Reticulated Brownish Erythema on the Lower Back

Article Type
Changed
Tue, 06/18/2024 - 12:04
Display Headline
Reticulated Brownish Erythema on the Lower Back

The Diagnosis: Erythema Ab Igne

Based on the patient's long-standing history of back pain treated with heating pads as well as the normal laboratory findings and skin examination, a diagnosis of erythema ab igne (EAI) was made.

Erythema ab igne presents as reticulated brownish erythema or hyperpigmentation on sites exposed to prolonged use of heat sources such as heating pads, laptops, and space heaters. Erythema ab igne most commonly affects the lower back, thighs, or legs1-6; however, EAI can appear on atypical sites such as the forehead and eyebrows due to newer technology (eg, virtual reality headsets).7 The level of heat required for EAI to occur is below the threshold for thermal burns (<45 °C [113 °F]).1 Erythema ab igne can occur at any age, and woman are more commonly affected than men.8 The pathophysiology currently is unknown; however, recurrent and prolonged heat exposure may damage superficial vessels. As a result, hemosiderin accumulates in the skin, and hyperpigmentation subsequently occurs.9

The diagnosis of EAI is clinical, and early stages of the rash present as blanching reticulated erythema in areas associated with heat exposure. If the offending source of heat is not removed, EAI can progress to nonblanching, fixed, hyperpigmented plaques with skin atrophy, bullae, or hyperkeratosis. Patients often are asymptomatic; however, mild burning may occur.2 Histopathology reveals cellular atypia, epidermal atrophy, dilation of dermal blood vessels, a minute inflammatory infiltrate, and keratinocyte apoptosis.10 Skin biopsy may be necessary in cases of suspected malignancy due to chronic heat exposure. Lesions that ulcerate or evolve should raise suspicion for malignancy.11 Squamous cell carcinoma is the most common malignancy associated with EAI; other malignancies that may manifest include basal cell carcinoma, Merkel cell carcinoma, or cutaneous marginal zone lymphoma.2,12-14

Erythema ab igne often is mistaken for livedo reticularis, which appears more erythematous without hyperpigmentation or epidermal changes and may be associated with a pathologic state.15 The differential diagnosis in our patient, who was in her 40s with a history of fatigue and joint pain, included livedo reticularis associated with lupus; however, the history of heating pad use, normal laboratory findings, and presence of epidermal changes suggested EAI. Lupus typically affects the hand and knee joints.16 Additionally, livedo reticularis more commonly appears on the legs.15

Other differentials for EAI include livedo racemosa, cutaneous T-cell lymphoma, and cutis marmorata. Livedo racemosa presents with broken rings of erythema in young to middle-aged women and primarily affects the trunk and proximal limbs. It is associated with an underlying condition such as polyarteritis nodosa and less commonly with lupus erythematosus with antiphospholipid or Sneddon syndrome.15,17 Cutaneous T-cell lymphoma typically manifests with poikilodermatous patches larger than the palm, especially in covered areas of skin.18 Cutis marmorata is transient and temperature dependent.9

The key intervention for EAI is removal of the offending heat source.2 Patients should be counseled that the erythema and hyperpigmentation may take months to years to resolve. Topical hydroquinone or tretinoin may be used in cases of persistent hyperpigmentation.19 Patients who continue to use heating pads for long-standing pain should be advised to limit their use to short intervals without occlusion. If malignancy is a concern, a biopsy should be performed.20

References
  1. Wipf AJ, Brown MR. Malignant transformation of erythema ab igne. JAAD Case Rep. 2022;26:85-87. doi:10.1016/j.jdcr.2022.06.018
  2. Sigmon JR, Cantrell J, Teague D, et al. Poorly differentiated carcinoma arising in the setting of erythema ab igne. Am J Dermatopathol. 2013;35:676-678. doi:10.1097/DAD.0b013e3182871648
  3. Patel DP. The evolving nomenclature of erythema ab igne-redness from fire. JAMA Dermatol. 2017;153:685. doi:10.1001/jamadermatol.2017.2021
  4. Arnold AW, Itin PH. Laptop computer-induced erythema ab igne in a child and review of the literature. Pediatrics. 2010;126:E1227-E1230. doi:10.1542/peds.2010-1390
  5. Riahi RR, Cohen PR. Laptop-induced erythema ab igne: report and review of literature. Dermatol Online J. 2012;18:5.
  6. Haleem Z, Philip J, Muhammad S. Erythema ab igne: a rare presentation of toasted skin syndrome with the use of a space heater. Cureus. 2021;13:e13401. doi:10.7759/cureus.13401
  7. Moreau T, Benzaquen M, Gueissaz F. Erythema ab igne after using a virtual reality headset: a new phenomenon to know. J Eur Acad Dermatol Venereol. 2022;36:E932-E933. doi:10.1111/jdv.18371
  8. Ozturk M, An I. Clinical features and etiology of patients with erythema ab igne: a retrospective multicenter study. J Cosmet Dermatol. 2020;19:1774-1779. doi:10.1111/jocd.13210
  9. Gmuca S, Yu J, Weiss PF, et al. Erythema ab igne in an adolescent with chronic pain: an alarming cutaneous eruption from heat exposure. Pediatr Emerg Care. 2020;36:E236-E238. doi:10.1097 /PEC.0000000000001460
  10. Wells A, Desai A, Rudnick EW, et al. Erythema ab igne with features resembling keratosis lichenoides chronica. J Cutan Pathol. 2021;48:151-153. doi:10.1111/cup.13885
  11. Milchak M, Smucker J, Chung CG, et al. Erythema ab igne due to heating pad use: a case report and review of clinical presentation, prevention, and complications. Case Rep Med. 2016;2016:1862480. doi:10.1155/2016/1862480
  12. Daneshvar E, Seraji S, Kamyab-Hesari K, et al. Basal cell carcinoma associated with erythema ab igne. Dermatol Online J. 2020;26:13030 /qt3kz985b4.
  13. Jones CS, Tyring SK, Lee PC, et al. Development of neuroendocrine (Merkel cell) carcinoma mixed with squamous cell carcinoma in erythema ab igne. Arch Dermatol. 1988;124:110-113.
  14. Wharton J, Roffwarg D, Miller J, et al. Cutaneous marginal zone lymphoma arising in the setting of erythema ab igne. J Am Acad Dermatol. 2010;62:1080-1081. doi:10.1016/j.jaad.2009.08.005
  15. Sajjan VV, Lunge S, Swamy MB, et al. Livedo reticularis: a review of the literature. Indian Dermatol Online J. 2015;6:315-321. doi:10.4103 /2229-5178.164493
  16. Grossman JM. Lupus arthritis. Best Pract Res Clin Rheumatol. 2009;23:495-506. doi:10.1016/j.berh.2009.04.003
  17. Aria AB, Chen L, Silapunt S. Erythema ab igne from heating pad use: a report of three clinical cases and a differential diagnosis. Cureus. 2018;10:E2635. doi:10.7759/cureus.2635
  18. Wilcox RA. Cutaneous T-cell lymphoma: 2017 update on diagnosis, risk-stratification, and management. Am J Hematol. 2017;92:1085-1102. doi:10.1002/ajh.24876
  19. Pennitz A, Kinberger M, Avila Valle G, et al. Self-applied topical interventions for melasma: a systematic review and meta-analysis of data from randomized, investigator-blinded clinical trials. Br J Dermatol. 2022;187:309-317.
  20. Sahl WJ, Taira JW. Erythema ab igne: treatment with 5-fluorouracil cream. J Am Acad Dermatol. 1992;27:109-110.
Article PDF
Author and Disclosure Information

Dr. Dao is from the Virginia Commonwealth University School of Medicine, Richmond. 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: Diem-Phuong D. Dao, MD, 1001 E Leigh St, 11th Floor, Richmond, VA 23219 (daopd@vcu.edu).

Issue
Cutis - 113(6)
Publications
Topics
Page Number
E8-E10
Sections
Author and Disclosure Information

Dr. Dao is from the Virginia Commonwealth University School of Medicine, Richmond. 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: Diem-Phuong D. Dao, MD, 1001 E Leigh St, 11th Floor, Richmond, VA 23219 (daopd@vcu.edu).

Author and Disclosure Information

Dr. Dao is from the Virginia Commonwealth University School of Medicine, Richmond. 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: Diem-Phuong D. Dao, MD, 1001 E Leigh St, 11th Floor, Richmond, VA 23219 (daopd@vcu.edu).

Article PDF
Article PDF
Related Articles

The Diagnosis: Erythema Ab Igne

Based on the patient's long-standing history of back pain treated with heating pads as well as the normal laboratory findings and skin examination, a diagnosis of erythema ab igne (EAI) was made.

Erythema ab igne presents as reticulated brownish erythema or hyperpigmentation on sites exposed to prolonged use of heat sources such as heating pads, laptops, and space heaters. Erythema ab igne most commonly affects the lower back, thighs, or legs1-6; however, EAI can appear on atypical sites such as the forehead and eyebrows due to newer technology (eg, virtual reality headsets).7 The level of heat required for EAI to occur is below the threshold for thermal burns (<45 °C [113 °F]).1 Erythema ab igne can occur at any age, and woman are more commonly affected than men.8 The pathophysiology currently is unknown; however, recurrent and prolonged heat exposure may damage superficial vessels. As a result, hemosiderin accumulates in the skin, and hyperpigmentation subsequently occurs.9

The diagnosis of EAI is clinical, and early stages of the rash present as blanching reticulated erythema in areas associated with heat exposure. If the offending source of heat is not removed, EAI can progress to nonblanching, fixed, hyperpigmented plaques with skin atrophy, bullae, or hyperkeratosis. Patients often are asymptomatic; however, mild burning may occur.2 Histopathology reveals cellular atypia, epidermal atrophy, dilation of dermal blood vessels, a minute inflammatory infiltrate, and keratinocyte apoptosis.10 Skin biopsy may be necessary in cases of suspected malignancy due to chronic heat exposure. Lesions that ulcerate or evolve should raise suspicion for malignancy.11 Squamous cell carcinoma is the most common malignancy associated with EAI; other malignancies that may manifest include basal cell carcinoma, Merkel cell carcinoma, or cutaneous marginal zone lymphoma.2,12-14

Erythema ab igne often is mistaken for livedo reticularis, which appears more erythematous without hyperpigmentation or epidermal changes and may be associated with a pathologic state.15 The differential diagnosis in our patient, who was in her 40s with a history of fatigue and joint pain, included livedo reticularis associated with lupus; however, the history of heating pad use, normal laboratory findings, and presence of epidermal changes suggested EAI. Lupus typically affects the hand and knee joints.16 Additionally, livedo reticularis more commonly appears on the legs.15

Other differentials for EAI include livedo racemosa, cutaneous T-cell lymphoma, and cutis marmorata. Livedo racemosa presents with broken rings of erythema in young to middle-aged women and primarily affects the trunk and proximal limbs. It is associated with an underlying condition such as polyarteritis nodosa and less commonly with lupus erythematosus with antiphospholipid or Sneddon syndrome.15,17 Cutaneous T-cell lymphoma typically manifests with poikilodermatous patches larger than the palm, especially in covered areas of skin.18 Cutis marmorata is transient and temperature dependent.9

The key intervention for EAI is removal of the offending heat source.2 Patients should be counseled that the erythema and hyperpigmentation may take months to years to resolve. Topical hydroquinone or tretinoin may be used in cases of persistent hyperpigmentation.19 Patients who continue to use heating pads for long-standing pain should be advised to limit their use to short intervals without occlusion. If malignancy is a concern, a biopsy should be performed.20

The Diagnosis: Erythema Ab Igne

Based on the patient's long-standing history of back pain treated with heating pads as well as the normal laboratory findings and skin examination, a diagnosis of erythema ab igne (EAI) was made.

Erythema ab igne presents as reticulated brownish erythema or hyperpigmentation on sites exposed to prolonged use of heat sources such as heating pads, laptops, and space heaters. Erythema ab igne most commonly affects the lower back, thighs, or legs1-6; however, EAI can appear on atypical sites such as the forehead and eyebrows due to newer technology (eg, virtual reality headsets).7 The level of heat required for EAI to occur is below the threshold for thermal burns (<45 °C [113 °F]).1 Erythema ab igne can occur at any age, and woman are more commonly affected than men.8 The pathophysiology currently is unknown; however, recurrent and prolonged heat exposure may damage superficial vessels. As a result, hemosiderin accumulates in the skin, and hyperpigmentation subsequently occurs.9

The diagnosis of EAI is clinical, and early stages of the rash present as blanching reticulated erythema in areas associated with heat exposure. If the offending source of heat is not removed, EAI can progress to nonblanching, fixed, hyperpigmented plaques with skin atrophy, bullae, or hyperkeratosis. Patients often are asymptomatic; however, mild burning may occur.2 Histopathology reveals cellular atypia, epidermal atrophy, dilation of dermal blood vessels, a minute inflammatory infiltrate, and keratinocyte apoptosis.10 Skin biopsy may be necessary in cases of suspected malignancy due to chronic heat exposure. Lesions that ulcerate or evolve should raise suspicion for malignancy.11 Squamous cell carcinoma is the most common malignancy associated with EAI; other malignancies that may manifest include basal cell carcinoma, Merkel cell carcinoma, or cutaneous marginal zone lymphoma.2,12-14

Erythema ab igne often is mistaken for livedo reticularis, which appears more erythematous without hyperpigmentation or epidermal changes and may be associated with a pathologic state.15 The differential diagnosis in our patient, who was in her 40s with a history of fatigue and joint pain, included livedo reticularis associated with lupus; however, the history of heating pad use, normal laboratory findings, and presence of epidermal changes suggested EAI. Lupus typically affects the hand and knee joints.16 Additionally, livedo reticularis more commonly appears on the legs.15

Other differentials for EAI include livedo racemosa, cutaneous T-cell lymphoma, and cutis marmorata. Livedo racemosa presents with broken rings of erythema in young to middle-aged women and primarily affects the trunk and proximal limbs. It is associated with an underlying condition such as polyarteritis nodosa and less commonly with lupus erythematosus with antiphospholipid or Sneddon syndrome.15,17 Cutaneous T-cell lymphoma typically manifests with poikilodermatous patches larger than the palm, especially in covered areas of skin.18 Cutis marmorata is transient and temperature dependent.9

The key intervention for EAI is removal of the offending heat source.2 Patients should be counseled that the erythema and hyperpigmentation may take months to years to resolve. Topical hydroquinone or tretinoin may be used in cases of persistent hyperpigmentation.19 Patients who continue to use heating pads for long-standing pain should be advised to limit their use to short intervals without occlusion. If malignancy is a concern, a biopsy should be performed.20

References
  1. Wipf AJ, Brown MR. Malignant transformation of erythema ab igne. JAAD Case Rep. 2022;26:85-87. doi:10.1016/j.jdcr.2022.06.018
  2. Sigmon JR, Cantrell J, Teague D, et al. Poorly differentiated carcinoma arising in the setting of erythema ab igne. Am J Dermatopathol. 2013;35:676-678. doi:10.1097/DAD.0b013e3182871648
  3. Patel DP. The evolving nomenclature of erythema ab igne-redness from fire. JAMA Dermatol. 2017;153:685. doi:10.1001/jamadermatol.2017.2021
  4. Arnold AW, Itin PH. Laptop computer-induced erythema ab igne in a child and review of the literature. Pediatrics. 2010;126:E1227-E1230. doi:10.1542/peds.2010-1390
  5. Riahi RR, Cohen PR. Laptop-induced erythema ab igne: report and review of literature. Dermatol Online J. 2012;18:5.
  6. Haleem Z, Philip J, Muhammad S. Erythema ab igne: a rare presentation of toasted skin syndrome with the use of a space heater. Cureus. 2021;13:e13401. doi:10.7759/cureus.13401
  7. Moreau T, Benzaquen M, Gueissaz F. Erythema ab igne after using a virtual reality headset: a new phenomenon to know. J Eur Acad Dermatol Venereol. 2022;36:E932-E933. doi:10.1111/jdv.18371
  8. Ozturk M, An I. Clinical features and etiology of patients with erythema ab igne: a retrospective multicenter study. J Cosmet Dermatol. 2020;19:1774-1779. doi:10.1111/jocd.13210
  9. Gmuca S, Yu J, Weiss PF, et al. Erythema ab igne in an adolescent with chronic pain: an alarming cutaneous eruption from heat exposure. Pediatr Emerg Care. 2020;36:E236-E238. doi:10.1097 /PEC.0000000000001460
  10. Wells A, Desai A, Rudnick EW, et al. Erythema ab igne with features resembling keratosis lichenoides chronica. J Cutan Pathol. 2021;48:151-153. doi:10.1111/cup.13885
  11. Milchak M, Smucker J, Chung CG, et al. Erythema ab igne due to heating pad use: a case report and review of clinical presentation, prevention, and complications. Case Rep Med. 2016;2016:1862480. doi:10.1155/2016/1862480
  12. Daneshvar E, Seraji S, Kamyab-Hesari K, et al. Basal cell carcinoma associated with erythema ab igne. Dermatol Online J. 2020;26:13030 /qt3kz985b4.
  13. Jones CS, Tyring SK, Lee PC, et al. Development of neuroendocrine (Merkel cell) carcinoma mixed with squamous cell carcinoma in erythema ab igne. Arch Dermatol. 1988;124:110-113.
  14. Wharton J, Roffwarg D, Miller J, et al. Cutaneous marginal zone lymphoma arising in the setting of erythema ab igne. J Am Acad Dermatol. 2010;62:1080-1081. doi:10.1016/j.jaad.2009.08.005
  15. Sajjan VV, Lunge S, Swamy MB, et al. Livedo reticularis: a review of the literature. Indian Dermatol Online J. 2015;6:315-321. doi:10.4103 /2229-5178.164493
  16. Grossman JM. Lupus arthritis. Best Pract Res Clin Rheumatol. 2009;23:495-506. doi:10.1016/j.berh.2009.04.003
  17. Aria AB, Chen L, Silapunt S. Erythema ab igne from heating pad use: a report of three clinical cases and a differential diagnosis. Cureus. 2018;10:E2635. doi:10.7759/cureus.2635
  18. Wilcox RA. Cutaneous T-cell lymphoma: 2017 update on diagnosis, risk-stratification, and management. Am J Hematol. 2017;92:1085-1102. doi:10.1002/ajh.24876
  19. Pennitz A, Kinberger M, Avila Valle G, et al. Self-applied topical interventions for melasma: a systematic review and meta-analysis of data from randomized, investigator-blinded clinical trials. Br J Dermatol. 2022;187:309-317.
  20. Sahl WJ, Taira JW. Erythema ab igne: treatment with 5-fluorouracil cream. J Am Acad Dermatol. 1992;27:109-110.
References
  1. Wipf AJ, Brown MR. Malignant transformation of erythema ab igne. JAAD Case Rep. 2022;26:85-87. doi:10.1016/j.jdcr.2022.06.018
  2. Sigmon JR, Cantrell J, Teague D, et al. Poorly differentiated carcinoma arising in the setting of erythema ab igne. Am J Dermatopathol. 2013;35:676-678. doi:10.1097/DAD.0b013e3182871648
  3. Patel DP. The evolving nomenclature of erythema ab igne-redness from fire. JAMA Dermatol. 2017;153:685. doi:10.1001/jamadermatol.2017.2021
  4. Arnold AW, Itin PH. Laptop computer-induced erythema ab igne in a child and review of the literature. Pediatrics. 2010;126:E1227-E1230. doi:10.1542/peds.2010-1390
  5. Riahi RR, Cohen PR. Laptop-induced erythema ab igne: report and review of literature. Dermatol Online J. 2012;18:5.
  6. Haleem Z, Philip J, Muhammad S. Erythema ab igne: a rare presentation of toasted skin syndrome with the use of a space heater. Cureus. 2021;13:e13401. doi:10.7759/cureus.13401
  7. Moreau T, Benzaquen M, Gueissaz F. Erythema ab igne after using a virtual reality headset: a new phenomenon to know. J Eur Acad Dermatol Venereol. 2022;36:E932-E933. doi:10.1111/jdv.18371
  8. Ozturk M, An I. Clinical features and etiology of patients with erythema ab igne: a retrospective multicenter study. J Cosmet Dermatol. 2020;19:1774-1779. doi:10.1111/jocd.13210
  9. Gmuca S, Yu J, Weiss PF, et al. Erythema ab igne in an adolescent with chronic pain: an alarming cutaneous eruption from heat exposure. Pediatr Emerg Care. 2020;36:E236-E238. doi:10.1097 /PEC.0000000000001460
  10. Wells A, Desai A, Rudnick EW, et al. Erythema ab igne with features resembling keratosis lichenoides chronica. J Cutan Pathol. 2021;48:151-153. doi:10.1111/cup.13885
  11. Milchak M, Smucker J, Chung CG, et al. Erythema ab igne due to heating pad use: a case report and review of clinical presentation, prevention, and complications. Case Rep Med. 2016;2016:1862480. doi:10.1155/2016/1862480
  12. Daneshvar E, Seraji S, Kamyab-Hesari K, et al. Basal cell carcinoma associated with erythema ab igne. Dermatol Online J. 2020;26:13030 /qt3kz985b4.
  13. Jones CS, Tyring SK, Lee PC, et al. Development of neuroendocrine (Merkel cell) carcinoma mixed with squamous cell carcinoma in erythema ab igne. Arch Dermatol. 1988;124:110-113.
  14. Wharton J, Roffwarg D, Miller J, et al. Cutaneous marginal zone lymphoma arising in the setting of erythema ab igne. J Am Acad Dermatol. 2010;62:1080-1081. doi:10.1016/j.jaad.2009.08.005
  15. Sajjan VV, Lunge S, Swamy MB, et al. Livedo reticularis: a review of the literature. Indian Dermatol Online J. 2015;6:315-321. doi:10.4103 /2229-5178.164493
  16. Grossman JM. Lupus arthritis. Best Pract Res Clin Rheumatol. 2009;23:495-506. doi:10.1016/j.berh.2009.04.003
  17. Aria AB, Chen L, Silapunt S. Erythema ab igne from heating pad use: a report of three clinical cases and a differential diagnosis. Cureus. 2018;10:E2635. doi:10.7759/cureus.2635
  18. Wilcox RA. Cutaneous T-cell lymphoma: 2017 update on diagnosis, risk-stratification, and management. Am J Hematol. 2017;92:1085-1102. doi:10.1002/ajh.24876
  19. Pennitz A, Kinberger M, Avila Valle G, et al. Self-applied topical interventions for melasma: a systematic review and meta-analysis of data from randomized, investigator-blinded clinical trials. Br J Dermatol. 2022;187:309-317.
  20. Sahl WJ, Taira JW. Erythema ab igne: treatment with 5-fluorouracil cream. J Am Acad Dermatol. 1992;27:109-110.
Issue
Cutis - 113(6)
Issue
Cutis - 113(6)
Page Number
E8-E10
Page Number
E8-E10
Publications
Publications
Topics
Article Type
Display Headline
Reticulated Brownish Erythema on the Lower Back
Display Headline
Reticulated Brownish Erythema on the Lower Back
Sections
Questionnaire Body

A 42-year-old woman presented with an asymptomatic, erythematous, lacelike rash on the lower back of 8 months’ duration that was first noticed by her husband. The patient had a long-standing history of chronic fatigue and lower back pain treated with acetaminophen, diclofenac gel, and heating pads. Physical examination revealed reticulated brownish erythema confined to the lower back. Laboratory findings were unremarkable.

Dao2_Quiz.jpg

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 06/14/2024 - 10:00
Un-Gate On Date
Fri, 06/14/2024 - 10:00
Use ProPublica
CFC Schedule Remove Status
Fri, 06/14/2024 - 10:00
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: 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

Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis

Article Type
Changed
Fri, 04/26/2024 - 09:13
Display Headline
Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis

Approximately 108 million individuals have been forcibly displaced across the globe as of 2022, 35 million of whom are formally designated as refugees.1,2 The United States has coordinated resettlement of more refugee populations than any other country; the most common countries of origin are the Democratic Republic of the Congo, Syria, Afghanistan, and Myanmar.3 In 2021, policy to increase the number of refugees resettled in the United States by more than 700% (from 15,000 up to 125,000) was established; since enactment, the United States has seen more than double the refugee arrivals in 2023 than the prior year, making medical care for this population increasingly relevant for the dermatologist.4

Understanding how to care for this population begins with an accurate understanding of the term refugee. The United Nations defines a refugee as a person who is unwilling or unable to return to their country of nationality because of persecution or well-founded fear of persecution due to race, religion, nationality, membership in a particular social group, or political opinion. This term grants a protected status under international law and encompasses access to travel assistance, housing, cultural orientation, and medical evaluation upon resettlement.5,6

The burden of treatable dermatologic conditions in refugee populations ranges from 19% to 96% in the literature7,8 and varies from inflammatory disorders to infectious and parasitic diseases.9 In one study of 6899 displaced individuals in Greece, the prevalence of dermatologic conditions was higher than traumatic injury, cardiac disease, psychological conditions, and dental disease.10

When outlining differential diagnoses for parasitic infestations of the skin that affect refugee populations, helpful considerations include the individual’s country of origin, route traveled, and method of travel.11 Parasitic infestations specifically are more common in refugee populations when there are barriers to basic hygiene, crowded living or travel conditions, or lack of access to health care, which they may experience at any point in their home country, during travel, or in resettlement housing.8

Even with limited examination and diagnostic resources, the skin is the most accessible first indication of patients’ overall well-being and often provides simple diagnostic clues—in combination with contextualization of the patient’s unique circumstances—necessary for successful diagnosis and treatment of scabies and pediculosis.12 The dermatologist working with refugee populations may be the first set of eyes available and trained to discern skin infestations and therefore has the potential to improve overall outcomes.

Some parasitic infestations in refugee populations may fall under the category of neglected tropical diseases, including scabies, ascariasis, trypanosomiasis, leishmaniasis, and schistosomiasis; they affect an estimated 1 billion individuals across the globe but historically have been underrepresented in the literature and in health policy due in part to limited access to care.13 This review will focus on infestations by the scabies mite (Sarcoptes scabiei var hominis) and the human louse, as these frequently are encountered, easily diagnosed, and treatable by trained clinicians, even in resource-limited settings.

Scabies

Scabies is a parasitic skin infestation caused by the 8-legged mite Sarcoptes scabiei var hominis. The female mite begins the infestation process via penetration of the epidermis, particularly the stratum corneum, and commences laying eggs (Figure 1). The subsequent larvae emerge 48 to 72 hours later and remain burrowed in the epidermis. The larvae mature over the next 10 to 14 days and continue the reproductive cycle.14,15 Symptoms of infestation occurs due to a hypersensitivity reaction to the mite and its by-products.16 Transmission of the mite primarily occurs via direct (skin-to-skin) contact with infected individuals or environmental surfaces for 24 to36 hours in specific conditions, though the latter source has been debated in the literature.

CT113004016_fig1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Sarcoptes%20scabiei%20mite%20(A)%2C%20ova%20(B)%2C%20and%20scybala%20(C)%20on%20microscopic%20evaluation.%3C%2Fp%3E

 

 

The method of transmission is particularly important when considering care for refugee populations. Scabies is found most often in those living in or traveling from tropical regions including East Asia, Southeast Asia, Oceania, and Latin America.17 In displaced or refugee populations, a lack of access to basic hygiene, extended travel in close quarters, and suboptimal health care access all may lead to an increased incidence of untreated scabies infestations.18 Scabies is more prevalent in children, with increased potential for secondary bacterial infections with Streptococcus and Staphylococcus species due to excoriation in unsanitary conditions. Secondary infection with Streptococcus pyogenes can lead to acute poststreptococcal glomerulonephritis, which accounts for a large burden of chronic kidney disease in affected populations.19 However, scabies may be found in any population, regardless of hygiene or health care access. Treating health care providers should keep a broad differential.

Presentation—The latency of scabies symptoms is 2 to 6 weeks in a primary outbreak and may be as short as 1 to 3 days with re-infestation, following the course of delayed-type hypersensitivity.20 The initial hallmark symptom is pruritus with increased severity in the evening. Visible lesions, excoriations, and burrows associated with scattered vesicles or pustules may be seen over the web spaces of the hands and feet, volar surfaces of the wrists, axillae, waist, genitalia, inner thighs, or buttocks.19 Chronic infestation often manifests with genital nodules. In populations with limited access to health care, there are reports of a sensitization phenomenon in which the individual may become less symptomatic after 4 to 6 weeks and yet be a potential carrier of the mite.21

Those with compromised immune function, such as individuals living with HIV or severe malnutrition, may present with crusted scabies, a variant that manifests as widespread hyperkeratotic scaling with more pronounced involvement of the head, neck, and acral areas. In contrast to classic scabies, crusted scabies is associated with minimal pruritus.22

Diagnosis—The diagnosis of scabies is largely clinical with confirmation through skin scrapings. The International Alliance for Control of Scabies has established diagnostic criteria that include a combination of clinical findings, history, and visualization of mites.23 A dermatologist working with refugee populations may employ any combination of history (eg, nocturnal itch, exposure to an affected individual) or clinical findings along with a high degree of suspicion in those with elevated risk. Visualization of mites is helpful to confirm the diagnosis and may be completed with the application of mineral oil at the terminal end of a burrow, skin scraping with a surgical blade or needle, and examination under light microscopy.

Treatment—First-line treatment for scabies consists of application of permethrin cream 5% on the skin of the neck to the soles of the feet, which is to be left on for 8 to 14 hours followed by rinsing. Re-application is recommended in 1 to 2 weeks. Oral ivermectin is a reasonable alternative to permethrin cream due to its low cost and easy administration in large affected groups. It is not labeled for use in pregnant women or children weighing less than 15 kg but has no selective fetal toxicity. Treatment of scabies with ivermectin has the benefit of treating many other parasitic infections. Both medications are on the World Health Organization Model List of Essential Medications and are widely available for treating providers, even in resource-limited settings.24

Much of the world still uses benzyl benzoate or precipitated sulfur ointment to treat scabies, and some botanicals used in folk medicine have genuine antiscabetic properties. Pruritus may persist for 1 to 4 weeks following treatment and does not indicate treatment failure. Topical camphor and menthol preparations, low-potency topical corticosteroids, or emollients all may be employed for relief.25Sarna is a Spanish term for scabies and has become the proprietary name for topical antipruritic agents. Additional methods of treatment and prevention include washing clothes and linens in hot water and drying on high heat. If machine washing is not available, clothing and linens may be sealed in a plastic bag for 72 hours.

Pediculosis

Pediculosis is an infestation caused by the ectoparasite Pediculus humanus, an obligate, sesame seed–sized louse that feeds exclusively on the blood of its host (Figure 2).26 Of the lice species, 2 require humans as hosts; one is P humanus and the other is Pthirus pubis (pubic lice). Pediculus humanus may be further classified into morphologies based largely on the affected area: body (P humanus corporis) or head (P humanus capitis), both of which will be discussed.27

CT113004016_fig2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20%3Cem%3EPediculus%20humanus%3C%2Fem%3E%20(louse)%2C%20adult%20form.%3C%2Fp%3E

 

 

Lice primarily attach to clothing and hair shafts, then transfer to the skin for blood feeds. Females lay eggs that hatch 6 to 10 days later, subsequently maturing into adults. The lifespan of these parasites with regular access to a host is 1 to 3 months for head lice and 18 days for body lice vs only 3 to 5 days without a host.28 Transmission of P humanus capitis primarily occurs via direct contact with affected individuals, either head-to-head contact or sharing of items such as brushes and headscarves; P humanus corporis also may be transmitted via direct contact with affected individuals or clothing.

Pediculosis is an important infestation to consider when providing care for refugee populations. Risk factors include lack of access to basic hygiene, including regular bathing or laundering of clothing, and crowded conditions that make direct person-to-person contact with affected individuals more likely.29 Body lice are associated more often with domestic turbulence and displaced populations30 in comparison to head lice, which have broad demographic variables, most often affecting females and children.28 Fatty acids in adult male sebum make the scalp less hospitable to lice.

Presentation—The most common clinical manifestation of pediculosis is pruritus. Cutaneous findings can include papules, wheals, or hemorrhagic puncta secondary to the louse bite. Due to the Tyndall effect of deep hemosiderin pigment, blue-grey macules termed maculae ceruleae (Figure 3) also may be present in chronic infestations of pediculosis pubis, in contrast to pediculosis capitis or corporis.31 Body louse infestation is associated with a general pruritus concentrated on the neck, shoulders, and waist—areas where clothing makes the most direct contact. Lesions may be visible and include eczematous patches with excoriation and possible secondary bacterial infection. Chronic infestation may exhibit lichenification or hyperpigmentation in associated areas. Head lice most often manifest with localized scalp pruritus and associated excoriation and cervical or occipital lymphadenopathy.32

CT113004016_fig3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Maculae%20ceruleae%E2%80%94blue-grey%20macules%E2%80%94may%20be%20present%20on%20the%20skin%20secondary%20to%20%3Cem%3EPediculosis%3C%2Fem%3E%20infestation.%3C%2Fp%3E

Diagnosis—The diagnosis of pediculosis is clinical, with confirmation requiring direct examination of the insect or nits (the egg case of the parasite)(Figure 4). Body lice and associated nits can be visualized on clothing seams near areas of highest body temperature, particularly the waistband. Head lice may be visualized crawling on hair shafts or on a louse comb. Nits are firmly attached to hair shafts and are visible to the naked eye, whereas pseudonits slide freely along the hair shaft and are not a manifestation of louse infestation (Figure 5).31

CT113004016_fig4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Pediculosis%20nits%E2%80%94the%20egg%20cases%20of%20the%20parasite%E2%80%94may%20firmly%20attach%20to%20the%20hair%20shaft.%3C%2Fp%3E

Treatment—Treatment varies by affected area. Pediculosis corporis may be treated with permethrin cream 5% applied to the entire body and left on for 8 to 10 hours, but this may not be necessary if facilities are available to wash and dry clothing.33 The use of oral ivermectin and permethrin-impregnated underwear both have been proposed.34,35 Treatment of pediculosis capitis may be accomplished with a variety of topical pediculicides including permethrin, pyrethrum with piperonyl butoxide, dimethicone, malathion, benzyl alcohol, spinosad, and topical ivermectin.22 Topical corticosteroids or emollients may be employed for residual pruritus.

CT113004016_fig5.jpg
%3Cp%3E%3Cstrong%3EFIGURE%205.%3C%2Fstrong%3E%20The%20pseudonit%20closely%20mimics%20pediculosis%20nits%20but%20consists%20of%20keratinized%20cell%20casts%20that%20are%20freely%20dislodged.%3C%2Fp%3E

Equally important is environmental elimination of infestation. Clothing should be discarded if possible or washed and dried using high heat. If neither approach is possible or appropriate, clothing may be sealed in a plastic bag for 2 weeks or treated with a pediculicide. Nit combing is an important adjunct in the treatment of pediculosis capitis.36 It is important to encourage return to work and/or school immediately after treatment. “No nit” policies are more harmful to education than helpful for prevention of investation.37

Pediculosis corporis may transmit infectious agents including Bartonella quintana, (trench fever, endocarditis, bacillary angiomatosis), Borrelia recurrentis (louse-borne relapsing fever), and Rickettsia prowazekii (epidemic typhus).31,38,39 Additionally, severe pediculosis infestations have the potential to cause chronic blood loss in affected populations. In a study of patients with active pediculosis infestation, mean hemoglobin values were found to be 2.5 g/dL lower than a matched population without infestation.40 It is important to consider pediculosis as a risk for iron-deficiency anemia in populations who are known to lack access to regular medical evaluation.41

 

 

Future Considerations

Increased access to tools and education for clinicians treating refugee populations is key to reducing the burden of parasitic skin disease and related morbidity and mortality in vulnerable groups both domestically and globally. One such tool, the Skin NTDs App, was launched by the World Health Organization in 2020. It is available for free for Android and iOS devices to assist clinicians in the field with the diagnosis and treatment of neglected tropical diseases—including scabies—that may affect refugee populations.42

Additionally, to both improve access and limit preventable sequelae, future investigations into appropriate models of community-based care are paramount. The model of community-based care is centered on the idea of care provision that prioritizes safety, accessibility, affordability, and acceptability in an environment closest to vulnerable populations. The largest dermatologic society, the International League of Dermatological Societies, formed a Migrant Health Dermatology Working Group that prioritizes understanding and improving care for refugee and migrant populations; this group hosted a summit in 2022, bringing together international subject matter leaders to discuss such models of care and set goals for the creation of tool kits for patients, frontline health care workers, and dermatologists.43

Conclusion

Improvement in dermatologic care of refugee populations includes provision of culturally and linguistically appropriate care by trained clinicians, adequate access to the most essential medications, and basic physical or legal access to health care systems in general.8,11,44 Parasitic infestations have the potential to remain asymptomatic for extended periods of time and result in spread to potentially nonendemic regions of resettlement.45 Additionally, the psychosocial well-being of refugee populations upon resettlement may be negatively affected by stigma of disease processes such as scabies and pediculosis, leading to additional barriers to successful re-entry into the patient’s new environment.46 Therefore, proper screening, diagnosis, and treatment of the most common parasitic infestations in this population have great potential to improve outcomes for large groups across the globe.

References
  1. Monin K, Batalova J, Lai T. Refugees and Asylees in the United States. Migration Information Source. Published May 13, 2021. Accessed April 4, 2024. https://www.migrationpolicy.org/article/refugees-and-asylees-united-states-2021
  2. UNHCR. Figures at a Glance. UNHCR USA. Update June 14, 2023. Accessed April 4, 2024. https://www.unhcr.org/en-us/figures-at-a-glance.html
  3. UNHCR. Refugee resettlement facts. Published October 2023. Accessed April 8, 2024. https://www.unhcr.org/us/media/refugee-resettlement-facts
  4. US Department of State. Report to Congress on Proposed Refugee Admissions for Fiscal Year 2024. Published November 3, 2023. Accessed April 8, 2024. https://www.state.gov/report-to-congress-on-proposed-refugee-admissions-for-fiscal-year-2024/
  5. UNHCR. Compact for Migration: Definitions. United Nations. Accessed April 4, 2024. https://refugeesmigrants.un.org/definitions
  6. United Nations High Commissioner for Refugees (UNHCR). Convention and Protocol Relating to the Status of Refugees. Published December 2010. Accessed January 11, 2024. https://www.unhcr.org/us/media/convention-and-protocol-relating-status-refugees
  7. Kibar Öztürk M. Skin diseases in rural Nyala, Sudan (in a rural hospital, in 12 orphanages, and in two refugee camps). Int J Dermatol. 2019;58:1341-1349. doi:10.1111/ijd.14619
  8. Padovese V, Knapp A. Challenges of managing skin diseases in refugees and migrants. Dermatol Clin. 2021;39:101-115. doi:10.1016/j.det.2020.08.010
  9. Saikal SL, Ge L, Mir A, et al. Skin disease profile of Syrian refugees in Jordan: a field-mission assessment. J Eur Acad Dermatol Venereol. 2020;34:419-425. doi:10.1111/jdv.15909
  10. Eonomopoulou A, Pavli A, Stasinopoulou P, et al. Migrant screening: lessons learned from the migrant holding level at the Greek-Turkish borders. J Infect Public Health. 2017;10:177-184. doi:10.1016/j.jiph.2016.04.012
  11. Marano N, Angelo KM, Merrill RD, et al. Expanding travel medicine in the 21st century to address the health needs of the world’s migrants.J Travel Med. 2018;25. doi:10.1093/jtm/tay067
  12. Hay RJ, Asiedu K. Skin-related neglected tropical diseases (skin NTDs)—a new challenge. Trop Med Infect Dis. 2018;4. doi:10.3390/tropicalmed4010004
  13. NIAID. Neglected tropical diseases. Updated July 11, 2016. Accessed April 4, 2024. https://www.niaid.nih.gov/research/neglected-tropical-diseases
  14. Arlian LG, Morgan MS. A review of Sarcoptes scabiei: past, present and future. Parasit Vectors. 2017;10:297. doi:10.1186/s13071-017-2234-1
  15. Arlian LG, Runyan RA, Achar S, et al. Survival and infectivity of Sarcoptes scabiei var. canis and var. hominis. J Am Acad Dermatol. 1984;11(2 pt 1):210-215. doi:10.1016/s0190-9622(84)70151-4
  16. Chandler DJ, Fuller LC. A review of scabies: an infestation more than skin deep. Dermatology. 2019;235:79-90. doi:10.1159/000495290
  17. Karimkhani C, Colombara DV, Drucker AM, et al. The global burden of scabies: a cross-sectional analysis from the Global Burden of Disease Study 2015. Lancet Infect Dis. 2017;17:1247-1254. doi:10.1016/S1473-3099(17)30483-8
  18. Romani L, Steer AC, Whitfeld MJ, et al. Prevalence of scabies and impetigo worldwide: a systematic review. Lancet Infect Dis. 2015;15:960-967. doi:10.1016/S1473-3099(15)00132-2
  19. Thomas C, Coates SJ, Engelman D, et al. Ectoparasites: scabies. J Am Acad Dermatol. 2020;82:533-548. doi:10.1016/j.jaad.2019.05.109
  20. Mellanby K, Johnson CG, Bartley WC. Treatment of scabies. Br Med J. 1942;2:1-4. doi:10.1136/bmj.2.4252.1
  21. Walton SF. The immunology of susceptibility and resistance to scabies. Parasit Immunol. 2010;32:532-540. doi:10.1111/j.1365-3024.2010.01218.x
  22. Coates SJ, Thomas C, Chosidow O, et al. Ectoparasites: pediculosis and tungiasis. J Am Acad Dermatol. 2020;82:551-569. doi:10.1016/j.jaad.2019.05.110
  23. Engelman D, Fuller LC, Steer AC; International Alliance for the Control of Scabies Delphi p. Consensus criteria for the diagnosis of scabies: a Delphi study of international experts. PLoS Negl Trop Dis. 2018;12:E0006549. doi:10.1371/journal.pntd.0006549
  24. World Health Organization. WHO Model Lists of Essential Medicines—23rd list, 2023. Updated July 26, 2023. Accessed April 8, 2024. https://www.who.int/publications/i/item/WHO-MHP-HPS-EML-2023.02
  25. Salavastru CM, Chosidow O, Boffa MJ, et al. European guideline for the management of scabies. J Eur Acad Dermatol Venereol. 2017;31:1248-1253. doi:10.1111/jdv.14351
  26. Badiaga S, Brouqui P. Human louse-transmitted infectious diseases. Clin Microbiol Infect. 2012;18:332-337. doi:10.1111/j.1469-0691.2012.03778.x
  27. Leo NP, Campbell NJH, Yang X, et al. Evidence from mitochondrial DNA that head lice and body lice of humans (Phthiraptera: Pediculidae) are conspecific. J Med Entomol. 2002;39:662-666. doi:10.1603/0022-2585-39.4.662
  28. Chosidow O. Scabies and pediculosis. Lancet. 2000;355:819-826. doi:10.1016/S0140-6736(99)09458-1
  29. Arnaud A, Chosidow O, Détrez M-A, et al. Prevalences of scabies and pediculosis corporis among homeless people in the Paris region: results from two randomized cross-sectional surveys (HYTPEAC study). Br J Dermatol. 2016;174:104-112. doi:10.1111/bjd.14226
  30. Brouqui P. Arthropod-borne diseases associated with political and social disorder. Annu Rev Entomol. 2011;56:357-374. doi:10.1146/annurev-ento-120709-144739
  31. Ko CJ, Elston DM. Pediculosis. J Am Acad Dermatol. 2004;50:1-12. doi:10.1016/S0190-9622(03)02729-4
  32. Bloomfield D. Head lice. Pediatr Rev. 2002;23:34-35; discussion 34-35. doi:10.1542/pir.23-1-34
  33. Stone SP GJ, Bacelieri RE. Scabies, other mites, and pediculosis. In: Wolf K GL, Katz SI, et al (eds). Fitzpatrick’s Dermatology in General Medicine. McGraw Hill; 2008:2029.
  34. Foucault C, Ranque S, Badiaga S, et al. Oral ivermectin in the treatment of body lice. J Infect Dis. 2006;193:474-476. doi:10.1086/499279
  35. Benkouiten S, Drali R, Badiaga S, et al. Effect of permethrin-impregnated underwear on body lice in sheltered homeless persons: a randomized controlled trial. JAMA Dermatol. 2014;150:273-279. doi:10.1001/jamadermatol.2013.6398
  36. CDC. Parasites: Treatment. Updated October 15, 2019. Accessed April 4, 2024. https://www.cdc.gov/parasites/lice/head/treatment.html
  37. Devore CD, Schutze GE; Council on School Health and Committee on Infectious Diseases, American Academy of Pediatrics. Head lice. Pediatrics. 2015;135:e1355-e1365. doi:10.1542/peds.2015-0746
  38. Ohl ME, Spach DH. Bartonella quintana and urban trench fever. Clin Infect Dis. 2000;31:131-135. doi:10.1086/313890
  39. Drali R, Sangaré AK, Boutellis A, et al. Bartonella quintana in body lice from scalp hair of homeless persons, France. Emerg Infect Dis. 2014;20:907-908. doi:10.3201/eid2005.131242
  40. Rudd N, Zakaria A, Kohn MA, et al. Association of body lice infestation with hemoglobin values in hospitalized dermatology patients. JAMA Dermatol. 2022;158:691-693. doi:10.1001/jamadermatol.2022.0818
  41. Guss DA, Koenig M, Castillo EM. Severe iron deficiency anemia and lice infestation. J Emergency Med. 2011;41:362-365. doi:10.1016/j.jemermed.2010.05.030
  42. Neglected tropical diseases of the skin: WHO launches mobile application to facilitate diagnosis. News release. World Health Organization; July 16, 2020. Accessed April 4, 2024. https://www.who.int/news/item/16-07-2020-neglected-tropical-diseases-of-the-skin-who-launches-mobile-application-to-facilitate-diagnosis
  43. Padovese V, Fuller LC, Griffiths CEM, et al; Migrant Health Dermatology Working Group of the International Foundation for Dermatology. Migrant skin health: perspectives from the Migrant Health Summit, Malta, 2022. Br J Dermatology. 2023;188:553-554. doi:10.1093/bjd/ljad001
  44. Knapp AP, Rehmus W, Chang AY. Skin diseases in displaced populations: a review of contributing factors, challenges, and approaches to care. Int J Dermatol. 2020;59:1299-1311. doi:10.1111/ijd.15063
  45. Norman FF, Comeche B, Chamorro S, et al. Overcoming challenges in the diagnosis and treatment of parasitic infectious diseases in migrants. Expert Rev Anti-infective Therapy. 2020;18:127-143. doi:10.1080/14787210.2020.1713099
  46. Skin NTDs: prioritizing integrated approaches to reduce suffering, psychosocial impact and stigmatization. News release. World Health Organization; October 29, 2020. Accessed April 4, 2024. https://www.who.int/news/item/29-10-2020-skin-ntds-prioritizing-integrated-approaches-to-reduce-suffering-psychosocial-impact-and-stigmatization
Article PDF
Author and Disclosure Information

Alexis G. Strahan is from the Mercer University School of Medicine, Savannah, Georgia. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

All images are in the public domain.

Correspondence: Alexis G. Strahan, MD, MSN, 55 Fruit St, Bartlett Hall 6R, Boston, MA 02114 (alexis.grabow.strahan@live.mercer.edu).

Issue
Cutis - 113(4)
Publications
Topics
Page Number
E16-E21
Sections
Author and Disclosure Information

Alexis G. Strahan is from the Mercer University School of Medicine, Savannah, Georgia. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

All images are in the public domain.

Correspondence: Alexis G. Strahan, MD, MSN, 55 Fruit St, Bartlett Hall 6R, Boston, MA 02114 (alexis.grabow.strahan@live.mercer.edu).

Author and Disclosure Information

Alexis G. Strahan is from the Mercer University School of Medicine, Savannah, Georgia. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

All images are in the public domain.

Correspondence: Alexis G. Strahan, MD, MSN, 55 Fruit St, Bartlett Hall 6R, Boston, MA 02114 (alexis.grabow.strahan@live.mercer.edu).

Article PDF
Article PDF

Approximately 108 million individuals have been forcibly displaced across the globe as of 2022, 35 million of whom are formally designated as refugees.1,2 The United States has coordinated resettlement of more refugee populations than any other country; the most common countries of origin are the Democratic Republic of the Congo, Syria, Afghanistan, and Myanmar.3 In 2021, policy to increase the number of refugees resettled in the United States by more than 700% (from 15,000 up to 125,000) was established; since enactment, the United States has seen more than double the refugee arrivals in 2023 than the prior year, making medical care for this population increasingly relevant for the dermatologist.4

Understanding how to care for this population begins with an accurate understanding of the term refugee. The United Nations defines a refugee as a person who is unwilling or unable to return to their country of nationality because of persecution or well-founded fear of persecution due to race, religion, nationality, membership in a particular social group, or political opinion. This term grants a protected status under international law and encompasses access to travel assistance, housing, cultural orientation, and medical evaluation upon resettlement.5,6

The burden of treatable dermatologic conditions in refugee populations ranges from 19% to 96% in the literature7,8 and varies from inflammatory disorders to infectious and parasitic diseases.9 In one study of 6899 displaced individuals in Greece, the prevalence of dermatologic conditions was higher than traumatic injury, cardiac disease, psychological conditions, and dental disease.10

When outlining differential diagnoses for parasitic infestations of the skin that affect refugee populations, helpful considerations include the individual’s country of origin, route traveled, and method of travel.11 Parasitic infestations specifically are more common in refugee populations when there are barriers to basic hygiene, crowded living or travel conditions, or lack of access to health care, which they may experience at any point in their home country, during travel, or in resettlement housing.8

Even with limited examination and diagnostic resources, the skin is the most accessible first indication of patients’ overall well-being and often provides simple diagnostic clues—in combination with contextualization of the patient’s unique circumstances—necessary for successful diagnosis and treatment of scabies and pediculosis.12 The dermatologist working with refugee populations may be the first set of eyes available and trained to discern skin infestations and therefore has the potential to improve overall outcomes.

Some parasitic infestations in refugee populations may fall under the category of neglected tropical diseases, including scabies, ascariasis, trypanosomiasis, leishmaniasis, and schistosomiasis; they affect an estimated 1 billion individuals across the globe but historically have been underrepresented in the literature and in health policy due in part to limited access to care.13 This review will focus on infestations by the scabies mite (Sarcoptes scabiei var hominis) and the human louse, as these frequently are encountered, easily diagnosed, and treatable by trained clinicians, even in resource-limited settings.

Scabies

Scabies is a parasitic skin infestation caused by the 8-legged mite Sarcoptes scabiei var hominis. The female mite begins the infestation process via penetration of the epidermis, particularly the stratum corneum, and commences laying eggs (Figure 1). The subsequent larvae emerge 48 to 72 hours later and remain burrowed in the epidermis. The larvae mature over the next 10 to 14 days and continue the reproductive cycle.14,15 Symptoms of infestation occurs due to a hypersensitivity reaction to the mite and its by-products.16 Transmission of the mite primarily occurs via direct (skin-to-skin) contact with infected individuals or environmental surfaces for 24 to36 hours in specific conditions, though the latter source has been debated in the literature.

CT113004016_fig1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Sarcoptes%20scabiei%20mite%20(A)%2C%20ova%20(B)%2C%20and%20scybala%20(C)%20on%20microscopic%20evaluation.%3C%2Fp%3E

 

 

The method of transmission is particularly important when considering care for refugee populations. Scabies is found most often in those living in or traveling from tropical regions including East Asia, Southeast Asia, Oceania, and Latin America.17 In displaced or refugee populations, a lack of access to basic hygiene, extended travel in close quarters, and suboptimal health care access all may lead to an increased incidence of untreated scabies infestations.18 Scabies is more prevalent in children, with increased potential for secondary bacterial infections with Streptococcus and Staphylococcus species due to excoriation in unsanitary conditions. Secondary infection with Streptococcus pyogenes can lead to acute poststreptococcal glomerulonephritis, which accounts for a large burden of chronic kidney disease in affected populations.19 However, scabies may be found in any population, regardless of hygiene or health care access. Treating health care providers should keep a broad differential.

Presentation—The latency of scabies symptoms is 2 to 6 weeks in a primary outbreak and may be as short as 1 to 3 days with re-infestation, following the course of delayed-type hypersensitivity.20 The initial hallmark symptom is pruritus with increased severity in the evening. Visible lesions, excoriations, and burrows associated with scattered vesicles or pustules may be seen over the web spaces of the hands and feet, volar surfaces of the wrists, axillae, waist, genitalia, inner thighs, or buttocks.19 Chronic infestation often manifests with genital nodules. In populations with limited access to health care, there are reports of a sensitization phenomenon in which the individual may become less symptomatic after 4 to 6 weeks and yet be a potential carrier of the mite.21

Those with compromised immune function, such as individuals living with HIV or severe malnutrition, may present with crusted scabies, a variant that manifests as widespread hyperkeratotic scaling with more pronounced involvement of the head, neck, and acral areas. In contrast to classic scabies, crusted scabies is associated with minimal pruritus.22

Diagnosis—The diagnosis of scabies is largely clinical with confirmation through skin scrapings. The International Alliance for Control of Scabies has established diagnostic criteria that include a combination of clinical findings, history, and visualization of mites.23 A dermatologist working with refugee populations may employ any combination of history (eg, nocturnal itch, exposure to an affected individual) or clinical findings along with a high degree of suspicion in those with elevated risk. Visualization of mites is helpful to confirm the diagnosis and may be completed with the application of mineral oil at the terminal end of a burrow, skin scraping with a surgical blade or needle, and examination under light microscopy.

Treatment—First-line treatment for scabies consists of application of permethrin cream 5% on the skin of the neck to the soles of the feet, which is to be left on for 8 to 14 hours followed by rinsing. Re-application is recommended in 1 to 2 weeks. Oral ivermectin is a reasonable alternative to permethrin cream due to its low cost and easy administration in large affected groups. It is not labeled for use in pregnant women or children weighing less than 15 kg but has no selective fetal toxicity. Treatment of scabies with ivermectin has the benefit of treating many other parasitic infections. Both medications are on the World Health Organization Model List of Essential Medications and are widely available for treating providers, even in resource-limited settings.24

Much of the world still uses benzyl benzoate or precipitated sulfur ointment to treat scabies, and some botanicals used in folk medicine have genuine antiscabetic properties. Pruritus may persist for 1 to 4 weeks following treatment and does not indicate treatment failure. Topical camphor and menthol preparations, low-potency topical corticosteroids, or emollients all may be employed for relief.25Sarna is a Spanish term for scabies and has become the proprietary name for topical antipruritic agents. Additional methods of treatment and prevention include washing clothes and linens in hot water and drying on high heat. If machine washing is not available, clothing and linens may be sealed in a plastic bag for 72 hours.

Pediculosis

Pediculosis is an infestation caused by the ectoparasite Pediculus humanus, an obligate, sesame seed–sized louse that feeds exclusively on the blood of its host (Figure 2).26 Of the lice species, 2 require humans as hosts; one is P humanus and the other is Pthirus pubis (pubic lice). Pediculus humanus may be further classified into morphologies based largely on the affected area: body (P humanus corporis) or head (P humanus capitis), both of which will be discussed.27

CT113004016_fig2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20%3Cem%3EPediculus%20humanus%3C%2Fem%3E%20(louse)%2C%20adult%20form.%3C%2Fp%3E

 

 

Lice primarily attach to clothing and hair shafts, then transfer to the skin for blood feeds. Females lay eggs that hatch 6 to 10 days later, subsequently maturing into adults. The lifespan of these parasites with regular access to a host is 1 to 3 months for head lice and 18 days for body lice vs only 3 to 5 days without a host.28 Transmission of P humanus capitis primarily occurs via direct contact with affected individuals, either head-to-head contact or sharing of items such as brushes and headscarves; P humanus corporis also may be transmitted via direct contact with affected individuals or clothing.

Pediculosis is an important infestation to consider when providing care for refugee populations. Risk factors include lack of access to basic hygiene, including regular bathing or laundering of clothing, and crowded conditions that make direct person-to-person contact with affected individuals more likely.29 Body lice are associated more often with domestic turbulence and displaced populations30 in comparison to head lice, which have broad demographic variables, most often affecting females and children.28 Fatty acids in adult male sebum make the scalp less hospitable to lice.

Presentation—The most common clinical manifestation of pediculosis is pruritus. Cutaneous findings can include papules, wheals, or hemorrhagic puncta secondary to the louse bite. Due to the Tyndall effect of deep hemosiderin pigment, blue-grey macules termed maculae ceruleae (Figure 3) also may be present in chronic infestations of pediculosis pubis, in contrast to pediculosis capitis or corporis.31 Body louse infestation is associated with a general pruritus concentrated on the neck, shoulders, and waist—areas where clothing makes the most direct contact. Lesions may be visible and include eczematous patches with excoriation and possible secondary bacterial infection. Chronic infestation may exhibit lichenification or hyperpigmentation in associated areas. Head lice most often manifest with localized scalp pruritus and associated excoriation and cervical or occipital lymphadenopathy.32

CT113004016_fig3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Maculae%20ceruleae%E2%80%94blue-grey%20macules%E2%80%94may%20be%20present%20on%20the%20skin%20secondary%20to%20%3Cem%3EPediculosis%3C%2Fem%3E%20infestation.%3C%2Fp%3E

Diagnosis—The diagnosis of pediculosis is clinical, with confirmation requiring direct examination of the insect or nits (the egg case of the parasite)(Figure 4). Body lice and associated nits can be visualized on clothing seams near areas of highest body temperature, particularly the waistband. Head lice may be visualized crawling on hair shafts or on a louse comb. Nits are firmly attached to hair shafts and are visible to the naked eye, whereas pseudonits slide freely along the hair shaft and are not a manifestation of louse infestation (Figure 5).31

CT113004016_fig4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Pediculosis%20nits%E2%80%94the%20egg%20cases%20of%20the%20parasite%E2%80%94may%20firmly%20attach%20to%20the%20hair%20shaft.%3C%2Fp%3E

Treatment—Treatment varies by affected area. Pediculosis corporis may be treated with permethrin cream 5% applied to the entire body and left on for 8 to 10 hours, but this may not be necessary if facilities are available to wash and dry clothing.33 The use of oral ivermectin and permethrin-impregnated underwear both have been proposed.34,35 Treatment of pediculosis capitis may be accomplished with a variety of topical pediculicides including permethrin, pyrethrum with piperonyl butoxide, dimethicone, malathion, benzyl alcohol, spinosad, and topical ivermectin.22 Topical corticosteroids or emollients may be employed for residual pruritus.

CT113004016_fig5.jpg
%3Cp%3E%3Cstrong%3EFIGURE%205.%3C%2Fstrong%3E%20The%20pseudonit%20closely%20mimics%20pediculosis%20nits%20but%20consists%20of%20keratinized%20cell%20casts%20that%20are%20freely%20dislodged.%3C%2Fp%3E

Equally important is environmental elimination of infestation. Clothing should be discarded if possible or washed and dried using high heat. If neither approach is possible or appropriate, clothing may be sealed in a plastic bag for 2 weeks or treated with a pediculicide. Nit combing is an important adjunct in the treatment of pediculosis capitis.36 It is important to encourage return to work and/or school immediately after treatment. “No nit” policies are more harmful to education than helpful for prevention of investation.37

Pediculosis corporis may transmit infectious agents including Bartonella quintana, (trench fever, endocarditis, bacillary angiomatosis), Borrelia recurrentis (louse-borne relapsing fever), and Rickettsia prowazekii (epidemic typhus).31,38,39 Additionally, severe pediculosis infestations have the potential to cause chronic blood loss in affected populations. In a study of patients with active pediculosis infestation, mean hemoglobin values were found to be 2.5 g/dL lower than a matched population without infestation.40 It is important to consider pediculosis as a risk for iron-deficiency anemia in populations who are known to lack access to regular medical evaluation.41

 

 

Future Considerations

Increased access to tools and education for clinicians treating refugee populations is key to reducing the burden of parasitic skin disease and related morbidity and mortality in vulnerable groups both domestically and globally. One such tool, the Skin NTDs App, was launched by the World Health Organization in 2020. It is available for free for Android and iOS devices to assist clinicians in the field with the diagnosis and treatment of neglected tropical diseases—including scabies—that may affect refugee populations.42

Additionally, to both improve access and limit preventable sequelae, future investigations into appropriate models of community-based care are paramount. The model of community-based care is centered on the idea of care provision that prioritizes safety, accessibility, affordability, and acceptability in an environment closest to vulnerable populations. The largest dermatologic society, the International League of Dermatological Societies, formed a Migrant Health Dermatology Working Group that prioritizes understanding and improving care for refugee and migrant populations; this group hosted a summit in 2022, bringing together international subject matter leaders to discuss such models of care and set goals for the creation of tool kits for patients, frontline health care workers, and dermatologists.43

Conclusion

Improvement in dermatologic care of refugee populations includes provision of culturally and linguistically appropriate care by trained clinicians, adequate access to the most essential medications, and basic physical or legal access to health care systems in general.8,11,44 Parasitic infestations have the potential to remain asymptomatic for extended periods of time and result in spread to potentially nonendemic regions of resettlement.45 Additionally, the psychosocial well-being of refugee populations upon resettlement may be negatively affected by stigma of disease processes such as scabies and pediculosis, leading to additional barriers to successful re-entry into the patient’s new environment.46 Therefore, proper screening, diagnosis, and treatment of the most common parasitic infestations in this population have great potential to improve outcomes for large groups across the globe.

Approximately 108 million individuals have been forcibly displaced across the globe as of 2022, 35 million of whom are formally designated as refugees.1,2 The United States has coordinated resettlement of more refugee populations than any other country; the most common countries of origin are the Democratic Republic of the Congo, Syria, Afghanistan, and Myanmar.3 In 2021, policy to increase the number of refugees resettled in the United States by more than 700% (from 15,000 up to 125,000) was established; since enactment, the United States has seen more than double the refugee arrivals in 2023 than the prior year, making medical care for this population increasingly relevant for the dermatologist.4

Understanding how to care for this population begins with an accurate understanding of the term refugee. The United Nations defines a refugee as a person who is unwilling or unable to return to their country of nationality because of persecution or well-founded fear of persecution due to race, religion, nationality, membership in a particular social group, or political opinion. This term grants a protected status under international law and encompasses access to travel assistance, housing, cultural orientation, and medical evaluation upon resettlement.5,6

The burden of treatable dermatologic conditions in refugee populations ranges from 19% to 96% in the literature7,8 and varies from inflammatory disorders to infectious and parasitic diseases.9 In one study of 6899 displaced individuals in Greece, the prevalence of dermatologic conditions was higher than traumatic injury, cardiac disease, psychological conditions, and dental disease.10

When outlining differential diagnoses for parasitic infestations of the skin that affect refugee populations, helpful considerations include the individual’s country of origin, route traveled, and method of travel.11 Parasitic infestations specifically are more common in refugee populations when there are barriers to basic hygiene, crowded living or travel conditions, or lack of access to health care, which they may experience at any point in their home country, during travel, or in resettlement housing.8

Even with limited examination and diagnostic resources, the skin is the most accessible first indication of patients’ overall well-being and often provides simple diagnostic clues—in combination with contextualization of the patient’s unique circumstances—necessary for successful diagnosis and treatment of scabies and pediculosis.12 The dermatologist working with refugee populations may be the first set of eyes available and trained to discern skin infestations and therefore has the potential to improve overall outcomes.

Some parasitic infestations in refugee populations may fall under the category of neglected tropical diseases, including scabies, ascariasis, trypanosomiasis, leishmaniasis, and schistosomiasis; they affect an estimated 1 billion individuals across the globe but historically have been underrepresented in the literature and in health policy due in part to limited access to care.13 This review will focus on infestations by the scabies mite (Sarcoptes scabiei var hominis) and the human louse, as these frequently are encountered, easily diagnosed, and treatable by trained clinicians, even in resource-limited settings.

Scabies

Scabies is a parasitic skin infestation caused by the 8-legged mite Sarcoptes scabiei var hominis. The female mite begins the infestation process via penetration of the epidermis, particularly the stratum corneum, and commences laying eggs (Figure 1). The subsequent larvae emerge 48 to 72 hours later and remain burrowed in the epidermis. The larvae mature over the next 10 to 14 days and continue the reproductive cycle.14,15 Symptoms of infestation occurs due to a hypersensitivity reaction to the mite and its by-products.16 Transmission of the mite primarily occurs via direct (skin-to-skin) contact with infected individuals or environmental surfaces for 24 to36 hours in specific conditions, though the latter source has been debated in the literature.

CT113004016_fig1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Sarcoptes%20scabiei%20mite%20(A)%2C%20ova%20(B)%2C%20and%20scybala%20(C)%20on%20microscopic%20evaluation.%3C%2Fp%3E

 

 

The method of transmission is particularly important when considering care for refugee populations. Scabies is found most often in those living in or traveling from tropical regions including East Asia, Southeast Asia, Oceania, and Latin America.17 In displaced or refugee populations, a lack of access to basic hygiene, extended travel in close quarters, and suboptimal health care access all may lead to an increased incidence of untreated scabies infestations.18 Scabies is more prevalent in children, with increased potential for secondary bacterial infections with Streptococcus and Staphylococcus species due to excoriation in unsanitary conditions. Secondary infection with Streptococcus pyogenes can lead to acute poststreptococcal glomerulonephritis, which accounts for a large burden of chronic kidney disease in affected populations.19 However, scabies may be found in any population, regardless of hygiene or health care access. Treating health care providers should keep a broad differential.

Presentation—The latency of scabies symptoms is 2 to 6 weeks in a primary outbreak and may be as short as 1 to 3 days with re-infestation, following the course of delayed-type hypersensitivity.20 The initial hallmark symptom is pruritus with increased severity in the evening. Visible lesions, excoriations, and burrows associated with scattered vesicles or pustules may be seen over the web spaces of the hands and feet, volar surfaces of the wrists, axillae, waist, genitalia, inner thighs, or buttocks.19 Chronic infestation often manifests with genital nodules. In populations with limited access to health care, there are reports of a sensitization phenomenon in which the individual may become less symptomatic after 4 to 6 weeks and yet be a potential carrier of the mite.21

Those with compromised immune function, such as individuals living with HIV or severe malnutrition, may present with crusted scabies, a variant that manifests as widespread hyperkeratotic scaling with more pronounced involvement of the head, neck, and acral areas. In contrast to classic scabies, crusted scabies is associated with minimal pruritus.22

Diagnosis—The diagnosis of scabies is largely clinical with confirmation through skin scrapings. The International Alliance for Control of Scabies has established diagnostic criteria that include a combination of clinical findings, history, and visualization of mites.23 A dermatologist working with refugee populations may employ any combination of history (eg, nocturnal itch, exposure to an affected individual) or clinical findings along with a high degree of suspicion in those with elevated risk. Visualization of mites is helpful to confirm the diagnosis and may be completed with the application of mineral oil at the terminal end of a burrow, skin scraping with a surgical blade or needle, and examination under light microscopy.

Treatment—First-line treatment for scabies consists of application of permethrin cream 5% on the skin of the neck to the soles of the feet, which is to be left on for 8 to 14 hours followed by rinsing. Re-application is recommended in 1 to 2 weeks. Oral ivermectin is a reasonable alternative to permethrin cream due to its low cost and easy administration in large affected groups. It is not labeled for use in pregnant women or children weighing less than 15 kg but has no selective fetal toxicity. Treatment of scabies with ivermectin has the benefit of treating many other parasitic infections. Both medications are on the World Health Organization Model List of Essential Medications and are widely available for treating providers, even in resource-limited settings.24

Much of the world still uses benzyl benzoate or precipitated sulfur ointment to treat scabies, and some botanicals used in folk medicine have genuine antiscabetic properties. Pruritus may persist for 1 to 4 weeks following treatment and does not indicate treatment failure. Topical camphor and menthol preparations, low-potency topical corticosteroids, or emollients all may be employed for relief.25Sarna is a Spanish term for scabies and has become the proprietary name for topical antipruritic agents. Additional methods of treatment and prevention include washing clothes and linens in hot water and drying on high heat. If machine washing is not available, clothing and linens may be sealed in a plastic bag for 72 hours.

Pediculosis

Pediculosis is an infestation caused by the ectoparasite Pediculus humanus, an obligate, sesame seed–sized louse that feeds exclusively on the blood of its host (Figure 2).26 Of the lice species, 2 require humans as hosts; one is P humanus and the other is Pthirus pubis (pubic lice). Pediculus humanus may be further classified into morphologies based largely on the affected area: body (P humanus corporis) or head (P humanus capitis), both of which will be discussed.27

CT113004016_fig2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20%3Cem%3EPediculus%20humanus%3C%2Fem%3E%20(louse)%2C%20adult%20form.%3C%2Fp%3E

 

 

Lice primarily attach to clothing and hair shafts, then transfer to the skin for blood feeds. Females lay eggs that hatch 6 to 10 days later, subsequently maturing into adults. The lifespan of these parasites with regular access to a host is 1 to 3 months for head lice and 18 days for body lice vs only 3 to 5 days without a host.28 Transmission of P humanus capitis primarily occurs via direct contact with affected individuals, either head-to-head contact or sharing of items such as brushes and headscarves; P humanus corporis also may be transmitted via direct contact with affected individuals or clothing.

Pediculosis is an important infestation to consider when providing care for refugee populations. Risk factors include lack of access to basic hygiene, including regular bathing or laundering of clothing, and crowded conditions that make direct person-to-person contact with affected individuals more likely.29 Body lice are associated more often with domestic turbulence and displaced populations30 in comparison to head lice, which have broad demographic variables, most often affecting females and children.28 Fatty acids in adult male sebum make the scalp less hospitable to lice.

Presentation—The most common clinical manifestation of pediculosis is pruritus. Cutaneous findings can include papules, wheals, or hemorrhagic puncta secondary to the louse bite. Due to the Tyndall effect of deep hemosiderin pigment, blue-grey macules termed maculae ceruleae (Figure 3) also may be present in chronic infestations of pediculosis pubis, in contrast to pediculosis capitis or corporis.31 Body louse infestation is associated with a general pruritus concentrated on the neck, shoulders, and waist—areas where clothing makes the most direct contact. Lesions may be visible and include eczematous patches with excoriation and possible secondary bacterial infection. Chronic infestation may exhibit lichenification or hyperpigmentation in associated areas. Head lice most often manifest with localized scalp pruritus and associated excoriation and cervical or occipital lymphadenopathy.32

CT113004016_fig3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Maculae%20ceruleae%E2%80%94blue-grey%20macules%E2%80%94may%20be%20present%20on%20the%20skin%20secondary%20to%20%3Cem%3EPediculosis%3C%2Fem%3E%20infestation.%3C%2Fp%3E

Diagnosis—The diagnosis of pediculosis is clinical, with confirmation requiring direct examination of the insect or nits (the egg case of the parasite)(Figure 4). Body lice and associated nits can be visualized on clothing seams near areas of highest body temperature, particularly the waistband. Head lice may be visualized crawling on hair shafts or on a louse comb. Nits are firmly attached to hair shafts and are visible to the naked eye, whereas pseudonits slide freely along the hair shaft and are not a manifestation of louse infestation (Figure 5).31

CT113004016_fig4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Pediculosis%20nits%E2%80%94the%20egg%20cases%20of%20the%20parasite%E2%80%94may%20firmly%20attach%20to%20the%20hair%20shaft.%3C%2Fp%3E

Treatment—Treatment varies by affected area. Pediculosis corporis may be treated with permethrin cream 5% applied to the entire body and left on for 8 to 10 hours, but this may not be necessary if facilities are available to wash and dry clothing.33 The use of oral ivermectin and permethrin-impregnated underwear both have been proposed.34,35 Treatment of pediculosis capitis may be accomplished with a variety of topical pediculicides including permethrin, pyrethrum with piperonyl butoxide, dimethicone, malathion, benzyl alcohol, spinosad, and topical ivermectin.22 Topical corticosteroids or emollients may be employed for residual pruritus.

CT113004016_fig5.jpg
%3Cp%3E%3Cstrong%3EFIGURE%205.%3C%2Fstrong%3E%20The%20pseudonit%20closely%20mimics%20pediculosis%20nits%20but%20consists%20of%20keratinized%20cell%20casts%20that%20are%20freely%20dislodged.%3C%2Fp%3E

Equally important is environmental elimination of infestation. Clothing should be discarded if possible or washed and dried using high heat. If neither approach is possible or appropriate, clothing may be sealed in a plastic bag for 2 weeks or treated with a pediculicide. Nit combing is an important adjunct in the treatment of pediculosis capitis.36 It is important to encourage return to work and/or school immediately after treatment. “No nit” policies are more harmful to education than helpful for prevention of investation.37

Pediculosis corporis may transmit infectious agents including Bartonella quintana, (trench fever, endocarditis, bacillary angiomatosis), Borrelia recurrentis (louse-borne relapsing fever), and Rickettsia prowazekii (epidemic typhus).31,38,39 Additionally, severe pediculosis infestations have the potential to cause chronic blood loss in affected populations. In a study of patients with active pediculosis infestation, mean hemoglobin values were found to be 2.5 g/dL lower than a matched population without infestation.40 It is important to consider pediculosis as a risk for iron-deficiency anemia in populations who are known to lack access to regular medical evaluation.41

 

 

Future Considerations

Increased access to tools and education for clinicians treating refugee populations is key to reducing the burden of parasitic skin disease and related morbidity and mortality in vulnerable groups both domestically and globally. One such tool, the Skin NTDs App, was launched by the World Health Organization in 2020. It is available for free for Android and iOS devices to assist clinicians in the field with the diagnosis and treatment of neglected tropical diseases—including scabies—that may affect refugee populations.42

Additionally, to both improve access and limit preventable sequelae, future investigations into appropriate models of community-based care are paramount. The model of community-based care is centered on the idea of care provision that prioritizes safety, accessibility, affordability, and acceptability in an environment closest to vulnerable populations. The largest dermatologic society, the International League of Dermatological Societies, formed a Migrant Health Dermatology Working Group that prioritizes understanding and improving care for refugee and migrant populations; this group hosted a summit in 2022, bringing together international subject matter leaders to discuss such models of care and set goals for the creation of tool kits for patients, frontline health care workers, and dermatologists.43

Conclusion

Improvement in dermatologic care of refugee populations includes provision of culturally and linguistically appropriate care by trained clinicians, adequate access to the most essential medications, and basic physical or legal access to health care systems in general.8,11,44 Parasitic infestations have the potential to remain asymptomatic for extended periods of time and result in spread to potentially nonendemic regions of resettlement.45 Additionally, the psychosocial well-being of refugee populations upon resettlement may be negatively affected by stigma of disease processes such as scabies and pediculosis, leading to additional barriers to successful re-entry into the patient’s new environment.46 Therefore, proper screening, diagnosis, and treatment of the most common parasitic infestations in this population have great potential to improve outcomes for large groups across the globe.

References
  1. Monin K, Batalova J, Lai T. Refugees and Asylees in the United States. Migration Information Source. Published May 13, 2021. Accessed April 4, 2024. https://www.migrationpolicy.org/article/refugees-and-asylees-united-states-2021
  2. UNHCR. Figures at a Glance. UNHCR USA. Update June 14, 2023. Accessed April 4, 2024. https://www.unhcr.org/en-us/figures-at-a-glance.html
  3. UNHCR. Refugee resettlement facts. Published October 2023. Accessed April 8, 2024. https://www.unhcr.org/us/media/refugee-resettlement-facts
  4. US Department of State. Report to Congress on Proposed Refugee Admissions for Fiscal Year 2024. Published November 3, 2023. Accessed April 8, 2024. https://www.state.gov/report-to-congress-on-proposed-refugee-admissions-for-fiscal-year-2024/
  5. UNHCR. Compact for Migration: Definitions. United Nations. Accessed April 4, 2024. https://refugeesmigrants.un.org/definitions
  6. United Nations High Commissioner for Refugees (UNHCR). Convention and Protocol Relating to the Status of Refugees. Published December 2010. Accessed January 11, 2024. https://www.unhcr.org/us/media/convention-and-protocol-relating-status-refugees
  7. Kibar Öztürk M. Skin diseases in rural Nyala, Sudan (in a rural hospital, in 12 orphanages, and in two refugee camps). Int J Dermatol. 2019;58:1341-1349. doi:10.1111/ijd.14619
  8. Padovese V, Knapp A. Challenges of managing skin diseases in refugees and migrants. Dermatol Clin. 2021;39:101-115. doi:10.1016/j.det.2020.08.010
  9. Saikal SL, Ge L, Mir A, et al. Skin disease profile of Syrian refugees in Jordan: a field-mission assessment. J Eur Acad Dermatol Venereol. 2020;34:419-425. doi:10.1111/jdv.15909
  10. Eonomopoulou A, Pavli A, Stasinopoulou P, et al. Migrant screening: lessons learned from the migrant holding level at the Greek-Turkish borders. J Infect Public Health. 2017;10:177-184. doi:10.1016/j.jiph.2016.04.012
  11. Marano N, Angelo KM, Merrill RD, et al. Expanding travel medicine in the 21st century to address the health needs of the world’s migrants.J Travel Med. 2018;25. doi:10.1093/jtm/tay067
  12. Hay RJ, Asiedu K. Skin-related neglected tropical diseases (skin NTDs)—a new challenge. Trop Med Infect Dis. 2018;4. doi:10.3390/tropicalmed4010004
  13. NIAID. Neglected tropical diseases. Updated July 11, 2016. Accessed April 4, 2024. https://www.niaid.nih.gov/research/neglected-tropical-diseases
  14. Arlian LG, Morgan MS. A review of Sarcoptes scabiei: past, present and future. Parasit Vectors. 2017;10:297. doi:10.1186/s13071-017-2234-1
  15. Arlian LG, Runyan RA, Achar S, et al. Survival and infectivity of Sarcoptes scabiei var. canis and var. hominis. J Am Acad Dermatol. 1984;11(2 pt 1):210-215. doi:10.1016/s0190-9622(84)70151-4
  16. Chandler DJ, Fuller LC. A review of scabies: an infestation more than skin deep. Dermatology. 2019;235:79-90. doi:10.1159/000495290
  17. Karimkhani C, Colombara DV, Drucker AM, et al. The global burden of scabies: a cross-sectional analysis from the Global Burden of Disease Study 2015. Lancet Infect Dis. 2017;17:1247-1254. doi:10.1016/S1473-3099(17)30483-8
  18. Romani L, Steer AC, Whitfeld MJ, et al. Prevalence of scabies and impetigo worldwide: a systematic review. Lancet Infect Dis. 2015;15:960-967. doi:10.1016/S1473-3099(15)00132-2
  19. Thomas C, Coates SJ, Engelman D, et al. Ectoparasites: scabies. J Am Acad Dermatol. 2020;82:533-548. doi:10.1016/j.jaad.2019.05.109
  20. Mellanby K, Johnson CG, Bartley WC. Treatment of scabies. Br Med J. 1942;2:1-4. doi:10.1136/bmj.2.4252.1
  21. Walton SF. The immunology of susceptibility and resistance to scabies. Parasit Immunol. 2010;32:532-540. doi:10.1111/j.1365-3024.2010.01218.x
  22. Coates SJ, Thomas C, Chosidow O, et al. Ectoparasites: pediculosis and tungiasis. J Am Acad Dermatol. 2020;82:551-569. doi:10.1016/j.jaad.2019.05.110
  23. Engelman D, Fuller LC, Steer AC; International Alliance for the Control of Scabies Delphi p. Consensus criteria for the diagnosis of scabies: a Delphi study of international experts. PLoS Negl Trop Dis. 2018;12:E0006549. doi:10.1371/journal.pntd.0006549
  24. World Health Organization. WHO Model Lists of Essential Medicines—23rd list, 2023. Updated July 26, 2023. Accessed April 8, 2024. https://www.who.int/publications/i/item/WHO-MHP-HPS-EML-2023.02
  25. Salavastru CM, Chosidow O, Boffa MJ, et al. European guideline for the management of scabies. J Eur Acad Dermatol Venereol. 2017;31:1248-1253. doi:10.1111/jdv.14351
  26. Badiaga S, Brouqui P. Human louse-transmitted infectious diseases. Clin Microbiol Infect. 2012;18:332-337. doi:10.1111/j.1469-0691.2012.03778.x
  27. Leo NP, Campbell NJH, Yang X, et al. Evidence from mitochondrial DNA that head lice and body lice of humans (Phthiraptera: Pediculidae) are conspecific. J Med Entomol. 2002;39:662-666. doi:10.1603/0022-2585-39.4.662
  28. Chosidow O. Scabies and pediculosis. Lancet. 2000;355:819-826. doi:10.1016/S0140-6736(99)09458-1
  29. Arnaud A, Chosidow O, Détrez M-A, et al. Prevalences of scabies and pediculosis corporis among homeless people in the Paris region: results from two randomized cross-sectional surveys (HYTPEAC study). Br J Dermatol. 2016;174:104-112. doi:10.1111/bjd.14226
  30. Brouqui P. Arthropod-borne diseases associated with political and social disorder. Annu Rev Entomol. 2011;56:357-374. doi:10.1146/annurev-ento-120709-144739
  31. Ko CJ, Elston DM. Pediculosis. J Am Acad Dermatol. 2004;50:1-12. doi:10.1016/S0190-9622(03)02729-4
  32. Bloomfield D. Head lice. Pediatr Rev. 2002;23:34-35; discussion 34-35. doi:10.1542/pir.23-1-34
  33. Stone SP GJ, Bacelieri RE. Scabies, other mites, and pediculosis. In: Wolf K GL, Katz SI, et al (eds). Fitzpatrick’s Dermatology in General Medicine. McGraw Hill; 2008:2029.
  34. Foucault C, Ranque S, Badiaga S, et al. Oral ivermectin in the treatment of body lice. J Infect Dis. 2006;193:474-476. doi:10.1086/499279
  35. Benkouiten S, Drali R, Badiaga S, et al. Effect of permethrin-impregnated underwear on body lice in sheltered homeless persons: a randomized controlled trial. JAMA Dermatol. 2014;150:273-279. doi:10.1001/jamadermatol.2013.6398
  36. CDC. Parasites: Treatment. Updated October 15, 2019. Accessed April 4, 2024. https://www.cdc.gov/parasites/lice/head/treatment.html
  37. Devore CD, Schutze GE; Council on School Health and Committee on Infectious Diseases, American Academy of Pediatrics. Head lice. Pediatrics. 2015;135:e1355-e1365. doi:10.1542/peds.2015-0746
  38. Ohl ME, Spach DH. Bartonella quintana and urban trench fever. Clin Infect Dis. 2000;31:131-135. doi:10.1086/313890
  39. Drali R, Sangaré AK, Boutellis A, et al. Bartonella quintana in body lice from scalp hair of homeless persons, France. Emerg Infect Dis. 2014;20:907-908. doi:10.3201/eid2005.131242
  40. Rudd N, Zakaria A, Kohn MA, et al. Association of body lice infestation with hemoglobin values in hospitalized dermatology patients. JAMA Dermatol. 2022;158:691-693. doi:10.1001/jamadermatol.2022.0818
  41. Guss DA, Koenig M, Castillo EM. Severe iron deficiency anemia and lice infestation. J Emergency Med. 2011;41:362-365. doi:10.1016/j.jemermed.2010.05.030
  42. Neglected tropical diseases of the skin: WHO launches mobile application to facilitate diagnosis. News release. World Health Organization; July 16, 2020. Accessed April 4, 2024. https://www.who.int/news/item/16-07-2020-neglected-tropical-diseases-of-the-skin-who-launches-mobile-application-to-facilitate-diagnosis
  43. Padovese V, Fuller LC, Griffiths CEM, et al; Migrant Health Dermatology Working Group of the International Foundation for Dermatology. Migrant skin health: perspectives from the Migrant Health Summit, Malta, 2022. Br J Dermatology. 2023;188:553-554. doi:10.1093/bjd/ljad001
  44. Knapp AP, Rehmus W, Chang AY. Skin diseases in displaced populations: a review of contributing factors, challenges, and approaches to care. Int J Dermatol. 2020;59:1299-1311. doi:10.1111/ijd.15063
  45. Norman FF, Comeche B, Chamorro S, et al. Overcoming challenges in the diagnosis and treatment of parasitic infectious diseases in migrants. Expert Rev Anti-infective Therapy. 2020;18:127-143. doi:10.1080/14787210.2020.1713099
  46. Skin NTDs: prioritizing integrated approaches to reduce suffering, psychosocial impact and stigmatization. News release. World Health Organization; October 29, 2020. Accessed April 4, 2024. https://www.who.int/news/item/29-10-2020-skin-ntds-prioritizing-integrated-approaches-to-reduce-suffering-psychosocial-impact-and-stigmatization
References
  1. Monin K, Batalova J, Lai T. Refugees and Asylees in the United States. Migration Information Source. Published May 13, 2021. Accessed April 4, 2024. https://www.migrationpolicy.org/article/refugees-and-asylees-united-states-2021
  2. UNHCR. Figures at a Glance. UNHCR USA. Update June 14, 2023. Accessed April 4, 2024. https://www.unhcr.org/en-us/figures-at-a-glance.html
  3. UNHCR. Refugee resettlement facts. Published October 2023. Accessed April 8, 2024. https://www.unhcr.org/us/media/refugee-resettlement-facts
  4. US Department of State. Report to Congress on Proposed Refugee Admissions for Fiscal Year 2024. Published November 3, 2023. Accessed April 8, 2024. https://www.state.gov/report-to-congress-on-proposed-refugee-admissions-for-fiscal-year-2024/
  5. UNHCR. Compact for Migration: Definitions. United Nations. Accessed April 4, 2024. https://refugeesmigrants.un.org/definitions
  6. United Nations High Commissioner for Refugees (UNHCR). Convention and Protocol Relating to the Status of Refugees. Published December 2010. Accessed January 11, 2024. https://www.unhcr.org/us/media/convention-and-protocol-relating-status-refugees
  7. Kibar Öztürk M. Skin diseases in rural Nyala, Sudan (in a rural hospital, in 12 orphanages, and in two refugee camps). Int J Dermatol. 2019;58:1341-1349. doi:10.1111/ijd.14619
  8. Padovese V, Knapp A. Challenges of managing skin diseases in refugees and migrants. Dermatol Clin. 2021;39:101-115. doi:10.1016/j.det.2020.08.010
  9. Saikal SL, Ge L, Mir A, et al. Skin disease profile of Syrian refugees in Jordan: a field-mission assessment. J Eur Acad Dermatol Venereol. 2020;34:419-425. doi:10.1111/jdv.15909
  10. Eonomopoulou A, Pavli A, Stasinopoulou P, et al. Migrant screening: lessons learned from the migrant holding level at the Greek-Turkish borders. J Infect Public Health. 2017;10:177-184. doi:10.1016/j.jiph.2016.04.012
  11. Marano N, Angelo KM, Merrill RD, et al. Expanding travel medicine in the 21st century to address the health needs of the world’s migrants.J Travel Med. 2018;25. doi:10.1093/jtm/tay067
  12. Hay RJ, Asiedu K. Skin-related neglected tropical diseases (skin NTDs)—a new challenge. Trop Med Infect Dis. 2018;4. doi:10.3390/tropicalmed4010004
  13. NIAID. Neglected tropical diseases. Updated July 11, 2016. Accessed April 4, 2024. https://www.niaid.nih.gov/research/neglected-tropical-diseases
  14. Arlian LG, Morgan MS. A review of Sarcoptes scabiei: past, present and future. Parasit Vectors. 2017;10:297. doi:10.1186/s13071-017-2234-1
  15. Arlian LG, Runyan RA, Achar S, et al. Survival and infectivity of Sarcoptes scabiei var. canis and var. hominis. J Am Acad Dermatol. 1984;11(2 pt 1):210-215. doi:10.1016/s0190-9622(84)70151-4
  16. Chandler DJ, Fuller LC. A review of scabies: an infestation more than skin deep. Dermatology. 2019;235:79-90. doi:10.1159/000495290
  17. Karimkhani C, Colombara DV, Drucker AM, et al. The global burden of scabies: a cross-sectional analysis from the Global Burden of Disease Study 2015. Lancet Infect Dis. 2017;17:1247-1254. doi:10.1016/S1473-3099(17)30483-8
  18. Romani L, Steer AC, Whitfeld MJ, et al. Prevalence of scabies and impetigo worldwide: a systematic review. Lancet Infect Dis. 2015;15:960-967. doi:10.1016/S1473-3099(15)00132-2
  19. Thomas C, Coates SJ, Engelman D, et al. Ectoparasites: scabies. J Am Acad Dermatol. 2020;82:533-548. doi:10.1016/j.jaad.2019.05.109
  20. Mellanby K, Johnson CG, Bartley WC. Treatment of scabies. Br Med J. 1942;2:1-4. doi:10.1136/bmj.2.4252.1
  21. Walton SF. The immunology of susceptibility and resistance to scabies. Parasit Immunol. 2010;32:532-540. doi:10.1111/j.1365-3024.2010.01218.x
  22. Coates SJ, Thomas C, Chosidow O, et al. Ectoparasites: pediculosis and tungiasis. J Am Acad Dermatol. 2020;82:551-569. doi:10.1016/j.jaad.2019.05.110
  23. Engelman D, Fuller LC, Steer AC; International Alliance for the Control of Scabies Delphi p. Consensus criteria for the diagnosis of scabies: a Delphi study of international experts. PLoS Negl Trop Dis. 2018;12:E0006549. doi:10.1371/journal.pntd.0006549
  24. World Health Organization. WHO Model Lists of Essential Medicines—23rd list, 2023. Updated July 26, 2023. Accessed April 8, 2024. https://www.who.int/publications/i/item/WHO-MHP-HPS-EML-2023.02
  25. Salavastru CM, Chosidow O, Boffa MJ, et al. European guideline for the management of scabies. J Eur Acad Dermatol Venereol. 2017;31:1248-1253. doi:10.1111/jdv.14351
  26. Badiaga S, Brouqui P. Human louse-transmitted infectious diseases. Clin Microbiol Infect. 2012;18:332-337. doi:10.1111/j.1469-0691.2012.03778.x
  27. Leo NP, Campbell NJH, Yang X, et al. Evidence from mitochondrial DNA that head lice and body lice of humans (Phthiraptera: Pediculidae) are conspecific. J Med Entomol. 2002;39:662-666. doi:10.1603/0022-2585-39.4.662
  28. Chosidow O. Scabies and pediculosis. Lancet. 2000;355:819-826. doi:10.1016/S0140-6736(99)09458-1
  29. Arnaud A, Chosidow O, Détrez M-A, et al. Prevalences of scabies and pediculosis corporis among homeless people in the Paris region: results from two randomized cross-sectional surveys (HYTPEAC study). Br J Dermatol. 2016;174:104-112. doi:10.1111/bjd.14226
  30. Brouqui P. Arthropod-borne diseases associated with political and social disorder. Annu Rev Entomol. 2011;56:357-374. doi:10.1146/annurev-ento-120709-144739
  31. Ko CJ, Elston DM. Pediculosis. J Am Acad Dermatol. 2004;50:1-12. doi:10.1016/S0190-9622(03)02729-4
  32. Bloomfield D. Head lice. Pediatr Rev. 2002;23:34-35; discussion 34-35. doi:10.1542/pir.23-1-34
  33. Stone SP GJ, Bacelieri RE. Scabies, other mites, and pediculosis. In: Wolf K GL, Katz SI, et al (eds). Fitzpatrick’s Dermatology in General Medicine. McGraw Hill; 2008:2029.
  34. Foucault C, Ranque S, Badiaga S, et al. Oral ivermectin in the treatment of body lice. J Infect Dis. 2006;193:474-476. doi:10.1086/499279
  35. Benkouiten S, Drali R, Badiaga S, et al. Effect of permethrin-impregnated underwear on body lice in sheltered homeless persons: a randomized controlled trial. JAMA Dermatol. 2014;150:273-279. doi:10.1001/jamadermatol.2013.6398
  36. CDC. Parasites: Treatment. Updated October 15, 2019. Accessed April 4, 2024. https://www.cdc.gov/parasites/lice/head/treatment.html
  37. Devore CD, Schutze GE; Council on School Health and Committee on Infectious Diseases, American Academy of Pediatrics. Head lice. Pediatrics. 2015;135:e1355-e1365. doi:10.1542/peds.2015-0746
  38. Ohl ME, Spach DH. Bartonella quintana and urban trench fever. Clin Infect Dis. 2000;31:131-135. doi:10.1086/313890
  39. Drali R, Sangaré AK, Boutellis A, et al. Bartonella quintana in body lice from scalp hair of homeless persons, France. Emerg Infect Dis. 2014;20:907-908. doi:10.3201/eid2005.131242
  40. Rudd N, Zakaria A, Kohn MA, et al. Association of body lice infestation with hemoglobin values in hospitalized dermatology patients. JAMA Dermatol. 2022;158:691-693. doi:10.1001/jamadermatol.2022.0818
  41. Guss DA, Koenig M, Castillo EM. Severe iron deficiency anemia and lice infestation. J Emergency Med. 2011;41:362-365. doi:10.1016/j.jemermed.2010.05.030
  42. Neglected tropical diseases of the skin: WHO launches mobile application to facilitate diagnosis. News release. World Health Organization; July 16, 2020. Accessed April 4, 2024. https://www.who.int/news/item/16-07-2020-neglected-tropical-diseases-of-the-skin-who-launches-mobile-application-to-facilitate-diagnosis
  43. Padovese V, Fuller LC, Griffiths CEM, et al; Migrant Health Dermatology Working Group of the International Foundation for Dermatology. Migrant skin health: perspectives from the Migrant Health Summit, Malta, 2022. Br J Dermatology. 2023;188:553-554. doi:10.1093/bjd/ljad001
  44. Knapp AP, Rehmus W, Chang AY. Skin diseases in displaced populations: a review of contributing factors, challenges, and approaches to care. Int J Dermatol. 2020;59:1299-1311. doi:10.1111/ijd.15063
  45. Norman FF, Comeche B, Chamorro S, et al. Overcoming challenges in the diagnosis and treatment of parasitic infectious diseases in migrants. Expert Rev Anti-infective Therapy. 2020;18:127-143. doi:10.1080/14787210.2020.1713099
  46. Skin NTDs: prioritizing integrated approaches to reduce suffering, psychosocial impact and stigmatization. News release. World Health Organization; October 29, 2020. Accessed April 4, 2024. https://www.who.int/news/item/29-10-2020-skin-ntds-prioritizing-integrated-approaches-to-reduce-suffering-psychosocial-impact-and-stigmatization
Issue
Cutis - 113(4)
Issue
Cutis - 113(4)
Page Number
E16-E21
Page Number
E16-E21
Publications
Publications
Topics
Article Type
Display Headline
Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis
Display Headline
Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis
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>Strahan</fileName> <TBEID>0C02F581.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F581</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Clinical Review</storyname> <articleType>1</articleType> <TBLocation>Published-CT</TBLocation> <QCDate/> <firstPublished>20240425T131253</firstPublished> <LastPublished>20240425T132642</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240425T131253</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Strahan</byline> <bylineText>Alexis G. Strahan, MD, MSN; Dirk M. Elston, MD</bylineText> <bylineFull>Strahan</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>E16-E21</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Approximately 108 million individuals have been forcibly displaced across the globe as of 2022, 35 million of whom are formally designated as refugees.1,2 The U</metaDescription> <articlePDF>301172</articlePDF> <teaserImage/> <title>Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>2</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>April</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>4</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2293</CMSID> <CMSID>2161</CMSID> </CMSIDs> <keywords> <keyword>infectious disease</keyword> <keyword> scabies</keyword> <keyword> pediculosis</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>April 2024</pubIssueName> <pubArticleType>Original Articles | 2161</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Original Article | 2293<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">49</term> </sections> <topics> <term canonical="true">234</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/1800270f.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis</title> <deck/> </itemMeta> <itemContent> <p class="abstract">There is a large burden of treatable dermatologic conditions in refugee populations. Parasitic infestations are particularly common when there are barriers to basic hygiene, crowded living or travel conditions, and lack of access to health care. Body lice are associated with anemia and can transmit a variety of diseases; chronic impetigo secondary to scabies is a leading cause of chronic kidney disease globally. Dermatologists have unique skills to identify skin infections, inflammatory diseases, and infestations. Appropriate dermatologic care has the potential to improve overall outcomes.</p> <p>Approximately 108 million individuals have been forcibly displaced across the globe as of 2022, 35 million of whom are formally designated as refugees.<sup>1,2</sup> The United States has coordinated resettlement of more refugee populations than any other country; the most common countries of origin are the Democratic Republic of the Congo, Syria, Afghanistan, and Myanmar.<sup>3</sup> In 2021, policy to increase the number of refugees resettled in the United States by more than 700% (from 15,000 up to 125,000) was established; since enactment, the United States has seen more than double the refugee arrivals in 2023 than the prior year, making medical care for this population increasingly relevant for the dermatologist.<sup>4</sup> </p> <p>Understanding how to care for this population begins with an accurate understanding of the term <i>refugee</i>. The United Nations defines a refugee as a person who is unwilling or unable to return to their country of nationality because of persecution or well-founded fear of persecution due to race, religion, nationality, membership in a particular social group, or political opinion. This term grants a protected status under international law and encompasses access to travel assistance, housing, cultural orientation, and medical evaluation upon resettlement.<sup>5,6</sup> <br/><br/>The burden of treatable dermatologic conditions in refugee populations ranges from 19% to 96% in the literature<sup>7,8</sup> and varies from inflammatory disorders to infectious and parasitic diseases.<sup>9</sup> In one study of 6899 displaced individuals in Greece, the prevalence of dermatologic conditions was higher than traumatic injury, cardiac disease, psychological conditions, and dental disease.<sup>10</sup> <br/><br/>When outlining differential diagnoses for parasitic infestations of the skin that affect refugee populations, helpful considerations include the individual’s country of origin, route traveled, and method of travel.<sup>11</sup> Parasitic infestations specifically are more common in refugee populations when there are barriers to basic hygiene, crowded living or travel conditions, or lack of access to health care, which they may experience at any point in their home country, during travel, or in resettlement housing.<sup>8</sup> <br/><br/>Even with limited examination and diagnostic resources, the skin is the most accessible first indication of patients’ overall well-being and often provides simple diagnostic clues—in combination with contextualization of the patient’s unique circumstances—necessary for successful diagnosis and treatment of scabies and pediculosis.<sup>12</sup> The dermatologist working with refugee populations may be the first set of eyes available and trained to discern skin infestations and therefore has the potential to improve overall outcomes. <br/><br/>Some parasitic infestations in refugee populations may fall under the category of neglected tropical diseases, including scabies, ascariasis, trypanosomiasis, leishmaniasis, and schistosomiasis; they affect an estimated 1 billion individuals across the globe but historically have been underrepresented in the literature and in health policy due in part to limited access to care.<sup>13</sup> This review will focus on infestations by the scabies mite (<i>Sarcoptes scabiei </i>var <i>hominis</i>)<i> </i>and the human louse, as these frequently are encountered, easily diagnosed, and treatable by trained clinicians, even in resource-limited settings. </p> <h3>Scabies </h3> <p>Scabies is a parasitic skin infestation caused by the 8-legged mite <i>Sarcoptes scabiei </i>var<i> hominis.</i> The female mite begins the infestation process via penetration of the epidermis, particularly the stratum corneum, and commences laying eggs (Figure 1). The subsequent larvae emerge 48 to 72 hours later and remain burrowed in the epidermis. The larvae mature over the next 10 to 14 days and continue the reproductive cycle.<sup>14,15</sup> Symptoms of infestation occurs due to a hypersensitivity reaction to the mite and its by-products.<sup>16</sup> Transmission of the mite primarily occurs via direct (skin-to-skin) contact with infected individuals or environmental surfaces for 24 to36 hours in specific conditions, though the latter source has been debated in the literature. </p> <p>The method of transmission is particularly important when considering care for refugee populations. Scabies is found most often in those living in or traveling from tropical regions including East Asia, Southeast Asia, Oceania, and Latin America.<sup>17</sup> In displaced or refugee populations, a lack of access to basic hygiene, extended travel in close quarters, and suboptimal health care access all may lead to an increased incidence of untreated scabies infestations.<sup>18</sup> Scabies is more prevalent in children, with increased potential for secondary bacterial infections with <i>Streptococcus</i> and <i>Staphylococcus</i> species due to excoriation in unsanitary conditions. Secondary infection with <i>Streptococcus pyogenes</i> can lead to acute poststreptococcal glomerulonephritis, which accounts for a large burden of chronic kidney disease in affected populations.<sup>19</sup> However, scabies may be found in any population, regardless of hygiene or health care access. Treating health care providers should keep a broad differential. <br/><br/><i>Presentation—</i>The latency of scabies symptoms is 2 to 6 weeks in a primary outbreak and may be as short as 1 to 3 days with re-infestation, following the course of delayed-type hypersensitivity.<sup>20</sup> The initial hallmark symptom is pruritus with increased severity in the evening. Visible lesions, excoriations, and burrows associated with scattered vesicles or pustules may be seen over the web spaces of the hands and feet, volar surfaces of the wrists, axillae, waist, genitalia, inner thighs, or buttocks.<sup>19</sup> Chronic infestation often manifests with genital nodules. In populations with limited access to health care, there are reports of a sensitization phenomenon in which the individual may become less symptomatic after 4 to 6 weeks and yet be a potential carrier of the mite.<sup>21</sup><i> <br/><br/></i>Those with compromised immune function, such as individuals living with HIV or severe malnutrition, may present with crusted scabies, a variant that manifests as widespread hyperkeratotic scaling with more pronounced involvement of the head, neck, and acral areas. In contrast to classic scabies, crusted scabies is associated with minimal pruritus.<sup>22</sup> <br/><br/><i>Diagnosis—</i>The diagnosis of scabies is largely clinical with confirmation through skin scrapings. The International Alliance for Control of Scabies has established diagnostic criteria that include a combination of clinical findings, history, and visualization of mites.<sup>23</sup> A dermatologist working with refugee populations may employ any combination of history (eg, nocturnal itch, exposure to an affected individual) or clinical findings along with a high degree of suspicion in those with elevated risk. Visualization of mites is helpful to confirm the diagnosis and may be completed with the application of mineral oil at the terminal end of a burrow, skin scraping with a surgical blade or needle, and examination under light microscopy. <br/><br/><i>Treatment—</i>First-line treatment for scabies consists of application of permethrin cream 5% on the skin of the neck to the soles of the feet, which is to be left on for 8 to 14 hours followed by rinsing. Re-application is recommended in 1 to 2 weeks. Oral ivermectin is a reasonable alternative to permethrin cream due to its low cost and easy administration in large affected groups. It is not labeled for use in pregnant women or children weighing less than 15 kg but has no selective fetal toxicity. Treatment of scabies with ivermectin has the benefit of treating many other parasitic infections. Both medications are on the <i>World Health Organization Model List of Essential Medications</i> and are widely available for treating providers, even in resource-limited settings.<sup>24</sup> <br/><br/>Much of the world still uses benzyl benzoate or precipitated sulfur ointment to treat scabies, and some botanicals used in folk medicine have genuine antiscabetic properties. Pruritus may persist for 1 to 4 weeks following treatment and does not indicate treatment failure. Topical camphor and menthol preparations, low-potency topical corticosteroids, or emollients all may be employed for relief.<sup>25</sup> <em>Sarna</em> is a Spanish term for scabies and has become the proprietary name for topical antipruritic agents. Additional methods of treatment and prevention include washing clothes and linens in hot water and drying on high heat. If machine washing is not available, clothing and linens may be sealed in a plastic bag for 72 hours. </p> <h3>Pediculosis </h3> <p>Pediculosis is an infestation caused by the ectoparasite <i>Pediculus humanus</i>, an obligate, sesame seed–sized louse that feeds exclusively on the blood of its host (Figure 2).<sup>26</sup> Of the lice species, 2 require humans as hosts; one is <i>P humanus</i> and the other is <i>Pthirus pubis</i> (pubic lice). <i>Pediculus humanus </i>may be further classified into morphologies based largely on the affected area: body (<i>P humanus</i> <i>corporis)</i> or head (<i>P humanus</i> <i>capitis)</i>, both of which will be discussed.<sup>27</sup> </p> <p>Lice primarily attach to clothing and hair shafts, then transfer to the skin for blood feeds. Females lay eggs that hatch 6 to 10 days later, subsequently maturing into adults. The lifespan of these parasites with regular access to a host is 1 to 3 months for head lice and 18 days for body lice vs only 3 to 5 days without a host.<sup>28</sup> Transmission of <i>P humanus capitis </i>primarily occurs via direct contact with affected individuals, either head-to-head contact or sharing of items such as brushes and headscarves; <i>P humanus corporis</i> also may be transmitted via direct contact with affected individuals or clothing. <br/><br/>Pediculosis is an important infestation to consider when providing care for refugee populations. Risk factors include lack of access to basic hygiene, including regular bathing or laundering of clothing, and crowded conditions that make direct person-to-person contact with affected individuals more likely.<sup>29</sup> Body lice are associated more often with domestic turbulence and displaced populations<sup>30</sup> in comparison to head lice, which have broad demographic variables, most often affecting females and children.<sup>28</sup> Fatty acids in adult male sebum make the scalp less hospitable to lice. <br/><br/><i>Presentation</i>—The most common clinical manifestation of pediculosis is pruritus. Cutaneous findings can include papules, wheals, or hemorrhagic puncta secondary to the louse bite. Due to the Tyndall effect of deep hemosiderin pigment, blue-grey macules termed <i>maculae ceruleae</i> (Figure 3) also may be present in chronic infestations of pediculosis pubis, in contrast to pediculosis capitis or corporis.<sup>31</sup> Body louse infestation is associated with a general pruritus concentrated on the neck, shoulders, and waist—areas where clothing makes the most direct contact. Lesions may be visible and include eczematous patches with excoriation and possible secondary bacterial infection. Chronic infestation may exhibit lichenification or hyperpigmentation in associated areas. Head lice most often manifest with localized scalp pruritus and associated excoriation and cervical or occipital lymphadenopathy.<sup>32</sup> <br/><br/><i>Diagnosis—</i>The diagnosis of pediculosis is clinical, with confirmation requiring direct examination of the insect or nits (the egg case of the parasite)(Figure 4). Body lice and associated nits can be visualized on clothing seams near areas of highest body temperature, particularly the waistband. Head lice may be visualized crawling on hair shafts or on a louse comb. Nits are firmly attached to hair shafts and are visible to the naked eye, whereas pseudonits slide freely along the hair shaft and are not a manifestation of louse infestation (Figure 5).<sup>31</sup> <br/><br/><i>Treatment—</i>Treatment varies by affected area. Pediculosis corporis may be treated with permethrin cream 5% applied to the entire body and left on for 8 to 10 hours, but this may not be necessary if facilities are available to wash and dry clothing.<sup>33</sup> The use of oral ivermectin and permethrin-impregnated underwear both have been proposed.<sup>34,35</sup> Treatment of pediculosis capitis may be accomplished with a variety of topical pediculicides including permethrin, pyrethrum with piperonyl butoxide, dimethicone, malathion, benzyl alcohol, spinosad, and topical ivermectin.<sup>22</sup> Topical corticosteroids or emollients may be employed for residual pruritus. <br/><br/>Equally important is environmental elimination of infestation. Clothing should be discarded if possible or washed and dried using high heat. If neither approach is possible or appropriate, clothing may be sealed in a plastic bag for 2 weeks or treated with a pediculicide. Nit combing is an important adjunct in the treatment of pediculosis capitis.<sup>36</sup> It is important to encourage return to work and/or school immediately after treatment. “No nit” policies are more harmful to education than helpful for prevention of investation.<sup>37<br/><br/></sup>Pediculosis corporis may transmit infectious agents including <i>Bartonella quintana</i>, (trench fever, endocarditis, bacillary angiomatosis), <i>Borrelia recurrentis</i> (louse-borne relapsing fever), and <i>Rickettsia prowazekii</i> (epidemic typhus).<sup>31,38,39</sup> Additionally, severe pediculosis infestations have the potential to cause chronic blood loss in affected populations. In a study of patients with active pediculosis infestation, mean hemoglobin values were found to be 2.5 g/dL lower than a matched population without infestation.<sup>40</sup> It is important to consider pediculosis as a risk for iron-deficiency anemia in populations who are known to lack access to regular medical evaluation.<sup>41</sup> </p> <h3>Future Considerations </h3> <p>Increased access to tools and education for clinicians treating refugee populations is key to reducing the burden of parasitic skin disease and related morbidity and mortality in vulnerable groups both domestically and globally. One such tool, the Skin NTDs App, was launched by the World Health Organization in 2020. It is available for free for Android and iOS devices to assist clinicians in the field with the diagnosis and treatment of neglected tropical diseases—including scabies—that may affect refugee populations.<sup>42</sup></p> <p>Additionally, to both improve access and limit preventable sequelae, future investigations into appropriate models of community-based care are paramount. The model of community-based care is centered on the idea of care provision that prioritizes safety, accessibility, affordability, and acceptability in an environment closest to vulnerable populations. The largest dermatologic society, the International League of Dermatological Societies, formed a Migrant Health Dermatology Working Group that prioritizes understanding and improving care for refugee and migrant populations; this group hosted a summit in 2022, bringing together international subject matter leaders to discuss such models of care and set goals for the creation of tool kits for patients, frontline health care workers, and dermatologists.<sup>43</sup></p> <h3>Conclusion</h3> <p>Improvement in dermatologic care of refugee populations includes provision of culturally and linguistically appropriate care by trained clinicians, adequate access to the most essential medications, and basic physical or legal access to health care systems in general.<sup>8,11,44</sup> Parasitic infestations have the potential to remain asymptomatic for extended periods of time and result in spread to potentially nonendemic regions of resettlement.<sup>45</sup> Additionally, the psychosocial well-being of refugee populations upon resettlement may be negatively affected by stigma of disease processes such as scabies and pediculosis, leading to additional barriers to successful re-entry into the patient’s new environment.<sup>46</sup> Therefore, proper screening, diagnosis, and treatment of the most common parasitic infestations in this population have great potential to improve outcomes for large groups across the globe.</p> <h2>References</h2> <p class="reference"> 1. Monin K, Batalova J, Lai T. Refugees and Asylees in the United States. Migration Information Source. Published May 13, 2021. Accessed April 4, 2024. https://www.migrationpolicy.org/article/refugees-and-asylees-united-states-2021<br/><br/> 2. UNHCR. Figures at a Glance. UNHCR USA. Update June 14, 2023. Accessed April 4, 2024. https://www.unhcr.org/en-us/figures-at-a-glance.html<br/><br/> 3. UNHCR. Refugee resettlement facts. Published October 2023. Accessed April 8, 2024. https://www.unhcr.org/us/media/refugee-resettlement-facts<br/><br/> 4. US Department of State. Report to Congress on Proposed Refugee Admissions for Fiscal Year 2024. Published November 3, 2023. Accessed April 8, 2024. https://www.state.gov/report-to-congress-on-proposed-refugee-admissions-for-fiscal-year-2024/ <br/><br/> 5. UNHCR. Compact for Migration: Definitions. United Nations. Accessed April 4, 2024. https://refugeesmigrants.un.org/definitions<br/><br/> 6. United Nations High Commissioner for Refugees (UNHCR). Convention and Protocol Relating to the Status of Refugees. Published December 2010. Accessed January 11, 2024. https://www.unhcr.org/us/media/convention-and-protocol-relating-status-refugees<br/><br/> 7. Kibar Öztürk M. Skin diseases in rural Nyala, Sudan (in a rural hospital, in 12 orphanages, and in two refugee camps). <i>Int J Dermatol</i>. 2019;58:1341-1349. doi:10.1111/ijd.14619<br/><br/> 8. Padovese V, Knapp A. Challenges of managing skin diseases in refugees and migrants. <i>Dermatol Clin</i>. 2021;39:101-115. doi:10.1016/j.det.2020.08.010<br/><br/> 9. Saikal SL, Ge L, Mir A, et al. Skin disease profile of Syrian refugees in Jordan: a field-mission assessment. <i>J Eur Acad Dermatol Venereol.</i> 2020;34:419-425. doi:10.1111/jdv.15909<br/><br/>10. Eonomopoulou A, Pavli A, Stasinopoulou P, et al. Migrant screening: lessons learned from the migrant holding level at the Greek-Turkish borders. <i>J Infect Public Health</i>. 2017;10:177-184. doi:10.1016/j.jiph.2016.04.012<br/><br/>11. Marano N, Angelo KM, Merrill RD, et al. Expanding travel medicine in the 21st century to address the health needs of the world’s migrants.<i>J Travel Med</i>. 2018;25. doi:10.1093/jtm/tay067<br/><br/>12. Hay RJ, Asiedu K. Skin-related neglected tropical diseases (skin NTDs)—a new challenge. <i>Trop Med Infect Dis</i>. 2018;4. doi:10.3390/tropicalmed4010004<br/><br/>13. NIAID. Neglected tropical diseases. Updated July 11, 2016. Accessed April 4, 2024. https://www.niaid.nih.gov/research/neglected-tropical-diseases<br/><br/>14. Arlian LG, Morgan MS. A review of Sarcoptes scabiei: past, present and future. <i>Parasit Vectors</i>. 2017;10:297. doi:10.1186/s13071-017-2234-1<br/><br/>15. Arlian LG, Runyan RA, Achar S, et al. Survival and infectivity of Sarcoptes scabiei var. canis and var. hominis. <i>J Am Acad Dermatol</i>. 1984;11(2 pt 1):210-215. doi:10.1016/s0190-9622(84)70151-4<br/><br/>16. Chandler DJ, Fuller LC. A review of scabies: an infestation more than skin deep. <i>Dermatology</i>. 2019;235:79-90. doi:10.1159/000495290<br/><br/>17. Karimkhani C, Colombara DV, Drucker AM, et al. The global burden of scabies: a cross-sectional analysis from the Global Burden of Disease Study 2015. <i>Lancet Infect Dis</i>. 2017;17:1247-1254. doi:10.1016/S1473-3099(17)30483-8<br/><br/>18. Romani L, Steer AC, Whitfeld MJ, et al. Prevalence of scabies and impetigo worldwide: a systematic review. <i>Lancet Infect Dis</i>. 2015;15:960-967. doi:10.1016/S1473-3099(15)00132-2<br/><br/>19. Thomas C, Coates SJ, Engelman D, et al. Ectoparasites: scabies. <i>J Am Acad Dermatol</i>. 2020;82:533-548. doi:10.1016/j.jaad.2019.05.109<br/><br/>20. Mellanby K, Johnson CG, Bartley WC. Treatment of scabies. <i>Br Med J</i>. 1942;2:1-4. doi:10.1136/bmj.2.4252.1<br/><br/>21. Walton SF. The immunology of susceptibility and resistance to scabies. <i>Parasit Immunol</i>. 2010;32:532-540. doi:10.1111/j.1365-3024.2010.01218.x<br/><br/>22. Coates SJ, Thomas C, Chosidow O, et al. Ectoparasites: pediculosis and tungiasis. <i>J Am Acad Dermatol</i>. 2020;82:551-569. doi:10.1016/j.jaad.2019.05.110</p> <p class="reference">23. Engelman D, Fuller LC, Steer AC; International Alliance for the Control of Scabies Delphi p. Consensus criteria for the diagnosis of scabies: a Delphi study of international experts. <i>PLoS Negl Trop Dis</i>. 2018;12:E0006549. doi:10.1371/journal.pntd.0006549<br/><br/>24. World Health Organization. WHO Model Lists of Essential Medicines—23rd list, 2023. Updated July 26, 2023. Accessed April 8, 2024. https://www.who.int/publications/i/item/WHO-MHP-HPS-EML-2023.02 <br/><br/>25. Salavastru CM, Chosidow O, Boffa MJ, et al. European guideline for the management of scabies. <i>J Eur Acad Dermatol Venereol</i>. 2017;31:1248-1253. doi:10.1111/jdv.14351<br/><br/>26. Badiaga S, Brouqui P. Human louse-transmitted infectious diseases. <i>Clin Microbiol Infect</i>. 2012;18:332-337. doi:10.1111/j.1469-0691.2012.03778.x<br/><br/>27. Leo NP, Campbell NJH, Yang X, et al. Evidence from mitochondrial DNA that head lice and body lice of humans (Phthiraptera: Pediculidae) are conspecific. <i>J Med Entomol. </i>2002;39:662-666. doi:10.1603/0022-2585-39.4.662<br/><br/>28. Chosidow O. Scabies and pediculosis. <i>Lancet</i>. 2000;355:819-826. doi:10.1016/S0140-6736(99)09458-1<br/><br/>29. Arnaud A, Chosidow O, Détrez M-A, et al. Prevalences of scabies and pediculosis corporis among homeless people in the Paris region: results from two randomized cross-sectional surveys (HYTPEAC study). <i>Br J Dermatol</i>. 2016;174:104-112. doi:10.1111/bjd.14226<br/><br/>30. Brouqui P. Arthropod-borne diseases associated with political and social disorder. <i>Annu Rev Entomol</i>. 2011;56:357-374. doi:10.1146/annurev-ento-120709-144739<br/><br/>31. Ko CJ, Elston DM. Pediculosis. <i>J Am Acad Dermatol</i>. 2004;50:1-12. doi:10.1016/S0190-9622(03)02729-4<br/><br/>32. Bloomfield D. Head lice. <i>Pediatr Rev</i>. 2002;23:34-35; discussion 34-35. doi:10.1542/pir.23-1-34<br/><br/>33. Stone SP GJ, Bacelieri RE. Scabies, other mites, and pediculosis. In: Wolf K GL, Katz SI, et al (eds). <i>Fitzpatrick’s Dermatology in General Medicine</i>. McGraw Hill; 2008:2029.<br/><br/>34. Foucault C, Ranque S, Badiaga S, et al. Oral ivermectin in the treatment of body lice. <i>J Infect Dis</i>. 2006;193:474-476. doi:10.1086/499279<br/><br/>35. Benkouiten S, Drali R, Badiaga S, et al. Effect of permethrin-impregnated underwear on body lice in sheltered homeless persons: a randomized controlled trial. <i>JAMA Dermatol. </i>2014;150:273-279. doi:10.1001/jamadermatol.2013.6398<br/><br/>36. CDC. Parasites: Treatment. Updated October 15, 2019. Accessed April 4, 2024. https://www.cdc.gov/parasites/lice/head/treatment.html<br/><br/>37. Devore CD, Schutze GE; Council on School Health and Committee on Infectious Diseases, American Academy of Pediatrics. Head lice. <i>Pediatrics</i>. 2015;135:e1355-e1365. doi:10.1542/peds.2015-0746<br/><br/>38. Ohl ME, Spach DH. Bartonella quintana and urban trench fever. <i>Clin Infect Dis</i>. 2000;31:131-135. doi:10.1086/313890<br/><br/>39. Drali R, Sangaré AK, Boutellis A, et al. Bartonella quintana in body lice from scalp hair of homeless persons, France. <i>Emerg Infect Dis</i>. 2014;20:907-908. doi:10.3201/eid2005.131242<br/><br/>40. Rudd N, Zakaria A, Kohn MA, et al. Association of body lice infestation with hemoglobin values in hospitalized dermatology patients. <i>JAMA Dermatol</i>. 2022;158:691-693. doi:10.1001/jamadermatol.2022.0818</p> <p class="reference">41. Guss DA, Koenig M, Castillo EM. Severe iron deficiency anemia and lice infestation. <i>J Emergency Med</i>. 2011;41:362-365. doi:10.1016/j.jemermed.2010.05.030<br/><br/>42. Neglected tropical diseases of the skin: WHO launches mobile application to facilitate diagnosis. News release. World Health Organization; July 16, 2020. Accessed April 4, 2024. https://www.who.int/news/item/16-07-2020-neglected-tropical-diseases-of-the-skin-who-launches-mobile-application-to-facilitate-diagnosis<br/><br/>43. Padovese V, Fuller LC, Griffiths CEM, et al; Migrant Health Dermatology Working Group of the International Foundation for Dermatology. Migrant skin health: perspectives from the Migrant Health Summit, Malta, 2022. <i>Br J Dermatology</i>. 2023;188:553-554. doi:10.1093/bjd/ljad001 <br/><br/>44. Knapp AP, Rehmus W, Chang AY. Skin diseases in displaced populations: a review of contributing factors, challenges, and approaches to care. <i>Int J Dermato</i>l. 2020;59:1299-1311. doi:10.1111/ijd.15063<br/><br/>45. Norman FF, Comeche B, Chamorro S, et al. Overcoming challenges in the diagnosis and treatment of parasitic infectious diseases in migrants. <i>Expert Rev Anti-infective Therapy. </i>2020;18:127-143. doi:10.1080/14787210.2020.1713099<br/><br/>46. Skin NTDs: prioritizing integrated approaches to reduce suffering, psychosocial impact and stigmatization. News release. World Health Organization; October 29, 2020. Accessed April 4, 2024. https://www.who.int/news/item/29-10-2020-skin-ntds-prioritizing-integrated-approaches-to-reduce-suffering-psychosocial-impact-and-stigmatization</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Alexis G. Strahan is from the Mercer University School of Medicine, Savannah, Georgia. 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/>All images are in the public domain. <br/><br/>Correspondence: Alexis G. Strahan, MD, MSN, 55 Fruit St, Bartlett Hall 6R, Boston, MA 02114 (<a href="mailto:alexis.grabow.strahan@live.mercer.edu">alexis.grabow.strahan@live.mercer.edu</a>). <br/><br/><em><hl name="17868"/>Cutis. </em>2024 April;113(4):E16-E21. doi:10.12788/cutis.0999 </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>War and natural disasters displace populations and disrupt infrastructure and access to medical care.</li> <li>Infestations and cutaneous infections are common among refugee populations, and impetigo often is a sign of underlying scabies infestation.</li> <li>Body lice are important disease vectors inrefugee populations. </li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • War and natural disasters displace populations and disrupt infrastructure and access to medical care.
  • Infestations and cutaneous infections are common among refugee populations, and impetigo often is a sign of underlying scabies infestation.
  • Body lice are important disease vectors inrefugee populations.
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

Asymptomatic Erythematous Plaque in an Outdoorsman

Article Type
Changed
Wed, 04/24/2024 - 10:16
Display Headline
Asymptomatic Erythematous Plaque in an Outdoorsman

The Diagnosis: Erythema Migrans

The patient was clinically diagnosed with erythema migrans. He did not recall a tick bite but spent a lot of time outdoors. He was treated with 10 days of doxycycline 100 mg twice daily with complete resolution of the rash.

Lyme disease is a spirochete infection caused by the Borrelia burgdorferi sensu lato species complex and transmitted by the Ixodidae tick family. It is the most common tick-borne disease in the United States and mostly is reported in the northeastern and upper midwestern states during the warmer seasons, but it is prevalent worldwide. In geographic areas where Lyme disease is common, the incidence is approximately 40 cases per 100,000 individuals.1 Our patient resided in coastal South Carolina. Lyme disease is more commonly reported in White individuals. The skin lesions may be more difficult to discern and diagnose in patients with darker skin types, leading to delayed diagnosis and treatment.2,3

Patients may be diagnosed with early localized, early disseminated, or late Lyme disease. Erythema migrans is the early localized form of the disease and is classically described as an erythematous targetlike plaque with raised borders arising at the site of the tick bite 1 to 2 weeks later.4 However, many patients simply have a homogeneous erythematous plaque with raised advancing borders ranging in size from 5 to 68 cm.5 In a 2022 study of 69 patients with suspected Lyme disease, only 35 (50.7%) were determined to truly have acute Lyme disease.6 Of them, only 2 (5.7%) had the classic ringwithin- a-ring pattern. Most plaques were uniform, pink, oval-shaped lesions with well-demarcated borders.6

The rash may present with a burning sensation, or patients may experience no symptoms at all, which can lead to delayed diagnosis and progression to late disease. Patients may develop malaise, fever, headache, body aches, or joint pain. Early disseminated disease manifests similarly. Patients with disseminated disease also may develop more serious complications, including lymphadenopathy; cranial nerve palsies; ocular involvement; meningitis; or cardiac abnormalities such as myocarditis, pericarditis, or arrhythmia. Late disease most often causes arthritis of the large joints, though it also can have cardiac or neurologic manifestations. Some patients with chronic disease—the majority of whom were diagnosed in Europe—may develop acrodermatitis chronica atrophicans with edematous blue-red plaques that become atrophic and hyperpigmented fibrotic plaques over the course of years.

Allergic contact dermatitis to a plant more likely would cause itchy or painful, oozy, weepy, vesicular lesions arranged in a linear pattern. A dermatophyte infection likely would cause a scaly eruption. Although our patient presented with a sharply demarcated, raised, erythematous lesion, the distribution did not follow normal clothing lines and would be unusual for a photosensitive drug eruption. Cellulitis likely would be associated with tenderness or warmth to the touch. Finally, southern tick-associated rash illness, which is associated with Amblyomma americanum (lone star tick) bites, may appear with a similar rash but few systemic symptoms. It also can be treated with tetracycline antibiotics.7

Our case in South Carolina demonstrates the importance of keeping Lyme disease in the differential. Clinicians should remember to ask patients about their travel history. In endemic areas, patients with erythema migrans can be started on treatment without waiting for serology. Patients with early Lyme disease may or may not have positive serologies at the time of presentation.6 Guidelines for the treatment of Lyme disease have been revised in recent years to decrease patient antibiotic exposure by reducing the number of days of antibiotic therapy.8 A recent randomized controlled trial found no significant difference in recurrence for patients treated with 7 days of doxycycline compared with 14 days.9 We typically prescribe a 10-day course of doxycycline, which also is adequate for concurrent rickettsial disease. Patients who develop malarialike symptoms should be evaluated for babesiosis, which is treated with clindamycin.

References
  1. Skar GL, Simonsen KA. Lyme disease. StatPearls [Internet]. Updated February 4, 2024. Accessed March 20, 2024. https://www.ncbi.nlm.nih.gov/books/NBK431066/
  2. Dennison R, Novak C, Rebman A, et al. Lyme disease with erythema migrans and seventh nerve palsy in an African-American man. Cureus. 2019;11:E6509.
  3. Bax CE, Clark AK, Oboite M, et al. A case of disseminated Lyme disease in a child with skin of color. Pediatr Dermatol. 2021;38 (suppl 2):140-141.
  4. Shah AS, Varatharaj Palraj BR. Multiple erythema migrans rashes characteristic of early disseminated lyme disease, before and after therapy. Mayo Clin Proc. 2019;94:172-173.
  5. Feder HM Jr, Abeles M, Bernstein M, et al. Diagnosis, treatment, and prognosis of erythema migrans and Lyme arthritis. Clin Dermatol. 2006;24:509-520.
  6. Schotthoefer AM, Green CB, Dempsey G, et al. The spectrum of erythema migrans in early Lyme disease: can we improve its recognition? Cureus. 2022;14:E30673.
  7. Strle F, Wormser GP. Early Lyme disease (erythema migrans) and its mimics (southern tick-associated rash illness and tick-associated rash illness). Infect Dis Clin North Am. 2022;36:523-539.
  8. Torbahn G, Hofmann H, Rücker G, et al. Efficacy and safety of antibiotic therapy in early cutaneous Lyme borreliosis: a network meta-analysis. JAMA Dermatol. 2018;154:1292-1303.
  9. Stupica D, Collinet-Adler S, Blagus R, et al. Treatment of erythema migrans with doxycycline for 7 days versus 14 days in Slovenia: a randomised open-label non-inferiority trial. Lancet Infect Dis. 2023;23:371-379.
Article PDF
Author and Disclosure Information

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.

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
183-184
Sections
Author and Disclosure Information

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.

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

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.

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
Related Articles

The Diagnosis: Erythema Migrans

The patient was clinically diagnosed with erythema migrans. He did not recall a tick bite but spent a lot of time outdoors. He was treated with 10 days of doxycycline 100 mg twice daily with complete resolution of the rash.

Lyme disease is a spirochete infection caused by the Borrelia burgdorferi sensu lato species complex and transmitted by the Ixodidae tick family. It is the most common tick-borne disease in the United States and mostly is reported in the northeastern and upper midwestern states during the warmer seasons, but it is prevalent worldwide. In geographic areas where Lyme disease is common, the incidence is approximately 40 cases per 100,000 individuals.1 Our patient resided in coastal South Carolina. Lyme disease is more commonly reported in White individuals. The skin lesions may be more difficult to discern and diagnose in patients with darker skin types, leading to delayed diagnosis and treatment.2,3

Patients may be diagnosed with early localized, early disseminated, or late Lyme disease. Erythema migrans is the early localized form of the disease and is classically described as an erythematous targetlike plaque with raised borders arising at the site of the tick bite 1 to 2 weeks later.4 However, many patients simply have a homogeneous erythematous plaque with raised advancing borders ranging in size from 5 to 68 cm.5 In a 2022 study of 69 patients with suspected Lyme disease, only 35 (50.7%) were determined to truly have acute Lyme disease.6 Of them, only 2 (5.7%) had the classic ringwithin- a-ring pattern. Most plaques were uniform, pink, oval-shaped lesions with well-demarcated borders.6

The rash may present with a burning sensation, or patients may experience no symptoms at all, which can lead to delayed diagnosis and progression to late disease. Patients may develop malaise, fever, headache, body aches, or joint pain. Early disseminated disease manifests similarly. Patients with disseminated disease also may develop more serious complications, including lymphadenopathy; cranial nerve palsies; ocular involvement; meningitis; or cardiac abnormalities such as myocarditis, pericarditis, or arrhythmia. Late disease most often causes arthritis of the large joints, though it also can have cardiac or neurologic manifestations. Some patients with chronic disease—the majority of whom were diagnosed in Europe—may develop acrodermatitis chronica atrophicans with edematous blue-red plaques that become atrophic and hyperpigmented fibrotic plaques over the course of years.

Allergic contact dermatitis to a plant more likely would cause itchy or painful, oozy, weepy, vesicular lesions arranged in a linear pattern. A dermatophyte infection likely would cause a scaly eruption. Although our patient presented with a sharply demarcated, raised, erythematous lesion, the distribution did not follow normal clothing lines and would be unusual for a photosensitive drug eruption. Cellulitis likely would be associated with tenderness or warmth to the touch. Finally, southern tick-associated rash illness, which is associated with Amblyomma americanum (lone star tick) bites, may appear with a similar rash but few systemic symptoms. It also can be treated with tetracycline antibiotics.7

Our case in South Carolina demonstrates the importance of keeping Lyme disease in the differential. Clinicians should remember to ask patients about their travel history. In endemic areas, patients with erythema migrans can be started on treatment without waiting for serology. Patients with early Lyme disease may or may not have positive serologies at the time of presentation.6 Guidelines for the treatment of Lyme disease have been revised in recent years to decrease patient antibiotic exposure by reducing the number of days of antibiotic therapy.8 A recent randomized controlled trial found no significant difference in recurrence for patients treated with 7 days of doxycycline compared with 14 days.9 We typically prescribe a 10-day course of doxycycline, which also is adequate for concurrent rickettsial disease. Patients who develop malarialike symptoms should be evaluated for babesiosis, which is treated with clindamycin.

The Diagnosis: Erythema Migrans

The patient was clinically diagnosed with erythema migrans. He did not recall a tick bite but spent a lot of time outdoors. He was treated with 10 days of doxycycline 100 mg twice daily with complete resolution of the rash.

Lyme disease is a spirochete infection caused by the Borrelia burgdorferi sensu lato species complex and transmitted by the Ixodidae tick family. It is the most common tick-borne disease in the United States and mostly is reported in the northeastern and upper midwestern states during the warmer seasons, but it is prevalent worldwide. In geographic areas where Lyme disease is common, the incidence is approximately 40 cases per 100,000 individuals.1 Our patient resided in coastal South Carolina. Lyme disease is more commonly reported in White individuals. The skin lesions may be more difficult to discern and diagnose in patients with darker skin types, leading to delayed diagnosis and treatment.2,3

Patients may be diagnosed with early localized, early disseminated, or late Lyme disease. Erythema migrans is the early localized form of the disease and is classically described as an erythematous targetlike plaque with raised borders arising at the site of the tick bite 1 to 2 weeks later.4 However, many patients simply have a homogeneous erythematous plaque with raised advancing borders ranging in size from 5 to 68 cm.5 In a 2022 study of 69 patients with suspected Lyme disease, only 35 (50.7%) were determined to truly have acute Lyme disease.6 Of them, only 2 (5.7%) had the classic ringwithin- a-ring pattern. Most plaques were uniform, pink, oval-shaped lesions with well-demarcated borders.6

The rash may present with a burning sensation, or patients may experience no symptoms at all, which can lead to delayed diagnosis and progression to late disease. Patients may develop malaise, fever, headache, body aches, or joint pain. Early disseminated disease manifests similarly. Patients with disseminated disease also may develop more serious complications, including lymphadenopathy; cranial nerve palsies; ocular involvement; meningitis; or cardiac abnormalities such as myocarditis, pericarditis, or arrhythmia. Late disease most often causes arthritis of the large joints, though it also can have cardiac or neurologic manifestations. Some patients with chronic disease—the majority of whom were diagnosed in Europe—may develop acrodermatitis chronica atrophicans with edematous blue-red plaques that become atrophic and hyperpigmented fibrotic plaques over the course of years.

Allergic contact dermatitis to a plant more likely would cause itchy or painful, oozy, weepy, vesicular lesions arranged in a linear pattern. A dermatophyte infection likely would cause a scaly eruption. Although our patient presented with a sharply demarcated, raised, erythematous lesion, the distribution did not follow normal clothing lines and would be unusual for a photosensitive drug eruption. Cellulitis likely would be associated with tenderness or warmth to the touch. Finally, southern tick-associated rash illness, which is associated with Amblyomma americanum (lone star tick) bites, may appear with a similar rash but few systemic symptoms. It also can be treated with tetracycline antibiotics.7

Our case in South Carolina demonstrates the importance of keeping Lyme disease in the differential. Clinicians should remember to ask patients about their travel history. In endemic areas, patients with erythema migrans can be started on treatment without waiting for serology. Patients with early Lyme disease may or may not have positive serologies at the time of presentation.6 Guidelines for the treatment of Lyme disease have been revised in recent years to decrease patient antibiotic exposure by reducing the number of days of antibiotic therapy.8 A recent randomized controlled trial found no significant difference in recurrence for patients treated with 7 days of doxycycline compared with 14 days.9 We typically prescribe a 10-day course of doxycycline, which also is adequate for concurrent rickettsial disease. Patients who develop malarialike symptoms should be evaluated for babesiosis, which is treated with clindamycin.

References
  1. Skar GL, Simonsen KA. Lyme disease. StatPearls [Internet]. Updated February 4, 2024. Accessed March 20, 2024. https://www.ncbi.nlm.nih.gov/books/NBK431066/
  2. Dennison R, Novak C, Rebman A, et al. Lyme disease with erythema migrans and seventh nerve palsy in an African-American man. Cureus. 2019;11:E6509.
  3. Bax CE, Clark AK, Oboite M, et al. A case of disseminated Lyme disease in a child with skin of color. Pediatr Dermatol. 2021;38 (suppl 2):140-141.
  4. Shah AS, Varatharaj Palraj BR. Multiple erythema migrans rashes characteristic of early disseminated lyme disease, before and after therapy. Mayo Clin Proc. 2019;94:172-173.
  5. Feder HM Jr, Abeles M, Bernstein M, et al. Diagnosis, treatment, and prognosis of erythema migrans and Lyme arthritis. Clin Dermatol. 2006;24:509-520.
  6. Schotthoefer AM, Green CB, Dempsey G, et al. The spectrum of erythema migrans in early Lyme disease: can we improve its recognition? Cureus. 2022;14:E30673.
  7. Strle F, Wormser GP. Early Lyme disease (erythema migrans) and its mimics (southern tick-associated rash illness and tick-associated rash illness). Infect Dis Clin North Am. 2022;36:523-539.
  8. Torbahn G, Hofmann H, Rücker G, et al. Efficacy and safety of antibiotic therapy in early cutaneous Lyme borreliosis: a network meta-analysis. JAMA Dermatol. 2018;154:1292-1303.
  9. Stupica D, Collinet-Adler S, Blagus R, et al. Treatment of erythema migrans with doxycycline for 7 days versus 14 days in Slovenia: a randomised open-label non-inferiority trial. Lancet Infect Dis. 2023;23:371-379.
References
  1. Skar GL, Simonsen KA. Lyme disease. StatPearls [Internet]. Updated February 4, 2024. Accessed March 20, 2024. https://www.ncbi.nlm.nih.gov/books/NBK431066/
  2. Dennison R, Novak C, Rebman A, et al. Lyme disease with erythema migrans and seventh nerve palsy in an African-American man. Cureus. 2019;11:E6509.
  3. Bax CE, Clark AK, Oboite M, et al. A case of disseminated Lyme disease in a child with skin of color. Pediatr Dermatol. 2021;38 (suppl 2):140-141.
  4. Shah AS, Varatharaj Palraj BR. Multiple erythema migrans rashes characteristic of early disseminated lyme disease, before and after therapy. Mayo Clin Proc. 2019;94:172-173.
  5. Feder HM Jr, Abeles M, Bernstein M, et al. Diagnosis, treatment, and prognosis of erythema migrans and Lyme arthritis. Clin Dermatol. 2006;24:509-520.
  6. Schotthoefer AM, Green CB, Dempsey G, et al. The spectrum of erythema migrans in early Lyme disease: can we improve its recognition? Cureus. 2022;14:E30673.
  7. Strle F, Wormser GP. Early Lyme disease (erythema migrans) and its mimics (southern tick-associated rash illness and tick-associated rash illness). Infect Dis Clin North Am. 2022;36:523-539.
  8. Torbahn G, Hofmann H, Rücker G, et al. Efficacy and safety of antibiotic therapy in early cutaneous Lyme borreliosis: a network meta-analysis. JAMA Dermatol. 2018;154:1292-1303.
  9. Stupica D, Collinet-Adler S, Blagus R, et al. Treatment of erythema migrans with doxycycline for 7 days versus 14 days in Slovenia: a randomised open-label non-inferiority trial. Lancet Infect Dis. 2023;23:371-379.
Issue
Cutis - 113(4)
Issue
Cutis - 113(4)
Page Number
183-184
Page Number
183-184
Publications
Publications
Topics
Article Type
Display Headline
Asymptomatic Erythematous Plaque in an Outdoorsman
Display Headline
Asymptomatic Erythematous Plaque in an Outdoorsman
Sections
Questionnaire Body

A middle-aged man presented with a well-demarcated, hyperpigmented, erythematous patch with an annular erythematous border that extended from the mid-back to the lower back. The patient was otherwise asymptomatic. He was an avid gardener who resided in South Carolina and had recently adopted 2 puppies.

Barker_asymptomatic.jpg

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 04/02/2024 - 10:45
Un-Gate On Date
Tue, 04/02/2024 - 10:45
Use ProPublica
CFC Schedule Remove Status
Tue, 04/02/2024 - 10:45
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
180026FC.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

Asymptomatic Violaceous Plaques on the Face and Back

Article Type
Changed
Wed, 03/13/2024 - 08:48
Display Headline
Asymptomatic Violaceous Plaques on the Face and Back

The Diagnosis: Cutaneous Sarcoidosis

A biopsy of a plaque on the back confirmed cutaneous sarcoidosis (CS). A chest radiograph demonstrated hilar nodes, and a referral was placed for comanagement with a pulmonologist. Histopathology was critical in making the diagnosis, with well-circumscribed noncaseating granulomas present in the dermis. The granulomas in CS often are described as naked, as there are minimal lymphocytes present and plasma cells normally are absent.1 Because the lungs are the most common site of involvement, a chest radiograph is necessary to examine for systemic sarcoidosis. Laboratory workup is used to evaluate for lymphopenia, hypercalcemia, elevated blood sedimentation rate, and elevated angiotensin- converting enzyme levels, which are common in systemic sarcoidosis.1

Sarcoidosis is a multisystemic granulomatous disorder with an unknown etiology. It is believed to develop in genetically predisposed individuals as a reaction to unidentified antigens in the environment.1 Helper T cells (TH1) respond to these environmental antigens in those who are susceptible, which leads to the disease process, but paradoxically, even with the elevation of cellular immune activity at the sites of the granulomatous inflammation, the peripheral immune response in these patients is suppressed as shown by lymphopenia.2

Cutaneous sarcoidosis is found in approximately one-third of patients with systemic sarcoidosis but can occur without systemic involvement.1,2 Sarcoidosis is reported worldwide and affects patients of all races and ethnicities, ages, and sexes but does have a higher prevalence among Black individuals in the United States, patients younger than 40 years (peak incidence, 20–29 years of age), and females.2 In 80% of patients, CS occurs before systemic sarcoidosis develops, or they may develop simultaneously.1

Cutaneous sarcoidosis has a wide range of clinical presentations that are classified as specific and nonspecific. Specific lesions in CS contain noncaseating granulomas while nonspecific lesions in CS appear as reactive processes.2 The most common specific presentation of CS includes papules that are brown in pigmentation in lighter skin tones and red to violaceous in darker skin tones (Figure). The most common nonspecific skin manifestation is erythema nodosum, which represents a hypersensitivity reaction. Cutaneous sarcoidosis can appear as hypopigmented or hyperpigmented patches or plaques.1

DeVore2_figure.jpg
%3Cp%3EIndurated%2C%20flesh-colored%20to%20violaceous%20plaques%20on%20the%20chin%20in%20a%20patient%20with%20cutaneous%20sarcoidosis.%3C%2Fp%3E

Treatments for CS vary based on the individual.1 For milder and more localized cases, topical or intralesional steroids may be used. If systemic sarcoidosis is suspected or if there is diffuse involvement of the skin, systemic steroids, antimalarials (eg, hydroxychloroquine), low-dose methotrexate, minocycline, allopurinol, azathioprine, isotretinoin, tumor necrosis factor α inhibitors, or psoralen plus long-wave UVA radiation may be used. If systemic sarcoidosis is present, referral to a pulmonologist is recommended for co-management.1

Cutaneous sarcoidosis is known as the “great imitator,” and there are multiple diseases to consider in the differential that are distinguished by the physical findings.1 In our case of a middle-aged Black woman with indurated plaques, a few diagnoses to consider were psoriasis, discoid lupus erythematosus (DLE), mycosis fungoides (MF), and tinea infection.

Psoriasis is a common disease, and 90% of patients have chronic plaquelike disease with well-demarcated erythematous plaques that have a silver-gray scale and a positive Auspitz sign (also known as pinpoint bleeding).3 Plaques often are distributed on the trunk, limb extensors, and scalp, along with nail changes. Some patients also have joint pain, indicating psoriatic arthritis. The etiology of psoriasis is unknown, but it develops due to unrestrained keratinocyte proliferation and defective differentiation, which leads to histopathology showing regular acanthosis and papillary dermal ectasia with rouleaux. Mild cases typically are treated with topical steroids or vitamin D, while more severe cases are treated with methotrexate, cyclosporine, retinoids, or biologics.3

Discoid lupus erythematosus occurs 4 times more often in Black patients than in White patients. Clinically, DLE begins as well-defined, erythematous, scaly patches that expand with hyperpigmentation at the periphery and leave an atrophic, scarred, hypopigmented center.4 It typically is localized to the head and neck, but in cases where it disseminates elsewhere on the body, the risk for systemic lupus erythematosus increases from 1.2% to 28%.5 Histopathology of DLE shows vacuolar degeneration of the basal cell layer in the epidermis along with patchy lymphocytic infiltrate in the dermis. Treatments range from topical steroids for mild cases to antimalarial agents, retinoids, anti-inflammatory drugs, and calcineurin inhibitors for more severe cases.4

Although there are multiple types of cutaneous T-cell lymphoma, the most common is MF, which traditionally is nonaggressive. The typical patient with MF is older than 60 years and presents with indolent, ongoing, flat to minimally indurated patches or plaques that have cigarette paper scale. As MF progresses, some plaques grow into tumors and can become more aggressive. Histologically, MF changes based on its clinical stage, with the initial phase showing epidermotropic atypical lymphocytes and later phases showing less epitheliotropic, larger, atypical lymphocytes. The treatment algorithm varies depending on cutaneous T-cell lymphoma staging.6

Tinea infections are caused by dermatophytes. In prepubertal children, they predominantly appear as tinea corporis (on the body) or tinea capitis (on the scalp), but in adults they appear as tinea cruris (on the groin), tinea pedis (on the feet), or tinea unguium (on the nails).7 Tinea infections classically are known to appear as an annular patch with an active erythematous scaling border and central clearing. The patches can be pruritic. Potassium hydroxide preparation of a skin scraping is a quick test to use in the office; if the results are inconclusive, a culture may be required. Treatment depends on the location of the infection but typically involves either topical or oral antifungal agents.7

References
  1. Tchernev G, Cardoso JC, Chokoeva AA, et al. The “mystery” of cutaneous sarcoidosis: facts and controversies. Int J Immunopathol Pharmacol. 2014;27:321-330. doi:10.1177/039463201402700302
  2. Ali MM, Atwan AA, Gonzalez ML. Cutaneous sarcoidosis: updates in the pathogenesis. J Eur Acad Dermatol Venereol. 2010;24:747-755. doi:10.1111/j.1468-3083.2009.03517.x
  3. Rendon A, Schäkel K. Psoriasis pathogenesis and treatment [published online March 23, 2019]. Int J Mol Sci. 2019;20:1475. doi:10.3390/ijms20061475
  4. McDaniel B, Sukumaran S, Koritala T, et al. Discoid lupus erythematosus. StatPearls [Internet]. StatPearls Publishing; 2023. Accessed December 11, 2023. https://www.ncbi.nlm.nih.gov/books/NBK493145/
  5. Bhat MR, Hulmani M, Dandakeri S, et al. Disseminated discoid lupus erythematosus leading to squamous cell carcinoma. Indian J Dermatol. 2012;57:158-161. doi:10.4103/0019-5154.94298
  6. Pulitzer M. Cutaneous T-cell Lymphoma. Clin Lab Med. 2017; 37:527-546. doi:10.1016/j.cll.2017.06.006
  7. Ely JW, Rosenfeld S, Seabury Stone M. Diagnosis and management of tinea infections. Am Fam Physician. 2014;90:702-710.
Article PDF
Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Dr. DeVore 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: Ansley C. DeVore, MD, Medical University of South Carolina, Department of Dermatology, 135 Rutledge Ave, 3rd Floor, Charleston, SC 29425 (devorea@musc.edu).

Issue
Cutis - 113(1)
Publications
Topics
Page Number
25,27-28
Sections
Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Dr. DeVore 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: Ansley C. DeVore, MD, Medical University of South Carolina, Department of Dermatology, 135 Rutledge Ave, 3rd Floor, Charleston, SC 29425 (devorea@musc.edu).

Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Dr. DeVore 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: Ansley C. DeVore, MD, Medical University of South Carolina, Department of Dermatology, 135 Rutledge Ave, 3rd Floor, Charleston, SC 29425 (devorea@musc.edu).

Article PDF
Article PDF
Related Articles

The Diagnosis: Cutaneous Sarcoidosis

A biopsy of a plaque on the back confirmed cutaneous sarcoidosis (CS). A chest radiograph demonstrated hilar nodes, and a referral was placed for comanagement with a pulmonologist. Histopathology was critical in making the diagnosis, with well-circumscribed noncaseating granulomas present in the dermis. The granulomas in CS often are described as naked, as there are minimal lymphocytes present and plasma cells normally are absent.1 Because the lungs are the most common site of involvement, a chest radiograph is necessary to examine for systemic sarcoidosis. Laboratory workup is used to evaluate for lymphopenia, hypercalcemia, elevated blood sedimentation rate, and elevated angiotensin- converting enzyme levels, which are common in systemic sarcoidosis.1

Sarcoidosis is a multisystemic granulomatous disorder with an unknown etiology. It is believed to develop in genetically predisposed individuals as a reaction to unidentified antigens in the environment.1 Helper T cells (TH1) respond to these environmental antigens in those who are susceptible, which leads to the disease process, but paradoxically, even with the elevation of cellular immune activity at the sites of the granulomatous inflammation, the peripheral immune response in these patients is suppressed as shown by lymphopenia.2

Cutaneous sarcoidosis is found in approximately one-third of patients with systemic sarcoidosis but can occur without systemic involvement.1,2 Sarcoidosis is reported worldwide and affects patients of all races and ethnicities, ages, and sexes but does have a higher prevalence among Black individuals in the United States, patients younger than 40 years (peak incidence, 20–29 years of age), and females.2 In 80% of patients, CS occurs before systemic sarcoidosis develops, or they may develop simultaneously.1

Cutaneous sarcoidosis has a wide range of clinical presentations that are classified as specific and nonspecific. Specific lesions in CS contain noncaseating granulomas while nonspecific lesions in CS appear as reactive processes.2 The most common specific presentation of CS includes papules that are brown in pigmentation in lighter skin tones and red to violaceous in darker skin tones (Figure). The most common nonspecific skin manifestation is erythema nodosum, which represents a hypersensitivity reaction. Cutaneous sarcoidosis can appear as hypopigmented or hyperpigmented patches or plaques.1

DeVore2_figure.jpg
%3Cp%3EIndurated%2C%20flesh-colored%20to%20violaceous%20plaques%20on%20the%20chin%20in%20a%20patient%20with%20cutaneous%20sarcoidosis.%3C%2Fp%3E

Treatments for CS vary based on the individual.1 For milder and more localized cases, topical or intralesional steroids may be used. If systemic sarcoidosis is suspected or if there is diffuse involvement of the skin, systemic steroids, antimalarials (eg, hydroxychloroquine), low-dose methotrexate, minocycline, allopurinol, azathioprine, isotretinoin, tumor necrosis factor α inhibitors, or psoralen plus long-wave UVA radiation may be used. If systemic sarcoidosis is present, referral to a pulmonologist is recommended for co-management.1

Cutaneous sarcoidosis is known as the “great imitator,” and there are multiple diseases to consider in the differential that are distinguished by the physical findings.1 In our case of a middle-aged Black woman with indurated plaques, a few diagnoses to consider were psoriasis, discoid lupus erythematosus (DLE), mycosis fungoides (MF), and tinea infection.

Psoriasis is a common disease, and 90% of patients have chronic plaquelike disease with well-demarcated erythematous plaques that have a silver-gray scale and a positive Auspitz sign (also known as pinpoint bleeding).3 Plaques often are distributed on the trunk, limb extensors, and scalp, along with nail changes. Some patients also have joint pain, indicating psoriatic arthritis. The etiology of psoriasis is unknown, but it develops due to unrestrained keratinocyte proliferation and defective differentiation, which leads to histopathology showing regular acanthosis and papillary dermal ectasia with rouleaux. Mild cases typically are treated with topical steroids or vitamin D, while more severe cases are treated with methotrexate, cyclosporine, retinoids, or biologics.3

Discoid lupus erythematosus occurs 4 times more often in Black patients than in White patients. Clinically, DLE begins as well-defined, erythematous, scaly patches that expand with hyperpigmentation at the periphery and leave an atrophic, scarred, hypopigmented center.4 It typically is localized to the head and neck, but in cases where it disseminates elsewhere on the body, the risk for systemic lupus erythematosus increases from 1.2% to 28%.5 Histopathology of DLE shows vacuolar degeneration of the basal cell layer in the epidermis along with patchy lymphocytic infiltrate in the dermis. Treatments range from topical steroids for mild cases to antimalarial agents, retinoids, anti-inflammatory drugs, and calcineurin inhibitors for more severe cases.4

Although there are multiple types of cutaneous T-cell lymphoma, the most common is MF, which traditionally is nonaggressive. The typical patient with MF is older than 60 years and presents with indolent, ongoing, flat to minimally indurated patches or plaques that have cigarette paper scale. As MF progresses, some plaques grow into tumors and can become more aggressive. Histologically, MF changes based on its clinical stage, with the initial phase showing epidermotropic atypical lymphocytes and later phases showing less epitheliotropic, larger, atypical lymphocytes. The treatment algorithm varies depending on cutaneous T-cell lymphoma staging.6

Tinea infections are caused by dermatophytes. In prepubertal children, they predominantly appear as tinea corporis (on the body) or tinea capitis (on the scalp), but in adults they appear as tinea cruris (on the groin), tinea pedis (on the feet), or tinea unguium (on the nails).7 Tinea infections classically are known to appear as an annular patch with an active erythematous scaling border and central clearing. The patches can be pruritic. Potassium hydroxide preparation of a skin scraping is a quick test to use in the office; if the results are inconclusive, a culture may be required. Treatment depends on the location of the infection but typically involves either topical or oral antifungal agents.7

The Diagnosis: Cutaneous Sarcoidosis

A biopsy of a plaque on the back confirmed cutaneous sarcoidosis (CS). A chest radiograph demonstrated hilar nodes, and a referral was placed for comanagement with a pulmonologist. Histopathology was critical in making the diagnosis, with well-circumscribed noncaseating granulomas present in the dermis. The granulomas in CS often are described as naked, as there are minimal lymphocytes present and plasma cells normally are absent.1 Because the lungs are the most common site of involvement, a chest radiograph is necessary to examine for systemic sarcoidosis. Laboratory workup is used to evaluate for lymphopenia, hypercalcemia, elevated blood sedimentation rate, and elevated angiotensin- converting enzyme levels, which are common in systemic sarcoidosis.1

Sarcoidosis is a multisystemic granulomatous disorder with an unknown etiology. It is believed to develop in genetically predisposed individuals as a reaction to unidentified antigens in the environment.1 Helper T cells (TH1) respond to these environmental antigens in those who are susceptible, which leads to the disease process, but paradoxically, even with the elevation of cellular immune activity at the sites of the granulomatous inflammation, the peripheral immune response in these patients is suppressed as shown by lymphopenia.2

Cutaneous sarcoidosis is found in approximately one-third of patients with systemic sarcoidosis but can occur without systemic involvement.1,2 Sarcoidosis is reported worldwide and affects patients of all races and ethnicities, ages, and sexes but does have a higher prevalence among Black individuals in the United States, patients younger than 40 years (peak incidence, 20–29 years of age), and females.2 In 80% of patients, CS occurs before systemic sarcoidosis develops, or they may develop simultaneously.1

Cutaneous sarcoidosis has a wide range of clinical presentations that are classified as specific and nonspecific. Specific lesions in CS contain noncaseating granulomas while nonspecific lesions in CS appear as reactive processes.2 The most common specific presentation of CS includes papules that are brown in pigmentation in lighter skin tones and red to violaceous in darker skin tones (Figure). The most common nonspecific skin manifestation is erythema nodosum, which represents a hypersensitivity reaction. Cutaneous sarcoidosis can appear as hypopigmented or hyperpigmented patches or plaques.1

DeVore2_figure.jpg
%3Cp%3EIndurated%2C%20flesh-colored%20to%20violaceous%20plaques%20on%20the%20chin%20in%20a%20patient%20with%20cutaneous%20sarcoidosis.%3C%2Fp%3E

Treatments for CS vary based on the individual.1 For milder and more localized cases, topical or intralesional steroids may be used. If systemic sarcoidosis is suspected or if there is diffuse involvement of the skin, systemic steroids, antimalarials (eg, hydroxychloroquine), low-dose methotrexate, minocycline, allopurinol, azathioprine, isotretinoin, tumor necrosis factor α inhibitors, or psoralen plus long-wave UVA radiation may be used. If systemic sarcoidosis is present, referral to a pulmonologist is recommended for co-management.1

Cutaneous sarcoidosis is known as the “great imitator,” and there are multiple diseases to consider in the differential that are distinguished by the physical findings.1 In our case of a middle-aged Black woman with indurated plaques, a few diagnoses to consider were psoriasis, discoid lupus erythematosus (DLE), mycosis fungoides (MF), and tinea infection.

Psoriasis is a common disease, and 90% of patients have chronic plaquelike disease with well-demarcated erythematous plaques that have a silver-gray scale and a positive Auspitz sign (also known as pinpoint bleeding).3 Plaques often are distributed on the trunk, limb extensors, and scalp, along with nail changes. Some patients also have joint pain, indicating psoriatic arthritis. The etiology of psoriasis is unknown, but it develops due to unrestrained keratinocyte proliferation and defective differentiation, which leads to histopathology showing regular acanthosis and papillary dermal ectasia with rouleaux. Mild cases typically are treated with topical steroids or vitamin D, while more severe cases are treated with methotrexate, cyclosporine, retinoids, or biologics.3

Discoid lupus erythematosus occurs 4 times more often in Black patients than in White patients. Clinically, DLE begins as well-defined, erythematous, scaly patches that expand with hyperpigmentation at the periphery and leave an atrophic, scarred, hypopigmented center.4 It typically is localized to the head and neck, but in cases where it disseminates elsewhere on the body, the risk for systemic lupus erythematosus increases from 1.2% to 28%.5 Histopathology of DLE shows vacuolar degeneration of the basal cell layer in the epidermis along with patchy lymphocytic infiltrate in the dermis. Treatments range from topical steroids for mild cases to antimalarial agents, retinoids, anti-inflammatory drugs, and calcineurin inhibitors for more severe cases.4

Although there are multiple types of cutaneous T-cell lymphoma, the most common is MF, which traditionally is nonaggressive. The typical patient with MF is older than 60 years and presents with indolent, ongoing, flat to minimally indurated patches or plaques that have cigarette paper scale. As MF progresses, some plaques grow into tumors and can become more aggressive. Histologically, MF changes based on its clinical stage, with the initial phase showing epidermotropic atypical lymphocytes and later phases showing less epitheliotropic, larger, atypical lymphocytes. The treatment algorithm varies depending on cutaneous T-cell lymphoma staging.6

Tinea infections are caused by dermatophytes. In prepubertal children, they predominantly appear as tinea corporis (on the body) or tinea capitis (on the scalp), but in adults they appear as tinea cruris (on the groin), tinea pedis (on the feet), or tinea unguium (on the nails).7 Tinea infections classically are known to appear as an annular patch with an active erythematous scaling border and central clearing. The patches can be pruritic. Potassium hydroxide preparation of a skin scraping is a quick test to use in the office; if the results are inconclusive, a culture may be required. Treatment depends on the location of the infection but typically involves either topical or oral antifungal agents.7

References
  1. Tchernev G, Cardoso JC, Chokoeva AA, et al. The “mystery” of cutaneous sarcoidosis: facts and controversies. Int J Immunopathol Pharmacol. 2014;27:321-330. doi:10.1177/039463201402700302
  2. Ali MM, Atwan AA, Gonzalez ML. Cutaneous sarcoidosis: updates in the pathogenesis. J Eur Acad Dermatol Venereol. 2010;24:747-755. doi:10.1111/j.1468-3083.2009.03517.x
  3. Rendon A, Schäkel K. Psoriasis pathogenesis and treatment [published online March 23, 2019]. Int J Mol Sci. 2019;20:1475. doi:10.3390/ijms20061475
  4. McDaniel B, Sukumaran S, Koritala T, et al. Discoid lupus erythematosus. StatPearls [Internet]. StatPearls Publishing; 2023. Accessed December 11, 2023. https://www.ncbi.nlm.nih.gov/books/NBK493145/
  5. Bhat MR, Hulmani M, Dandakeri S, et al. Disseminated discoid lupus erythematosus leading to squamous cell carcinoma. Indian J Dermatol. 2012;57:158-161. doi:10.4103/0019-5154.94298
  6. Pulitzer M. Cutaneous T-cell Lymphoma. Clin Lab Med. 2017; 37:527-546. doi:10.1016/j.cll.2017.06.006
  7. Ely JW, Rosenfeld S, Seabury Stone M. Diagnosis and management of tinea infections. Am Fam Physician. 2014;90:702-710.
References
  1. Tchernev G, Cardoso JC, Chokoeva AA, et al. The “mystery” of cutaneous sarcoidosis: facts and controversies. Int J Immunopathol Pharmacol. 2014;27:321-330. doi:10.1177/039463201402700302
  2. Ali MM, Atwan AA, Gonzalez ML. Cutaneous sarcoidosis: updates in the pathogenesis. J Eur Acad Dermatol Venereol. 2010;24:747-755. doi:10.1111/j.1468-3083.2009.03517.x
  3. Rendon A, Schäkel K. Psoriasis pathogenesis and treatment [published online March 23, 2019]. Int J Mol Sci. 2019;20:1475. doi:10.3390/ijms20061475
  4. McDaniel B, Sukumaran S, Koritala T, et al. Discoid lupus erythematosus. StatPearls [Internet]. StatPearls Publishing; 2023. Accessed December 11, 2023. https://www.ncbi.nlm.nih.gov/books/NBK493145/
  5. Bhat MR, Hulmani M, Dandakeri S, et al. Disseminated discoid lupus erythematosus leading to squamous cell carcinoma. Indian J Dermatol. 2012;57:158-161. doi:10.4103/0019-5154.94298
  6. Pulitzer M. Cutaneous T-cell Lymphoma. Clin Lab Med. 2017; 37:527-546. doi:10.1016/j.cll.2017.06.006
  7. Ely JW, Rosenfeld S, Seabury Stone M. Diagnosis and management of tinea infections. Am Fam Physician. 2014;90:702-710.
Issue
Cutis - 113(1)
Issue
Cutis - 113(1)
Page Number
25,27-28
Page Number
25,27-28
Publications
Publications
Topics
Article Type
Display Headline
Asymptomatic Violaceous Plaques on the Face and Back
Display Headline
Asymptomatic Violaceous Plaques on the Face and Back
Sections
Questionnaire Body

A 35-year-old Black woman presented to dermatology as a new patient for evaluation of an asymptomatic rash that had enlarged and spread to involve both the face and back over the last 4 months. She had not tried any treatments. She had no notable medical history and was uncertain of her family history. Physical examination showed indurated, flesh-colored to violaceous plaques around the alar-facial groove (top), nasal tip, chin, and back (bottom). The mucosae and nails were not involved.

CT113001025_Quiz_top_bottom.jpg

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 01/05/2024 - 11:00
Un-Gate On Date
Fri, 01/05/2024 - 11:00
Use ProPublica
CFC Schedule Remove Status
Fri, 01/05/2024 - 11:00
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
1800269F.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