.uptodate.com/contents/poison-ivy-toxicodendron-dermatitis
  23. Klein PA, Clark RA. An evidence-based review of the efficacy of antihistamines in relieving pruritus in atopic dermatitis. Arch Dermatol. 1999;135:1522-1525. doi:10.1001/archderm.135.12.1522
  24. He A, Feldman SR, Fleischer AB Jr. An assessment of the use of antihistamines in the management of atopic dermatitis. J Am Acad Dermatol. 2018;79:92-96. doi:10.1016/j.jaad.2017.12.077
  25. van Zuuren EJ, Apfelbacher CJ, Fedorowicz Z, et al. No high level evidence to support the use of oral H1 antihistamines as monotherapy for eczema: a summary of a Cochrane systematic review. Syst Rev. 2014;3:25. doi:10.1186/2046-4053-3-25
  26. Neill BC, Neill JA, Brauker J, et al. Postexposure prevention of Toxicodendron dermatitis by early forceful unidirectional washing with liquid dishwashing soap. J Am Acad Dermatol. 2019;81:E25. doi:10.1016/j.jaad.2017.12.081
  27. Stibich AS, Yagan M, Sharma V, et al. Cost-effective post-exposure prevention of poison ivy dermatitis. Int J Dermatol. 2000;39:515-518. doi:10.1046/j.1365-4362.2000.00003.x
  28. Davila A, Laurora M, Fulton J, et al. A new topical agent, Zanfel, ameliorates urushiol-induced Toxicodendron allergic contact dermatitis [abstract]. Ann Emerg Med. 2003;42:S98.
Issue
Cutis - 113(1)
Issue
Cutis - 113(1)
Page Number
E11-E14
Page Number
E11-E14
Publications
Publications
Topics
Article Type
Display Headline
Botanical Briefs: Contact Dermatitis Induced by Western Poison Ivy (Toxicodendron rydbergii)
Display Headline
Botanical Briefs: Contact Dermatitis Induced by Western Poison Ivy (Toxicodendron rydbergii)
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Afvari</fileName> <TBEID>0C02F07B.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F07B</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Afvari</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240104T143002</firstPublished> <LastPublished>20240104T143002</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240104T143001</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Shawn Afvari, BS; Dirk M. Elston, MD; Thomas W. McGovern, MD</byline> <bylineText>Shawn Afvari, BS; Dirk M. Elston, MD; Thomas W. McGovern, MD</bylineText> <bylineFull>Shawn Afvari, BS; Dirk M. Elston, MD; Thomas W. McGovern, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>E11-E14</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Western poison ivy (Toxicodendron rydbergii) is responsible for many of the cases of Toxicodendron contact dermatitis (TCD) reported in the western and northern</metaDescription> <articlePDF>299857</articlePDF> <teaserImage/> <title>Botanical Briefs: Contact Dermatitis Induced by Western Poison Ivy (Toxicodendron rydbergii)</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>January</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>1</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2165</CMSID> </CMSIDs> <keywords> <keyword>contact dermatitis</keyword> <keyword> western poison ivy</keyword> <keyword> toxicodendron rydbergii</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>January 2024</pubIssueName> <pubArticleType>Audio | 2165</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">60</term> </sections> <topics> <term canonical="true">199</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002699.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Botanical Briefs: Contact Dermatitis Induced by Western Poison Ivy (Toxicodendron rydbergii)</title> <deck/> </itemMeta> <itemContent> <p class="abstract">“Leaves of three, leave it be” serves as an apt caution for avoiding poison ivy <em>(Toxicodendron </em>species) and its dermatitis-inducing sap. <em>Toxicodendron</em> contact dermatitis (TCD) poses a notable burden to the American health care system by accounting for half a million reported cases of allergic contact dermatitis (ACD) annually. Identifying and avoiding physical contact with the western poison ivy<em> (Toxicodendron rydbergii)</em> plant prevails as the chief method of preventing TCD. This article discusses common features of <em>T rydbergii</em> as well as clinical manifestations and treatment options following exposure to this allergenic plant. </p> <p> <em><em>Cutis</em>. 2024;113:E11-E14.</em> </p> <h3>Clinical Importance </h3> <p>Western poison ivy (<i>Toxicodendron rydbergii</i>) is responsible for many of the cases of <i>Toxicodendron</i> contact dermatitis (TCD) reported in the western and northern United States. <i>Toxicodendron</i> plants cause more cases of allergic contact dermatitis (ACD) in North America than any other allergen<sup>1</sup>; 9 million Americans present to physician offices and 1.6 million present to emergency departments annually for ACD, emphasizing the notable medical burden of this condition.<sup>2,3</sup> Exposure to urushiol, a plant resin containing potent allergens, precipitates this form of ACD.</p> <p>An estimated 50% to 75% of adults in the United States demonstrate clinical sensitivity and exhibit ACD following contact with <i>T rydbergii</i>.<sup>4</sup> Campers, hikers, firefighters, and forest workers often risk increased exposure through physical contact or aerosolized allergens in smoke. According to the Centers for Disease Control and Prevention, the incidence of visits to US emergency departments for TCD nearly doubled from 2002 to 2012,<sup>5</sup> which may be explained by atmospheric CO<sub>2</sub> levels that both promote increased growth of <i>Toxicodendron</i> species and augment their toxicity.<sup>6</sup></p> <h3>Cutaneous Manifestations </h3> <p>The clinical presentation of <i>T rydbergii</i> contact dermatitis is similar to other allergenic members of the <i>Toxicodendron</i> genus. Patients sensitive to urushiol typically develop a pruritic erythematous rash within 1 to 2 days of exposure (range, 5 hours to 15 days).<sup>7</sup> Erythematous and edematous streaks initially manifest on the extremities and often progress to bullae and oozing papulovesicles. In early disease, patients also may display black lesions on or near the rash<sup>8</sup> (so-called black-dot dermatitis) caused by oxidized urushiol deposited on the skin—an uncommon yet classic presentation of TCD. Generally, symptoms resolve without complications and with few sequalae, though hyperpigmentation or a secondary infection can develop on or near affected areas.<sup>9,10</sup> </p> <h3>Taxonomy</h3> <p>The <i>Toxicodendron</i> genus belongs to the Anacardiaceae family, which includes pistachios, mangos, and cashews, and causes more cases of ACD than every other plant combined.<sup>4</sup> (Shelled pistachios and cashews do not possess cross-reacting allergens and should not worry consumers; mango skin does contain urushiol.) </p> <p><i>Toxicodendron</i> (formerly part of the <i>Rhus</i> genus) includes several species of poison oak, poison ivy, and poison sumac and can be found in shrubs (<i>T rydbergii</i> and <i>Toxicodendron diversilobum</i>), vines (<i>Toxicodendron radicans</i> and <i>Toxicodendron pubescens</i>), and trees (<i>Toxicodendron vernix</i>). In addition, <i>Toxicodendron</i> taxa can hybridize with other taxa in close geographic proximity to form morphologic intermediates. Some individual plants have features of multiple species.<sup>11</sup> </p> <h3>Etymology</h3> <p>The common name of <i>T rydbergii</i>—western poison ivy—misleads the public; the plant contains no poison that can cause death and does not grow as ivy by wrapping around trees, as <i>T radicans </i>and English ivy (<i>Hedera helix</i>) do. Its formal genus, <i>Toxicodendron</i>, means “poison tree” in Greek and was given its generic name by the English botanist Phillip Miller in 1768,<sup>12</sup> which caused the renaming of <i>Rhus rydbergii</i> as <i>T rydbergii</i>. The species name honors Per Axel Rydberg, a 19th and 20th century Swedish-American botanist. </p> <h3>Distribution</h3> <p><i>Toxicodendron rydbergii</i> grows in California and other states in the western half of the United States as well as the states bordering Canada and Mexico. In Canada, it reigns as the most dominant form of poison ivy.<sup>13</sup> Hikers and campers find <i>T rydbergii </i>in a variety of areas, including roadsides, river bottoms, sandy shores, talus slopes, precipices, and floodplains.<sup>11</sup> This taxon grows under a variety of conditions and in distinct regions, and it thrives in both full sun or shade. </p> <h3>Identifying Features </h3> <p><i>Toxicodendron rydbergii </i>turns red earlier than most plants; early red summer leaves should serve as a warning sign to hikers from a distance (Figure 1). It displays trifoliate ovate leaves (ie, each leaf contains 3 leaflets) on a dwarf nonclimbing shrub (Figure 2). Although the plant shares common features with its cousin <i>T radicans</i> (eastern poison ivy), <i>T rydbergii</i> is easily distinguished by its thicker stems, absence of aerial rootlets (abundant in <i>T radicans</i>), and short (approximately 1 meter) height.<sup>4</sup> </p> <p>Curly hairs occupy the underside of <i>T rydbergii </i>leaflets and along the midrib; leaflet margins appear lobed or rounded. Lenticels appear as small holes in the bark that turn gray in the cold and become brighter come spring.<sup>13<br/><br/></sup>The plant bears glabrous long petioles (leaf stems) and densely grouped clusters of yellow flowers. In autumn, the globose fruit—formed in clusters between each twig and leaf petiole (known as an axillary position)—change from yellow-green to tan (Figure 3). When urushiol exudes from damaged leaflets or other plant parts, it oxidizes on exposure to air and creates hardened black deposits on the plant. Even when grown in garden pots, <i>T rydbergii</i> maintains its distinguishing features.<sup>11</sup> </p> <h3>Dermatitis-Inducing Plant Parts</h3> <p>All parts of <i>T rydbergii</i> including leaves, stems, roots, and fruit contain the allergenic sap throughout the year.<sup>14</sup> A person must damage or bruise the plant for urushiol to be released and produce its allergenic effects; softly brushing against undamaged plants typically does not induce dermatitis.<sup>4</sup> </p> <h3>Pathophysiology of Urushiol </h3> <p>Urushiol, a pale yellow, oily mixture of organic compounds conserved throughout all <i>Toxicodendron</i> species, contains highly allergenic alkyl catechols. These catechols possess hydroxyl groups at positions 1 and 2 on a benzene ring; the hydrocarbon side chain of poison ivies (typically 15–carbon atoms long) attaches at position 3.<sup>15</sup> The catechols and the aliphatic side chain contribute to the plant’s antigenic and dermatitis-inducing properties.<sup>16</sup> </p> <p>The high lipophilicity of urushiol allows for rapid and unforgiving absorption into the skin, notwithstanding attempts to wash it off. Upon direct contact, catechols of urushiol penetrate the epidermis and become oxidized to quinone intermediates that bind to antigen-presenting cells in the epidermis and dermis. Epidermal Langerhans cells and dermal macrophages internalize and present the antigen to CD4<span class="caption"><sup>+</sup></span> T cells in nearby lymph nodes. This sequence results in production of inflammatory mediators, clonal expansion of T-effector and T-memory cells specific to the allergenic catechols, and an ensuing cytotoxic response against epidermal cells and the dermal vasculature. Keratinocytes and monocytes mediate the inflammatory response by releasing other cytokines.<sup>4,17</sup> <br/><br/>Sensitization to urushiol generally occurs at 8 to 14 years of age; therefore, infants have lower susceptibility to dermatitis upon contact with <i>T rydbergii</i>.<sup>18</sup> Most animals do not experience sensitization upon contact; in fact, birds and forest animals consume the urushiol-rich fruit of <i>T rydbergii </i>without harm.<sup>3</sup> </p> <h3>Prevention and Treatment</h3> <p><i>Toxicodendron</i> dermatitis typically lasts 1 to 3 weeks but can remain for as long as 6 weeks without treatment.<sup>19</sup> Recognition and physical avoidance of the plant provides the most promising preventive strategy. Immediate rinsing with soap and water can prevent TCD by breaking down urushiol and its allergenic components; however, this is an option for only a short time, as the skin absorbs 50% of urushiol within 10 minutes after contact.<sup>20</sup> Nevertheless, patients must seize the earliest opportunity to wash off the affected area and remove any residual urushiol. Patients must be cautious when removing and washing clothing to prevent further contact. </p> <p>Most health care providers treat TCD with a corticosteroid to reduce inflammation and intense pruritus. A high-potency topical corticosteroid (eg, clobetasol) may prove effective in providing early therapeutic relief in mild disease.<sup>21</sup> A short course of a systemic steroid quickly and effectively quenches intense itching and should not be limited to what the clinician considers severe disease. Do not underestimate the patient’s symptoms with this eruption. <br/><br/>Prednisone dosing begins at 1 mg/kg daily and is then tapered slowly over 2 weeks (no shorter a time) for an optimal treatment course of 15 days.<sup>22</sup> Prescribing an inadequate dosage and course of a corticosteroid leaves the patient susceptible to rebound dermatitis—and loss of trust in their provider.<br/><br/> Intramuscular injection of the long-acting corticosteroid triamcinolone acetonide with rapid-onset betamethasone provides rapid relief and fewer adverse effects than an oral corticosteroid.<sup>22</sup> Despite the long-standing use of sedating oral antihistamines by clinicians, these drugs provide no benefit for pruritus or sleep because the histamine does not cause the itching of TCD, and antihistamines disrupt normal sleep architecture.<sup>23-25</sup> <br/><br/> Patients can consider several over-the-counter products that have varying degrees of efficacy.<sup>4,26</sup> The few products for which prospective studies support their use include Tecnu (Tec Laboraties Inc), Zanfel (RhusTox), and the well-known soaps Dial (Henkel Corporation) and Goop (Critzas Industries, Inc).<sup>27,28</sup> <br/><br/>Aside from treating the direct effects of TCD, clinicians also must take note of any look for signs of secondary infection and occasionally should consider supplementing treatment with an antibiotic. </p> <h2>REFERENCES</h2> <p class="reference"> 1. Lofgran T, Mahabal GD. <i>Toxicodendron</i> toxicity. <i>StatPearls [Internet]</i>. Updated May 16, 2023. Accessed December 23, 2023. <span class="apple-converted-space">https://www.ncbi.nlm.nih.gov/books/NBK557866/<br/><br/></span> 2. The Lewin Group. <i>The Burden of Skin Diseases 2005</i>. Society for Investigative Dermatology and American Academy of Dermatology Association; 2005:37-40. Accessed December 26, 2023. https://www.lewin.com/content/dam/Lewin/Resources/Site_Sections/Publications/april2005skindisease.pdf <br/><br/> 3. Monroe J. Toxicodendron contact dermatitis: a case report and brief review. <i>J Clin Aesthet Dermatol</i>. 2020;13(9 Suppl 1):S29-S34.<br/><br/> 4. Gladman AC. Toxicodendron dermatitis: poison ivy, oak, and sumac. <i>Wilderness Environ Med</i>. 2006;17:120-128. doi:10.1580/pr31-05.1<br/><br/> 5. Fretwell S. Poison ivy cases on the rise. <i>The State</i>. Updated May 15,2017. Accessed December 26, 2023. https://www.thestate.com/news/local/article150403932.html<br/><br/> 6. Mohan JE, Ziska LH, Schlesinger WH, et al. Biomass and toxicity responses of poison ivy (<i>Toxicodendron radicans</i>) to elevated atmospheric CO<sub>2</sub>. <i>Proc Natl Acad Sci U S A</i>. 2006;103:9086-9089. doi:10.1073/pnas.0602392103<br/><br/> 7. Williams JV, Light J, Marks JG Jr. Individual variations in allergic contact dermatitis from urushiol. <i>Arch Dermatol</i>. 1999;135:1002-1003. doi:10.1001/archderm.135.8.1002<br/><br/> 8. Kurlan JG, Lucky AW. Black spot poison ivy: a report of 5 cases and a review of the literature. <i>J Am Acad Dermatol</i>. 2001;45:246-249. doi:10.1067/mjd.2001.114295<br/><br/> 9. Fisher AA. Poison ivy/oak/sumac. part II: specific features. <i>Cutis</i>. 1996;58:22-24.<br/><br/>10. Brook I, Frazier EH, Yeager JK. Microbiology of infected poison ivy dermatitis. <i>Br J Dermatol</i>. 2000;142:943-946. doi:10.1046/j.1365-2133.2000.03475.x<br/><br/>11. Gillis WT. The systematics and ecology of poison-ivy and the poison-oaks (Toxicodendron, Anacardiaceae). <i>Rhodora</i>. 1971;73:370-443.<br/><br/>12. Reveal JL. Typification of six Philip Miller names of temperate North American <i>Toxicodendron</i> (<i>Anacardiaceae</i>) with proposals (999-1000) to reject <i>T. crenatum</i> and <i>T. volubile</i>. <i>TAXON</i>. 1991;40:333-335. doi:10.2307/1222994 <br/><br/>13. Guin JD, Gillis WT, Beaman JH. Recognizing the Toxicodendrons (poison ivy, poison oak, and poison sumac). <i>J Am Acad Dermatol</i>. 1981;4:99-114. doi:10.1016/s0190-9622(81)70014-8<br/><br/>14. Lee NP, Arriola ER. Poison ivy, oak, and sumac dermatitis. <i>West J Med</i>. 1999;171:354-355.<br/><br/>15. Marks JG Jr, Anderson BE, DeLeo VA, eds. <i>Contact and Occupational Dermatology</i>. Jaypee Brothers Medical Publishers Ltd; 2016.<br/><br/>16. Dawson CR. The chemistry of poison ivy. <i>Trans N Y Acad Sci</i>. 1956;18:427-443. doi:10.1111/j.2164-0947.1956.tb00465.x<br/><br/>17. Kalish RS. Recent developments in the pathogenesis of allergic contact dermatitis. <i>Arch Dermatol</i>. 1991;127:1558-1563.<br/><br/>18. Fisher AA, Mitchell J. Toxicodendron plants and spices. In: Rietschel RL, Fowler JF Jr. <i>Fisher’s Contact Dermatitis.</i> 4th ed. Williams &amp; Wilkins; 1995:461-523.<br/><br/>19. Labib A, Yosipovitch G. Itchy Toxicodendron plant dermatitis. <i>Allergies</i>. 2022;2:16-22. doi:10.3390/allergies2010002 <br/><br/>20. Fisher AA. Poison ivy/oak dermatitis part I: prevention—soap and water, topical barriers, hyposensitization. <i>Cutis</i>. 1996;57:384-386.<br/><br/>21. Kim Y, Flamm A, ElSohly MA, et al. Poison ivy, oak, and sumac dermatitis: what is known and what is new? 2019;30:183-190. <span class="citation-doi">doi:10.1097/DER.0000000000000472<br/><br/></span>22. Prok L, McGovern T. Poison ivy (<i>Toxicodendron</i>) dermatitis. UpToDate. Updated October 16, 2023. Accessed December 26, 2023. https://www.uptodate.com/contents/poison-ivy-toxicodendron-dermatitis<br/><br/>23. Klein PA, Clark RA. An evidence-based review of the efficacy of antihistamines in relieving pruritus in atopic dermatitis. <i>Arch Dermatol</i>. 1999;135:1522-1525. doi:10.1001/archderm.135.12.1522<br/><br/>24. He A, Feldman SR, Fleischer AB Jr. An assessment of the use of antihistamines in the management of atopic dermatitis. <i>J Am Acad Dermatol</i>. 2018;79:92-96. doi:10.1016/j.jaad.2017.12.077<br/><br/>25. van Zuuren EJ, Apfelbacher CJ, Fedorowicz Z, et al. No high level evidence to support the use of oral H1 antihistamines as monotherapy for eczema: a summary of a Cochrane systematic review. <i>Syst Rev</i>. 2014;3:25. doi:10.1186/2046-4053-3-25<br/><br/>26. Neill BC, Neill JA, Brauker J, et al. Postexposure prevention of <i>Toxicodendron</i> dermatitis by early forceful unidirectional washing with liquid dishwashing soap. <i>J Am Acad Dermatol</i>. 2019;81:E25. doi:10.1016/j.jaad.2017.12.081<br/><br/>27. Stibich AS, Yagan M, Sharma V, et al. Cost-effective post-exposure prevention of poison ivy dermatitis. <i>Int J Dermatol</i>. 2000;39:515-518. doi:10.1046/j.1365-4362.2000.00003.x<br/><br/>28. Davila A, Laurora M, Fulton J, et al. A new topical agent, Zanfel, ameliorates urushiol-induced <i>Toxicodendron</i> allergic contact dermatitis [abstract]. <i>Ann Emerg Med</i>. 2003;42:S98.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">PRACTICE <strong>POINTS</strong></p> <ul class="insidebody"> <li>Western poison ivy<em> (Toxicodendron rydbergii)</em> accounts for many of the cases of <em>Toxicodendron</em> contact dermatitis (TCD) in the western and northern United States. Individuals in these regions should be educated on how to identify<em> T rydbergii</em> to avoid TCD. </li> <li>Dermatologists should include TCD in the differential diagnosis when a patient presents with an erythematous pruritic rash in a linear pattern with sharp borders. </li> </ul> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Shawn Afvari is from New York Medical College School of Medicine, Valhalla. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana. </p> <p class="disclosure">The authors report no conflict of interest. <br/><br/>Correspondence: Shawn Afvari, BS (safvari@student.nymc.edu). <br/><br/>doi:10.12788/cutis.0936</p> </itemContent> </newsItem> </itemSet></root>
Inside the Article

PRACTICE POINTS

  • Western poison ivy (Toxicodendron rydbergii) accounts for many of the cases of Toxicodendron contact dermatitis (TCD) in the western and northern United States. Individuals in these regions should be educated on how to identify T rydbergii to avoid TCD.
  • Dermatologists should include TCD in the differential diagnosis when a patient presents with an erythematous pruritic rash in a linear pattern with sharp borders.
  • Most patients who experience intense itching and pain from TCD benefit greatly from prompt treatment with an oral or intramuscular corticosteroid. Topical steroids rarely provide relief; oral antihistamines provide no benefit.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
18002699.SIG
Disable zoom
Off

What’s Eating You? Update on the Sticktight Flea (Echidnophaga gallinacea)

Article Type
Changed
Thu, 12/14/2023 - 11:04
Display Headline
What’s Eating You? Update on the Sticktight Flea (Echidnophaga gallinacea)

Fleas (order Siphonaptera) are vectors for various diseases, such as plague (as carriers of Yersinia pestis) and rickettsial infections.1-4 The sticktight flea (Echidnophaga gallinacea) commonly is seen on birds and mammals, including ground squirrels, dogs, cats, and rodents, and can attach to its host for days at a time by burrowing its head into the skin. Similar to other fleas, the sticktight flea needs a blood supply to reproduce.5 Therefore, it is important to study the sticktight flea, its habitat, and infection patterns to improve public health and prevent infestation.

Identification

Echidnophaga gallinacea is named for the female flea’s behavior—it “sticks tight” to the surface of the host by embedding its head into the skin for days at a time.5 The sticktight flea and the rat flea (Xenopsylla cheopis) can be differentiated by the sticktight’s reduced thorax and lack of a pleural rod (the vertical ridge that divides the mesosternum above the second pair of legs)(Figure, A and B). The sticktight flea can be differentiated from the dog flea (Ctenocephalides canis) and the cat flea (Ctenocephalides felis) by its lack of genal ctenidia (horizontal combs in the mustache area) and pronotal ctenidia (vertical combs behind the head)(Figure, B and C).6,7 Other defining features of E gallinacea include 2 pairs of large postantennal setae (hairs) on its anteriorly flattened head; a C-shaped reproductive organ known as the spermatheca; and broad maxillary lacinia (Figure, C).8

CT112006015_e_ABC.jpg
%3Cp%3EA-C%2C%20Anatomy%20of%20the%20sticktight%20flea%20(%3Cem%3EEchidnophaga%20gallinacea%3C%2Fem%3E)%2C%20rat%20flea%20(%3Cem%3EXenopsylla%20cheopis%3C%2Fem%3E)%2C%20and%20cat%20flea%20(%3Cem%3ECtenocephalides%20felis%3C%2Fem%3E)%2C%20respectively.%20The%20rat%20flea%20has%20a%20pleural%20rod%20and%20the%20cat%20flea%20has%20genal%20and%20pronotal%20ctenidia%20(combs)%2C%20which%20are%20absent%20in%20%3Cem%3EE%20gallinacean%3C%2Fem%3E.%3C%2Fp%3E

Habitat, Seasonality, and Behavior

Echidnophaga gallinacea commonly infests the comb, wattles, and surrounding ears of chickens; the flea also has been found on dogs, cats, rodents, and other species of birds.9 The sticktight flea is more prevalent in summer and autumn, which may explain its predominance in warmer climates, including California, Florida, Mexico, Egypt, Africa, and Iran.1,9-11

When a female sticktight flea begins to feed, it stays on the host for days at a time, waiting for a male.5 The female deposits its fertilized eggs in nests on the host or in lesions caused by infestation. Eventually, eggs hatch and fall into soil, where they lay dormant or grow to adulthood.5

Cutaneous Reaction to Infestation

Flea bites cause a hypersensitivity reaction, with pruritic pustules and erythematous papules that have a central punctum.12 In a reported case in Los Angeles, California, a female sticktight flea buried itself into the cheek of a young boy for more than 12 hours. The lesion was not marked by surrounding erythema, tenderness, pruritus, or swelling; however, several days after the flea was removed, erythema developed at the site then spontaneously resolved.7 In a study of dogs that were infested with E gallinacea, the flea never disengaged to attach to a human; when the flea was deliberately placed on a human, it fed and left hastily.11

Management

Because E gallinacea burrows its head into the skin, the best removal method is applying slow gentle traction under sterile conditions to ensure removal of mouthparts.7 An oral antihistamine can be administered or a topical antihistamine or corticosteroid can be applied to the affected area.12 Flea infestation should be treated with an insecticide. Affected animals should be treated by a veterinarian using a pesticide, such as fipronil, selamectin, imidacloprid, metaflumizone, nitenpyram, lufenuron, methoprene, or pyriproxyfen.13

References
  1. Hubbart JA, Jachowski DS, Eads DA. Seasonal and among-site variation in the occurrence and abundance of fleas on California ground squirrels (Otospermophilus beecheyi). J Vector Ecol. 2011;36:117-123. doi:10.1111/j.1948-7134.2011.00148.x
  2. Jiang J, Maina AN, Knobel DL, et al. Molecular detection of Rickettsia felis and Candidatus Rickettsia asemboensis in fleas from human habitats, Asembo, Kenya. Vector Borne Zoonotic Dis. 2013;13:550-558. doi:10.1089/vbz.2012.1123
  3. López-Pérez AM, Chaves A, Sánchez-Montes S, et al. Diversity of rickettsiae in domestic, synanthropic, and sylvatic mammals and their ectoparasites in a spotted fever-epidemic region at the western US-Mexico border. Transbound Emerg Dis. 2022;69:609-622. doi:10.1111/tbed.14027
  4. Ehlers J, Krüger A, Rakotondranary SJ, et al. Molecular detection of Rickettsia spp., Borrelia spp., Bartonella spp. and Yersinia pestis in ectoparasites of endemic and domestic animals in southwest Madagascar. Acta Trop. 2020;205:105339. doi:10.1016/j.actatropica.2020.105339
  5. Boughton RK, Atwell JW, Schoech SJ. An introduced generalist parasite, the sticktight flea (Echidnophaga gallinacea), and its pathology in the threatened Florida scrub-jay (Aphelocoma coerulescens). J Parasitol. 2006;92:941-948. doi:10.1645/GE-769R.1
  6. Bitam I, Dittmar K, Parola P, et al. Fleas and flea-borne diseases. Int J Infect Dis. 2010;14:e667-e676. doi:10.1016/j.ijid.2009.11.011
  7. Linardi PM, Santos JLC. Ctenocephalides felis felis vs. Ctenocephalides canis (Siphonaptera: Pulicidae): some issues in correctly identify these species. Rev Bras Parasitol Vet. 2012;21:345-354. doi:10.1590/s1984-29612012000400002
  8. Carlson JC, Fox MS. A sticktight flea removed from the cheek of a two-year-old boy from Los Angeles. Dermatol Online J. 2009;15:4. https://doi.org/10.5070/D36vb8p1b1
  9. Mirzaei M, Ghashghaei O, Yakhchali M. Prevalence of ectoparasites of indigenous chickens from Dalahu region, Kermanshah province, Iran. Turkiye Parazitol Derg. 2016;40:13-16. doi:10.5152/tpd.2016.4185
  10. Farid DS, Sallam NH, Eldein AMS, et al. Cross-sectional seasonal prevalence and relative risk of ectoparasitic infestations of rodents in North Sinai, Egypt. Vet World. 2021;14:2996-3006. doi:10.14202/vetworld.2021.2996-3006
  11. Harman DW, Halliwell RE, Greiner EC. Flea species from dogs and cats in north-central Florida. Vet Parasitol. 1987;23:135-140. doi:10.1016/0304-4017(87)90031-8
  12. Anderson J, Paterek E. Flea bites. StatPearls [Internet]. StatPearls Publishing; 2023. Updated August 8, 2023. Accessed November 27, 2023. https://www.ncbi.nlm.nih.gov/books/NBK541118/
  13. Gyimesi ZS, Hayden ER, Greiner EC. Sticktight flea (Echidnophaga gallinacea) infestation in a Victoria crowned pigeon (Goura victoria). J Zoo Wildl Med. 2007;38:594-596. doi:10.1638/2007-0062.1
Article PDF
Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Penny Lane Huebsch is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

The images are in the public domain.

Correspondence: Penny Lane Huebsch, MS, 96 Jonathan Lucas St, Ste 601, Charleston, SC 29425 (huebsch@musc.edu).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
E15-E16
Sections
Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Penny Lane Huebsch is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

The images are in the public domain.

Correspondence: Penny Lane Huebsch, MS, 96 Jonathan Lucas St, Ste 601, Charleston, SC 29425 (huebsch@musc.edu).

Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Penny Lane Huebsch is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

The images are in the public domain.

Correspondence: Penny Lane Huebsch, MS, 96 Jonathan Lucas St, Ste 601, Charleston, SC 29425 (huebsch@musc.edu).

Article PDF
Article PDF

Fleas (order Siphonaptera) are vectors for various diseases, such as plague (as carriers of Yersinia pestis) and rickettsial infections.1-4 The sticktight flea (Echidnophaga gallinacea) commonly is seen on birds and mammals, including ground squirrels, dogs, cats, and rodents, and can attach to its host for days at a time by burrowing its head into the skin. Similar to other fleas, the sticktight flea needs a blood supply to reproduce.5 Therefore, it is important to study the sticktight flea, its habitat, and infection patterns to improve public health and prevent infestation.

Identification

Echidnophaga gallinacea is named for the female flea’s behavior—it “sticks tight” to the surface of the host by embedding its head into the skin for days at a time.5 The sticktight flea and the rat flea (Xenopsylla cheopis) can be differentiated by the sticktight’s reduced thorax and lack of a pleural rod (the vertical ridge that divides the mesosternum above the second pair of legs)(Figure, A and B). The sticktight flea can be differentiated from the dog flea (Ctenocephalides canis) and the cat flea (Ctenocephalides felis) by its lack of genal ctenidia (horizontal combs in the mustache area) and pronotal ctenidia (vertical combs behind the head)(Figure, B and C).6,7 Other defining features of E gallinacea include 2 pairs of large postantennal setae (hairs) on its anteriorly flattened head; a C-shaped reproductive organ known as the spermatheca; and broad maxillary lacinia (Figure, C).8

CT112006015_e_ABC.jpg
%3Cp%3EA-C%2C%20Anatomy%20of%20the%20sticktight%20flea%20(%3Cem%3EEchidnophaga%20gallinacea%3C%2Fem%3E)%2C%20rat%20flea%20(%3Cem%3EXenopsylla%20cheopis%3C%2Fem%3E)%2C%20and%20cat%20flea%20(%3Cem%3ECtenocephalides%20felis%3C%2Fem%3E)%2C%20respectively.%20The%20rat%20flea%20has%20a%20pleural%20rod%20and%20the%20cat%20flea%20has%20genal%20and%20pronotal%20ctenidia%20(combs)%2C%20which%20are%20absent%20in%20%3Cem%3EE%20gallinacean%3C%2Fem%3E.%3C%2Fp%3E

Habitat, Seasonality, and Behavior

Echidnophaga gallinacea commonly infests the comb, wattles, and surrounding ears of chickens; the flea also has been found on dogs, cats, rodents, and other species of birds.9 The sticktight flea is more prevalent in summer and autumn, which may explain its predominance in warmer climates, including California, Florida, Mexico, Egypt, Africa, and Iran.1,9-11

When a female sticktight flea begins to feed, it stays on the host for days at a time, waiting for a male.5 The female deposits its fertilized eggs in nests on the host or in lesions caused by infestation. Eventually, eggs hatch and fall into soil, where they lay dormant or grow to adulthood.5

Cutaneous Reaction to Infestation

Flea bites cause a hypersensitivity reaction, with pruritic pustules and erythematous papules that have a central punctum.12 In a reported case in Los Angeles, California, a female sticktight flea buried itself into the cheek of a young boy for more than 12 hours. The lesion was not marked by surrounding erythema, tenderness, pruritus, or swelling; however, several days after the flea was removed, erythema developed at the site then spontaneously resolved.7 In a study of dogs that were infested with E gallinacea, the flea never disengaged to attach to a human; when the flea was deliberately placed on a human, it fed and left hastily.11

Management

Because E gallinacea burrows its head into the skin, the best removal method is applying slow gentle traction under sterile conditions to ensure removal of mouthparts.7 An oral antihistamine can be administered or a topical antihistamine or corticosteroid can be applied to the affected area.12 Flea infestation should be treated with an insecticide. Affected animals should be treated by a veterinarian using a pesticide, such as fipronil, selamectin, imidacloprid, metaflumizone, nitenpyram, lufenuron, methoprene, or pyriproxyfen.13

Fleas (order Siphonaptera) are vectors for various diseases, such as plague (as carriers of Yersinia pestis) and rickettsial infections.1-4 The sticktight flea (Echidnophaga gallinacea) commonly is seen on birds and mammals, including ground squirrels, dogs, cats, and rodents, and can attach to its host for days at a time by burrowing its head into the skin. Similar to other fleas, the sticktight flea needs a blood supply to reproduce.5 Therefore, it is important to study the sticktight flea, its habitat, and infection patterns to improve public health and prevent infestation.

Identification

Echidnophaga gallinacea is named for the female flea’s behavior—it “sticks tight” to the surface of the host by embedding its head into the skin for days at a time.5 The sticktight flea and the rat flea (Xenopsylla cheopis) can be differentiated by the sticktight’s reduced thorax and lack of a pleural rod (the vertical ridge that divides the mesosternum above the second pair of legs)(Figure, A and B). The sticktight flea can be differentiated from the dog flea (Ctenocephalides canis) and the cat flea (Ctenocephalides felis) by its lack of genal ctenidia (horizontal combs in the mustache area) and pronotal ctenidia (vertical combs behind the head)(Figure, B and C).6,7 Other defining features of E gallinacea include 2 pairs of large postantennal setae (hairs) on its anteriorly flattened head; a C-shaped reproductive organ known as the spermatheca; and broad maxillary lacinia (Figure, C).8

CT112006015_e_ABC.jpg
%3Cp%3EA-C%2C%20Anatomy%20of%20the%20sticktight%20flea%20(%3Cem%3EEchidnophaga%20gallinacea%3C%2Fem%3E)%2C%20rat%20flea%20(%3Cem%3EXenopsylla%20cheopis%3C%2Fem%3E)%2C%20and%20cat%20flea%20(%3Cem%3ECtenocephalides%20felis%3C%2Fem%3E)%2C%20respectively.%20The%20rat%20flea%20has%20a%20pleural%20rod%20and%20the%20cat%20flea%20has%20genal%20and%20pronotal%20ctenidia%20(combs)%2C%20which%20are%20absent%20in%20%3Cem%3EE%20gallinacean%3C%2Fem%3E.%3C%2Fp%3E

Habitat, Seasonality, and Behavior

Echidnophaga gallinacea commonly infests the comb, wattles, and surrounding ears of chickens; the flea also has been found on dogs, cats, rodents, and other species of birds.9 The sticktight flea is more prevalent in summer and autumn, which may explain its predominance in warmer climates, including California, Florida, Mexico, Egypt, Africa, and Iran.1,9-11

When a female sticktight flea begins to feed, it stays on the host for days at a time, waiting for a male.5 The female deposits its fertilized eggs in nests on the host or in lesions caused by infestation. Eventually, eggs hatch and fall into soil, where they lay dormant or grow to adulthood.5

Cutaneous Reaction to Infestation

Flea bites cause a hypersensitivity reaction, with pruritic pustules and erythematous papules that have a central punctum.12 In a reported case in Los Angeles, California, a female sticktight flea buried itself into the cheek of a young boy for more than 12 hours. The lesion was not marked by surrounding erythema, tenderness, pruritus, or swelling; however, several days after the flea was removed, erythema developed at the site then spontaneously resolved.7 In a study of dogs that were infested with E gallinacea, the flea never disengaged to attach to a human; when the flea was deliberately placed on a human, it fed and left hastily.11

Management

Because E gallinacea burrows its head into the skin, the best removal method is applying slow gentle traction under sterile conditions to ensure removal of mouthparts.7 An oral antihistamine can be administered or a topical antihistamine or corticosteroid can be applied to the affected area.12 Flea infestation should be treated with an insecticide. Affected animals should be treated by a veterinarian using a pesticide, such as fipronil, selamectin, imidacloprid, metaflumizone, nitenpyram, lufenuron, methoprene, or pyriproxyfen.13

References
  1. Hubbart JA, Jachowski DS, Eads DA. Seasonal and among-site variation in the occurrence and abundance of fleas on California ground squirrels (Otospermophilus beecheyi). J Vector Ecol. 2011;36:117-123. doi:10.1111/j.1948-7134.2011.00148.x
  2. Jiang J, Maina AN, Knobel DL, et al. Molecular detection of Rickettsia felis and Candidatus Rickettsia asemboensis in fleas from human habitats, Asembo, Kenya. Vector Borne Zoonotic Dis. 2013;13:550-558. doi:10.1089/vbz.2012.1123
  3. López-Pérez AM, Chaves A, Sánchez-Montes S, et al. Diversity of rickettsiae in domestic, synanthropic, and sylvatic mammals and their ectoparasites in a spotted fever-epidemic region at the western US-Mexico border. Transbound Emerg Dis. 2022;69:609-622. doi:10.1111/tbed.14027
  4. Ehlers J, Krüger A, Rakotondranary SJ, et al. Molecular detection of Rickettsia spp., Borrelia spp., Bartonella spp. and Yersinia pestis in ectoparasites of endemic and domestic animals in southwest Madagascar. Acta Trop. 2020;205:105339. doi:10.1016/j.actatropica.2020.105339
  5. Boughton RK, Atwell JW, Schoech SJ. An introduced generalist parasite, the sticktight flea (Echidnophaga gallinacea), and its pathology in the threatened Florida scrub-jay (Aphelocoma coerulescens). J Parasitol. 2006;92:941-948. doi:10.1645/GE-769R.1
  6. Bitam I, Dittmar K, Parola P, et al. Fleas and flea-borne diseases. Int J Infect Dis. 2010;14:e667-e676. doi:10.1016/j.ijid.2009.11.011
  7. Linardi PM, Santos JLC. Ctenocephalides felis felis vs. Ctenocephalides canis (Siphonaptera: Pulicidae): some issues in correctly identify these species. Rev Bras Parasitol Vet. 2012;21:345-354. doi:10.1590/s1984-29612012000400002
  8. Carlson JC, Fox MS. A sticktight flea removed from the cheek of a two-year-old boy from Los Angeles. Dermatol Online J. 2009;15:4. https://doi.org/10.5070/D36vb8p1b1
  9. Mirzaei M, Ghashghaei O, Yakhchali M. Prevalence of ectoparasites of indigenous chickens from Dalahu region, Kermanshah province, Iran. Turkiye Parazitol Derg. 2016;40:13-16. doi:10.5152/tpd.2016.4185
  10. Farid DS, Sallam NH, Eldein AMS, et al. Cross-sectional seasonal prevalence and relative risk of ectoparasitic infestations of rodents in North Sinai, Egypt. Vet World. 2021;14:2996-3006. doi:10.14202/vetworld.2021.2996-3006
  11. Harman DW, Halliwell RE, Greiner EC. Flea species from dogs and cats in north-central Florida. Vet Parasitol. 1987;23:135-140. doi:10.1016/0304-4017(87)90031-8
  12. Anderson J, Paterek E. Flea bites. StatPearls [Internet]. StatPearls Publishing; 2023. Updated August 8, 2023. Accessed November 27, 2023. https://www.ncbi.nlm.nih.gov/books/NBK541118/
  13. Gyimesi ZS, Hayden ER, Greiner EC. Sticktight flea (Echidnophaga gallinacea) infestation in a Victoria crowned pigeon (Goura victoria). J Zoo Wildl Med. 2007;38:594-596. doi:10.1638/2007-0062.1
References
  1. Hubbart JA, Jachowski DS, Eads DA. Seasonal and among-site variation in the occurrence and abundance of fleas on California ground squirrels (Otospermophilus beecheyi). J Vector Ecol. 2011;36:117-123. doi:10.1111/j.1948-7134.2011.00148.x
  2. Jiang J, Maina AN, Knobel DL, et al. Molecular detection of Rickettsia felis and Candidatus Rickettsia asemboensis in fleas from human habitats, Asembo, Kenya. Vector Borne Zoonotic Dis. 2013;13:550-558. doi:10.1089/vbz.2012.1123
  3. López-Pérez AM, Chaves A, Sánchez-Montes S, et al. Diversity of rickettsiae in domestic, synanthropic, and sylvatic mammals and their ectoparasites in a spotted fever-epidemic region at the western US-Mexico border. Transbound Emerg Dis. 2022;69:609-622. doi:10.1111/tbed.14027
  4. Ehlers J, Krüger A, Rakotondranary SJ, et al. Molecular detection of Rickettsia spp., Borrelia spp., Bartonella spp. and Yersinia pestis in ectoparasites of endemic and domestic animals in southwest Madagascar. Acta Trop. 2020;205:105339. doi:10.1016/j.actatropica.2020.105339
  5. Boughton RK, Atwell JW, Schoech SJ. An introduced generalist parasite, the sticktight flea (Echidnophaga gallinacea), and its pathology in the threatened Florida scrub-jay (Aphelocoma coerulescens). J Parasitol. 2006;92:941-948. doi:10.1645/GE-769R.1
  6. Bitam I, Dittmar K, Parola P, et al. Fleas and flea-borne diseases. Int J Infect Dis. 2010;14:e667-e676. doi:10.1016/j.ijid.2009.11.011
  7. Linardi PM, Santos JLC. Ctenocephalides felis felis vs. Ctenocephalides canis (Siphonaptera: Pulicidae): some issues in correctly identify these species. Rev Bras Parasitol Vet. 2012;21:345-354. doi:10.1590/s1984-29612012000400002
  8. Carlson JC, Fox MS. A sticktight flea removed from the cheek of a two-year-old boy from Los Angeles. Dermatol Online J. 2009;15:4. https://doi.org/10.5070/D36vb8p1b1
  9. Mirzaei M, Ghashghaei O, Yakhchali M. Prevalence of ectoparasites of indigenous chickens from Dalahu region, Kermanshah province, Iran. Turkiye Parazitol Derg. 2016;40:13-16. doi:10.5152/tpd.2016.4185
  10. Farid DS, Sallam NH, Eldein AMS, et al. Cross-sectional seasonal prevalence and relative risk of ectoparasitic infestations of rodents in North Sinai, Egypt. Vet World. 2021;14:2996-3006. doi:10.14202/vetworld.2021.2996-3006
  11. Harman DW, Halliwell RE, Greiner EC. Flea species from dogs and cats in north-central Florida. Vet Parasitol. 1987;23:135-140. doi:10.1016/0304-4017(87)90031-8
  12. Anderson J, Paterek E. Flea bites. StatPearls [Internet]. StatPearls Publishing; 2023. Updated August 8, 2023. Accessed November 27, 2023. https://www.ncbi.nlm.nih.gov/books/NBK541118/
  13. Gyimesi ZS, Hayden ER, Greiner EC. Sticktight flea (Echidnophaga gallinacea) infestation in a Victoria crowned pigeon (Goura victoria). J Zoo Wildl Med. 2007;38:594-596. doi:10.1638/2007-0062.1
Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
E15-E16
Page Number
E15-E16
Publications
Publications
Topics
Article Type
Display Headline
What’s Eating You? Update on the Sticktight Flea (Echidnophaga gallinacea)
Display Headline
What’s Eating You? Update on the Sticktight Flea (Echidnophaga gallinacea)
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Huebsch</fileName> <TBEID>0C02EF2C.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02EF2C</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Huebsch</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20231214T103107</firstPublished> <LastPublished>20231214T103107</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20231214T103106</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Penny Lane Huebsch, MS; Dirk M. Elston, MD</byline> <bylineText>Penny Lane Huebsch, MS; Dirk M. Elston, MD</bylineText> <bylineFull>Penny Lane Huebsch, MS; Dirk M. Elston, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>E15-E16</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Fleas (order Siphonaptera) are vectors for various diseases, such as plague (as carriers of Yersinia pestis) and rickettsial infections.1-4 The sticktight flea </metaDescription> <articlePDF>299678</articlePDF> <teaserImage/> <title>What’s Eating You? Update on the Sticktight Flea (Echidnophaga gallinacea)</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2023</pubPubdateYear> <pubPubdateMonth>December</pubPubdateMonth> <pubPubdateDay/> <pubVolume>112</pubVolume> <pubNumber>6</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2159</CMSID> </CMSIDs> <keywords> <keyword>infectious disease</keyword> <keyword> sticktight flea</keyword> <keyword> echidnophaga gallinacea</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>December 2023</pubIssueName> <pubArticleType>Departments | 2159</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">60</term> </sections> <topics> <term canonical="true">234</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002684.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>What’s Eating You? Update on the Sticktight Flea (Echidnophaga gallinacea)</title> <deck/> </itemMeta> <itemContent> <p class="abstract">The sticktight flea (<i>Echidnophaga gallinacea</i>), a carrier of plague(<i>Yersinia pestis</i>), rickettsial infections, and other diseases, can be found in warm climates. The flea attaches to a host by embedding its head in the skin for days at a time. Most human infestations occur in individuals who handle infested animals. The sticktight flea can cause delayed erythema of the skin surrounding the embedded head. Common treatments include oral or topical antihistamines or a topical corticosteroid applied to the affected area.</p> <p> <em><i>Cutis.</i> 2023;112:E15-E16.</em> </p> <p>Fleas (order Siphonaptera) are vectors for various diseases, such as plague (as carriers of <i>Yersinia pestis</i>) and<i> </i>rickettsial infections<i>.</i><sup>1-4</sup><i> </i>The sticktight flea (<i>Echidnophaga gallinacea</i>) commonly is seen on birds and mammals, including ground squirrels, dogs, cats, and rodents, and can attach to its host for days at a time by burrowing its head into the skin. Similar to other fleas, the sticktight flea needs a blood supply to reproduce.<sup>5</sup> Therefore, it is important to study the sticktight flea, its habitat, and infection patterns to improve public health and prevent infestation.</p> <h3>Identification</h3> <p><i>Echidnophaga gallinacea</i> is named for the female flea’s behavior—it “sticks tight” to the surface of the host by embedding its head into the skin for days at a time.<sup>5</sup> The sticktight flea and the rat flea (<i>Xenopsylla cheopis</i>) can be differentiated by the sticktight’s reduced thorax and lack of a pleural rod (the vertical ridge that divides the mesosternum above the second pair of legs)(Figure, A and B). The sticktight flea can be differentiated from the dog flea (<i>Ctenocephalides canis</i>) and the cat flea (<i>Ctenocephalides felis</i>) by its lack of genal ctenidia (horizontal combs in the mustache area) and pronotal ctenidia (vertical combs behind the head)(Figure, B and C).<sup>6,7</sup> Other defining features of <i>E gallinacea</i> include 2 pairs of large postantennal setae (hairs) on its anteriorly flattened head; a <i>C</i>-shaped reproductive organ known as the spermatheca; and broad maxillary lacinia (Figure, C).<sup>8</sup> </p> <h3>Habitat, Seasonality, and Behavior</h3> <p><i>Echidnophaga gallinacea</i> commonly infests the comb, wattles, and surrounding ears of chickens; the flea also has been found on dogs, cats, rodents, and other species of birds.<sup>9</sup> The sticktight flea is more prevalent in summer and autumn, which may explain its predominance in warmer climates, including California, Florida, Mexico, Egypt, Africa, and Iran.<sup>1,9-11</sup> </p> <p>When a female sticktight flea begins to feed, it stays on the host for days at a time, waiting for a male.<sup>5</sup> The female deposits its fertilized eggs in nests on the host or in lesions caused by infestation. Eventually, eggs hatch and fall into soil, where they lay dormant or grow to adulthood.<sup>5</sup></p> <h3>Cutaneous Reaction to Infestation</h3> <p>Flea bites cause a hypersensitivity reaction, with pruritic pustules and erythematous papules that have a central punctum.<sup>12</sup> In a reported case in Los Angeles, California, a female sticktight flea buried itself into the cheek of a young boy for more than 12 hours. The lesion was not marked by surrounding erythema, tenderness, pruritus, or swelling; however, several days after the flea was removed, erythema developed at the site then spontaneously resolved.<sup>7</sup> In a study of dogs that were infested with <i>E gallinacea</i>, the flea never disengaged to attach to a human; when the flea was deliberately placed on a human, it fed and left hastily.<sup>11</sup> </p> <h3>Management</h3> <p>Because <i>E gallinacea</i> burrows its head into the skin, the best removal method is applying slow gentle traction under sterile conditions to ensure removal of mouthparts.<sup>7</sup> An oral antihistamine can be administered or a topical antihistamine or corticosteroid can be applied to the affected area.<sup>12</sup> Flea infestation should be treated with an insecticide. Affected animals should be treated by a veterinarian using a pesticide, such as fipronil, selamectin, imidacloprid, metaflumizone, nitenpyram, lufenuron, methoprene, or pyriproxyfen.<sup>13</sup></p> <h2>REFERENCES</h2> <p class="reference"> 1. Hubbart JA, Jachowski DS, Eads DA. Seasonal and among-site variation in the occurrence and abundance of fleas on California ground squirrels (<i>Otospermophilus beecheyi</i>). <i>J Vector Ecol</i>. 2011;36:117-123. doi:10.1111/j.1948-7134.2011.00148.x<br/><br/> 2. Jiang J, Maina AN, Knobel DL, et al. Molecular detection of <i>Rickettsia felis</i> and <i>Candidatus Rickettsia</i> asemboensis in fleas from human habitats, Asembo, Kenya. <i>Vector Borne Zoonotic Dis</i>. 2013;13:550-558. doi:10.1089/vbz.2012.1123<br/><br/> 3. López-Pérez AM, Chaves A, Sánchez-Montes S, et al. Diversity of rickettsiae in domestic, synanthropic, and sylvatic mammals and their ectoparasites in a spotted fever-epidemic region at the western US-Mexico border. <i>Transbound Emerg Dis</i>. 2022;69:609-622. doi:10.1111/tbed.14027<br/><br/> 4. Ehlers J, Krüger A, Rakotondranary SJ, et al. Molecular detection of <i>Rickettsia spp</i>., <i>Borrelia spp</i>., <i>Bartonella spp</i>. and <i>Yersinia pestis</i> in ectoparasites of endemic and domestic animals in southwest Madagascar. <i>Acta Trop</i>. 2020;205:105339. doi:10.1016/j.actatropica.2020.105339<br/><br/> 5. Boughton RK, Atwell JW, Schoech SJ. An introduced generalist parasite, the sticktight flea (<i>Echidnophaga gallinacea</i>), and its pathology in the threatened Florida scrub-jay (<i>Aphelocoma coerulescens</i>). <i>J Parasitol</i>. 2006;92:941-948. doi:10.1645/GE-769R.1<br/><br/> 6. Bitam I, Dittmar K, Parola P, et al. Fleas and flea-borne diseases. <i>Int J Infect Dis</i>. 2010;14:e667-e676. doi:10.1016/j.ijid.2009.11.011<br/><br/> 7. Linardi PM, Santos JLC. <em>Ctenocephalides felis felis</em> vs. <em>Ctenocephalides canis</em> (Siphonaptera: Pulicidae): some issues in correctly identify these species. <i>Rev Bras Parasitol Vet</i>. 2012;21:345-354. doi:10.1590/s1984-29612012000400002 <br/><br/> 8. Carlson JC, Fox MS. A sticktight flea removed from the cheek of a two-year-old boy from Los Angeles. <i>Dermatol Online J</i>. 2009;15:4. https://doi.org/10.5070/D36vb8p1b1<br/><br/> 9. Mirzaei M, Ghashghaei O, Yakhchali M. Prevalence of ectoparasites of indigenous chickens from Dalahu region, Kermanshah province, Iran. <i>Turkiye Parazitol Derg</i>. 2016;40:13-16. doi:10.5152/tpd.2016.4185<br/><br/>10. Farid DS, Sallam NH, Eldein AMS, et al. Cross-sectional seasonal prevalence and relative risk of ectoparasitic infestations of rodents in North Sinai, Egypt. <i>Vet World</i>. 2021;14:2996-3006. doi:10.14202/vetworld.2021.2996-3006<br/><br/>11. Harman DW, Halliwell RE, Greiner EC. Flea species from dogs and cats in north-central Florida. <i>Vet Parasitol</i>. 1987;23:135-140. doi:10.1016/0304-4017(87)90031-8<br/><br/>12. Anderson J, Paterek E. Flea bites. <i>StatPearls </i>[Internet]. StatPearls Publishing; 2023. Updated August 8, 2023. Accessed November 27, 2023. https://www.ncbi.nlm.nih.gov/books/NBK541118/<br/><br/>13. Gyimesi ZS, Hayden ER, Greiner EC. Sticktight flea (<i>Echidnophaga gallinacea</i>) infestation in a Victoria crowned pigeon (<i>Goura victoria</i>). <i>J Zoo Wildl Med</i>. 2007;38:594-596. doi:10.1638/2007-0062.1</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">From the Medical University of South Carolina, Charleston. Penny Lane Huebsch is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.</p> <p class="disclosure">The authors report no conflict of interest.<br/><br/>The images are in the public domain.<br/><br/>Correspondence: Penny Lane Huebsch, MS, 96 Jonathan Lucas St, Ste 601, Charleston, SC 29425 (huebsch@musc.edu).<br/><br/>doi:10.12788/cutis.0916</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>The sticktight flea (<i>Echidnophaga gallinacea</i>) attaches to its host by embedding its head in the skin for days at a time.</li> <li>Unlike other fleas that bite and run, the sticktight flea can be identified dermoscopically.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • The sticktight flea (Echidnophaga gallinacea) attaches to its host by embedding its head in the skin for days at a time.
  • Unlike other fleas that bite and run, the sticktight flea can be identified dermoscopically.
  • The sticktight flea serves as a vector for plague as a carrier of Yersinia pestis, rickettsial infections, and other diseases.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
18002684.SIG
Disable zoom
Off

Botanical Briefs: Australian Stinging Tree (Dendrocnide moroides)

Article Type
Changed
Tue, 10/31/2023 - 09:34
Display Headline
Botanical Briefs: Australian Stinging Tree (Dendrocnide moroides)

Clinical Importance

Dendrocnide moroides is arguably the most brutal of stinging plants, even leading to death in dogs, horses, and humans in rare cases.1-3 Commonly called gympie-gympie (based on its discovery by gold miners near the town of Gympie in Queensland, Australia), D moroides also has been referred to as the mulberrylike stinging tree or stinger.2,4-6

Family and Nomenclature

The Australian stinging tree belongs to the family Urticaceae (known as the nettle family) within the order Rosales.1,2,3,5 Urticaceae is derived from the Latin term urere (to burn)—an apt description of the clinical experience of patients with D moroides–induced urticaria.

Urticaceae includes 54 genera, comprising herbs, shrubs, small trees, and vines found predominantly in tropical regions. Dendrocnide comprises approximately 40 species, all commonly known in Australia as stinging trees.2,7,8

Distribution

Dendrocnide moroides is found in the rainforests of Australia and Southeast Asia.2 Because the plant has a strong need for sunlight and wind protection, it typically is found in light-filled gaps within the rainforest, in moist ravines, along the edges of creeks, and on land bordering the rainforest.3,6

Appearance

Although D moroides is referred to as a tree, it is an understory shrub that typically grows to 3 m, with heart-shaped, serrated, dark green leaves that are 50-cm wide (Figure 1).6 The leaves are produced consistently through the year, with variable growth depending on the season.9

DeVore_stinging_tree_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Leaf%20and%20fruit%20of%20%3Cem%3EDendrocnide%20moroides%3C%2Fem%3E.%3C%2Fp%3E

The plant is covered in what appears to be soft downy fur made up of trichomes (or plant hairs).1,6 The density of the hairs on leaves decreases as they age.2,9 The fruit, which is actually edible (if one is careful to avoid hairs), appears similar to red to dark purple raspberries growing on long stems.5,6

Cutaneous Manifestations

Symptoms of contact with the stems and leaves of D moroides range from slight irritation to serious neurologic disorders, including neuropathy. The severity of the reaction depends on the person, how much skin was contacted, and how one came into contact with the plant.1,5 Upon touch, there is an immediate reaction, with burning, urticaria, and edema. Pain increases, peaking 30 minutes later; then the pain slowly subsides.1 Tachycardia and throbbing regional lymphadenopathy can occur for 1 to 4 hours.1,6

 

 

Cutaneous Findings—Examination reveals immediate piloerection, erythema due to arteriolar dilation, and local swelling.2 These findings may disappear after 1 hour or last as long as 24 hours.1 Although objective signs may fade, subjective pain, pruritus, and burning can persist for months.3

Dermatitis-Inducing Plant Parts

After contact with the stems or leaves, the sharp trichomes become embedded in the skin, making them difficult to remove.1 The toxins are contained in siliceous hairs that the human body cannot break down.3 Symptoms can be experienced for as long as 1 year after contact, especially when the skin is pressed firmly or washed with hot or cold water.3,6 Because the plant’s hairs are shed continuously, being in close proximity to D moroides for longer than 20 minutes can lead to extreme sneezing, nosebleeds, and major respiratory damage from inhaling hairs.1,6,9

The stinging hairs of D moroides differ from irritant hairs on other plants because they contain physiologically active substances. Stinging hairs are classified as either a hypodermic syringe, which expels liquid only, or as a tragia-type syringe, in which liquid and sharp crystals are injected.

The Australian stinging tree falls into the first of these 2 groups (Figure 2)1; the sharp tip of the hair breaks on contact, leading to expulsion of the toxin into skin.1,4 The hairs function as a defense against mammalian herbivores but typically have no impact on pests.1 Nocturnal beetles and on occasion possums and red-legged pademelons dare to eat D moroides.3,6

DeVore_stinging_tree_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Stinging%20hairs%20resembling%20hypodermic%20syringes%20of%20%3Cem%3EDendrocnide%20moroides%3C%2Fem%3E.%26nbsp%3B%3C%2Fp%3E

The Irritant

Initially, formic acid was proposed as the irritant chemical in D moroides1; other candidates have included neurotransmitters, such as histamine, acetylcholine, and serotonin, as well as inorganic ions, such as potassium. These compounds may play a role but none explain the persistent sensory effects and years-long stable nature of the toxin.1,4

The most likely culprit irritant is a member of a newly discovered family of neurotoxins, the gympietides. These knot-shaped chemicals, found in D moroides and some spider venoms, have the ability to activate voltage-gated sodium channels of cutaneous neurons and cause local cutaneous vasodilation by stimulating neurotransmitter release.4 These neurotoxins not only generate pain but also suppress the mechanism used to interrupt those pain signals.10 Synthesized gympietides can replicate the effects of natural contact, indicating that they are the primary active toxins. These toxins are ultrastable, thus producing lasting effects.1

Although much is understood about the evolution and distribution of D moroides and the ecological role that it plays, there is still more to learn about the plant’s toxicology.

 

 

Prevention and Treatment

Prevention—Dendrocnide moroides dermatitis is best prevented by avoiding contact with the plant and related species, as well as wearing upper body clothing with long sleeves, pants, and boots, though plant hairs can still penetrate garments and sting.2,3

Therapy—There is no reversal therapy of D moroides dermatitis but symptoms can be managed.4 For pain, analgesics, such as opioids, have been used; on occasion, however, pain is so intense that even morphine does not help.4,10

Systemic or topical corticosteroids are the main therapy for many forms of plant-induced dermatitis because they are able to decrease cytokine production and stop lymphocyte production. Adding an oral antihistamine can alleviate histamine-mediated pruritus but not pruritus that is mediated by other chemicals.11

Other methods of relieving symptoms of D moroides dermatitis have been proposed or reported anecdotally. Diluted hydrochloric acid can be applied to the skin to denature remaining toxin.4 The sap of Alocasia brisbanensis (the cunjevoi plant) can be rubbed on affected areas to provide a cooling effect, but do not allow A brisbanensis sap to enter the mouth, as it contains calcium oxalate, a toxic irritant found in dumb cane (Dieffenbachia species). The roots of the Australian stinging tree also can be ground and made into a paste, which is applied to the skin.3 However, given the stability of the toxin, we do not recommend these remedies.

Instead, heavy-duty masking tape or hot wax can be applied to remove plant hairs from the skin. The most successful method of removing plant hair is hair removal wax strips, which are considered an essential component of a first aid kit where D moroides is found.3

References
  1. Ensikat H-J, Wessely H, Engeser M, et al. Distribution, ecology, chemistry and toxicology of plant stinging hairs. Toxins (Basel). 2021;13:141. doi:10.3390/toxins13020141
  2. Schmitt C, Parola P, de Haro L. Painful sting after exposure to Dendrocnide sp: two case reports. Wilderness Environ Med. 2013;24:471-473. doi:10.1016/j.wem.2013.03.021
  3. Hurley M. Selective stingers. ECOS. 2000;105:18-23. Accessed October 13, 2023. https://www.writingclearscience.com.au/wp-content/uploads/2015/06/stingers.pdf
  4. Gilding EK, Jami S, Deuis JR, et al. Neurotoxic peptides from the venom of the giant Australian stinging tree. Sci Adv. 2020;6:eabb8828. doi:10.1126/sciadv.abb8828
  5. Dendrocnide moroides. James Cook University Australia website. Accessed Accessed October 13, 2023. https://www.jcu.edu.au/discover-nature-at-jcu/plants/plants-by-scientific-name2/dendrocnide-moroides
  6. Hurley M. ‘The worst kind of pain you can imagine’—what it’s like to be stung by a stinging tree. The Conversation. September 28, 2018. Accessed October 13, 2023. https://theconversation.com/the-worst-kind-of-pain-you-can-imagine-what-its-like-to-be-stung-by-a-stinging-tree-103220
  7. Urticaceae: plant family. Britannica [Internet]. Accessed October 13, 2023. https://www.britannica.com/plant/Urticaceae
  8. Stinging trees (genus Dendrocnide). iNaturalist.ca [Internet]. Accessed October 13, 2023. https://inaturalist.ca/taxa/129502-Dendrocnide
  9. Hurley M. Growth dynamics and leaf quality of the stinging trees Dendrocnide moroides and Dendrocnide cordifolia (family Urticaceae) in Australian tropical rainforest: implications for herbivores. Aust J Bot. 2000;48:191-201. doi:10.1071/BT98006
  10. How the giant stinging tree of Australia can inflict months of agony. Nature. September 17, 2020. Accessed October 13, 2023. https://www.nature.com/articles/d41586-020-02668-9
  11. Chang Y-T, Shen J-J, Wong W-R, et al. Alternative therapy for autosensitization dermatitis. Chang Gung Med J. 2009;32:668-673.
Article PDF
Author and Disclosure Information

Dr. DeVore is from the Medical University of South Carolina, Charleston. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.

The authors report no conflict of interest.

Correspondence: Ansley C. DeVore, MD, 363 Twin Oaks Dr, Spartanburg, SC 29306 (devorea@musc.edu).

Issue
Cutis - 112(5)
Publications
Topics
Page Number
250-252
Sections
Author and Disclosure Information

Dr. DeVore is from the Medical University of South Carolina, Charleston. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.

The authors report no conflict of interest.

Correspondence: Ansley C. DeVore, MD, 363 Twin Oaks Dr, Spartanburg, SC 29306 (devorea@musc.edu).

Author and Disclosure Information

Dr. DeVore is from the Medical University of South Carolina, Charleston. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.

The authors report no conflict of interest.

Correspondence: Ansley C. DeVore, MD, 363 Twin Oaks Dr, Spartanburg, SC 29306 (devorea@musc.edu).

Article PDF
Article PDF

Clinical Importance

Dendrocnide moroides is arguably the most brutal of stinging plants, even leading to death in dogs, horses, and humans in rare cases.1-3 Commonly called gympie-gympie (based on its discovery by gold miners near the town of Gympie in Queensland, Australia), D moroides also has been referred to as the mulberrylike stinging tree or stinger.2,4-6

Family and Nomenclature

The Australian stinging tree belongs to the family Urticaceae (known as the nettle family) within the order Rosales.1,2,3,5 Urticaceae is derived from the Latin term urere (to burn)—an apt description of the clinical experience of patients with D moroides–induced urticaria.

Urticaceae includes 54 genera, comprising herbs, shrubs, small trees, and vines found predominantly in tropical regions. Dendrocnide comprises approximately 40 species, all commonly known in Australia as stinging trees.2,7,8

Distribution

Dendrocnide moroides is found in the rainforests of Australia and Southeast Asia.2 Because the plant has a strong need for sunlight and wind protection, it typically is found in light-filled gaps within the rainforest, in moist ravines, along the edges of creeks, and on land bordering the rainforest.3,6

Appearance

Although D moroides is referred to as a tree, it is an understory shrub that typically grows to 3 m, with heart-shaped, serrated, dark green leaves that are 50-cm wide (Figure 1).6 The leaves are produced consistently through the year, with variable growth depending on the season.9

DeVore_stinging_tree_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Leaf%20and%20fruit%20of%20%3Cem%3EDendrocnide%20moroides%3C%2Fem%3E.%3C%2Fp%3E

The plant is covered in what appears to be soft downy fur made up of trichomes (or plant hairs).1,6 The density of the hairs on leaves decreases as they age.2,9 The fruit, which is actually edible (if one is careful to avoid hairs), appears similar to red to dark purple raspberries growing on long stems.5,6

Cutaneous Manifestations

Symptoms of contact with the stems and leaves of D moroides range from slight irritation to serious neurologic disorders, including neuropathy. The severity of the reaction depends on the person, how much skin was contacted, and how one came into contact with the plant.1,5 Upon touch, there is an immediate reaction, with burning, urticaria, and edema. Pain increases, peaking 30 minutes later; then the pain slowly subsides.1 Tachycardia and throbbing regional lymphadenopathy can occur for 1 to 4 hours.1,6

 

 

Cutaneous Findings—Examination reveals immediate piloerection, erythema due to arteriolar dilation, and local swelling.2 These findings may disappear after 1 hour or last as long as 24 hours.1 Although objective signs may fade, subjective pain, pruritus, and burning can persist for months.3

Dermatitis-Inducing Plant Parts

After contact with the stems or leaves, the sharp trichomes become embedded in the skin, making them difficult to remove.1 The toxins are contained in siliceous hairs that the human body cannot break down.3 Symptoms can be experienced for as long as 1 year after contact, especially when the skin is pressed firmly or washed with hot or cold water.3,6 Because the plant’s hairs are shed continuously, being in close proximity to D moroides for longer than 20 minutes can lead to extreme sneezing, nosebleeds, and major respiratory damage from inhaling hairs.1,6,9

The stinging hairs of D moroides differ from irritant hairs on other plants because they contain physiologically active substances. Stinging hairs are classified as either a hypodermic syringe, which expels liquid only, or as a tragia-type syringe, in which liquid and sharp crystals are injected.

The Australian stinging tree falls into the first of these 2 groups (Figure 2)1; the sharp tip of the hair breaks on contact, leading to expulsion of the toxin into skin.1,4 The hairs function as a defense against mammalian herbivores but typically have no impact on pests.1 Nocturnal beetles and on occasion possums and red-legged pademelons dare to eat D moroides.3,6

DeVore_stinging_tree_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Stinging%20hairs%20resembling%20hypodermic%20syringes%20of%20%3Cem%3EDendrocnide%20moroides%3C%2Fem%3E.%26nbsp%3B%3C%2Fp%3E

The Irritant

Initially, formic acid was proposed as the irritant chemical in D moroides1; other candidates have included neurotransmitters, such as histamine, acetylcholine, and serotonin, as well as inorganic ions, such as potassium. These compounds may play a role but none explain the persistent sensory effects and years-long stable nature of the toxin.1,4

The most likely culprit irritant is a member of a newly discovered family of neurotoxins, the gympietides. These knot-shaped chemicals, found in D moroides and some spider venoms, have the ability to activate voltage-gated sodium channels of cutaneous neurons and cause local cutaneous vasodilation by stimulating neurotransmitter release.4 These neurotoxins not only generate pain but also suppress the mechanism used to interrupt those pain signals.10 Synthesized gympietides can replicate the effects of natural contact, indicating that they are the primary active toxins. These toxins are ultrastable, thus producing lasting effects.1

Although much is understood about the evolution and distribution of D moroides and the ecological role that it plays, there is still more to learn about the plant’s toxicology.

 

 

Prevention and Treatment

Prevention—Dendrocnide moroides dermatitis is best prevented by avoiding contact with the plant and related species, as well as wearing upper body clothing with long sleeves, pants, and boots, though plant hairs can still penetrate garments and sting.2,3

Therapy—There is no reversal therapy of D moroides dermatitis but symptoms can be managed.4 For pain, analgesics, such as opioids, have been used; on occasion, however, pain is so intense that even morphine does not help.4,10

Systemic or topical corticosteroids are the main therapy for many forms of plant-induced dermatitis because they are able to decrease cytokine production and stop lymphocyte production. Adding an oral antihistamine can alleviate histamine-mediated pruritus but not pruritus that is mediated by other chemicals.11

Other methods of relieving symptoms of D moroides dermatitis have been proposed or reported anecdotally. Diluted hydrochloric acid can be applied to the skin to denature remaining toxin.4 The sap of Alocasia brisbanensis (the cunjevoi plant) can be rubbed on affected areas to provide a cooling effect, but do not allow A brisbanensis sap to enter the mouth, as it contains calcium oxalate, a toxic irritant found in dumb cane (Dieffenbachia species). The roots of the Australian stinging tree also can be ground and made into a paste, which is applied to the skin.3 However, given the stability of the toxin, we do not recommend these remedies.

Instead, heavy-duty masking tape or hot wax can be applied to remove plant hairs from the skin. The most successful method of removing plant hair is hair removal wax strips, which are considered an essential component of a first aid kit where D moroides is found.3

Clinical Importance

Dendrocnide moroides is arguably the most brutal of stinging plants, even leading to death in dogs, horses, and humans in rare cases.1-3 Commonly called gympie-gympie (based on its discovery by gold miners near the town of Gympie in Queensland, Australia), D moroides also has been referred to as the mulberrylike stinging tree or stinger.2,4-6

Family and Nomenclature

The Australian stinging tree belongs to the family Urticaceae (known as the nettle family) within the order Rosales.1,2,3,5 Urticaceae is derived from the Latin term urere (to burn)—an apt description of the clinical experience of patients with D moroides–induced urticaria.

Urticaceae includes 54 genera, comprising herbs, shrubs, small trees, and vines found predominantly in tropical regions. Dendrocnide comprises approximately 40 species, all commonly known in Australia as stinging trees.2,7,8

Distribution

Dendrocnide moroides is found in the rainforests of Australia and Southeast Asia.2 Because the plant has a strong need for sunlight and wind protection, it typically is found in light-filled gaps within the rainforest, in moist ravines, along the edges of creeks, and on land bordering the rainforest.3,6

Appearance

Although D moroides is referred to as a tree, it is an understory shrub that typically grows to 3 m, with heart-shaped, serrated, dark green leaves that are 50-cm wide (Figure 1).6 The leaves are produced consistently through the year, with variable growth depending on the season.9

DeVore_stinging_tree_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Leaf%20and%20fruit%20of%20%3Cem%3EDendrocnide%20moroides%3C%2Fem%3E.%3C%2Fp%3E

The plant is covered in what appears to be soft downy fur made up of trichomes (or plant hairs).1,6 The density of the hairs on leaves decreases as they age.2,9 The fruit, which is actually edible (if one is careful to avoid hairs), appears similar to red to dark purple raspberries growing on long stems.5,6

Cutaneous Manifestations

Symptoms of contact with the stems and leaves of D moroides range from slight irritation to serious neurologic disorders, including neuropathy. The severity of the reaction depends on the person, how much skin was contacted, and how one came into contact with the plant.1,5 Upon touch, there is an immediate reaction, with burning, urticaria, and edema. Pain increases, peaking 30 minutes later; then the pain slowly subsides.1 Tachycardia and throbbing regional lymphadenopathy can occur for 1 to 4 hours.1,6

 

 

Cutaneous Findings—Examination reveals immediate piloerection, erythema due to arteriolar dilation, and local swelling.2 These findings may disappear after 1 hour or last as long as 24 hours.1 Although objective signs may fade, subjective pain, pruritus, and burning can persist for months.3

Dermatitis-Inducing Plant Parts

After contact with the stems or leaves, the sharp trichomes become embedded in the skin, making them difficult to remove.1 The toxins are contained in siliceous hairs that the human body cannot break down.3 Symptoms can be experienced for as long as 1 year after contact, especially when the skin is pressed firmly or washed with hot or cold water.3,6 Because the plant’s hairs are shed continuously, being in close proximity to D moroides for longer than 20 minutes can lead to extreme sneezing, nosebleeds, and major respiratory damage from inhaling hairs.1,6,9

The stinging hairs of D moroides differ from irritant hairs on other plants because they contain physiologically active substances. Stinging hairs are classified as either a hypodermic syringe, which expels liquid only, or as a tragia-type syringe, in which liquid and sharp crystals are injected.

The Australian stinging tree falls into the first of these 2 groups (Figure 2)1; the sharp tip of the hair breaks on contact, leading to expulsion of the toxin into skin.1,4 The hairs function as a defense against mammalian herbivores but typically have no impact on pests.1 Nocturnal beetles and on occasion possums and red-legged pademelons dare to eat D moroides.3,6

DeVore_stinging_tree_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Stinging%20hairs%20resembling%20hypodermic%20syringes%20of%20%3Cem%3EDendrocnide%20moroides%3C%2Fem%3E.%26nbsp%3B%3C%2Fp%3E

The Irritant

Initially, formic acid was proposed as the irritant chemical in D moroides1; other candidates have included neurotransmitters, such as histamine, acetylcholine, and serotonin, as well as inorganic ions, such as potassium. These compounds may play a role but none explain the persistent sensory effects and years-long stable nature of the toxin.1,4

The most likely culprit irritant is a member of a newly discovered family of neurotoxins, the gympietides. These knot-shaped chemicals, found in D moroides and some spider venoms, have the ability to activate voltage-gated sodium channels of cutaneous neurons and cause local cutaneous vasodilation by stimulating neurotransmitter release.4 These neurotoxins not only generate pain but also suppress the mechanism used to interrupt those pain signals.10 Synthesized gympietides can replicate the effects of natural contact, indicating that they are the primary active toxins. These toxins are ultrastable, thus producing lasting effects.1

Although much is understood about the evolution and distribution of D moroides and the ecological role that it plays, there is still more to learn about the plant’s toxicology.

 

 

Prevention and Treatment

Prevention—Dendrocnide moroides dermatitis is best prevented by avoiding contact with the plant and related species, as well as wearing upper body clothing with long sleeves, pants, and boots, though plant hairs can still penetrate garments and sting.2,3

Therapy—There is no reversal therapy of D moroides dermatitis but symptoms can be managed.4 For pain, analgesics, such as opioids, have been used; on occasion, however, pain is so intense that even morphine does not help.4,10

Systemic or topical corticosteroids are the main therapy for many forms of plant-induced dermatitis because they are able to decrease cytokine production and stop lymphocyte production. Adding an oral antihistamine can alleviate histamine-mediated pruritus but not pruritus that is mediated by other chemicals.11

Other methods of relieving symptoms of D moroides dermatitis have been proposed or reported anecdotally. Diluted hydrochloric acid can be applied to the skin to denature remaining toxin.4 The sap of Alocasia brisbanensis (the cunjevoi plant) can be rubbed on affected areas to provide a cooling effect, but do not allow A brisbanensis sap to enter the mouth, as it contains calcium oxalate, a toxic irritant found in dumb cane (Dieffenbachia species). The roots of the Australian stinging tree also can be ground and made into a paste, which is applied to the skin.3 However, given the stability of the toxin, we do not recommend these remedies.

Instead, heavy-duty masking tape or hot wax can be applied to remove plant hairs from the skin. The most successful method of removing plant hair is hair removal wax strips, which are considered an essential component of a first aid kit where D moroides is found.3

References
  1. Ensikat H-J, Wessely H, Engeser M, et al. Distribution, ecology, chemistry and toxicology of plant stinging hairs. Toxins (Basel). 2021;13:141. doi:10.3390/toxins13020141
  2. Schmitt C, Parola P, de Haro L. Painful sting after exposure to Dendrocnide sp: two case reports. Wilderness Environ Med. 2013;24:471-473. doi:10.1016/j.wem.2013.03.021
  3. Hurley M. Selective stingers. ECOS. 2000;105:18-23. Accessed October 13, 2023. https://www.writingclearscience.com.au/wp-content/uploads/2015/06/stingers.pdf
  4. Gilding EK, Jami S, Deuis JR, et al. Neurotoxic peptides from the venom of the giant Australian stinging tree. Sci Adv. 2020;6:eabb8828. doi:10.1126/sciadv.abb8828
  5. Dendrocnide moroides. James Cook University Australia website. Accessed Accessed October 13, 2023. https://www.jcu.edu.au/discover-nature-at-jcu/plants/plants-by-scientific-name2/dendrocnide-moroides
  6. Hurley M. ‘The worst kind of pain you can imagine’—what it’s like to be stung by a stinging tree. The Conversation. September 28, 2018. Accessed October 13, 2023. https://theconversation.com/the-worst-kind-of-pain-you-can-imagine-what-its-like-to-be-stung-by-a-stinging-tree-103220
  7. Urticaceae: plant family. Britannica [Internet]. Accessed October 13, 2023. https://www.britannica.com/plant/Urticaceae
  8. Stinging trees (genus Dendrocnide). iNaturalist.ca [Internet]. Accessed October 13, 2023. https://inaturalist.ca/taxa/129502-Dendrocnide
  9. Hurley M. Growth dynamics and leaf quality of the stinging trees Dendrocnide moroides and Dendrocnide cordifolia (family Urticaceae) in Australian tropical rainforest: implications for herbivores. Aust J Bot. 2000;48:191-201. doi:10.1071/BT98006
  10. How the giant stinging tree of Australia can inflict months of agony. Nature. September 17, 2020. Accessed October 13, 2023. https://www.nature.com/articles/d41586-020-02668-9
  11. Chang Y-T, Shen J-J, Wong W-R, et al. Alternative therapy for autosensitization dermatitis. Chang Gung Med J. 2009;32:668-673.
References
  1. Ensikat H-J, Wessely H, Engeser M, et al. Distribution, ecology, chemistry and toxicology of plant stinging hairs. Toxins (Basel). 2021;13:141. doi:10.3390/toxins13020141
  2. Schmitt C, Parola P, de Haro L. Painful sting after exposure to Dendrocnide sp: two case reports. Wilderness Environ Med. 2013;24:471-473. doi:10.1016/j.wem.2013.03.021
  3. Hurley M. Selective stingers. ECOS. 2000;105:18-23. Accessed October 13, 2023. https://www.writingclearscience.com.au/wp-content/uploads/2015/06/stingers.pdf
  4. Gilding EK, Jami S, Deuis JR, et al. Neurotoxic peptides from the venom of the giant Australian stinging tree. Sci Adv. 2020;6:eabb8828. doi:10.1126/sciadv.abb8828
  5. Dendrocnide moroides. James Cook University Australia website. Accessed Accessed October 13, 2023. https://www.jcu.edu.au/discover-nature-at-jcu/plants/plants-by-scientific-name2/dendrocnide-moroides
  6. Hurley M. ‘The worst kind of pain you can imagine’—what it’s like to be stung by a stinging tree. The Conversation. September 28, 2018. Accessed October 13, 2023. https://theconversation.com/the-worst-kind-of-pain-you-can-imagine-what-its-like-to-be-stung-by-a-stinging-tree-103220
  7. Urticaceae: plant family. Britannica [Internet]. Accessed October 13, 2023. https://www.britannica.com/plant/Urticaceae
  8. Stinging trees (genus Dendrocnide). iNaturalist.ca [Internet]. Accessed October 13, 2023. https://inaturalist.ca/taxa/129502-Dendrocnide
  9. Hurley M. Growth dynamics and leaf quality of the stinging trees Dendrocnide moroides and Dendrocnide cordifolia (family Urticaceae) in Australian tropical rainforest: implications for herbivores. Aust J Bot. 2000;48:191-201. doi:10.1071/BT98006
  10. How the giant stinging tree of Australia can inflict months of agony. Nature. September 17, 2020. Accessed October 13, 2023. https://www.nature.com/articles/d41586-020-02668-9
  11. Chang Y-T, Shen J-J, Wong W-R, et al. Alternative therapy for autosensitization dermatitis. Chang Gung Med J. 2009;32:668-673.
Issue
Cutis - 112(5)
Issue
Cutis - 112(5)
Page Number
250-252
Page Number
250-252
Publications
Publications
Topics
Article Type
Display Headline
Botanical Briefs: Australian Stinging Tree (Dendrocnide moroides)
Display Headline
Botanical Briefs: Australian Stinging Tree (Dendrocnide moroides)
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>DeVore stinging tree</fileName> <TBEID>0C02E7C8.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02E7C8</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Devore stinging tree</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20231030T110243</firstPublished> <LastPublished>20231030T110243</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20231030T110243</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Ansley C. DeVore, MD; Thomas W. McGovern, MD</byline> <bylineText>Ansley C. DeVore, MD; Thomas W. McGovern, MD</bylineText> <bylineFull>Ansley C. DeVore, MD; Thomas W. McGovern, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>250-252</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Dendrocnide moroides is arguably the most brutal of stinging plants, even leading to death in dogs, horses, and humans in rare cases.1-3 Commonly called gympie-</metaDescription> <articlePDF>298804</articlePDF> <teaserImage/> <title>Botanical Briefs: Australian Stinging Tree (Dendrocnide moroides)</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2023</pubPubdateYear> <pubPubdateMonth>November</pubPubdateMonth> <pubPubdateDay/> <pubVolume>112</pubVolume> <pubNumber>5</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2159</CMSID> </CMSIDs> <keywords> <keyword>contact dermatitis</keyword> <keyword> Australian Stinging&#13;Tree</keyword> <keyword> Dendrocnide moroides</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>November 2023</pubIssueName> <pubArticleType>Departments | 2159</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">60</term> </sections> <topics> <term canonical="true">199</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180025e4.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Botanical Briefs: Australian Stinging Tree (Dendrocnide moroides)</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><em>Dendrocnide moroides</em> (also known as gympie-gympie, mulberrylike stinging tree, or stinger) is arguably the most brutal of stinging plants, even leading to death in dogs, horses, and humans in rare cases. They can be recognized as shrubs with heart-shaped, serrated, dark green leaves that are covered in what appears to be soft downy fur with red to dark purple raspberries growing on long stems. After contact, there is immediate piloerection and local swelling, which may disappear after 1 hour or last as long as 24 hours, but the subjective pain, pruritus, and burning can persist for months. One can only treat conservatively with symptom management, and the most successful method of removing plant hair is hair removal wax strips, which are considered an essential component of a first aid kit where <em>D moroides</em> is found.</p> <p> <em><em>Cutis.</em> 2023;112:250-252.</em> </p> <h3>Clinical Importance</h3> <p><i>Dendrocnide moroides</i> is arguably the most brutal of stinging plants, even leading to death in dogs, horses, and humans in rare cases.<sup>1-3</sup> Commonly called <i>gympie-gympie</i> (based on its discovery by gold miners near the town of Gympie in Queensland, Australia), <i>D moroides </i>also has been referred to as the <i>mulberrylike stinging tree</i> or <i>stinger</i>.<sup>2,4-6</sup></p> <h3>Family and Nomenclature</h3> <p>The Australian stinging tree belongs to the family Urticaceae (known as the nettle family) within the order Rosales.<sup>1,2,3,5</sup> Urticaceae is derived from the Latin term <i>urere </i>(to burn)—an apt description of the clinical experience of patients with <i>D moroides</i>–induced urticaria.</p> <p>Urticaceae includes 54 genera, comprising herbs, shrubs, small trees, and vines found predominantly in tropical regions. <i>Dendrocnide</i> comprises approximately 40 species, all commonly known in Australia as stinging trees.<sup>2,7,8</sup> </p> <h3>Distribution</h3> <p><i>Dendrocnide moroides</i> is found in the rainforests of Australia and Southeast Asia.<sup>2</sup> Because the plant has a strong need for sunlight and wind protection, it typically is found in light-filled gaps within the rainforest, in moist ravines, along the edges of creeks, and on land bordering the rainforest.<sup>3,6</sup></p> <h3>Appearance</h3> <p>Although <i>D moroides</i> is referred to as a tree,<i> </i>it is an understory shrub that typically grows to 3 m, with heart-shaped, serrated, dark green leaves that are 50-cm wide (Figure 1).<sup>6</sup> The leaves are produced consistently through the year, with variable growth depending on the season.<sup>9</sup></p> <p>The plant is covered in what appears to be soft downy fur made up of trichomes (or plant hairs).<sup>1,6</sup> The density of the hairs on leaves decreases as they age.<sup>2,9</sup> The fruit, which is actually edible (if one is careful to avoid hairs), appears similar to red to dark purple raspberries growing on long stems.<sup>5,6</sup> </p> <h3>Cutaneous Manifestations</h3> <p>Symptoms of contact with the stems and leaves of <i>D moroides</i> range from slight irritation to serious neurologic disorders, including neuropathy. The severity of the reaction depends on the person, how much skin was contacted, and how one came into contact with the plant.<sup>1,5</sup> Upon touch, there is an immediate reaction, with burning, urticaria, and edema. Pain increases, peaking 30 minutes later; then the pain slowly subsides.<sup>1</sup> Tachycardia and throbbing regional lymphadenopathy can occur for 1 to 4 hours.<sup>1,6</sup> </p> <p><i>Cutaneous Findings—</i>Examination reveals immediate piloerection, erythema due to arteriolar dilation, and local swelling.<sup>2</sup> These findings may disappear after 1 hour or last as long as 24 hours.<sup>1</sup> Although objective signs may fade, subjective pain, pruritus, and burning can persist for months.<sup>3</sup> </p> <h3>Dermatitis-Inducing Plant Parts</h3> <p>After contact with the stems or leaves, the sharp trichomes become embedded in the skin, making them difficult to remove.<sup>1</sup> The toxins are contained in siliceous hairs that the human body cannot break down.<sup>3</sup> Symptoms can be experienced for as long as 1 year after contact, especially when the skin is pressed firmly or washed with hot or cold water.<sup>3,6</sup> Because the plant’s hairs are shed continuously, being in close proximity to <i>D moroides</i> for longer than 20 minutes can lead to extreme sneezing, nosebleeds, and major respiratory damage from inhaling hairs.<sup>1,6,9</sup></p> <p>The stinging hairs of <i>D moroides</i> differ from irritant hairs on other plants because they contain physiologically active substances. Stinging hairs are classified as either a hypodermic syringe, which expels liquid only, or as a tragia-type syringe, in which liquid and sharp crystals are injected. <br/><br/>The Australian stinging tree<i> </i>falls into the first of these 2 groups (Figure 2)<sup>1</sup>; the sharp tip of the hair breaks on contact, leading to expulsion of the toxin into skin.<sup>1,4</sup> The hairs function as a defense against mammalian herbivores but typically have no impact on pests.<sup>1</sup> Nocturnal beetles and on occasion possums and red-legged pademelons dare to eat <i>D moroides.</i><sup>3,6</sup> </p> <h3>The Irritant </h3> <p>Initially, formic acid was proposed as the irritant chemical in <i>D moroides</i><sup>1</sup>; other candidates have included neurotransmitters, such as histamine, acetylcholine, and serotonin, as well as inorganic ions, such as potassium. These compounds may play a role but none explain the persistent sensory effects and years-long stable nature of the toxin.<sup>1,4</sup> </p> <p>The most likely culprit irritant is a member of a newly discovered family of neurotoxins, the gympietides. These knot-shaped chemicals, found in <i>D moroides </i>and some spider venoms, have the ability to activate voltage-gated sodium channels of cutaneous neurons and cause local cutaneous vasodilation by stimulating neurotransmitter release.<sup>4</sup> These neurotoxins not only generate pain but also suppress the mechanism used to interrupt those pain signals.<sup>10</sup> Synthesized gympietides can replicate the effects of natural contact, indicating that they are the primary active toxins. These toxins are ultrastable, thus producing lasting effects.<sup>1<br/><br/></sup>Although much is understood about the evolution and distribution of <i>D moroides</i> and the ecological role that it plays, there is still more to learn about the plant’s toxicology.</p> <h3>Prevention and Treatment</h3> <p><i>Prevention—Dendrocnide moroides</i> dermatitis is best prevented by avoiding contact with the plant and related species, as well as wearing upper body clothing with long sleeves, pants, and boots, though plant hairs can still penetrate garments and sting.<sup>2,3</sup> </p> <p><i>Therapy—</i>There is no reversal therapy of<i> D moroides </i>dermatitis but symptoms can be managed.<sup>4</sup> For pain, analgesics, such as opioids, have been used; on occasion, however, pain is so intense that even morphine does not help.<sup>4,10</sup> <br/><br/>Systemic or topical corticosteroids are the main therapy for many forms of plant-induced dermatitis because they are able to decrease cytokine production and stop lymphocyte production. Adding an oral antihistamine can alleviate histamine-mediated pruritus but not pruritus that is mediated by other chemicals.<sup>11</sup> <br/><br/>Other methods of relieving symptoms of <i>D moroides </i>dermatitis have been proposed or reported anecdotally. Diluted hydrochloric acid can be applied to the skin to denature remaining toxin.<sup>4</sup> The sap of <i>Alocasia brisbanensis</i> (the cunjevoi plant) can be rubbed on affected areas to provide a cooling effect, but do not allow <i>A brisbanensis</i> sap to enter the mouth, as it contains calcium oxalate, a toxic irritant found in dumb cane (<i>Dieffenbachia </i>species). The roots of the Australian stinging tree also can be ground and made into a paste, which is applied to the skin.<sup>3</sup> However, given the stability of the toxin, we do not recommend these remedies.<br/><br/>Instead, heavy-duty masking tape or hot wax can be applied to remove plant hairs from the skin. The most successful method of removing plant hair is hair removal wax strips, which are considered an essential component of a first aid kit where <i>D moroides</i> is found.<sup>3</sup></p> <h2>REFERENCES</h2> <p class="reference"> 1. Ensikat H-J, Wessely H, Engeser M, et al. Distribution, ecology, chemistry and toxicology of plant stinging hairs. <i>Toxins (Basel). </i>2021;13:141. doi:10.3390/toxins13020141<br/><br/> 2. Schmitt C, Parola P, de Haro L. Painful sting after exposure to <i>Dendrocnide</i> sp: two case reports. <i>Wilderness Environ Med.</i> 2013;24:471-473. doi:10.1016/j.wem.2013.03.021<br/><br/> 3. Hurley M. Selective stingers. <i>ECOS</i>. 2000;105:18-23. Accessed October 13, 2023. https://www.writingclearscience.com.au/wp-content/uploads/2015/06/stingers.pdf <br/><br/> 4. Gilding EK, Jami S, Deuis JR, et al. Neurotoxic peptides from the venom of the giant Australian stinging tree. <i>Sci Adv.</i> 2020;6:eabb8828. doi:10.1126/sciadv.abb8828<br/><br/> 5. <i>Dendrocnide moroides</i>. James Cook University Australia website. Accessed Accessed October 13, 2023. https://www.jcu.edu.au/discover-nature-at-jcu/plants/plants-by-scientific-name2/dendrocnide-moroides<br/><br/> 6. Hurley M. ‘The worst kind of pain you can imagine’—what it’s like to be stung by a stinging tree. <i>The Conversation</i>. September 28, 2018. Accessed October 13, 2023. https://theconversation.com/the-worst-kind-of-pain-you-can-imagine-what-its-like-to-be-stung-by-a-stinging-tree-103220 <br/><br/> 7. <i>Urticaceae</i>: plant family. <i>Britannica</i> <i>[Internet]</i>. Accessed October 13, 2023. https://www.britannica.com/plant/Urticaceae <br/><br/> 8. Stinging trees (genus <i>Dendrocnide</i>). iNaturalist.ca [Internet]. Accessed October 13, 2023. https://inaturalist.ca/taxa/129502-Dendrocnide <br/><br/> 9. Hurley M. Growth dynamics and leaf quality of the stinging trees <i>Dendrocnide moroides</i> and <i>Dendrocnide cordifolia</i> (family Urticaceae) in Australian tropical rainforest: implications for herbivores. <i>Aust J Bot.</i> 2000;48:191-201. doi:10.1071/BT98006<br/><br/>10. How the giant stinging tree of Australia can inflict months of agony. <i>Nature</i>. September 17, 2020. Accessed October 13, 2023. https://www.nature.com/articles/d41586-020-02668-9<br/><br/>11. Chang Y-T, Shen J-J, Wong W-R, et al. Alternative therapy for autosensitization dermatitis. <i>Chang Gung Med J</i>. 2009;32:668-673.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Dr. DeVore is from the Medical University of South Carolina, Charleston. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.</p> <p class="disclosure">The authors report no conflict of interest.<br/><br/>Correspondence: Ansley C. DeVore, MD, 363 Twin Oaks Dr, Spartanburg, SC 29306 (devorea@musc.edu).<br/><br/>doi:10.12788/cutis.0885</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li><em>Dendrocnide moroides</em> is arguably the most brutal of stinging plants, even leading to death in dogs, horses, and humans in rare cases.</li> <li>Clinical observations after contact reveal immediate piloerection and local swelling, which may disappear after 1 hour or last as long as 24 hours, but subjective pain, pruritus, and burning can persist for months.</li> <li>The most successful method of removing plant hair is hair removal wax strips, which are considered an essential component of a first aid kit where <em>D moroides</em> is found.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • Dendrocnide moroides is arguably the most brutal of stinging plants, even leading to death in dogs, horses, and humans in rare cases.
  • Clinical observations after contact reveal immediate piloerection and local swelling, which may disappear after 1 hour or last as long as 24 hours, but subjective pain, pruritus, and burning can persist for months.
  • The most successful method of removing plant hair is hair removal wax strips, which are considered an essential component of a first aid kit where D moroides is found.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
180025E4.SIG
Disable zoom
Off