Aquatic Antagonists: Sponge Dermatitis

Article Type
Changed
Mon, 01/11/2021 - 17:01

Sponges are among the oldest animals on earth, appearing more than 640 million years ago before the Cambrian explosion, a period when most major animal phyla appeared in the fossil records.1 More than 10,000 species of sponges have been identified worldwide and are distributed from polar to tropical regions in both marine (Figure 1) and freshwater (Figure 2) environments. They inhabit both shallow waters as well as depths of more than 2800 m, with shallower sponges tending to be more vibrantly colored than their deeper counterparts. The wide-ranging habitats of sponges have led to size variations from as small as 0.05 mm to more than 3 m in height.2 Their taxonomic phylum, Porifera (meaning pore bearers), is derived from the millions of pores lining the surface of the sponge that are used to filter planktonic organisms.3 Flagellated epithelioid cells called choanocytes line the internal chambers of sponges, creating a water current that promotes filter feeding as well as nutrient absorption across their microvilli.4 The body walls of many sponges consist of a collagenous skeleton made up of spongin and spicules of silicon dioxide (silica) or calcium carbonate embedded in the spongin connective tissue matrix.5 Bath sponges lack silica spicules.

Figure 1. Marine sponges. A, Tedania ignis (fire sponge). Photograph courtesy of Vidal Haddad Jr, MD, PhD (Botucatu, São Paulo, Brazil). B, Agelas conifera (brown tube sponge). Photograph courtesy of Dirk M. Elston, MD (Charleston, South Carolina).

Figure 2. Cauxi sponge, a type of freshwater sponge. Photograph courtesy of Vidal Haddad Jr, MD, PhD (Botucatu, São Paulo, Brazil).

Sponges have been used in medicine for centuries. The first use in Western culture was recorded in 405 bce in The Frogs, a comedy by Aristophanes in which a sponge was placed on a character’s heart following a syncopal episode. Additionally, in many Hippocratic writings, the use of sponges is outlined in the treatment of a variety of ailments. Similarly, the ancient Chinese and Greeks used burnt sponge and seaweed as a source of iodine to treat goiters.6,7 Modern research focuses on the use of sponge metabolites for their antineoplastic, antimicrobial, and anti-inflammatory effects.8 Identification of spongouridine and spongothymidine from the sponge Tectitethya crypta led to the development of cytarabine and gemcitabine8 as well as the discovery of the antiviral agent vidarabine.9 The monoclonal antibody assay for the detection of shellfish poisoning was prepared using the sponge Halichondria okadai.10

Mechanisms and Symptoms of Injury

Bathing sponges (silk sponges) derived from Spongia officinalis are harmless. Other sponges can exert their damaging effects through a variety of mechanisms that lead to dermatologic manifestations (eTable). Some species of sponges produce and secrete toxic metabolites (eg, crinotoxins) onto the body surface or into the surrounding water. They also are capable of synthesizing a mucous slime that can be irritating to human skin. Direct trauma also can be caused by fragments of the silica or calcium carbonate sponge skeleton penetrating the skin. Stinging members of the phylum Cnidaria can colonize the sponge, leading to injury when a human handles the sponge.25-27

Sponge dermatitis can be divided into 2 major categories: an initial pruritic dermatitis (Figure 3) that occurs within 20 minutes to a few hours after contact and a delayed irritant dermatitis caused by penetration of the spicules and chemical agents into skin.28 Importantly, different species can lead to varying manifestations.

Figure 3. Initial pruritic eczematous plaques with erythema and edema after handling a toxic marine sponge. Photograph courtesy of Vidal Haddad Jr, MD, PhD (Botucatu, São Paulo, Brazil).


The initial pruritic dermatitis is characterized by itching and burning that progresses to local edema, vesiculation, joint swelling, and stiffness. Because most contact with sponges occurs with handling, joint immobility may ensue within 24 hours of the encounter. Rarely, larger areas of the skin are affected, and fever, chills, malaise, dizziness, nausea, purulent bullae, muscle cramps, and formication may occur.28 Anaphylactic reactions have been described in a small subset of patients. There have even been reports of delayed (ie, 1–2 weeks following exposure) erythema multiforme, livedo reticularis, purpura, and dyshidrotic eczema.16,20,29 The irritant dermatitis caused by spicule trauma is due to a foreign body reaction that can be exacerbated by toxins entering the skin. In severe cases, desquamation, recurrent eczema, and arthralgia can occur.30 In general, more mild cases should self-resolve within 3 to 7 days. Dermatologic conditions also can be caused by organisms that inhabit sponges and as a result produce a dermatitis when the sponge is handled, including sponge divers disease (maladie des plongeurs), a necrotic dermatitis caused by stinging Cnidaria species.31 Dogger Bank itch, first described as a dermatitis caused by sensitization to (2-hydroxyethyl) dimethylsulfoxonium chloride, initially was isolated from the sea chervil (a type of Bryozoan); however, that same chemical also was later found in sponges, producing the same dermatitis after handling the sponge.32 Freshwater sponges also have been reported to be injurious and exist worldwide. In contrast to marine sponges, lesions from freshwater sponges are disseminated pruritic erythematous papules with ulcerations, crusts, and secondary infections.22 The disseminated nature of the dermatitis caused by freshwater sponges is due to contact with the spicules of dead sponges that are dispersed throughout the water rather than from direct handling. Sponge dermatitis occurs mostly in sponge collectors, divers, trawlers, and biology students and has been reported extensively in the United States, Caribbean Islands, Australia, New Zealand, and Brazil.18,27,33,34

Management

Treatment should consist of an initial decontamination; the skin should be dried, and adhesive tape or rubber cement should be utilized to remove any spicules embedded in the skin. Diluted vinegar soaks should be initiated for 10 to 30 minutes on the affected area(s) 3 or 4 times daily.19 The initial decontamination should occur immediately, as delay may lead to persistent purulent bullae that may take months to heal. Topical steroids may be used following the initial decontamination to help relieve inflammation. Antihistamines and nonsteroidal anti-inflammatory drugs may be used to alleviate pruritus and pain, respectively. Severe cases may require systemic glucocorticoids. Additionally, immunization status against tetanus toxoid should be assessed.35 In the event of an anaphylactic reaction, it is important to maintain a patent airway and normalized blood pressure through the use of intramuscular epinephrine.36 Frequent follow-up is warranted, as serious secondary infections can develop.37 Patients also should be counseled on the potential for delayed dermatologic reactions, including erythema multiforme. Contact between humans and coastal environments has been increasing in the last few decades; therefore, an increase in contact with sponges is to be expected.22

References
  1. Gold DA, Grabenstatter J, de Mendoza A, et al. Sterol and genomic analyses validate the sponge biomarker hypothesis. Proc Natl Acad Sci U S A. 2016;113:2684-2689.
  2. Bonamonte D, Filoni A, Verni P, et al. Dermatitis caused by sponges. In: Bonamonte D, Angelini G, eds. Aquatic Dermatology. 2nd ed. Springer; 2016:121-126.
  3. Marsh LM, Slack-Smith S, Gurry DL. Field Guide to Sea Stingers and Other Venomous and Poisonous Marine Invertebrates. 2nd ed. Western Australian Museum; 2010.
  4. Eid E, Al-Tawaha M. A Guide to Harmful and Toxic Creatures in the Gulf of Aqaba Jordan. The Royal Marine Conservation Society of Jordan; 2016.
  5. Reese E, Depenbrock P. Water envenomations and stings. Curr Sports Med Rep. 2014;13:126-131.
  6. Dormandy TL. Trace element analysis of hair. Br Med J (Clin Res Ed). 1986;293:975-976.
  7. Voultsiadou E. Sponges: an historical survey of their knowledge in Greek antiquity. J Mar Biol Assoc UK. 2007;87:1757-1763.
  8. Senthilkumar K, Kim SK. Marine invertebrate natural products for anti-inflammatory and chronic diseases [published online December 31, 2013]. Evid Based Complement Alternat Med. doi:10.1155/2013/572859
  9. Sagar S, Kaur M, Minneman KP. Antiviral lead compounds from marine sponges. Mar Drugs. 2010;8:2619-2638.
  10. Usagawa T, Nishimura M, Itoh Y, et al. Preparation of monoclonal antibodies against okadaic acid prepared from the sponge Halichondria okadai. Toxicon. 1989;27:1323-1330.
  11. Elston DM. Aquatic antagonists: sponge dermatitis. Cutis. 2007;80:279-280.
  12. Parra-Velandia FJ, Zea S, Van Soest RW. Reef sponges of the genus Agelas (Porifera: Demospongiae) from the Greater Caribbean. Zootaxa. 2014;3794:301-343.
  13. Hooper JN, Capon RJ, Hodder RA. A new species of toxic marine sponge (Porifera: Demospongiae: Poecilosclerida) from northwest Australia. The Beagle, Records of the Northern Territory Museum of Arts and sciences. 1991;8:27-36.
  14. Burnett JW, Calton GJ, Morgan RJ. Dermatitis due to stinging sponges. Cutis. 1987;39:476.
  15. Kizer KW. Marine envenomations. J Toxicol Clin Toxicol. 1983;21:527-555.
  16. Isbister GK, Hooper JN. Clinical effects of stings by sponges of the genus Tedania and a review of sponge stings worldwide. Toxicon. 2005;46:782-785.
  17. Fromont J, Abdo DA. New species of Haliclona (Demospongiae: Haplosclerida: Chalinidae) from Western Australia. Zootaxa. 2014;3835:97-109.
  18. Flachsenberger W, Holmes NJ, Leigh C, et al. Properties of the extract and spicules of the dermatitis inducing sponge Neofibularia mordens Hartman. J Toxicol Clin Toxicol. 1987;25:255-272.
  19. Southcott RV, Coulter JR. The effects of the southern Australian marine stinging sponges, Neofibularia mordens and Lissodendoryx sp. Med J Aust. 1971;2:895-901.
  20. Yaffee HS, Stargardter F. Erythema multiforme from Tedania ignis. report of a case and an experimental study of the mechanism of cutaneous irritation from the fire sponge. Arch Dermatol. 1963;87:601-604.
  21. Yaffee HS. Irritation from red sponge. N Engl J Med. 1970;282:51.
  22. Haddad V Jr. Environmental dermatology: skin manifestations of injuries caused by invertebrate aquatic animals. An Bras Dermatol. 2013;88:496-506.
  23. Volkmer-Ribeiro C, Lenzi HL, Orefice F, et al. Freshwater sponge spicules: a new agent of ocular pathology. Mem Inst Oswaldo Cruz. 2006;101:899-903.
  24. Cruz AA, Alencar VM, Medina NH, et al. Dangerous waters: outbreak of eye lesions caused by fresh water sponge spicules. Eye (Lond). 2013;27:398-402.
  25. Haddad V Jr. Clinical and therapeutic aspects of envenomations caused by sponges and jellyfish. In: Gopalakrishnakone P, Haddad V Jr, Kem WR, et al, eds. Marine and Freshwater Toxins. Springer; 2016:317-325.
  26. Haddad V Jr, Lupi O, Lonza JP, et al. Tropical dermatology: marine and aquatic dermatology. J Am Acad Dermatol. 2009;61:733-750.
  27. Gaastra MT. Aquatic skin disorders. In: Faber WR, Hay RJ, Naafs B, eds. Imported Skin Diseases. 2nd ed. Wiley; 2012:283-292.
  28. Auerbach P. Envenomation by aquatic invertebrates. In: Auerbach P, ed. Wilderness Medicine. 6th ed. Elsevier Mosby; 2011;1596-1627.
  29. Sims JK, Irei MY. Human Hawaiian marine sponge poisoning. Hawaii Med J. 1979;38:263-270.
  30. Haddad V Jr. Aquatic animals of medical importance in Brazil. Rev Soc Bras Med Trop. 2003;36:591-597.
  31. Tlougan BE, Podjasek JO, Adams BB. Aquatic sports dermatoses. part 2—in the water: saltwater dermatoses. Int J Dermatol. 2010;49:994-1002.
  32. Warabi K, Nakao Y, Matsunaga S, et al. Dogger Bank itch revisited: isolation of (2-hydroxyethyl) dimethylsulfoxonium chloride as a cytotoxic constituent from the marine sponge Theonella aff. mirabilis. Comp Biochem Physiol B Biochem Mol Biol. 2001;128:27-30.
  33. Southcott R. Human injuries from invertebrate animals in the Australian seas. Clin Toxicol. 1970;3:617-636.
  34. Russell FE. Sponge injury—traumatic, toxic or allergic? N Engl J Med. 1970;282:753-754.
  35. Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337.
  36. Muraro A, Roberts G, Worm M, et al. Anaphylaxis: guidelines from the European Academy of Allergy and Clinical Immunology. Allergy. 2014;69:1026-1045.
  37. Kizer K, Auerbach P, Dwyer B. Marine envenomations: not just a problem of the tropics. Emerg Med Rep. 1985;6:129-135.
Article PDF
Author and Disclosure Information

Dr. Cahn is from the Memorial Sloan Kettering Cancer Center, New York, New York. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Brian A. Cahn, MD, 1275 York Ave, New York, NY 10065 (briancahn1489@gmail.com).

Issue
Cutis - 107(1)
Publications
Topics
Page Number
34-36, E5
Sections
Author and Disclosure Information

Dr. Cahn is from the Memorial Sloan Kettering Cancer Center, New York, New York. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Brian A. Cahn, MD, 1275 York Ave, New York, NY 10065 (briancahn1489@gmail.com).

Author and Disclosure Information

Dr. Cahn is from the Memorial Sloan Kettering Cancer Center, New York, New York. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Brian A. Cahn, MD, 1275 York Ave, New York, NY 10065 (briancahn1489@gmail.com).

Article PDF
Article PDF
Related Articles

Sponges are among the oldest animals on earth, appearing more than 640 million years ago before the Cambrian explosion, a period when most major animal phyla appeared in the fossil records.1 More than 10,000 species of sponges have been identified worldwide and are distributed from polar to tropical regions in both marine (Figure 1) and freshwater (Figure 2) environments. They inhabit both shallow waters as well as depths of more than 2800 m, with shallower sponges tending to be more vibrantly colored than their deeper counterparts. The wide-ranging habitats of sponges have led to size variations from as small as 0.05 mm to more than 3 m in height.2 Their taxonomic phylum, Porifera (meaning pore bearers), is derived from the millions of pores lining the surface of the sponge that are used to filter planktonic organisms.3 Flagellated epithelioid cells called choanocytes line the internal chambers of sponges, creating a water current that promotes filter feeding as well as nutrient absorption across their microvilli.4 The body walls of many sponges consist of a collagenous skeleton made up of spongin and spicules of silicon dioxide (silica) or calcium carbonate embedded in the spongin connective tissue matrix.5 Bath sponges lack silica spicules.

Figure 1. Marine sponges. A, Tedania ignis (fire sponge). Photograph courtesy of Vidal Haddad Jr, MD, PhD (Botucatu, São Paulo, Brazil). B, Agelas conifera (brown tube sponge). Photograph courtesy of Dirk M. Elston, MD (Charleston, South Carolina).

Figure 2. Cauxi sponge, a type of freshwater sponge. Photograph courtesy of Vidal Haddad Jr, MD, PhD (Botucatu, São Paulo, Brazil).

Sponges have been used in medicine for centuries. The first use in Western culture was recorded in 405 bce in The Frogs, a comedy by Aristophanes in which a sponge was placed on a character’s heart following a syncopal episode. Additionally, in many Hippocratic writings, the use of sponges is outlined in the treatment of a variety of ailments. Similarly, the ancient Chinese and Greeks used burnt sponge and seaweed as a source of iodine to treat goiters.6,7 Modern research focuses on the use of sponge metabolites for their antineoplastic, antimicrobial, and anti-inflammatory effects.8 Identification of spongouridine and spongothymidine from the sponge Tectitethya crypta led to the development of cytarabine and gemcitabine8 as well as the discovery of the antiviral agent vidarabine.9 The monoclonal antibody assay for the detection of shellfish poisoning was prepared using the sponge Halichondria okadai.10

Mechanisms and Symptoms of Injury

Bathing sponges (silk sponges) derived from Spongia officinalis are harmless. Other sponges can exert their damaging effects through a variety of mechanisms that lead to dermatologic manifestations (eTable). Some species of sponges produce and secrete toxic metabolites (eg, crinotoxins) onto the body surface or into the surrounding water. They also are capable of synthesizing a mucous slime that can be irritating to human skin. Direct trauma also can be caused by fragments of the silica or calcium carbonate sponge skeleton penetrating the skin. Stinging members of the phylum Cnidaria can colonize the sponge, leading to injury when a human handles the sponge.25-27

Sponge dermatitis can be divided into 2 major categories: an initial pruritic dermatitis (Figure 3) that occurs within 20 minutes to a few hours after contact and a delayed irritant dermatitis caused by penetration of the spicules and chemical agents into skin.28 Importantly, different species can lead to varying manifestations.

Figure 3. Initial pruritic eczematous plaques with erythema and edema after handling a toxic marine sponge. Photograph courtesy of Vidal Haddad Jr, MD, PhD (Botucatu, São Paulo, Brazil).


The initial pruritic dermatitis is characterized by itching and burning that progresses to local edema, vesiculation, joint swelling, and stiffness. Because most contact with sponges occurs with handling, joint immobility may ensue within 24 hours of the encounter. Rarely, larger areas of the skin are affected, and fever, chills, malaise, dizziness, nausea, purulent bullae, muscle cramps, and formication may occur.28 Anaphylactic reactions have been described in a small subset of patients. There have even been reports of delayed (ie, 1–2 weeks following exposure) erythema multiforme, livedo reticularis, purpura, and dyshidrotic eczema.16,20,29 The irritant dermatitis caused by spicule trauma is due to a foreign body reaction that can be exacerbated by toxins entering the skin. In severe cases, desquamation, recurrent eczema, and arthralgia can occur.30 In general, more mild cases should self-resolve within 3 to 7 days. Dermatologic conditions also can be caused by organisms that inhabit sponges and as a result produce a dermatitis when the sponge is handled, including sponge divers disease (maladie des plongeurs), a necrotic dermatitis caused by stinging Cnidaria species.31 Dogger Bank itch, first described as a dermatitis caused by sensitization to (2-hydroxyethyl) dimethylsulfoxonium chloride, initially was isolated from the sea chervil (a type of Bryozoan); however, that same chemical also was later found in sponges, producing the same dermatitis after handling the sponge.32 Freshwater sponges also have been reported to be injurious and exist worldwide. In contrast to marine sponges, lesions from freshwater sponges are disseminated pruritic erythematous papules with ulcerations, crusts, and secondary infections.22 The disseminated nature of the dermatitis caused by freshwater sponges is due to contact with the spicules of dead sponges that are dispersed throughout the water rather than from direct handling. Sponge dermatitis occurs mostly in sponge collectors, divers, trawlers, and biology students and has been reported extensively in the United States, Caribbean Islands, Australia, New Zealand, and Brazil.18,27,33,34

Management

Treatment should consist of an initial decontamination; the skin should be dried, and adhesive tape or rubber cement should be utilized to remove any spicules embedded in the skin. Diluted vinegar soaks should be initiated for 10 to 30 minutes on the affected area(s) 3 or 4 times daily.19 The initial decontamination should occur immediately, as delay may lead to persistent purulent bullae that may take months to heal. Topical steroids may be used following the initial decontamination to help relieve inflammation. Antihistamines and nonsteroidal anti-inflammatory drugs may be used to alleviate pruritus and pain, respectively. Severe cases may require systemic glucocorticoids. Additionally, immunization status against tetanus toxoid should be assessed.35 In the event of an anaphylactic reaction, it is important to maintain a patent airway and normalized blood pressure through the use of intramuscular epinephrine.36 Frequent follow-up is warranted, as serious secondary infections can develop.37 Patients also should be counseled on the potential for delayed dermatologic reactions, including erythema multiforme. Contact between humans and coastal environments has been increasing in the last few decades; therefore, an increase in contact with sponges is to be expected.22

Sponges are among the oldest animals on earth, appearing more than 640 million years ago before the Cambrian explosion, a period when most major animal phyla appeared in the fossil records.1 More than 10,000 species of sponges have been identified worldwide and are distributed from polar to tropical regions in both marine (Figure 1) and freshwater (Figure 2) environments. They inhabit both shallow waters as well as depths of more than 2800 m, with shallower sponges tending to be more vibrantly colored than their deeper counterparts. The wide-ranging habitats of sponges have led to size variations from as small as 0.05 mm to more than 3 m in height.2 Their taxonomic phylum, Porifera (meaning pore bearers), is derived from the millions of pores lining the surface of the sponge that are used to filter planktonic organisms.3 Flagellated epithelioid cells called choanocytes line the internal chambers of sponges, creating a water current that promotes filter feeding as well as nutrient absorption across their microvilli.4 The body walls of many sponges consist of a collagenous skeleton made up of spongin and spicules of silicon dioxide (silica) or calcium carbonate embedded in the spongin connective tissue matrix.5 Bath sponges lack silica spicules.

Figure 1. Marine sponges. A, Tedania ignis (fire sponge). Photograph courtesy of Vidal Haddad Jr, MD, PhD (Botucatu, São Paulo, Brazil). B, Agelas conifera (brown tube sponge). Photograph courtesy of Dirk M. Elston, MD (Charleston, South Carolina).

Figure 2. Cauxi sponge, a type of freshwater sponge. Photograph courtesy of Vidal Haddad Jr, MD, PhD (Botucatu, São Paulo, Brazil).

Sponges have been used in medicine for centuries. The first use in Western culture was recorded in 405 bce in The Frogs, a comedy by Aristophanes in which a sponge was placed on a character’s heart following a syncopal episode. Additionally, in many Hippocratic writings, the use of sponges is outlined in the treatment of a variety of ailments. Similarly, the ancient Chinese and Greeks used burnt sponge and seaweed as a source of iodine to treat goiters.6,7 Modern research focuses on the use of sponge metabolites for their antineoplastic, antimicrobial, and anti-inflammatory effects.8 Identification of spongouridine and spongothymidine from the sponge Tectitethya crypta led to the development of cytarabine and gemcitabine8 as well as the discovery of the antiviral agent vidarabine.9 The monoclonal antibody assay for the detection of shellfish poisoning was prepared using the sponge Halichondria okadai.10

Mechanisms and Symptoms of Injury

Bathing sponges (silk sponges) derived from Spongia officinalis are harmless. Other sponges can exert their damaging effects through a variety of mechanisms that lead to dermatologic manifestations (eTable). Some species of sponges produce and secrete toxic metabolites (eg, crinotoxins) onto the body surface or into the surrounding water. They also are capable of synthesizing a mucous slime that can be irritating to human skin. Direct trauma also can be caused by fragments of the silica or calcium carbonate sponge skeleton penetrating the skin. Stinging members of the phylum Cnidaria can colonize the sponge, leading to injury when a human handles the sponge.25-27

Sponge dermatitis can be divided into 2 major categories: an initial pruritic dermatitis (Figure 3) that occurs within 20 minutes to a few hours after contact and a delayed irritant dermatitis caused by penetration of the spicules and chemical agents into skin.28 Importantly, different species can lead to varying manifestations.

Figure 3. Initial pruritic eczematous plaques with erythema and edema after handling a toxic marine sponge. Photograph courtesy of Vidal Haddad Jr, MD, PhD (Botucatu, São Paulo, Brazil).


The initial pruritic dermatitis is characterized by itching and burning that progresses to local edema, vesiculation, joint swelling, and stiffness. Because most contact with sponges occurs with handling, joint immobility may ensue within 24 hours of the encounter. Rarely, larger areas of the skin are affected, and fever, chills, malaise, dizziness, nausea, purulent bullae, muscle cramps, and formication may occur.28 Anaphylactic reactions have been described in a small subset of patients. There have even been reports of delayed (ie, 1–2 weeks following exposure) erythema multiforme, livedo reticularis, purpura, and dyshidrotic eczema.16,20,29 The irritant dermatitis caused by spicule trauma is due to a foreign body reaction that can be exacerbated by toxins entering the skin. In severe cases, desquamation, recurrent eczema, and arthralgia can occur.30 In general, more mild cases should self-resolve within 3 to 7 days. Dermatologic conditions also can be caused by organisms that inhabit sponges and as a result produce a dermatitis when the sponge is handled, including sponge divers disease (maladie des plongeurs), a necrotic dermatitis caused by stinging Cnidaria species.31 Dogger Bank itch, first described as a dermatitis caused by sensitization to (2-hydroxyethyl) dimethylsulfoxonium chloride, initially was isolated from the sea chervil (a type of Bryozoan); however, that same chemical also was later found in sponges, producing the same dermatitis after handling the sponge.32 Freshwater sponges also have been reported to be injurious and exist worldwide. In contrast to marine sponges, lesions from freshwater sponges are disseminated pruritic erythematous papules with ulcerations, crusts, and secondary infections.22 The disseminated nature of the dermatitis caused by freshwater sponges is due to contact with the spicules of dead sponges that are dispersed throughout the water rather than from direct handling. Sponge dermatitis occurs mostly in sponge collectors, divers, trawlers, and biology students and has been reported extensively in the United States, Caribbean Islands, Australia, New Zealand, and Brazil.18,27,33,34

Management

Treatment should consist of an initial decontamination; the skin should be dried, and adhesive tape or rubber cement should be utilized to remove any spicules embedded in the skin. Diluted vinegar soaks should be initiated for 10 to 30 minutes on the affected area(s) 3 or 4 times daily.19 The initial decontamination should occur immediately, as delay may lead to persistent purulent bullae that may take months to heal. Topical steroids may be used following the initial decontamination to help relieve inflammation. Antihistamines and nonsteroidal anti-inflammatory drugs may be used to alleviate pruritus and pain, respectively. Severe cases may require systemic glucocorticoids. Additionally, immunization status against tetanus toxoid should be assessed.35 In the event of an anaphylactic reaction, it is important to maintain a patent airway and normalized blood pressure through the use of intramuscular epinephrine.36 Frequent follow-up is warranted, as serious secondary infections can develop.37 Patients also should be counseled on the potential for delayed dermatologic reactions, including erythema multiforme. Contact between humans and coastal environments has been increasing in the last few decades; therefore, an increase in contact with sponges is to be expected.22

References
  1. Gold DA, Grabenstatter J, de Mendoza A, et al. Sterol and genomic analyses validate the sponge biomarker hypothesis. Proc Natl Acad Sci U S A. 2016;113:2684-2689.
  2. Bonamonte D, Filoni A, Verni P, et al. Dermatitis caused by sponges. In: Bonamonte D, Angelini G, eds. Aquatic Dermatology. 2nd ed. Springer; 2016:121-126.
  3. Marsh LM, Slack-Smith S, Gurry DL. Field Guide to Sea Stingers and Other Venomous and Poisonous Marine Invertebrates. 2nd ed. Western Australian Museum; 2010.
  4. Eid E, Al-Tawaha M. A Guide to Harmful and Toxic Creatures in the Gulf of Aqaba Jordan. The Royal Marine Conservation Society of Jordan; 2016.
  5. Reese E, Depenbrock P. Water envenomations and stings. Curr Sports Med Rep. 2014;13:126-131.
  6. Dormandy TL. Trace element analysis of hair. Br Med J (Clin Res Ed). 1986;293:975-976.
  7. Voultsiadou E. Sponges: an historical survey of their knowledge in Greek antiquity. J Mar Biol Assoc UK. 2007;87:1757-1763.
  8. Senthilkumar K, Kim SK. Marine invertebrate natural products for anti-inflammatory and chronic diseases [published online December 31, 2013]. Evid Based Complement Alternat Med. doi:10.1155/2013/572859
  9. Sagar S, Kaur M, Minneman KP. Antiviral lead compounds from marine sponges. Mar Drugs. 2010;8:2619-2638.
  10. Usagawa T, Nishimura M, Itoh Y, et al. Preparation of monoclonal antibodies against okadaic acid prepared from the sponge Halichondria okadai. Toxicon. 1989;27:1323-1330.
  11. Elston DM. Aquatic antagonists: sponge dermatitis. Cutis. 2007;80:279-280.
  12. Parra-Velandia FJ, Zea S, Van Soest RW. Reef sponges of the genus Agelas (Porifera: Demospongiae) from the Greater Caribbean. Zootaxa. 2014;3794:301-343.
  13. Hooper JN, Capon RJ, Hodder RA. A new species of toxic marine sponge (Porifera: Demospongiae: Poecilosclerida) from northwest Australia. The Beagle, Records of the Northern Territory Museum of Arts and sciences. 1991;8:27-36.
  14. Burnett JW, Calton GJ, Morgan RJ. Dermatitis due to stinging sponges. Cutis. 1987;39:476.
  15. Kizer KW. Marine envenomations. J Toxicol Clin Toxicol. 1983;21:527-555.
  16. Isbister GK, Hooper JN. Clinical effects of stings by sponges of the genus Tedania and a review of sponge stings worldwide. Toxicon. 2005;46:782-785.
  17. Fromont J, Abdo DA. New species of Haliclona (Demospongiae: Haplosclerida: Chalinidae) from Western Australia. Zootaxa. 2014;3835:97-109.
  18. Flachsenberger W, Holmes NJ, Leigh C, et al. Properties of the extract and spicules of the dermatitis inducing sponge Neofibularia mordens Hartman. J Toxicol Clin Toxicol. 1987;25:255-272.
  19. Southcott RV, Coulter JR. The effects of the southern Australian marine stinging sponges, Neofibularia mordens and Lissodendoryx sp. Med J Aust. 1971;2:895-901.
  20. Yaffee HS, Stargardter F. Erythema multiforme from Tedania ignis. report of a case and an experimental study of the mechanism of cutaneous irritation from the fire sponge. Arch Dermatol. 1963;87:601-604.
  21. Yaffee HS. Irritation from red sponge. N Engl J Med. 1970;282:51.
  22. Haddad V Jr. Environmental dermatology: skin manifestations of injuries caused by invertebrate aquatic animals. An Bras Dermatol. 2013;88:496-506.
  23. Volkmer-Ribeiro C, Lenzi HL, Orefice F, et al. Freshwater sponge spicules: a new agent of ocular pathology. Mem Inst Oswaldo Cruz. 2006;101:899-903.
  24. Cruz AA, Alencar VM, Medina NH, et al. Dangerous waters: outbreak of eye lesions caused by fresh water sponge spicules. Eye (Lond). 2013;27:398-402.
  25. Haddad V Jr. Clinical and therapeutic aspects of envenomations caused by sponges and jellyfish. In: Gopalakrishnakone P, Haddad V Jr, Kem WR, et al, eds. Marine and Freshwater Toxins. Springer; 2016:317-325.
  26. Haddad V Jr, Lupi O, Lonza JP, et al. Tropical dermatology: marine and aquatic dermatology. J Am Acad Dermatol. 2009;61:733-750.
  27. Gaastra MT. Aquatic skin disorders. In: Faber WR, Hay RJ, Naafs B, eds. Imported Skin Diseases. 2nd ed. Wiley; 2012:283-292.
  28. Auerbach P. Envenomation by aquatic invertebrates. In: Auerbach P, ed. Wilderness Medicine. 6th ed. Elsevier Mosby; 2011;1596-1627.
  29. Sims JK, Irei MY. Human Hawaiian marine sponge poisoning. Hawaii Med J. 1979;38:263-270.
  30. Haddad V Jr. Aquatic animals of medical importance in Brazil. Rev Soc Bras Med Trop. 2003;36:591-597.
  31. Tlougan BE, Podjasek JO, Adams BB. Aquatic sports dermatoses. part 2—in the water: saltwater dermatoses. Int J Dermatol. 2010;49:994-1002.
  32. Warabi K, Nakao Y, Matsunaga S, et al. Dogger Bank itch revisited: isolation of (2-hydroxyethyl) dimethylsulfoxonium chloride as a cytotoxic constituent from the marine sponge Theonella aff. mirabilis. Comp Biochem Physiol B Biochem Mol Biol. 2001;128:27-30.
  33. Southcott R. Human injuries from invertebrate animals in the Australian seas. Clin Toxicol. 1970;3:617-636.
  34. Russell FE. Sponge injury—traumatic, toxic or allergic? N Engl J Med. 1970;282:753-754.
  35. Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337.
  36. Muraro A, Roberts G, Worm M, et al. Anaphylaxis: guidelines from the European Academy of Allergy and Clinical Immunology. Allergy. 2014;69:1026-1045.
  37. Kizer K, Auerbach P, Dwyer B. Marine envenomations: not just a problem of the tropics. Emerg Med Rep. 1985;6:129-135.
References
  1. Gold DA, Grabenstatter J, de Mendoza A, et al. Sterol and genomic analyses validate the sponge biomarker hypothesis. Proc Natl Acad Sci U S A. 2016;113:2684-2689.
  2. Bonamonte D, Filoni A, Verni P, et al. Dermatitis caused by sponges. In: Bonamonte D, Angelini G, eds. Aquatic Dermatology. 2nd ed. Springer; 2016:121-126.
  3. Marsh LM, Slack-Smith S, Gurry DL. Field Guide to Sea Stingers and Other Venomous and Poisonous Marine Invertebrates. 2nd ed. Western Australian Museum; 2010.
  4. Eid E, Al-Tawaha M. A Guide to Harmful and Toxic Creatures in the Gulf of Aqaba Jordan. The Royal Marine Conservation Society of Jordan; 2016.
  5. Reese E, Depenbrock P. Water envenomations and stings. Curr Sports Med Rep. 2014;13:126-131.
  6. Dormandy TL. Trace element analysis of hair. Br Med J (Clin Res Ed). 1986;293:975-976.
  7. Voultsiadou E. Sponges: an historical survey of their knowledge in Greek antiquity. J Mar Biol Assoc UK. 2007;87:1757-1763.
  8. Senthilkumar K, Kim SK. Marine invertebrate natural products for anti-inflammatory and chronic diseases [published online December 31, 2013]. Evid Based Complement Alternat Med. doi:10.1155/2013/572859
  9. Sagar S, Kaur M, Minneman KP. Antiviral lead compounds from marine sponges. Mar Drugs. 2010;8:2619-2638.
  10. Usagawa T, Nishimura M, Itoh Y, et al. Preparation of monoclonal antibodies against okadaic acid prepared from the sponge Halichondria okadai. Toxicon. 1989;27:1323-1330.
  11. Elston DM. Aquatic antagonists: sponge dermatitis. Cutis. 2007;80:279-280.
  12. Parra-Velandia FJ, Zea S, Van Soest RW. Reef sponges of the genus Agelas (Porifera: Demospongiae) from the Greater Caribbean. Zootaxa. 2014;3794:301-343.
  13. Hooper JN, Capon RJ, Hodder RA. A new species of toxic marine sponge (Porifera: Demospongiae: Poecilosclerida) from northwest Australia. The Beagle, Records of the Northern Territory Museum of Arts and sciences. 1991;8:27-36.
  14. Burnett JW, Calton GJ, Morgan RJ. Dermatitis due to stinging sponges. Cutis. 1987;39:476.
  15. Kizer KW. Marine envenomations. J Toxicol Clin Toxicol. 1983;21:527-555.
  16. Isbister GK, Hooper JN. Clinical effects of stings by sponges of the genus Tedania and a review of sponge stings worldwide. Toxicon. 2005;46:782-785.
  17. Fromont J, Abdo DA. New species of Haliclona (Demospongiae: Haplosclerida: Chalinidae) from Western Australia. Zootaxa. 2014;3835:97-109.
  18. Flachsenberger W, Holmes NJ, Leigh C, et al. Properties of the extract and spicules of the dermatitis inducing sponge Neofibularia mordens Hartman. J Toxicol Clin Toxicol. 1987;25:255-272.
  19. Southcott RV, Coulter JR. The effects of the southern Australian marine stinging sponges, Neofibularia mordens and Lissodendoryx sp. Med J Aust. 1971;2:895-901.
  20. Yaffee HS, Stargardter F. Erythema multiforme from Tedania ignis. report of a case and an experimental study of the mechanism of cutaneous irritation from the fire sponge. Arch Dermatol. 1963;87:601-604.
  21. Yaffee HS. Irritation from red sponge. N Engl J Med. 1970;282:51.
  22. Haddad V Jr. Environmental dermatology: skin manifestations of injuries caused by invertebrate aquatic animals. An Bras Dermatol. 2013;88:496-506.
  23. Volkmer-Ribeiro C, Lenzi HL, Orefice F, et al. Freshwater sponge spicules: a new agent of ocular pathology. Mem Inst Oswaldo Cruz. 2006;101:899-903.
  24. Cruz AA, Alencar VM, Medina NH, et al. Dangerous waters: outbreak of eye lesions caused by fresh water sponge spicules. Eye (Lond). 2013;27:398-402.
  25. Haddad V Jr. Clinical and therapeutic aspects of envenomations caused by sponges and jellyfish. In: Gopalakrishnakone P, Haddad V Jr, Kem WR, et al, eds. Marine and Freshwater Toxins. Springer; 2016:317-325.
  26. Haddad V Jr, Lupi O, Lonza JP, et al. Tropical dermatology: marine and aquatic dermatology. J Am Acad Dermatol. 2009;61:733-750.
  27. Gaastra MT. Aquatic skin disorders. In: Faber WR, Hay RJ, Naafs B, eds. Imported Skin Diseases. 2nd ed. Wiley; 2012:283-292.
  28. Auerbach P. Envenomation by aquatic invertebrates. In: Auerbach P, ed. Wilderness Medicine. 6th ed. Elsevier Mosby; 2011;1596-1627.
  29. Sims JK, Irei MY. Human Hawaiian marine sponge poisoning. Hawaii Med J. 1979;38:263-270.
  30. Haddad V Jr. Aquatic animals of medical importance in Brazil. Rev Soc Bras Med Trop. 2003;36:591-597.
  31. Tlougan BE, Podjasek JO, Adams BB. Aquatic sports dermatoses. part 2—in the water: saltwater dermatoses. Int J Dermatol. 2010;49:994-1002.
  32. Warabi K, Nakao Y, Matsunaga S, et al. Dogger Bank itch revisited: isolation of (2-hydroxyethyl) dimethylsulfoxonium chloride as a cytotoxic constituent from the marine sponge Theonella aff. mirabilis. Comp Biochem Physiol B Biochem Mol Biol. 2001;128:27-30.
  33. Southcott R. Human injuries from invertebrate animals in the Australian seas. Clin Toxicol. 1970;3:617-636.
  34. Russell FE. Sponge injury—traumatic, toxic or allergic? N Engl J Med. 1970;282:753-754.
  35. Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337.
  36. Muraro A, Roberts G, Worm M, et al. Anaphylaxis: guidelines from the European Academy of Allergy and Clinical Immunology. Allergy. 2014;69:1026-1045.
  37. Kizer K, Auerbach P, Dwyer B. Marine envenomations: not just a problem of the tropics. Emerg Med Rep. 1985;6:129-135.
Issue
Cutis - 107(1)
Issue
Cutis - 107(1)
Page Number
34-36, E5
Page Number
34-36, E5
Publications
Publications
Topics
Article Type
Sections
Inside the Article

Practice Points

  • Sponges exist in both marine and freshwater environments throughout the world.
  • Immediate management of sponge dermatitis should include decontamination by removing the sponge spicules with tape or rubber cement followed by dilute vinegar soaks.
  • Topical steroids may be used only after initial decontamination. Use of oral steroids may be needed for more severe reactions.
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
Article PDF Media

What’s Eating You? Human Flea (Pulex irritans)

Article Type
Changed
Thu, 06/03/2021 - 15:49
Display Headline
What’s Eating You? Human Flea (Pulex irritans)

 

Characteristics

The ubiquitous human flea, Pulex irritans, is a hematophagous wingless ectoparasite in the order Siphonaptera (wingless siphon) that survives by consuming the blood of its mammalian and avian hosts. Due to diseases such as the bubonic plague, fleas have claimed more victims than all the wars ever fought; in the 14th century, the Black Death caused more than 200 million deaths. Fleas fossilized in amber have been found to be 200 million years old and closely resemble the modern human flea, demonstrating the resilience of the species.

The adult human flea is a small, reddish brown, laterally compressed, wingless insect that is approximately 2- to 3.5-mm long (females, 2.5–3.5 mm; males, 2–2.5 mm) and enclosed by a tough cuticle. Compared to the dog flea (Ctenocephalides canis) and cat flea (Ctenocephalides felis), P irritans has no combs or ctenidia (Figure 1). Fleas have large powerful hind legs enabling them to jump horizontally or vertically 200 times their body length (equivalent to a 6-foot human jumping 1200 feet) using stored muscle energy in a pad on the hind legs composed of the elastic protein resilin.1 They feed off a wide variety of hosts, including humans, pigs, cats, dogs, goats, sheep, cattle, chickens, owls, foxes, rabbits, mice, and feral cats. The flea’s mouthparts are highly specialized for piercing the skin and sucking its blood meal via direct capillary cannulation.

Figure 1. Pulex irritans anatomy. A reddish brown flea lacking characteristic features from most other flea species including a comb and pleural rod.

Life Cycle

There are 4 stages of the flea life cycle: egg, larva, pupa, and adult. Most adult flea species mate on the host; the female will lay an average of 4 to 8 small white eggs on the host after each blood meal, laying more than 400 eggs during her lifetime. The eggs then drop from the host and hatch in approximately 4 to 6 days to become larvae. The active larvae feed on available organic matter in their environment, such as their parents’ feces and detritus, while undergoing 3 molts within 1 week to several months.2 The larva then spins a silken cocoon from modified salivary glands to form the pupa. In favorable conditions, the pupa lasts only a few weeks; however, it can last for a year or more in unfavorable conditions. Triggers for emergence of the adult flea from the pupa include high humidity, warm temperatures, increased levels of carbon dioxide, and vibrations including sound. An adult P irritans flea can live for a few weeks to more than 1.5 years in favorable conditions of lower air temperature, high relative humidity, and access to a host.3

Related Diseases

Pulex irritans can be a vector for several human diseases. Yersinia pestis is a gram-negative bacteria that causes plague, a highly virulent disease that killed millions of people during its 3 largest human pandemics. The black rat (Rattus rattus) and the oriental rat flea (Xenopsylla cheopis) have been implicated as initial vectors; however, transmission may be human-to-human with pneumonic plague, and septicemic plague may be spread via Pulex fleas or body lice.4,5 In 1971, Y pestis was isolated from P irritans on a dog in the home of a plague patient in Kayenta, Arizona.6Yersinia pestis bacterial DNA also was extracted from P irritans during a plague outbreak in Madagascar in 20147 and was implicated in epidemiologic studies of plague in Tanzania from 1986 to 2004, suggesting it also plays a role in endemic disease.8

Bartonellosis is an emerging disease caused by different species of the gram-negative intracellular bacteria of the genus Bartonella transmitted by lice, ticks, and fleas. Bartonella quintana causes trench fever primarily transmitted by the human body louse, Pediculus humanus corporis, and resulted in more than 1 million cases during World War I. Trench fever is characterized by headache, fever, dizziness, and shin pain that lasts 1 to 3 days and recurs in cycles every 4 to 6 days. Other clinical manifestations of B quintana include chronic bacteremia, endocarditis, lymphadenopathy, and bacillary angiomatosis.9Bartonella henselae causes cat scratch fever, characterized by lymphadenopathy, fever, headache, joint pain, and lethargy from infected cat scratches or the bite of an infected flea. Bartonella rochalimae also has been found to cause a trench fever–like bacteremia.10Bartonella species have been found in P irritans, and the flea is implicated as a vector of bartonellosis in humans.11-15



Rickettsioses are worldwide diseases caused by the gram-negative intracellular bacteria of the genus Rickettsia transmitted to humans via hematophagous arthropods. The rickettsiae traditionally have been classified into the spotted fever or typhus groups. The spotted fever group (ie, Rocky Mountain spotted fever, Mediterranean spotted fever) is transmitted via ticks. The typhus group is transmitted via lice (epidemic typhus) and fleas (endemic or murine typhus). Murine typhus can be caused by Rickettsia typhi in warm coastal areas around the world where the main mammal reservoir is the rat and the rat flea vector X cheopis. Clinical signs of infection are abrupt onset of fever, headaches, myalgia, malaise, and chills, with a truncal maculopapular rash progressing peripherally several days after the initial clinical signs. Rash is present in up to 50% of cases.16Rickettsia felis is an emerging flea-borne pathogen causing an acute febrile illness usually transmitted via the cat flea C felis.17Rickettsia species DNA have been found to be present in P irritans from dogs18 and livestock19 and pose a risk for causing rickettsioses in humans.

Environmental Treatment and Prevention

Flea bites present as intense, pruritic, urticarial to vesicular papules that usually are located on the lower extremities but also can be present on exposed areas of the upper extremities and hands (Figure 2). Human fleas infest clothing, and bites can be widespread. Topical antipruritics and corticosteroids can be used for controlling itch and the intense cutaneous inflammatory response. The flea host should be identified in areas of the home, school, farm, work, or local environment. House pets should be examined and treated by a veterinarian. The pet’s bedding should be washed and dried at high temperatures, and carpets and floors should be routinely vacuumed or cleaned to remove eggs, larvae, flea feces, and/or pupae. The killing of adult fleas with insecticidal products (eg, imidacloprid, fipronil, spinosad, selamectin, lufenuron, ivermectin) is the primary method of flea control. Use of insect growth regulators such as pyriproxyfen inhibits adult reproduction and blocks the organogenesis of immature larval stages via hormonal or enzymatic actions.20 The combination of an insecticide and an insect growth regulator appears to be most effective in their synergistic actions against adult fleas and larvae. There have been reports of insecticidal resistance in the flea population, especially with pyrethroids.21,22 A professional exterminator and veterinarian should be consulted. In recalcitrant cases, evaluation for other wild mammals or birds should be performed in unoccupied areas of the home such as the attic, crawl spaces, and basements, as well as inside walls.

Figure 2. Vesicular papules on an exposed area of the arm from flea bites (Pulex irritans).


 

Conclusion

The human flea, P irritans, is an important vector in the transmission of human diseases such as the bubonic plague, bartonellosis, and rickettsioses. Flea bites present as intensely pruritic, urticarial to vesicular papules that most commonly present on the lower extremities. Flea bites can be treated with topical steroids, and fleas can be controlled by a combination of insecticidal products and insect growth regulators.

References
  1. Burrow M. How fleas jump. J Exp Biol. 2009;18:2881-2883.
  2. Buckland PC, Sandler JP. A biogeography of the human flea, Pulex irritans L (Siphonaptera: Pulicidae). J Biogeogr. 1989;16:115-120.
  3. Krasnov BR. Life cycles. In: Krasnov BR, ed. Functional and Evolutional Ecology of Fleas. Cambridge, MA: Cambridge Univ Press; 2008:45-67.
  4. Dean KR, Krauer F, Walloe L, et al. Human ectoparasites and the spread of plague in Europe during the second pandemic. Proc Natl Acad Sci U S A. 2018;115:1304-1309.
  5. Hufthammer AK, Walloe L. Rats cannot have been intermediate hosts for Yersinia pestis during medieval plague epidemics in Northern Europe. J Archeol Sci. 2013;40:1752-1759.
  6. Archibald WS, Kunitz SJ. Detection of plague by testing serums of dogs on the Navajo Reservation. HSMHA Health Rep. 1971;86:377-380.
  7. Ratovonjato J, Rajerison M, Rahelinirina S, et al. Yersinia pestis in Pulex irritans fleas during plague outbreak, Madagascar. Emerg Infect Dis. 2014;20:1414-1415.
  8. Laudisoit A, Leirs H, Makundi RH, et al. Plague and the human flea, Tanzania. Emerg Infect Dis. 2007;13:687-693.
  9. Foucault C, Brouqui P, Raoult D. Bartonella quintana characteristics and clinical management. Emerg Infect Dis. 2006;12:217-223.
  10. Eremeeva ME, Gerns HL, Lydy SL, et al. Bacteremia, fever, and splenomegaly caused by a newly recognized bartonella species. N Engl J Med. 2007; 356:2381-2387.11.
  11. Marquez FJ, Millan J, Rodriguez-Liebana JJ, et al. Detection and identification of Bartonella sp. in fleas from carnivorous mammals in Andalusia, Spain. Med Vet Entomol. 2009;23:393-398.
  12. Perez-Martinez L, Venzal JM, Portillo A, et al. Bartonella rochalimae and other Bartonella spp. in fleas, Chile. Emerg Infect Dis. 2009;15:1150-1152.
  13. Sofer S, Gutierrez DM, Mumcuoglu KY, et al. Molecular detection of zoonotic bartonellae (B. henselae, B. elizabethae and B. rochalimae) in fleas collected from dogs in Israel. Med Vet Entomol. 2015;29:344-348.
  14. Zouari S, Khrouf F, M’ghirbi Y, et al. First molecular detection and characterization of zoonotic Bartonella species in fleas infesting domestic animals in Tunisia. Parasit Vectors. 2017;10:436.
  15. Rolain JM, Bourry, O, Davoust B, et al. Bartonella quintana and Rickettsia felis in Gabon. Emerg Infect Dis. 2005;11:1742-1744.
  16. Tsioutis C, Zafeiri M, Avramopoulos A, et al. Clinical and laboratory characteristics, epidemiology, and outcomes of murine typhus: a systematic review. Acta Trop. 2017;166:16-24.
  17. Brown L, Macaluso KR. Rickettsia felis, an emerging flea-borne rickettsiosis. Curr Trop Med Rep. 2016;3:27-39.
  18. Oteo JA, Portillo A, Potero F, et al. ‘Candidatus Rickettsia asemboensis’ and Wolbachia spp. in Ctenocephalides felis and Pulex irritans fleas removed from dogs in Ecuador. Parasit Vectors. 2014;7:455.
  19. Ghavami MB, Mirzadeh H, Mohammadi J, et al. Molecular survey of ITS spacer and Rickettsia infection in human flea, Pulex irritans. Parasitol Res. 2018;117:1433-1442.
  20. Traversa D. Fleas infesting pets in the era of emerging extra-intestinal nematodes. Parasit Vectors. 2013;6:59.
  21. Rust MK. Insecticide resistance in fleas. Insects. 2016;7:10.
  22. Ghavami MB, Haghi FP, Alibabaei Z, et al. First report of target site insensitivity to pyrethroids in human flea, Pulex irritans (Siphonaptera: Pulicidae). Pest Biochem Physiol. 2018;146:97-105.
Article PDF
Author and Disclosure Information

Ms. O’Donnell is from Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

The images are in the public domain.

Correspondence: Megan O’Donnell, BS, 1025 Walnut St #100, Philadelphia, PA 19107 (mco003@jefferson.edu).

Issue
Cutis - 106(5)
Publications
Topics
Page Number
233-235
Sections
Author and Disclosure Information

Ms. O’Donnell is from Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

The images are in the public domain.

Correspondence: Megan O’Donnell, BS, 1025 Walnut St #100, Philadelphia, PA 19107 (mco003@jefferson.edu).

Author and Disclosure Information

Ms. O’Donnell is from Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

The images are in the public domain.

Correspondence: Megan O’Donnell, BS, 1025 Walnut St #100, Philadelphia, PA 19107 (mco003@jefferson.edu).

Article PDF
Article PDF

 

Characteristics

The ubiquitous human flea, Pulex irritans, is a hematophagous wingless ectoparasite in the order Siphonaptera (wingless siphon) that survives by consuming the blood of its mammalian and avian hosts. Due to diseases such as the bubonic plague, fleas have claimed more victims than all the wars ever fought; in the 14th century, the Black Death caused more than 200 million deaths. Fleas fossilized in amber have been found to be 200 million years old and closely resemble the modern human flea, demonstrating the resilience of the species.

The adult human flea is a small, reddish brown, laterally compressed, wingless insect that is approximately 2- to 3.5-mm long (females, 2.5–3.5 mm; males, 2–2.5 mm) and enclosed by a tough cuticle. Compared to the dog flea (Ctenocephalides canis) and cat flea (Ctenocephalides felis), P irritans has no combs or ctenidia (Figure 1). Fleas have large powerful hind legs enabling them to jump horizontally or vertically 200 times their body length (equivalent to a 6-foot human jumping 1200 feet) using stored muscle energy in a pad on the hind legs composed of the elastic protein resilin.1 They feed off a wide variety of hosts, including humans, pigs, cats, dogs, goats, sheep, cattle, chickens, owls, foxes, rabbits, mice, and feral cats. The flea’s mouthparts are highly specialized for piercing the skin and sucking its blood meal via direct capillary cannulation.

Figure 1. Pulex irritans anatomy. A reddish brown flea lacking characteristic features from most other flea species including a comb and pleural rod.

Life Cycle

There are 4 stages of the flea life cycle: egg, larva, pupa, and adult. Most adult flea species mate on the host; the female will lay an average of 4 to 8 small white eggs on the host after each blood meal, laying more than 400 eggs during her lifetime. The eggs then drop from the host and hatch in approximately 4 to 6 days to become larvae. The active larvae feed on available organic matter in their environment, such as their parents’ feces and detritus, while undergoing 3 molts within 1 week to several months.2 The larva then spins a silken cocoon from modified salivary glands to form the pupa. In favorable conditions, the pupa lasts only a few weeks; however, it can last for a year or more in unfavorable conditions. Triggers for emergence of the adult flea from the pupa include high humidity, warm temperatures, increased levels of carbon dioxide, and vibrations including sound. An adult P irritans flea can live for a few weeks to more than 1.5 years in favorable conditions of lower air temperature, high relative humidity, and access to a host.3

Related Diseases

Pulex irritans can be a vector for several human diseases. Yersinia pestis is a gram-negative bacteria that causes plague, a highly virulent disease that killed millions of people during its 3 largest human pandemics. The black rat (Rattus rattus) and the oriental rat flea (Xenopsylla cheopis) have been implicated as initial vectors; however, transmission may be human-to-human with pneumonic plague, and septicemic plague may be spread via Pulex fleas or body lice.4,5 In 1971, Y pestis was isolated from P irritans on a dog in the home of a plague patient in Kayenta, Arizona.6Yersinia pestis bacterial DNA also was extracted from P irritans during a plague outbreak in Madagascar in 20147 and was implicated in epidemiologic studies of plague in Tanzania from 1986 to 2004, suggesting it also plays a role in endemic disease.8

Bartonellosis is an emerging disease caused by different species of the gram-negative intracellular bacteria of the genus Bartonella transmitted by lice, ticks, and fleas. Bartonella quintana causes trench fever primarily transmitted by the human body louse, Pediculus humanus corporis, and resulted in more than 1 million cases during World War I. Trench fever is characterized by headache, fever, dizziness, and shin pain that lasts 1 to 3 days and recurs in cycles every 4 to 6 days. Other clinical manifestations of B quintana include chronic bacteremia, endocarditis, lymphadenopathy, and bacillary angiomatosis.9Bartonella henselae causes cat scratch fever, characterized by lymphadenopathy, fever, headache, joint pain, and lethargy from infected cat scratches or the bite of an infected flea. Bartonella rochalimae also has been found to cause a trench fever–like bacteremia.10Bartonella species have been found in P irritans, and the flea is implicated as a vector of bartonellosis in humans.11-15



Rickettsioses are worldwide diseases caused by the gram-negative intracellular bacteria of the genus Rickettsia transmitted to humans via hematophagous arthropods. The rickettsiae traditionally have been classified into the spotted fever or typhus groups. The spotted fever group (ie, Rocky Mountain spotted fever, Mediterranean spotted fever) is transmitted via ticks. The typhus group is transmitted via lice (epidemic typhus) and fleas (endemic or murine typhus). Murine typhus can be caused by Rickettsia typhi in warm coastal areas around the world where the main mammal reservoir is the rat and the rat flea vector X cheopis. Clinical signs of infection are abrupt onset of fever, headaches, myalgia, malaise, and chills, with a truncal maculopapular rash progressing peripherally several days after the initial clinical signs. Rash is present in up to 50% of cases.16Rickettsia felis is an emerging flea-borne pathogen causing an acute febrile illness usually transmitted via the cat flea C felis.17Rickettsia species DNA have been found to be present in P irritans from dogs18 and livestock19 and pose a risk for causing rickettsioses in humans.

Environmental Treatment and Prevention

Flea bites present as intense, pruritic, urticarial to vesicular papules that usually are located on the lower extremities but also can be present on exposed areas of the upper extremities and hands (Figure 2). Human fleas infest clothing, and bites can be widespread. Topical antipruritics and corticosteroids can be used for controlling itch and the intense cutaneous inflammatory response. The flea host should be identified in areas of the home, school, farm, work, or local environment. House pets should be examined and treated by a veterinarian. The pet’s bedding should be washed and dried at high temperatures, and carpets and floors should be routinely vacuumed or cleaned to remove eggs, larvae, flea feces, and/or pupae. The killing of adult fleas with insecticidal products (eg, imidacloprid, fipronil, spinosad, selamectin, lufenuron, ivermectin) is the primary method of flea control. Use of insect growth regulators such as pyriproxyfen inhibits adult reproduction and blocks the organogenesis of immature larval stages via hormonal or enzymatic actions.20 The combination of an insecticide and an insect growth regulator appears to be most effective in their synergistic actions against adult fleas and larvae. There have been reports of insecticidal resistance in the flea population, especially with pyrethroids.21,22 A professional exterminator and veterinarian should be consulted. In recalcitrant cases, evaluation for other wild mammals or birds should be performed in unoccupied areas of the home such as the attic, crawl spaces, and basements, as well as inside walls.

Figure 2. Vesicular papules on an exposed area of the arm from flea bites (Pulex irritans).


 

Conclusion

The human flea, P irritans, is an important vector in the transmission of human diseases such as the bubonic plague, bartonellosis, and rickettsioses. Flea bites present as intensely pruritic, urticarial to vesicular papules that most commonly present on the lower extremities. Flea bites can be treated with topical steroids, and fleas can be controlled by a combination of insecticidal products and insect growth regulators.

 

Characteristics

The ubiquitous human flea, Pulex irritans, is a hematophagous wingless ectoparasite in the order Siphonaptera (wingless siphon) that survives by consuming the blood of its mammalian and avian hosts. Due to diseases such as the bubonic plague, fleas have claimed more victims than all the wars ever fought; in the 14th century, the Black Death caused more than 200 million deaths. Fleas fossilized in amber have been found to be 200 million years old and closely resemble the modern human flea, demonstrating the resilience of the species.

The adult human flea is a small, reddish brown, laterally compressed, wingless insect that is approximately 2- to 3.5-mm long (females, 2.5–3.5 mm; males, 2–2.5 mm) and enclosed by a tough cuticle. Compared to the dog flea (Ctenocephalides canis) and cat flea (Ctenocephalides felis), P irritans has no combs or ctenidia (Figure 1). Fleas have large powerful hind legs enabling them to jump horizontally or vertically 200 times their body length (equivalent to a 6-foot human jumping 1200 feet) using stored muscle energy in a pad on the hind legs composed of the elastic protein resilin.1 They feed off a wide variety of hosts, including humans, pigs, cats, dogs, goats, sheep, cattle, chickens, owls, foxes, rabbits, mice, and feral cats. The flea’s mouthparts are highly specialized for piercing the skin and sucking its blood meal via direct capillary cannulation.

Figure 1. Pulex irritans anatomy. A reddish brown flea lacking characteristic features from most other flea species including a comb and pleural rod.

Life Cycle

There are 4 stages of the flea life cycle: egg, larva, pupa, and adult. Most adult flea species mate on the host; the female will lay an average of 4 to 8 small white eggs on the host after each blood meal, laying more than 400 eggs during her lifetime. The eggs then drop from the host and hatch in approximately 4 to 6 days to become larvae. The active larvae feed on available organic matter in their environment, such as their parents’ feces and detritus, while undergoing 3 molts within 1 week to several months.2 The larva then spins a silken cocoon from modified salivary glands to form the pupa. In favorable conditions, the pupa lasts only a few weeks; however, it can last for a year or more in unfavorable conditions. Triggers for emergence of the adult flea from the pupa include high humidity, warm temperatures, increased levels of carbon dioxide, and vibrations including sound. An adult P irritans flea can live for a few weeks to more than 1.5 years in favorable conditions of lower air temperature, high relative humidity, and access to a host.3

Related Diseases

Pulex irritans can be a vector for several human diseases. Yersinia pestis is a gram-negative bacteria that causes plague, a highly virulent disease that killed millions of people during its 3 largest human pandemics. The black rat (Rattus rattus) and the oriental rat flea (Xenopsylla cheopis) have been implicated as initial vectors; however, transmission may be human-to-human with pneumonic plague, and septicemic plague may be spread via Pulex fleas or body lice.4,5 In 1971, Y pestis was isolated from P irritans on a dog in the home of a plague patient in Kayenta, Arizona.6Yersinia pestis bacterial DNA also was extracted from P irritans during a plague outbreak in Madagascar in 20147 and was implicated in epidemiologic studies of plague in Tanzania from 1986 to 2004, suggesting it also plays a role in endemic disease.8

Bartonellosis is an emerging disease caused by different species of the gram-negative intracellular bacteria of the genus Bartonella transmitted by lice, ticks, and fleas. Bartonella quintana causes trench fever primarily transmitted by the human body louse, Pediculus humanus corporis, and resulted in more than 1 million cases during World War I. Trench fever is characterized by headache, fever, dizziness, and shin pain that lasts 1 to 3 days and recurs in cycles every 4 to 6 days. Other clinical manifestations of B quintana include chronic bacteremia, endocarditis, lymphadenopathy, and bacillary angiomatosis.9Bartonella henselae causes cat scratch fever, characterized by lymphadenopathy, fever, headache, joint pain, and lethargy from infected cat scratches or the bite of an infected flea. Bartonella rochalimae also has been found to cause a trench fever–like bacteremia.10Bartonella species have been found in P irritans, and the flea is implicated as a vector of bartonellosis in humans.11-15



Rickettsioses are worldwide diseases caused by the gram-negative intracellular bacteria of the genus Rickettsia transmitted to humans via hematophagous arthropods. The rickettsiae traditionally have been classified into the spotted fever or typhus groups. The spotted fever group (ie, Rocky Mountain spotted fever, Mediterranean spotted fever) is transmitted via ticks. The typhus group is transmitted via lice (epidemic typhus) and fleas (endemic or murine typhus). Murine typhus can be caused by Rickettsia typhi in warm coastal areas around the world where the main mammal reservoir is the rat and the rat flea vector X cheopis. Clinical signs of infection are abrupt onset of fever, headaches, myalgia, malaise, and chills, with a truncal maculopapular rash progressing peripherally several days after the initial clinical signs. Rash is present in up to 50% of cases.16Rickettsia felis is an emerging flea-borne pathogen causing an acute febrile illness usually transmitted via the cat flea C felis.17Rickettsia species DNA have been found to be present in P irritans from dogs18 and livestock19 and pose a risk for causing rickettsioses in humans.

Environmental Treatment and Prevention

Flea bites present as intense, pruritic, urticarial to vesicular papules that usually are located on the lower extremities but also can be present on exposed areas of the upper extremities and hands (Figure 2). Human fleas infest clothing, and bites can be widespread. Topical antipruritics and corticosteroids can be used for controlling itch and the intense cutaneous inflammatory response. The flea host should be identified in areas of the home, school, farm, work, or local environment. House pets should be examined and treated by a veterinarian. The pet’s bedding should be washed and dried at high temperatures, and carpets and floors should be routinely vacuumed or cleaned to remove eggs, larvae, flea feces, and/or pupae. The killing of adult fleas with insecticidal products (eg, imidacloprid, fipronil, spinosad, selamectin, lufenuron, ivermectin) is the primary method of flea control. Use of insect growth regulators such as pyriproxyfen inhibits adult reproduction and blocks the organogenesis of immature larval stages via hormonal or enzymatic actions.20 The combination of an insecticide and an insect growth regulator appears to be most effective in their synergistic actions against adult fleas and larvae. There have been reports of insecticidal resistance in the flea population, especially with pyrethroids.21,22 A professional exterminator and veterinarian should be consulted. In recalcitrant cases, evaluation for other wild mammals or birds should be performed in unoccupied areas of the home such as the attic, crawl spaces, and basements, as well as inside walls.

Figure 2. Vesicular papules on an exposed area of the arm from flea bites (Pulex irritans).


 

Conclusion

The human flea, P irritans, is an important vector in the transmission of human diseases such as the bubonic plague, bartonellosis, and rickettsioses. Flea bites present as intensely pruritic, urticarial to vesicular papules that most commonly present on the lower extremities. Flea bites can be treated with topical steroids, and fleas can be controlled by a combination of insecticidal products and insect growth regulators.

References
  1. Burrow M. How fleas jump. J Exp Biol. 2009;18:2881-2883.
  2. Buckland PC, Sandler JP. A biogeography of the human flea, Pulex irritans L (Siphonaptera: Pulicidae). J Biogeogr. 1989;16:115-120.
  3. Krasnov BR. Life cycles. In: Krasnov BR, ed. Functional and Evolutional Ecology of Fleas. Cambridge, MA: Cambridge Univ Press; 2008:45-67.
  4. Dean KR, Krauer F, Walloe L, et al. Human ectoparasites and the spread of plague in Europe during the second pandemic. Proc Natl Acad Sci U S A. 2018;115:1304-1309.
  5. Hufthammer AK, Walloe L. Rats cannot have been intermediate hosts for Yersinia pestis during medieval plague epidemics in Northern Europe. J Archeol Sci. 2013;40:1752-1759.
  6. Archibald WS, Kunitz SJ. Detection of plague by testing serums of dogs on the Navajo Reservation. HSMHA Health Rep. 1971;86:377-380.
  7. Ratovonjato J, Rajerison M, Rahelinirina S, et al. Yersinia pestis in Pulex irritans fleas during plague outbreak, Madagascar. Emerg Infect Dis. 2014;20:1414-1415.
  8. Laudisoit A, Leirs H, Makundi RH, et al. Plague and the human flea, Tanzania. Emerg Infect Dis. 2007;13:687-693.
  9. Foucault C, Brouqui P, Raoult D. Bartonella quintana characteristics and clinical management. Emerg Infect Dis. 2006;12:217-223.
  10. Eremeeva ME, Gerns HL, Lydy SL, et al. Bacteremia, fever, and splenomegaly caused by a newly recognized bartonella species. N Engl J Med. 2007; 356:2381-2387.11.
  11. Marquez FJ, Millan J, Rodriguez-Liebana JJ, et al. Detection and identification of Bartonella sp. in fleas from carnivorous mammals in Andalusia, Spain. Med Vet Entomol. 2009;23:393-398.
  12. Perez-Martinez L, Venzal JM, Portillo A, et al. Bartonella rochalimae and other Bartonella spp. in fleas, Chile. Emerg Infect Dis. 2009;15:1150-1152.
  13. Sofer S, Gutierrez DM, Mumcuoglu KY, et al. Molecular detection of zoonotic bartonellae (B. henselae, B. elizabethae and B. rochalimae) in fleas collected from dogs in Israel. Med Vet Entomol. 2015;29:344-348.
  14. Zouari S, Khrouf F, M’ghirbi Y, et al. First molecular detection and characterization of zoonotic Bartonella species in fleas infesting domestic animals in Tunisia. Parasit Vectors. 2017;10:436.
  15. Rolain JM, Bourry, O, Davoust B, et al. Bartonella quintana and Rickettsia felis in Gabon. Emerg Infect Dis. 2005;11:1742-1744.
  16. Tsioutis C, Zafeiri M, Avramopoulos A, et al. Clinical and laboratory characteristics, epidemiology, and outcomes of murine typhus: a systematic review. Acta Trop. 2017;166:16-24.
  17. Brown L, Macaluso KR. Rickettsia felis, an emerging flea-borne rickettsiosis. Curr Trop Med Rep. 2016;3:27-39.
  18. Oteo JA, Portillo A, Potero F, et al. ‘Candidatus Rickettsia asemboensis’ and Wolbachia spp. in Ctenocephalides felis and Pulex irritans fleas removed from dogs in Ecuador. Parasit Vectors. 2014;7:455.
  19. Ghavami MB, Mirzadeh H, Mohammadi J, et al. Molecular survey of ITS spacer and Rickettsia infection in human flea, Pulex irritans. Parasitol Res. 2018;117:1433-1442.
  20. Traversa D. Fleas infesting pets in the era of emerging extra-intestinal nematodes. Parasit Vectors. 2013;6:59.
  21. Rust MK. Insecticide resistance in fleas. Insects. 2016;7:10.
  22. Ghavami MB, Haghi FP, Alibabaei Z, et al. First report of target site insensitivity to pyrethroids in human flea, Pulex irritans (Siphonaptera: Pulicidae). Pest Biochem Physiol. 2018;146:97-105.
References
  1. Burrow M. How fleas jump. J Exp Biol. 2009;18:2881-2883.
  2. Buckland PC, Sandler JP. A biogeography of the human flea, Pulex irritans L (Siphonaptera: Pulicidae). J Biogeogr. 1989;16:115-120.
  3. Krasnov BR. Life cycles. In: Krasnov BR, ed. Functional and Evolutional Ecology of Fleas. Cambridge, MA: Cambridge Univ Press; 2008:45-67.
  4. Dean KR, Krauer F, Walloe L, et al. Human ectoparasites and the spread of plague in Europe during the second pandemic. Proc Natl Acad Sci U S A. 2018;115:1304-1309.
  5. Hufthammer AK, Walloe L. Rats cannot have been intermediate hosts for Yersinia pestis during medieval plague epidemics in Northern Europe. J Archeol Sci. 2013;40:1752-1759.
  6. Archibald WS, Kunitz SJ. Detection of plague by testing serums of dogs on the Navajo Reservation. HSMHA Health Rep. 1971;86:377-380.
  7. Ratovonjato J, Rajerison M, Rahelinirina S, et al. Yersinia pestis in Pulex irritans fleas during plague outbreak, Madagascar. Emerg Infect Dis. 2014;20:1414-1415.
  8. Laudisoit A, Leirs H, Makundi RH, et al. Plague and the human flea, Tanzania. Emerg Infect Dis. 2007;13:687-693.
  9. Foucault C, Brouqui P, Raoult D. Bartonella quintana characteristics and clinical management. Emerg Infect Dis. 2006;12:217-223.
  10. Eremeeva ME, Gerns HL, Lydy SL, et al. Bacteremia, fever, and splenomegaly caused by a newly recognized bartonella species. N Engl J Med. 2007; 356:2381-2387.11.
  11. Marquez FJ, Millan J, Rodriguez-Liebana JJ, et al. Detection and identification of Bartonella sp. in fleas from carnivorous mammals in Andalusia, Spain. Med Vet Entomol. 2009;23:393-398.
  12. Perez-Martinez L, Venzal JM, Portillo A, et al. Bartonella rochalimae and other Bartonella spp. in fleas, Chile. Emerg Infect Dis. 2009;15:1150-1152.
  13. Sofer S, Gutierrez DM, Mumcuoglu KY, et al. Molecular detection of zoonotic bartonellae (B. henselae, B. elizabethae and B. rochalimae) in fleas collected from dogs in Israel. Med Vet Entomol. 2015;29:344-348.
  14. Zouari S, Khrouf F, M’ghirbi Y, et al. First molecular detection and characterization of zoonotic Bartonella species in fleas infesting domestic animals in Tunisia. Parasit Vectors. 2017;10:436.
  15. Rolain JM, Bourry, O, Davoust B, et al. Bartonella quintana and Rickettsia felis in Gabon. Emerg Infect Dis. 2005;11:1742-1744.
  16. Tsioutis C, Zafeiri M, Avramopoulos A, et al. Clinical and laboratory characteristics, epidemiology, and outcomes of murine typhus: a systematic review. Acta Trop. 2017;166:16-24.
  17. Brown L, Macaluso KR. Rickettsia felis, an emerging flea-borne rickettsiosis. Curr Trop Med Rep. 2016;3:27-39.
  18. Oteo JA, Portillo A, Potero F, et al. ‘Candidatus Rickettsia asemboensis’ and Wolbachia spp. in Ctenocephalides felis and Pulex irritans fleas removed from dogs in Ecuador. Parasit Vectors. 2014;7:455.
  19. Ghavami MB, Mirzadeh H, Mohammadi J, et al. Molecular survey of ITS spacer and Rickettsia infection in human flea, Pulex irritans. Parasitol Res. 2018;117:1433-1442.
  20. Traversa D. Fleas infesting pets in the era of emerging extra-intestinal nematodes. Parasit Vectors. 2013;6:59.
  21. Rust MK. Insecticide resistance in fleas. Insects. 2016;7:10.
  22. Ghavami MB, Haghi FP, Alibabaei Z, et al. First report of target site insensitivity to pyrethroids in human flea, Pulex irritans (Siphonaptera: Pulicidae). Pest Biochem Physiol. 2018;146:97-105.
Issue
Cutis - 106(5)
Issue
Cutis - 106(5)
Page Number
233-235
Page Number
233-235
Publications
Publications
Topics
Article Type
Display Headline
What’s Eating You? Human Flea (Pulex irritans)
Display Headline
What’s Eating You? Human Flea (Pulex irritans)
Sections
Inside the Article

Practice Points

  • The human flea, Pulex irritans, is a vector for various human diseases including the bubonic plague, bartonellosis, and rickettsioses.
  • Presenting symptoms of flea bites include intensely pruritic, urticarial to vesicular papules on exposed areas of skin.
  • The primary method of flea control includes a combination of insecticidal products and insect growth regulators.
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

What’s Eating You? Oriental Rat Flea (Xenopsylla cheopis)

Article Type
Changed
Wed, 09/16/2020 - 09:29
Display Headline
What’s Eating You? Oriental Rat Flea (Xenopsylla cheopis)

A dult Siphonaptera (fleas) are highly adapted to life on the surface of their hosts. Their small 2- to 10-mm bodies are laterally flattened and wingless. They utilize particularly strong hind legs for jumping up to 150 times their body length and backward-directed spines on their legs and bodies for moving forward through fur, hair, and feathers. Xenopsylla cheopis , the oriental rat flea, lacks pronotal and genal combs and has a mesopleuron divided by internal scleritinization (Figure). These features differentiate the species from its close relatives, Ctenocephalides (cat and dog fleas), which have both sets of combs, as well as Pulex irritans (human flea), which do not have a divided mesopleuron. 1,2

Xenopsylla cheopis.

Flea-borne infections are extremely important to public health and are present throughout the world. Further, humidity and warmth are essential for the life cycle of many species of fleas. Predicted global warming likely will increase their distribution, allowing the spread of diseases they carry into previously untouched areas.1 Therefore, it is important to continue to examine species that carry particularly dangerous pathogens, such as X cheopis.

Disease Vector

Xenopsylla cheopis primarily is known for being a vector in the transmission of Yersinia pestis, the etiologic agent of the plague. Plague occurs in 3 forms: bubonic, pneumonic, and septicemic. It has caused major epidemics throughout history, the most widely known being the Black Death, which lasted for 130 years, beginning in the 1330s in China and spreading into Europe where it wiped out one-third of the population. However, bubonic plague is thought to have caused documented outbreaks as early as 320 bce, and it still remains endemic today.3,4

Between January 2010 and December 2015, 3248 cases of plague in humans were reported, resulting in 584 deaths worldwide.5 It is thought that the plague originated in Central Asia, and this area still is a focus of disease. However, the at-risk population is reduced to breeders and hunters of gerbils and marmots, the main reservoirs in the area. In Africa, 4 countries still regularly report cases, with Madagascar being the most severely affected country in the world.5 The Democratic Republic of the Congo, Uganda, and Tanzania also are affected. The Americas also experience the plague. There are sporadic cases of bubonic plague in the northwest corner of Peru, mostly in rural areas. In the western United States, plague circulates among wild rodents, resulting in several reported cases each year, with the most recent confirmed case noted in California in August 2020.5,6 Further adding to its relevance, Y pestis is one of several organisms most likely to be used as a biologic weapon.3,4

Due to the historical and continued significance of Y pestis, many studies have been performed over the decades regarding its association with X cheopis. It has been discovered that fleas transmit the bacteria to the host in 2 ways. The most well-defined form of transmission occurs after an incubation period of Y pestis in the flea for 7 to 31 days. During this time, the bacteria form a dense biofilm on a valve in the flea foregut—the proventriculus—interfering with its function, which allows regurgitation of the blood and the bacteria it contains into the bite site and consequently disease transmission. The proventriculus can become completely blocked in some fleas, preventing any blood from reaching the midgut and causing starvation. In these scenarios, the flea will make continuous attempts to feed, increasing transmission.7 The hemin storage gene, hms, encoding the second messenger molecule cyclic di-GMP plays a critical role in biofilm formation and proventricular blockage.8 The phosphoheptose isomerase gene, GmhA, also has been elucidated as crucial in late transmission due to its role in biofilm formation.9 Early-phase transmission, or biofilm-independent transmission, has been documented to occur as early as 3 hours after infection of the flea but can occur for up to 4 days.10 Historically, the importance of early-phase transmission has been overlooked. Research has shown that it likely is crucial to the epizootic transmission of the plague.10 As a result, the search has begun for genes that contribute to the maintenance of Y pestis in the flea vector during the first 4 days of colonization. It is thought that a key evolutionary development was the selective loss-of-function mutation in a gene essential for the activity of urease, an enzyme that causes acute oral toxicity and mortality in fleas.11,12 The Yersinia murine toxin gene, Ymt, also allows for early survival of Y pestis in the flea midgut by producing a phospholipase D that protects the bacteria from toxic by-products produced during digestion of blood.11,13 In addition, gene products that function in lipid A modification are crucial for the ability of Y pestis to resist the action of cationic antimicrobial peptides it produces, such as cecropin A and polymyxin B.13

Murine typhus, an acute febrile illness caused by Rickettsia typhi, is another disease that can be spread by oriental rat fleas. It has a worldwide distribution. In the United States, R typhi–induced rickettsia mainly is concentrated in suburban areas of Texas and California where it is thought to be mainly spread by Ctenocephalides, but it also is found in Hawaii where spread by X cheopis has been documented.14,15 The most common symptoms of rickettsia include fever, headache, arthralgia, and a characteristic rash that is pruritic and maculopapular, starting on the trunk and spreading peripherally but sparing the palms and soles. This rash occurs about a week after the onset of fever.14Rickettsia felis also has been isolated in the oriental rat flea. However, only a handful of cases of human disease caused by this bacterium have been reported throughout the world, with clinical similarity to murine typhus likely leading to underestimation of disease prevalence.15Bartonella and other species of bacteria also have been documented to be spread by X cheopis.16 Unfortunately, the interactions of X cheopis with these other bacteria are not as well studied as its relationship with Y pestis.

Adverse Reactions

A flea bite itself can cause discomfort. It begins as a punctate hemorrhagic area that develops a surrounding wheal within 30 minutes. Over the course of 1 to 2 days, a delayed reaction occurs and there is a transition to an extremely pruritic, papular lesion. Bites often occur in clusters and can persist for weeks.1

Prevention and Treatment

Control of host animals via extermination and proper sanitation can secondarily reduce the population of X cheopis. Direct pesticide control of the flea population also has been suggested to reduce flea-borne disease. However, insecticides cause a selective pressure on the flea population, leading to populations that are resistant to them. For example, the flea population in Madagascar developed resistance to DDT (dichlorodiphenyltrichloroethane), dieldrin, deltamethrin, and cyfluthrin after their widespread use.17 Further, a recent study revealed resistance of X cheopis populations to alphacypermethrin, lambda-cyhalothrin, and etofenprox, none of which were used in mass vector control, indicating that some cross-resistance mechanism between these and the extensively used insecticides may exist. With the development of widespread resistance to most pesticides, flea control in endemic areas is difficult. Insecticide targeting to fleas on rodents (eg, rodent bait containing insecticide) can allow for more targeted insecticide treatment, limiting the development of resistance.17 Recent development of a maceration protocol used to detect zoonotic pathogens in fleas in the field also will allow management with pesticides to be targeted geographically and temporally where infected vectors are located.18 Research of the interaction between vector, pathogen, and insect microbiome also should continue, as it may allow for development of biopesticides, limiting the use of chemical pesticides all together. The strategy is based on the introduction of microorganisms that can reduce vector lifespan or their ability to transmit pathogens.17

When flea-transmitted diseases do occur, treatment with antibiotics is advised. Early treatment of the plague with effective antibiotics such as streptomycin, gentamicin, tetracycline, or chloramphenicol for a minimum of 10 days is critical for survival. Additionally, patients with bubonic plague should be isolated for at least 2 days after administration of antibiotics, while patients with the pneumonic form should be isolated for 4 days into therapy to prevent the spread of disease. Prophylactic therapy for individuals who come into contact with infected individuals also is advised.4 Patients with murine typhus typically respond to doxycycline, tetracycline, or fluoroquinolones. The few cases of R felis–induced disease have responded to doxycycline. Of note, short courses of treatment of doxycycline are appropriate and safe in young children. The short (3–7 day) nature of the course limits the chances of teeth staining.14

References
  1. Bitam I, Dittmar K, Parola P, et al. Flea and flea-borne diseases. Int J Infect Dis. 2010;14:E667-E676.
  2. Mathison BA, Pritt BS. Laboratory identification of arthropod ectoparasites. Clin Microbiol Rev. 2014;27:48-67.
  3. Ligon BL. Plague: a review of its history and potential as a biological weapon. Semin Pediatr Infect Dis. 2006;17:161-170.
  4. Josko D. Yersinia pestis: still a plague in the 21st century. Clin Lab Sci. 2004;17:25-29.
  5. Plague around the world, 2010–2015. Wkly Epidemiol Rec. 2016;91:89-93.
  6. Sullivan K. California confirms first human case of the plague in 5 years: what to know. NBC News website. https://www.nbcnews.com/health/health-news/california-confirms-first-human-case-bubonic-plague-5-years-what-n1237306. Published August 19, 2020. Accessed August 24, 2020.
  7. Hinnebusch BJ, Bland DM, Bosio CF, et al. Comparative ability of Oropsylla and Xenopsylla cheopis fleas to transmit Yersinia pestis by two different mechanisms. PLOS Negl Trop Dis. 2017;11:e0005276.
  8. Bobrov AG, Kirillina O, Vadyvaloo V, et al. The Yersinia pestis HmsCDE regulatory system is essential for blockage of the oriental rat flea (Xenopsylla cheopis), a classic plague vector. Environ Microbiol. 2015;17:947-959.
  9. Darby C, Ananth SL, Tan L, et al. Identification of gmhA, a Yersina pestis gene required for flea blockage, by using a Caenorhabditis elegans biofilm system. Infect Immun. 2005;73:7236-7242.
  10. Eisen RJ, Dennis DT, Gage KL. The role of early-phase transmission in the spread of Yersinia pestis. J Med Entomol. 2015;52:1183-1192.
  11. Carniel E. Subtle genetic modifications transformed an enteropathogen into a flea-borne pathogen. Proc Natl Acad Sci U S A. 2014;111:18409-18410.
  12. Chouikha I, Hinnebusch BJ. Silencing urease: a key evolutionary step that facilitated the adaptation of Yersinia pestis to the flea-borne transmission route. Proc Natl Acad Sci U S A. 2014;111:18709-19714.
  13. Aoyagi KL, Brooks BD, Bearden SW, et al. LPS modification promotes maintenance of Yersinia pestis in fleas. Microbiology. 2015;161:628-638.
  14. Civen R, Ngo V. Murine typhus: an unrecognized suburban vectorborne disease. Clin Infect Dis. 2008;46:913-918.
  15. Eremeeva ME, Warashina WR, Sturgeon MM, et al. Rickettsia typhi and R. felis in rat fleas (Xenopsylla cheopis), Oahu, Hawaii. Emerg Infect Dis. 2018;14:1613-1615.
  16. Billeter SA, Gundi VAKB, Rood MP, et al. Molecular detection and identification of Bartonella species in Xenopsylla cheopis fleas (Siphonaptera: Pulicidae) collected from Rattus norvecus rats in Los Angeles, California. Appl Environ Microbiol. 2011;77:7850-7852.
  17. Miarinjara A, Boyer S. Current perspectives on plague vector control in Madagascar: susceptibility status of Xenopsylla cheopis to 12 insecticides. PLOS Negl Trop Dis. 2016;10:e0004414.
  18. Harrison GF, Scheirer JL, Melanson VR. Development and validation of an arthropod maceration protocol for zoonotic pathogen detection in mosquitoes and fleas. J Vector Ecol. 2014;40:83-89.
Article PDF
Author and Disclosure Information

Dr. Wells is from the Department of Internal Medicine, University of Virginia, Charlottesville. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

The image is in the public domain.

Correspondence: Leah Ellis Wells, MD, University Medical Associates, UVA Jefferson Park Ave, Medical Office Building, 3rd Floor, 1222 Jefferson Park Ave, Charlottesville, VA 22903 (leah.ellis.wells@gmail.com).

Issue
Cutis - 106(3)
Publications
Topics
Page Number
124-126
Sections
Author and Disclosure Information

Dr. Wells is from the Department of Internal Medicine, University of Virginia, Charlottesville. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

The image is in the public domain.

Correspondence: Leah Ellis Wells, MD, University Medical Associates, UVA Jefferson Park Ave, Medical Office Building, 3rd Floor, 1222 Jefferson Park Ave, Charlottesville, VA 22903 (leah.ellis.wells@gmail.com).

Author and Disclosure Information

Dr. Wells is from the Department of Internal Medicine, University of Virginia, Charlottesville. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

The image is in the public domain.

Correspondence: Leah Ellis Wells, MD, University Medical Associates, UVA Jefferson Park Ave, Medical Office Building, 3rd Floor, 1222 Jefferson Park Ave, Charlottesville, VA 22903 (leah.ellis.wells@gmail.com).

Article PDF
Article PDF

A dult Siphonaptera (fleas) are highly adapted to life on the surface of their hosts. Their small 2- to 10-mm bodies are laterally flattened and wingless. They utilize particularly strong hind legs for jumping up to 150 times their body length and backward-directed spines on their legs and bodies for moving forward through fur, hair, and feathers. Xenopsylla cheopis , the oriental rat flea, lacks pronotal and genal combs and has a mesopleuron divided by internal scleritinization (Figure). These features differentiate the species from its close relatives, Ctenocephalides (cat and dog fleas), which have both sets of combs, as well as Pulex irritans (human flea), which do not have a divided mesopleuron. 1,2

Xenopsylla cheopis.

Flea-borne infections are extremely important to public health and are present throughout the world. Further, humidity and warmth are essential for the life cycle of many species of fleas. Predicted global warming likely will increase their distribution, allowing the spread of diseases they carry into previously untouched areas.1 Therefore, it is important to continue to examine species that carry particularly dangerous pathogens, such as X cheopis.

Disease Vector

Xenopsylla cheopis primarily is known for being a vector in the transmission of Yersinia pestis, the etiologic agent of the plague. Plague occurs in 3 forms: bubonic, pneumonic, and septicemic. It has caused major epidemics throughout history, the most widely known being the Black Death, which lasted for 130 years, beginning in the 1330s in China and spreading into Europe where it wiped out one-third of the population. However, bubonic plague is thought to have caused documented outbreaks as early as 320 bce, and it still remains endemic today.3,4

Between January 2010 and December 2015, 3248 cases of plague in humans were reported, resulting in 584 deaths worldwide.5 It is thought that the plague originated in Central Asia, and this area still is a focus of disease. However, the at-risk population is reduced to breeders and hunters of gerbils and marmots, the main reservoirs in the area. In Africa, 4 countries still regularly report cases, with Madagascar being the most severely affected country in the world.5 The Democratic Republic of the Congo, Uganda, and Tanzania also are affected. The Americas also experience the plague. There are sporadic cases of bubonic plague in the northwest corner of Peru, mostly in rural areas. In the western United States, plague circulates among wild rodents, resulting in several reported cases each year, with the most recent confirmed case noted in California in August 2020.5,6 Further adding to its relevance, Y pestis is one of several organisms most likely to be used as a biologic weapon.3,4

Due to the historical and continued significance of Y pestis, many studies have been performed over the decades regarding its association with X cheopis. It has been discovered that fleas transmit the bacteria to the host in 2 ways. The most well-defined form of transmission occurs after an incubation period of Y pestis in the flea for 7 to 31 days. During this time, the bacteria form a dense biofilm on a valve in the flea foregut—the proventriculus—interfering with its function, which allows regurgitation of the blood and the bacteria it contains into the bite site and consequently disease transmission. The proventriculus can become completely blocked in some fleas, preventing any blood from reaching the midgut and causing starvation. In these scenarios, the flea will make continuous attempts to feed, increasing transmission.7 The hemin storage gene, hms, encoding the second messenger molecule cyclic di-GMP plays a critical role in biofilm formation and proventricular blockage.8 The phosphoheptose isomerase gene, GmhA, also has been elucidated as crucial in late transmission due to its role in biofilm formation.9 Early-phase transmission, or biofilm-independent transmission, has been documented to occur as early as 3 hours after infection of the flea but can occur for up to 4 days.10 Historically, the importance of early-phase transmission has been overlooked. Research has shown that it likely is crucial to the epizootic transmission of the plague.10 As a result, the search has begun for genes that contribute to the maintenance of Y pestis in the flea vector during the first 4 days of colonization. It is thought that a key evolutionary development was the selective loss-of-function mutation in a gene essential for the activity of urease, an enzyme that causes acute oral toxicity and mortality in fleas.11,12 The Yersinia murine toxin gene, Ymt, also allows for early survival of Y pestis in the flea midgut by producing a phospholipase D that protects the bacteria from toxic by-products produced during digestion of blood.11,13 In addition, gene products that function in lipid A modification are crucial for the ability of Y pestis to resist the action of cationic antimicrobial peptides it produces, such as cecropin A and polymyxin B.13

Murine typhus, an acute febrile illness caused by Rickettsia typhi, is another disease that can be spread by oriental rat fleas. It has a worldwide distribution. In the United States, R typhi–induced rickettsia mainly is concentrated in suburban areas of Texas and California where it is thought to be mainly spread by Ctenocephalides, but it also is found in Hawaii where spread by X cheopis has been documented.14,15 The most common symptoms of rickettsia include fever, headache, arthralgia, and a characteristic rash that is pruritic and maculopapular, starting on the trunk and spreading peripherally but sparing the palms and soles. This rash occurs about a week after the onset of fever.14Rickettsia felis also has been isolated in the oriental rat flea. However, only a handful of cases of human disease caused by this bacterium have been reported throughout the world, with clinical similarity to murine typhus likely leading to underestimation of disease prevalence.15Bartonella and other species of bacteria also have been documented to be spread by X cheopis.16 Unfortunately, the interactions of X cheopis with these other bacteria are not as well studied as its relationship with Y pestis.

Adverse Reactions

A flea bite itself can cause discomfort. It begins as a punctate hemorrhagic area that develops a surrounding wheal within 30 minutes. Over the course of 1 to 2 days, a delayed reaction occurs and there is a transition to an extremely pruritic, papular lesion. Bites often occur in clusters and can persist for weeks.1

Prevention and Treatment

Control of host animals via extermination and proper sanitation can secondarily reduce the population of X cheopis. Direct pesticide control of the flea population also has been suggested to reduce flea-borne disease. However, insecticides cause a selective pressure on the flea population, leading to populations that are resistant to them. For example, the flea population in Madagascar developed resistance to DDT (dichlorodiphenyltrichloroethane), dieldrin, deltamethrin, and cyfluthrin after their widespread use.17 Further, a recent study revealed resistance of X cheopis populations to alphacypermethrin, lambda-cyhalothrin, and etofenprox, none of which were used in mass vector control, indicating that some cross-resistance mechanism between these and the extensively used insecticides may exist. With the development of widespread resistance to most pesticides, flea control in endemic areas is difficult. Insecticide targeting to fleas on rodents (eg, rodent bait containing insecticide) can allow for more targeted insecticide treatment, limiting the development of resistance.17 Recent development of a maceration protocol used to detect zoonotic pathogens in fleas in the field also will allow management with pesticides to be targeted geographically and temporally where infected vectors are located.18 Research of the interaction between vector, pathogen, and insect microbiome also should continue, as it may allow for development of biopesticides, limiting the use of chemical pesticides all together. The strategy is based on the introduction of microorganisms that can reduce vector lifespan or their ability to transmit pathogens.17

When flea-transmitted diseases do occur, treatment with antibiotics is advised. Early treatment of the plague with effective antibiotics such as streptomycin, gentamicin, tetracycline, or chloramphenicol for a minimum of 10 days is critical for survival. Additionally, patients with bubonic plague should be isolated for at least 2 days after administration of antibiotics, while patients with the pneumonic form should be isolated for 4 days into therapy to prevent the spread of disease. Prophylactic therapy for individuals who come into contact with infected individuals also is advised.4 Patients with murine typhus typically respond to doxycycline, tetracycline, or fluoroquinolones. The few cases of R felis–induced disease have responded to doxycycline. Of note, short courses of treatment of doxycycline are appropriate and safe in young children. The short (3–7 day) nature of the course limits the chances of teeth staining.14

A dult Siphonaptera (fleas) are highly adapted to life on the surface of their hosts. Their small 2- to 10-mm bodies are laterally flattened and wingless. They utilize particularly strong hind legs for jumping up to 150 times their body length and backward-directed spines on their legs and bodies for moving forward through fur, hair, and feathers. Xenopsylla cheopis , the oriental rat flea, lacks pronotal and genal combs and has a mesopleuron divided by internal scleritinization (Figure). These features differentiate the species from its close relatives, Ctenocephalides (cat and dog fleas), which have both sets of combs, as well as Pulex irritans (human flea), which do not have a divided mesopleuron. 1,2

Xenopsylla cheopis.

Flea-borne infections are extremely important to public health and are present throughout the world. Further, humidity and warmth are essential for the life cycle of many species of fleas. Predicted global warming likely will increase their distribution, allowing the spread of diseases they carry into previously untouched areas.1 Therefore, it is important to continue to examine species that carry particularly dangerous pathogens, such as X cheopis.

Disease Vector

Xenopsylla cheopis primarily is known for being a vector in the transmission of Yersinia pestis, the etiologic agent of the plague. Plague occurs in 3 forms: bubonic, pneumonic, and septicemic. It has caused major epidemics throughout history, the most widely known being the Black Death, which lasted for 130 years, beginning in the 1330s in China and spreading into Europe where it wiped out one-third of the population. However, bubonic plague is thought to have caused documented outbreaks as early as 320 bce, and it still remains endemic today.3,4

Between January 2010 and December 2015, 3248 cases of plague in humans were reported, resulting in 584 deaths worldwide.5 It is thought that the plague originated in Central Asia, and this area still is a focus of disease. However, the at-risk population is reduced to breeders and hunters of gerbils and marmots, the main reservoirs in the area. In Africa, 4 countries still regularly report cases, with Madagascar being the most severely affected country in the world.5 The Democratic Republic of the Congo, Uganda, and Tanzania also are affected. The Americas also experience the plague. There are sporadic cases of bubonic plague in the northwest corner of Peru, mostly in rural areas. In the western United States, plague circulates among wild rodents, resulting in several reported cases each year, with the most recent confirmed case noted in California in August 2020.5,6 Further adding to its relevance, Y pestis is one of several organisms most likely to be used as a biologic weapon.3,4

Due to the historical and continued significance of Y pestis, many studies have been performed over the decades regarding its association with X cheopis. It has been discovered that fleas transmit the bacteria to the host in 2 ways. The most well-defined form of transmission occurs after an incubation period of Y pestis in the flea for 7 to 31 days. During this time, the bacteria form a dense biofilm on a valve in the flea foregut—the proventriculus—interfering with its function, which allows regurgitation of the blood and the bacteria it contains into the bite site and consequently disease transmission. The proventriculus can become completely blocked in some fleas, preventing any blood from reaching the midgut and causing starvation. In these scenarios, the flea will make continuous attempts to feed, increasing transmission.7 The hemin storage gene, hms, encoding the second messenger molecule cyclic di-GMP plays a critical role in biofilm formation and proventricular blockage.8 The phosphoheptose isomerase gene, GmhA, also has been elucidated as crucial in late transmission due to its role in biofilm formation.9 Early-phase transmission, or biofilm-independent transmission, has been documented to occur as early as 3 hours after infection of the flea but can occur for up to 4 days.10 Historically, the importance of early-phase transmission has been overlooked. Research has shown that it likely is crucial to the epizootic transmission of the plague.10 As a result, the search has begun for genes that contribute to the maintenance of Y pestis in the flea vector during the first 4 days of colonization. It is thought that a key evolutionary development was the selective loss-of-function mutation in a gene essential for the activity of urease, an enzyme that causes acute oral toxicity and mortality in fleas.11,12 The Yersinia murine toxin gene, Ymt, also allows for early survival of Y pestis in the flea midgut by producing a phospholipase D that protects the bacteria from toxic by-products produced during digestion of blood.11,13 In addition, gene products that function in lipid A modification are crucial for the ability of Y pestis to resist the action of cationic antimicrobial peptides it produces, such as cecropin A and polymyxin B.13

Murine typhus, an acute febrile illness caused by Rickettsia typhi, is another disease that can be spread by oriental rat fleas. It has a worldwide distribution. In the United States, R typhi–induced rickettsia mainly is concentrated in suburban areas of Texas and California where it is thought to be mainly spread by Ctenocephalides, but it also is found in Hawaii where spread by X cheopis has been documented.14,15 The most common symptoms of rickettsia include fever, headache, arthralgia, and a characteristic rash that is pruritic and maculopapular, starting on the trunk and spreading peripherally but sparing the palms and soles. This rash occurs about a week after the onset of fever.14Rickettsia felis also has been isolated in the oriental rat flea. However, only a handful of cases of human disease caused by this bacterium have been reported throughout the world, with clinical similarity to murine typhus likely leading to underestimation of disease prevalence.15Bartonella and other species of bacteria also have been documented to be spread by X cheopis.16 Unfortunately, the interactions of X cheopis with these other bacteria are not as well studied as its relationship with Y pestis.

Adverse Reactions

A flea bite itself can cause discomfort. It begins as a punctate hemorrhagic area that develops a surrounding wheal within 30 minutes. Over the course of 1 to 2 days, a delayed reaction occurs and there is a transition to an extremely pruritic, papular lesion. Bites often occur in clusters and can persist for weeks.1

Prevention and Treatment

Control of host animals via extermination and proper sanitation can secondarily reduce the population of X cheopis. Direct pesticide control of the flea population also has been suggested to reduce flea-borne disease. However, insecticides cause a selective pressure on the flea population, leading to populations that are resistant to them. For example, the flea population in Madagascar developed resistance to DDT (dichlorodiphenyltrichloroethane), dieldrin, deltamethrin, and cyfluthrin after their widespread use.17 Further, a recent study revealed resistance of X cheopis populations to alphacypermethrin, lambda-cyhalothrin, and etofenprox, none of which were used in mass vector control, indicating that some cross-resistance mechanism between these and the extensively used insecticides may exist. With the development of widespread resistance to most pesticides, flea control in endemic areas is difficult. Insecticide targeting to fleas on rodents (eg, rodent bait containing insecticide) can allow for more targeted insecticide treatment, limiting the development of resistance.17 Recent development of a maceration protocol used to detect zoonotic pathogens in fleas in the field also will allow management with pesticides to be targeted geographically and temporally where infected vectors are located.18 Research of the interaction between vector, pathogen, and insect microbiome also should continue, as it may allow for development of biopesticides, limiting the use of chemical pesticides all together. The strategy is based on the introduction of microorganisms that can reduce vector lifespan or their ability to transmit pathogens.17

When flea-transmitted diseases do occur, treatment with antibiotics is advised. Early treatment of the plague with effective antibiotics such as streptomycin, gentamicin, tetracycline, or chloramphenicol for a minimum of 10 days is critical for survival. Additionally, patients with bubonic plague should be isolated for at least 2 days after administration of antibiotics, while patients with the pneumonic form should be isolated for 4 days into therapy to prevent the spread of disease. Prophylactic therapy for individuals who come into contact with infected individuals also is advised.4 Patients with murine typhus typically respond to doxycycline, tetracycline, or fluoroquinolones. The few cases of R felis–induced disease have responded to doxycycline. Of note, short courses of treatment of doxycycline are appropriate and safe in young children. The short (3–7 day) nature of the course limits the chances of teeth staining.14

References
  1. Bitam I, Dittmar K, Parola P, et al. Flea and flea-borne diseases. Int J Infect Dis. 2010;14:E667-E676.
  2. Mathison BA, Pritt BS. Laboratory identification of arthropod ectoparasites. Clin Microbiol Rev. 2014;27:48-67.
  3. Ligon BL. Plague: a review of its history and potential as a biological weapon. Semin Pediatr Infect Dis. 2006;17:161-170.
  4. Josko D. Yersinia pestis: still a plague in the 21st century. Clin Lab Sci. 2004;17:25-29.
  5. Plague around the world, 2010–2015. Wkly Epidemiol Rec. 2016;91:89-93.
  6. Sullivan K. California confirms first human case of the plague in 5 years: what to know. NBC News website. https://www.nbcnews.com/health/health-news/california-confirms-first-human-case-bubonic-plague-5-years-what-n1237306. Published August 19, 2020. Accessed August 24, 2020.
  7. Hinnebusch BJ, Bland DM, Bosio CF, et al. Comparative ability of Oropsylla and Xenopsylla cheopis fleas to transmit Yersinia pestis by two different mechanisms. PLOS Negl Trop Dis. 2017;11:e0005276.
  8. Bobrov AG, Kirillina O, Vadyvaloo V, et al. The Yersinia pestis HmsCDE regulatory system is essential for blockage of the oriental rat flea (Xenopsylla cheopis), a classic plague vector. Environ Microbiol. 2015;17:947-959.
  9. Darby C, Ananth SL, Tan L, et al. Identification of gmhA, a Yersina pestis gene required for flea blockage, by using a Caenorhabditis elegans biofilm system. Infect Immun. 2005;73:7236-7242.
  10. Eisen RJ, Dennis DT, Gage KL. The role of early-phase transmission in the spread of Yersinia pestis. J Med Entomol. 2015;52:1183-1192.
  11. Carniel E. Subtle genetic modifications transformed an enteropathogen into a flea-borne pathogen. Proc Natl Acad Sci U S A. 2014;111:18409-18410.
  12. Chouikha I, Hinnebusch BJ. Silencing urease: a key evolutionary step that facilitated the adaptation of Yersinia pestis to the flea-borne transmission route. Proc Natl Acad Sci U S A. 2014;111:18709-19714.
  13. Aoyagi KL, Brooks BD, Bearden SW, et al. LPS modification promotes maintenance of Yersinia pestis in fleas. Microbiology. 2015;161:628-638.
  14. Civen R, Ngo V. Murine typhus: an unrecognized suburban vectorborne disease. Clin Infect Dis. 2008;46:913-918.
  15. Eremeeva ME, Warashina WR, Sturgeon MM, et al. Rickettsia typhi and R. felis in rat fleas (Xenopsylla cheopis), Oahu, Hawaii. Emerg Infect Dis. 2018;14:1613-1615.
  16. Billeter SA, Gundi VAKB, Rood MP, et al. Molecular detection and identification of Bartonella species in Xenopsylla cheopis fleas (Siphonaptera: Pulicidae) collected from Rattus norvecus rats in Los Angeles, California. Appl Environ Microbiol. 2011;77:7850-7852.
  17. Miarinjara A, Boyer S. Current perspectives on plague vector control in Madagascar: susceptibility status of Xenopsylla cheopis to 12 insecticides. PLOS Negl Trop Dis. 2016;10:e0004414.
  18. Harrison GF, Scheirer JL, Melanson VR. Development and validation of an arthropod maceration protocol for zoonotic pathogen detection in mosquitoes and fleas. J Vector Ecol. 2014;40:83-89.
References
  1. Bitam I, Dittmar K, Parola P, et al. Flea and flea-borne diseases. Int J Infect Dis. 2010;14:E667-E676.
  2. Mathison BA, Pritt BS. Laboratory identification of arthropod ectoparasites. Clin Microbiol Rev. 2014;27:48-67.
  3. Ligon BL. Plague: a review of its history and potential as a biological weapon. Semin Pediatr Infect Dis. 2006;17:161-170.
  4. Josko D. Yersinia pestis: still a plague in the 21st century. Clin Lab Sci. 2004;17:25-29.
  5. Plague around the world, 2010–2015. Wkly Epidemiol Rec. 2016;91:89-93.
  6. Sullivan K. California confirms first human case of the plague in 5 years: what to know. NBC News website. https://www.nbcnews.com/health/health-news/california-confirms-first-human-case-bubonic-plague-5-years-what-n1237306. Published August 19, 2020. Accessed August 24, 2020.
  7. Hinnebusch BJ, Bland DM, Bosio CF, et al. Comparative ability of Oropsylla and Xenopsylla cheopis fleas to transmit Yersinia pestis by two different mechanisms. PLOS Negl Trop Dis. 2017;11:e0005276.
  8. Bobrov AG, Kirillina O, Vadyvaloo V, et al. The Yersinia pestis HmsCDE regulatory system is essential for blockage of the oriental rat flea (Xenopsylla cheopis), a classic plague vector. Environ Microbiol. 2015;17:947-959.
  9. Darby C, Ananth SL, Tan L, et al. Identification of gmhA, a Yersina pestis gene required for flea blockage, by using a Caenorhabditis elegans biofilm system. Infect Immun. 2005;73:7236-7242.
  10. Eisen RJ, Dennis DT, Gage KL. The role of early-phase transmission in the spread of Yersinia pestis. J Med Entomol. 2015;52:1183-1192.
  11. Carniel E. Subtle genetic modifications transformed an enteropathogen into a flea-borne pathogen. Proc Natl Acad Sci U S A. 2014;111:18409-18410.
  12. Chouikha I, Hinnebusch BJ. Silencing urease: a key evolutionary step that facilitated the adaptation of Yersinia pestis to the flea-borne transmission route. Proc Natl Acad Sci U S A. 2014;111:18709-19714.
  13. Aoyagi KL, Brooks BD, Bearden SW, et al. LPS modification promotes maintenance of Yersinia pestis in fleas. Microbiology. 2015;161:628-638.
  14. Civen R, Ngo V. Murine typhus: an unrecognized suburban vectorborne disease. Clin Infect Dis. 2008;46:913-918.
  15. Eremeeva ME, Warashina WR, Sturgeon MM, et al. Rickettsia typhi and R. felis in rat fleas (Xenopsylla cheopis), Oahu, Hawaii. Emerg Infect Dis. 2018;14:1613-1615.
  16. Billeter SA, Gundi VAKB, Rood MP, et al. Molecular detection and identification of Bartonella species in Xenopsylla cheopis fleas (Siphonaptera: Pulicidae) collected from Rattus norvecus rats in Los Angeles, California. Appl Environ Microbiol. 2011;77:7850-7852.
  17. Miarinjara A, Boyer S. Current perspectives on plague vector control in Madagascar: susceptibility status of Xenopsylla cheopis to 12 insecticides. PLOS Negl Trop Dis. 2016;10:e0004414.
  18. Harrison GF, Scheirer JL, Melanson VR. Development and validation of an arthropod maceration protocol for zoonotic pathogen detection in mosquitoes and fleas. J Vector Ecol. 2014;40:83-89.
Issue
Cutis - 106(3)
Issue
Cutis - 106(3)
Page Number
124-126
Page Number
124-126
Publications
Publications
Topics
Article Type
Display Headline
What’s Eating You? Oriental Rat Flea (Xenopsylla cheopis)
Display Headline
What’s Eating You? Oriental Rat Flea (Xenopsylla cheopis)
Sections
Inside the Article

Practice Points

  • Xenopsylla cheopis, the oriental rat flea, is most known for carrying Yersinia pestis, the causative agent of the plague; however, it also is a vector for other bacteria, such as Rickettsia typhi, the species responsible for most cases of murine typhus.
  • Despite the perception that it largely is a historical illness, modern outbreaks of plague occur in many parts of the world each year. Because fleas thrive in warm humid weather, global warming threatens the spread of the oriental rat flea and its diseases into previously unaffected parts of the world.
  • There has been an effort to control oriental rat flea populations, which unfortunately has been complicated by pesticide resistance in many flea populations. It is important to continue to research the oriental rat flea and the bacterial species it carries in the hopes of finding better methods of controlling the pests and therefore decreasing illness in humans.
  • Health care providers should be vigilant in identifying symptoms of flea-borne illnesses. If a patient is displaying symptoms, prompt recognition and antibiotic therapy is critical, particularly for the plague.
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
Article PDF Media

What’s Eating You? Megalopyge opercularis

Article Type
Changed
Tue, 08/18/2020 - 11:06
Display Headline
What’s Eating You? Megalopyge opercularis

Lepidoptera is the second largest order of the class Insecta and comprises approximately 160,000 species of butterflies and moths classified among approximately 124 families and subfamilies. Venomous properties have been identified in 12 of these families, posing a serious threat to human health. 1

The clinical manifestations from Lepidoptera envenomation can range from general systemic symptoms such as fever and abdominal distress; to more complex focal affections including hemorrhage, ophthalmologic lesions, and irritation of the respiratory tracts; to less severe reactions of the skin, which are the most common presentation.1

Terminology

Lepidopterism is the term used to address a clinical spectrum of systemic manifestations from direct contact with venomous butterflies or moths and/or their products.2 Conversely, erucism is a term used to describe localized cutaneous reactions after direct contact with toxins from caterpillars.

Lepidopterism is derived from the Greek roots lepis, meaning scale, and pteron, meaning wing. The term erucism stems from the Latin word eruca, which means larva.2

Ideally, lepidopterism should refer solely to reactions from butterflies and moths—adult forms of insects with scaly wings—while erucism should refer to reactions from contact with caterpillars—the larval form of butterflies and moths.

In common use, lepidopterism can describe any reaction from caterpillars, moths, or adult butterflies, as well as any case of Lepidoptera exposure with only systemic manifestations, regardless of cutaneous findings. Concurrently, erucism has been defined as either any reaction from caterpillars or any skin reaction from contact with caterpillars or moths.2



Because caterpillars are the larval form of butterflies and moths, caterpillar-associated skin reactions also have been conveniently denominated caterpillar dermatitis.1 Henceforth in this article, both terms erucism and caterpillar dermatitis are used interchangeably.

Caterpillar Envenomation

Caterpillars cause the vast majority of adverse events from lepidopteran exposures.2 Envenomation by caterpillars might stand as the world’s most common envenomation given the larvae proximity to humans.3 Although involvement of internal organs (eg, renal failure), cerebral hemorrhage, and joint lesions can occur, skin manifestations are more predominant with the majority of species. Initial localized pain, edema, and erythema usually are present at the site of direct contact and subsequently progress toward maculopapular to bullous lesions, erosions, petechiae, necrosis, and ulceration depending on the offending species.1,4

Megalopyge opercularis

In the United States, more than 50 species of caterpillars have been identified as poisonous or venomous.Megalopyge opercularis (Figure 1), the larval form of the flannel moth, is an important cause of caterpillar-associated dermatitis in the southern United States.6,7 Megalopyge opercularis also is commonly known as the puss caterpillar, opossum bug, wooly slug, el perrito, tree asp, or Italian asp.6 This lepidopteran insect is mainly found in the southeastern and southcentral United States, with noted particular abundance in Texas, Louisiana, and Florida.6,8 The puss caterpillar has 2 generations per year; the first develops during the months of June to July, and the second develops from September to October, carrying seasonal health hazards.6,8

Figure 1. A and B, Larval stage of Megalopyge opercularis.

 

 

Megalopyge opercularis is tapered at the ends and can measure 2.5 to 3.5×1 cm at maturity. It is covered by silky, long-streaked, wavy hairs that may appear single colored or as a mix of colors—from white to gray to brown—forming a mid-dorsal crest.6 Beneath this furry coat, rows of short sharp spines are hidden. Upon contact with the human skin, these spines will break and discharge venom.1,6,8 Toxins contained within the hollow spines are thought to be produced by specialized basal cells, but there still is little knowledge about the dynamics and composition of the venom.1

Clinical Manifestations

The severity of the reaction depends on the caterpillar’s size and the extent of contact.1,4 Contact with M opercularis instantly presents with a throbbing or burning pain that may be followed by localized erythema and rash.1,6 A characteristic gridlike pattern of erythematous macules develops, reflecting each site of puncture from the insect’s spines (Figure 2).8,9 Skin lesions can progress from erythematous macules to hemorrhagic vesicles or pustules, usually self-resolving after a few days. The reaction also can present with radiating pain to regional lymph nodes and numbness of the affected area.1,6,8 Moreover, some patients may report urticaria and pruritus.9

Figure 2. Gridlike pattern of hemorrhagic papules and crusts on the palmar aspect of the right hand following Megalopyge opercularis envenomation.

Envenomation by a puss caterpillar also can present with systemic manifestations including fever, headache, nausea, vomiting, shocklike symptoms, and seizures.1,6,7 Anaphylactic reaction is rare but also can present.7 Uncommon cases have been reported with severe abdominal pain and muscle spasm mimicking acute appendicitis and latrodectism, respectively.7,9

Diagnosis

The diagnosis of M opercularis envenomation is made clinically based on the morphology of the skin lesions and a history of probable exposure. Coexistent leukocytosis is likely, but laboratory testing is not warranted, as it is both nonspecific and insensitive.9

Management/Treatment

The most commonly reported immediate approaches to treatment involve attempts to remove the spines from the skin with tape (stripping), application of ice packs over the affected area, oral antihistamines, topical and intralesional anesthetics, regional nerve block, and oral analgesics.6,9 There have been several cases detailing the successful use of parenteral calcium gluconate,5,7 and diazepam has been used to treat severe muscle spasms. Anaphylactic reactions should be managed in a controlled monitored setting with subcutaneous epinephrine.7 Despite their common use, some data suggest that ice packs and mid- to high-potency topical steroids are ineffective.9

Incidence

From 2001 to 2005, a mean average of 94,552 annual cases of animal bites and stings were reported to poison control centers in the United States, of which 2094 were linked to caterpillars in this 5-year period.10 There were 3484 M opercularis caterpillar stings reported to the Texas Poison Center Network from 2000 to 2016.5,6 Given their ability to sting throughout their life cycle, thousands of M opercularis caterpillar stings can occur each year.1,6 Existing literature on M opercularis caterpillar stings mainly involves case reports with affections of the skin and oral mucosa, self-reported envenomation, and case studies.5,6,8

Although multiple health concerns associated with caterpillar envenomation have been reported worldwide, the lack of official epidemiologic reports highly suggests that this problem remains underestimated. There also may be many unreported cases because certain reactions are mild or self-limited and can even go unnoticed.11 Nonetheless, there is an evident rise of cases reported in the United States. According to the 2018 annual report of the American Association of Poison Control Centers, there were 2815 case mentions from caterpillar envenomation.12

In 1921 and 1952, some public schools in Texas were temporarily closed due to outbreaks of puss caterpillar–associated dermatitis.8 Similar outbreaks also have been reported in South Carolina, Virginia, and Oklahoma.9 Emerging data suggest that plant oil products and the pesticide cypermethrin may be helpful in controlling local infestations of the puss caterpillar.8

References
  1. Villas-Boas IM, Bonfa G, Tambourgi DV. Venomous caterpillars: from inoculation apparatus to venom composition and envenomation. Toxicon. 2018;153:39-52.
  2. Hossler EW. Caterpillars and moths: part I. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:1-10; quiz 11-12.
  3. Haddad Junior V, Amorim PC, Haddad Junior WT, et al. Venomous and poisonous arthropods: identification, clinical manifestations of envenomation, and treatments used in human injuries. Rev Soc Bras Med Trop. 2015;48:650-657.
  4. Haddad V Jr, Cardoso JL, Lupi O, et al. Tropical dermatology: venomous arthropods and human skin: part I. Insecta. J Am Acad Dermatol. 2012;67:331.e1-331.e14; quiz 345.
  5. Pappano DA, Trout Fryxell R, Warren M. Oral mucosal envenomation of an infant by a puss caterpillar. Pediatr Emerg Care. 2017;33:424-426.
  6. Forrester MB. Megalopyge opercularis caterpillar stings reported to Texas poison centers. Wilderness Environ Med. 2018;29:215-220.
  7. Hossler EW. Caterpillars and moths: part II. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:13-28; quiz 29-30.
  8. Eagleman DM. Envenomation by the asp caterpillar (Megalopyge opercularis). Clin Toxicol (Phila). 2008;46:201-205.
  9. Greene SC, Carey JM. Puss caterpillar envenomation: erucism mimicking appendicitis in a young child [published online May 23, 2018]. Pediatr Emerg Care. doi:10.1097/PEC.0000000000001514.
  10. Langley RL. Animal bites and stings reported by United States Poison Control Centers, 2001-2005. Wilderness Environ Med. 2008;19:7-14.
  11. Seldeslachts A, Peigneur S, Tytgat J. Caterpillar venom: a health hazard of the 21st century [published online May 30, 2020]. Biomedicines. doi:10.3390/biomedicines8060143.
  12. Gummin DD, Mowry JB, Spyker DA, et al. 2018 annual report of the American Association of Poison Control Centers’ National Poison Data System (NPDS): 36th annual report. Clin Toxicol (Phila). 2019;57:1220-1413.
Article PDF
Author and Disclosure Information

From the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

The images are in the public domain.

Correspondence: Melba Estrella, MD, Rutledge Tower, 135 Rutledge Ave, Charleston SC 29425 (estrelme@musc.edu).

Issue
Cutis - 106(1)
Publications
Topics
Page Number
23-24, 27
Sections
Author and Disclosure Information

From the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

The images are in the public domain.

Correspondence: Melba Estrella, MD, Rutledge Tower, 135 Rutledge Ave, Charleston SC 29425 (estrelme@musc.edu).

Author and Disclosure Information

From the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

The images are in the public domain.

Correspondence: Melba Estrella, MD, Rutledge Tower, 135 Rutledge Ave, Charleston SC 29425 (estrelme@musc.edu).

Article PDF
Article PDF

Lepidoptera is the second largest order of the class Insecta and comprises approximately 160,000 species of butterflies and moths classified among approximately 124 families and subfamilies. Venomous properties have been identified in 12 of these families, posing a serious threat to human health. 1

The clinical manifestations from Lepidoptera envenomation can range from general systemic symptoms such as fever and abdominal distress; to more complex focal affections including hemorrhage, ophthalmologic lesions, and irritation of the respiratory tracts; to less severe reactions of the skin, which are the most common presentation.1

Terminology

Lepidopterism is the term used to address a clinical spectrum of systemic manifestations from direct contact with venomous butterflies or moths and/or their products.2 Conversely, erucism is a term used to describe localized cutaneous reactions after direct contact with toxins from caterpillars.

Lepidopterism is derived from the Greek roots lepis, meaning scale, and pteron, meaning wing. The term erucism stems from the Latin word eruca, which means larva.2

Ideally, lepidopterism should refer solely to reactions from butterflies and moths—adult forms of insects with scaly wings—while erucism should refer to reactions from contact with caterpillars—the larval form of butterflies and moths.

In common use, lepidopterism can describe any reaction from caterpillars, moths, or adult butterflies, as well as any case of Lepidoptera exposure with only systemic manifestations, regardless of cutaneous findings. Concurrently, erucism has been defined as either any reaction from caterpillars or any skin reaction from contact with caterpillars or moths.2



Because caterpillars are the larval form of butterflies and moths, caterpillar-associated skin reactions also have been conveniently denominated caterpillar dermatitis.1 Henceforth in this article, both terms erucism and caterpillar dermatitis are used interchangeably.

Caterpillar Envenomation

Caterpillars cause the vast majority of adverse events from lepidopteran exposures.2 Envenomation by caterpillars might stand as the world’s most common envenomation given the larvae proximity to humans.3 Although involvement of internal organs (eg, renal failure), cerebral hemorrhage, and joint lesions can occur, skin manifestations are more predominant with the majority of species. Initial localized pain, edema, and erythema usually are present at the site of direct contact and subsequently progress toward maculopapular to bullous lesions, erosions, petechiae, necrosis, and ulceration depending on the offending species.1,4

Megalopyge opercularis

In the United States, more than 50 species of caterpillars have been identified as poisonous or venomous.Megalopyge opercularis (Figure 1), the larval form of the flannel moth, is an important cause of caterpillar-associated dermatitis in the southern United States.6,7 Megalopyge opercularis also is commonly known as the puss caterpillar, opossum bug, wooly slug, el perrito, tree asp, or Italian asp.6 This lepidopteran insect is mainly found in the southeastern and southcentral United States, with noted particular abundance in Texas, Louisiana, and Florida.6,8 The puss caterpillar has 2 generations per year; the first develops during the months of June to July, and the second develops from September to October, carrying seasonal health hazards.6,8

Figure 1. A and B, Larval stage of Megalopyge opercularis.

 

 

Megalopyge opercularis is tapered at the ends and can measure 2.5 to 3.5×1 cm at maturity. It is covered by silky, long-streaked, wavy hairs that may appear single colored or as a mix of colors—from white to gray to brown—forming a mid-dorsal crest.6 Beneath this furry coat, rows of short sharp spines are hidden. Upon contact with the human skin, these spines will break and discharge venom.1,6,8 Toxins contained within the hollow spines are thought to be produced by specialized basal cells, but there still is little knowledge about the dynamics and composition of the venom.1

Clinical Manifestations

The severity of the reaction depends on the caterpillar’s size and the extent of contact.1,4 Contact with M opercularis instantly presents with a throbbing or burning pain that may be followed by localized erythema and rash.1,6 A characteristic gridlike pattern of erythematous macules develops, reflecting each site of puncture from the insect’s spines (Figure 2).8,9 Skin lesions can progress from erythematous macules to hemorrhagic vesicles or pustules, usually self-resolving after a few days. The reaction also can present with radiating pain to regional lymph nodes and numbness of the affected area.1,6,8 Moreover, some patients may report urticaria and pruritus.9

Figure 2. Gridlike pattern of hemorrhagic papules and crusts on the palmar aspect of the right hand following Megalopyge opercularis envenomation.

Envenomation by a puss caterpillar also can present with systemic manifestations including fever, headache, nausea, vomiting, shocklike symptoms, and seizures.1,6,7 Anaphylactic reaction is rare but also can present.7 Uncommon cases have been reported with severe abdominal pain and muscle spasm mimicking acute appendicitis and latrodectism, respectively.7,9

Diagnosis

The diagnosis of M opercularis envenomation is made clinically based on the morphology of the skin lesions and a history of probable exposure. Coexistent leukocytosis is likely, but laboratory testing is not warranted, as it is both nonspecific and insensitive.9

Management/Treatment

The most commonly reported immediate approaches to treatment involve attempts to remove the spines from the skin with tape (stripping), application of ice packs over the affected area, oral antihistamines, topical and intralesional anesthetics, regional nerve block, and oral analgesics.6,9 There have been several cases detailing the successful use of parenteral calcium gluconate,5,7 and diazepam has been used to treat severe muscle spasms. Anaphylactic reactions should be managed in a controlled monitored setting with subcutaneous epinephrine.7 Despite their common use, some data suggest that ice packs and mid- to high-potency topical steroids are ineffective.9

Incidence

From 2001 to 2005, a mean average of 94,552 annual cases of animal bites and stings were reported to poison control centers in the United States, of which 2094 were linked to caterpillars in this 5-year period.10 There were 3484 M opercularis caterpillar stings reported to the Texas Poison Center Network from 2000 to 2016.5,6 Given their ability to sting throughout their life cycle, thousands of M opercularis caterpillar stings can occur each year.1,6 Existing literature on M opercularis caterpillar stings mainly involves case reports with affections of the skin and oral mucosa, self-reported envenomation, and case studies.5,6,8

Although multiple health concerns associated with caterpillar envenomation have been reported worldwide, the lack of official epidemiologic reports highly suggests that this problem remains underestimated. There also may be many unreported cases because certain reactions are mild or self-limited and can even go unnoticed.11 Nonetheless, there is an evident rise of cases reported in the United States. According to the 2018 annual report of the American Association of Poison Control Centers, there were 2815 case mentions from caterpillar envenomation.12

In 1921 and 1952, some public schools in Texas were temporarily closed due to outbreaks of puss caterpillar–associated dermatitis.8 Similar outbreaks also have been reported in South Carolina, Virginia, and Oklahoma.9 Emerging data suggest that plant oil products and the pesticide cypermethrin may be helpful in controlling local infestations of the puss caterpillar.8

Lepidoptera is the second largest order of the class Insecta and comprises approximately 160,000 species of butterflies and moths classified among approximately 124 families and subfamilies. Venomous properties have been identified in 12 of these families, posing a serious threat to human health. 1

The clinical manifestations from Lepidoptera envenomation can range from general systemic symptoms such as fever and abdominal distress; to more complex focal affections including hemorrhage, ophthalmologic lesions, and irritation of the respiratory tracts; to less severe reactions of the skin, which are the most common presentation.1

Terminology

Lepidopterism is the term used to address a clinical spectrum of systemic manifestations from direct contact with venomous butterflies or moths and/or their products.2 Conversely, erucism is a term used to describe localized cutaneous reactions after direct contact with toxins from caterpillars.

Lepidopterism is derived from the Greek roots lepis, meaning scale, and pteron, meaning wing. The term erucism stems from the Latin word eruca, which means larva.2

Ideally, lepidopterism should refer solely to reactions from butterflies and moths—adult forms of insects with scaly wings—while erucism should refer to reactions from contact with caterpillars—the larval form of butterflies and moths.

In common use, lepidopterism can describe any reaction from caterpillars, moths, or adult butterflies, as well as any case of Lepidoptera exposure with only systemic manifestations, regardless of cutaneous findings. Concurrently, erucism has been defined as either any reaction from caterpillars or any skin reaction from contact with caterpillars or moths.2



Because caterpillars are the larval form of butterflies and moths, caterpillar-associated skin reactions also have been conveniently denominated caterpillar dermatitis.1 Henceforth in this article, both terms erucism and caterpillar dermatitis are used interchangeably.

Caterpillar Envenomation

Caterpillars cause the vast majority of adverse events from lepidopteran exposures.2 Envenomation by caterpillars might stand as the world’s most common envenomation given the larvae proximity to humans.3 Although involvement of internal organs (eg, renal failure), cerebral hemorrhage, and joint lesions can occur, skin manifestations are more predominant with the majority of species. Initial localized pain, edema, and erythema usually are present at the site of direct contact and subsequently progress toward maculopapular to bullous lesions, erosions, petechiae, necrosis, and ulceration depending on the offending species.1,4

Megalopyge opercularis

In the United States, more than 50 species of caterpillars have been identified as poisonous or venomous.Megalopyge opercularis (Figure 1), the larval form of the flannel moth, is an important cause of caterpillar-associated dermatitis in the southern United States.6,7 Megalopyge opercularis also is commonly known as the puss caterpillar, opossum bug, wooly slug, el perrito, tree asp, or Italian asp.6 This lepidopteran insect is mainly found in the southeastern and southcentral United States, with noted particular abundance in Texas, Louisiana, and Florida.6,8 The puss caterpillar has 2 generations per year; the first develops during the months of June to July, and the second develops from September to October, carrying seasonal health hazards.6,8

Figure 1. A and B, Larval stage of Megalopyge opercularis.

 

 

Megalopyge opercularis is tapered at the ends and can measure 2.5 to 3.5×1 cm at maturity. It is covered by silky, long-streaked, wavy hairs that may appear single colored or as a mix of colors—from white to gray to brown—forming a mid-dorsal crest.6 Beneath this furry coat, rows of short sharp spines are hidden. Upon contact with the human skin, these spines will break and discharge venom.1,6,8 Toxins contained within the hollow spines are thought to be produced by specialized basal cells, but there still is little knowledge about the dynamics and composition of the venom.1

Clinical Manifestations

The severity of the reaction depends on the caterpillar’s size and the extent of contact.1,4 Contact with M opercularis instantly presents with a throbbing or burning pain that may be followed by localized erythema and rash.1,6 A characteristic gridlike pattern of erythematous macules develops, reflecting each site of puncture from the insect’s spines (Figure 2).8,9 Skin lesions can progress from erythematous macules to hemorrhagic vesicles or pustules, usually self-resolving after a few days. The reaction also can present with radiating pain to regional lymph nodes and numbness of the affected area.1,6,8 Moreover, some patients may report urticaria and pruritus.9

Figure 2. Gridlike pattern of hemorrhagic papules and crusts on the palmar aspect of the right hand following Megalopyge opercularis envenomation.

Envenomation by a puss caterpillar also can present with systemic manifestations including fever, headache, nausea, vomiting, shocklike symptoms, and seizures.1,6,7 Anaphylactic reaction is rare but also can present.7 Uncommon cases have been reported with severe abdominal pain and muscle spasm mimicking acute appendicitis and latrodectism, respectively.7,9

Diagnosis

The diagnosis of M opercularis envenomation is made clinically based on the morphology of the skin lesions and a history of probable exposure. Coexistent leukocytosis is likely, but laboratory testing is not warranted, as it is both nonspecific and insensitive.9

Management/Treatment

The most commonly reported immediate approaches to treatment involve attempts to remove the spines from the skin with tape (stripping), application of ice packs over the affected area, oral antihistamines, topical and intralesional anesthetics, regional nerve block, and oral analgesics.6,9 There have been several cases detailing the successful use of parenteral calcium gluconate,5,7 and diazepam has been used to treat severe muscle spasms. Anaphylactic reactions should be managed in a controlled monitored setting with subcutaneous epinephrine.7 Despite their common use, some data suggest that ice packs and mid- to high-potency topical steroids are ineffective.9

Incidence

From 2001 to 2005, a mean average of 94,552 annual cases of animal bites and stings were reported to poison control centers in the United States, of which 2094 were linked to caterpillars in this 5-year period.10 There were 3484 M opercularis caterpillar stings reported to the Texas Poison Center Network from 2000 to 2016.5,6 Given their ability to sting throughout their life cycle, thousands of M opercularis caterpillar stings can occur each year.1,6 Existing literature on M opercularis caterpillar stings mainly involves case reports with affections of the skin and oral mucosa, self-reported envenomation, and case studies.5,6,8

Although multiple health concerns associated with caterpillar envenomation have been reported worldwide, the lack of official epidemiologic reports highly suggests that this problem remains underestimated. There also may be many unreported cases because certain reactions are mild or self-limited and can even go unnoticed.11 Nonetheless, there is an evident rise of cases reported in the United States. According to the 2018 annual report of the American Association of Poison Control Centers, there were 2815 case mentions from caterpillar envenomation.12

In 1921 and 1952, some public schools in Texas were temporarily closed due to outbreaks of puss caterpillar–associated dermatitis.8 Similar outbreaks also have been reported in South Carolina, Virginia, and Oklahoma.9 Emerging data suggest that plant oil products and the pesticide cypermethrin may be helpful in controlling local infestations of the puss caterpillar.8

References
  1. Villas-Boas IM, Bonfa G, Tambourgi DV. Venomous caterpillars: from inoculation apparatus to venom composition and envenomation. Toxicon. 2018;153:39-52.
  2. Hossler EW. Caterpillars and moths: part I. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:1-10; quiz 11-12.
  3. Haddad Junior V, Amorim PC, Haddad Junior WT, et al. Venomous and poisonous arthropods: identification, clinical manifestations of envenomation, and treatments used in human injuries. Rev Soc Bras Med Trop. 2015;48:650-657.
  4. Haddad V Jr, Cardoso JL, Lupi O, et al. Tropical dermatology: venomous arthropods and human skin: part I. Insecta. J Am Acad Dermatol. 2012;67:331.e1-331.e14; quiz 345.
  5. Pappano DA, Trout Fryxell R, Warren M. Oral mucosal envenomation of an infant by a puss caterpillar. Pediatr Emerg Care. 2017;33:424-426.
  6. Forrester MB. Megalopyge opercularis caterpillar stings reported to Texas poison centers. Wilderness Environ Med. 2018;29:215-220.
  7. Hossler EW. Caterpillars and moths: part II. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:13-28; quiz 29-30.
  8. Eagleman DM. Envenomation by the asp caterpillar (Megalopyge opercularis). Clin Toxicol (Phila). 2008;46:201-205.
  9. Greene SC, Carey JM. Puss caterpillar envenomation: erucism mimicking appendicitis in a young child [published online May 23, 2018]. Pediatr Emerg Care. doi:10.1097/PEC.0000000000001514.
  10. Langley RL. Animal bites and stings reported by United States Poison Control Centers, 2001-2005. Wilderness Environ Med. 2008;19:7-14.
  11. Seldeslachts A, Peigneur S, Tytgat J. Caterpillar venom: a health hazard of the 21st century [published online May 30, 2020]. Biomedicines. doi:10.3390/biomedicines8060143.
  12. Gummin DD, Mowry JB, Spyker DA, et al. 2018 annual report of the American Association of Poison Control Centers’ National Poison Data System (NPDS): 36th annual report. Clin Toxicol (Phila). 2019;57:1220-1413.
References
  1. Villas-Boas IM, Bonfa G, Tambourgi DV. Venomous caterpillars: from inoculation apparatus to venom composition and envenomation. Toxicon. 2018;153:39-52.
  2. Hossler EW. Caterpillars and moths: part I. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:1-10; quiz 11-12.
  3. Haddad Junior V, Amorim PC, Haddad Junior WT, et al. Venomous and poisonous arthropods: identification, clinical manifestations of envenomation, and treatments used in human injuries. Rev Soc Bras Med Trop. 2015;48:650-657.
  4. Haddad V Jr, Cardoso JL, Lupi O, et al. Tropical dermatology: venomous arthropods and human skin: part I. Insecta. J Am Acad Dermatol. 2012;67:331.e1-331.e14; quiz 345.
  5. Pappano DA, Trout Fryxell R, Warren M. Oral mucosal envenomation of an infant by a puss caterpillar. Pediatr Emerg Care. 2017;33:424-426.
  6. Forrester MB. Megalopyge opercularis caterpillar stings reported to Texas poison centers. Wilderness Environ Med. 2018;29:215-220.
  7. Hossler EW. Caterpillars and moths: part II. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:13-28; quiz 29-30.
  8. Eagleman DM. Envenomation by the asp caterpillar (Megalopyge opercularis). Clin Toxicol (Phila). 2008;46:201-205.
  9. Greene SC, Carey JM. Puss caterpillar envenomation: erucism mimicking appendicitis in a young child [published online May 23, 2018]. Pediatr Emerg Care. doi:10.1097/PEC.0000000000001514.
  10. Langley RL. Animal bites and stings reported by United States Poison Control Centers, 2001-2005. Wilderness Environ Med. 2008;19:7-14.
  11. Seldeslachts A, Peigneur S, Tytgat J. Caterpillar venom: a health hazard of the 21st century [published online May 30, 2020]. Biomedicines. doi:10.3390/biomedicines8060143.
  12. Gummin DD, Mowry JB, Spyker DA, et al. 2018 annual report of the American Association of Poison Control Centers’ National Poison Data System (NPDS): 36th annual report. Clin Toxicol (Phila). 2019;57:1220-1413.
Issue
Cutis - 106(1)
Issue
Cutis - 106(1)
Page Number
23-24, 27
Page Number
23-24, 27
Publications
Publications
Topics
Article Type
Display Headline
What’s Eating You? Megalopyge opercularis
Display Headline
What’s Eating You? Megalopyge opercularis
Sections
Inside the Article

Practice Points

  • Megalopyge opercularis is the most widely distributed caterpillar species in the Americas, and envenomation by it can occur year-round.
  • Skin reactions to M opercularis stings can present as maculopapular dermatitis, eczematous eruptions, or urticarial reactions.
  • During the initial presentation, patients experience intense throbbing pain, yet the severity of symptoms depends on the caterpillar’s size and the extent of contact.
  • A history of caterpillar exposure helps with diagnosis, and treatment remains empiric.
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
Article PDF Media

What’s Eating You? Bark Scorpions (Centruroides exilicauda and Centruroides sculpturatus)

Article Type
Changed
Mon, 05/11/2020 - 23:11
Display Headline
What’s Eating You? Bark Scorpions (Centruroides exilicauda and Centruroides sculpturatus)

Epidemiology and Identification

Centruroides is a common genus of bark scorpions in the United States with at least 21 species considered to be medically important, including the closely related Centruroides exilicauda and Centruroides sculpturatus.1 Scorpions can be recognized by a bulbous sac and pointed stinger at the end of a tail-like abdomen. They also have long lobsterlike pedipalps (pincers) for grasping their prey. Identifying characteristics for C exilicauda and C sculpturatus include a small, slender, yellow to light brown or tan body typically measuring 1.3 to 7.6 cm in length with a subaculear tooth or tubercle at the base of the stinger, a characteristic that is common to all Centruroides species (Figure).2 Some variability in size has been shown, with smaller scorpions found in increased elevations and cooler temperatures.1,3 Both C exilicauda and C sculpturatus are found in northern Mexico as well as the southwestern United States (eg, Arizona, New Mexico, Texas, California, Nevada).1 They have a preference for residing in or around trees and often are found on the underside of bark, stones, or tables as well as inside shoes or small cracks and crevices. Scorpions typically sting in self-defense, and stings commonly occur when humans attempt to move tables, put on shoes, or walk barefoot in scorpion-infested areas. Most stings occur from the end of spring through the end summer, but many may go unreported.1,4

Bark scorpion (Centruroides sculpturatus).

The venom of the Centruroides genus includes peptides and proteins that play a fundamental role in toxic activity by impairing potassium, sodium, and calcium ion channels.1,3 Toxins have been shown to be species specific, functioning either in capturing prey or deterring predators. Intraspecies variability in toxins has been demonstrated, which may complicate the production of adequate antivenin.3 Many have thought that C exilicauda Wood and C sculpturatus Ewing are the same species, and the names have been used synonymously in the past; however, genetic and biochemical studies of their venom components have shown that they are distinct species and that C sculpturatus is the more dangerous of the two.5 The median lethal dose 50% of C sculpturatus was found to be 22.7 μg in CD1 mice.6

Envenomation and Clinical Manifestations

Stings from C exilicauda and C sculpturatus have been shown to cause fatality in children more often than in adults.7 In the United States, Arizona has the highest frequency of serious symptoms of envenomation as well as the highest hospital and intensive care unit admission rates.6 Envenomation results in an immediate sharp burning pain followed by numbness.4 Wounds can produce some regional lymph node swelling, ecchymosis, paresthesia, and lymphangitis. More often than not, however, wounds have little to no inflammation and are characterized only by pain.4 The puncture wound is too small to be seen, and C exilicauda and C sculpturatus venom do not cause local tissue destruction, an important factor in distinguishing it from other scorpion envenomations.

More severe complications that may follow are caused by the neurotoxin released by Centruroides stings. The toxin components can increase the duration and amplitude of the neuronal action potential and enhance the release of neurotransmitters such as acetylcholine and norepinephrine.8 Stings can lead to cranial nerve dysfunction and somatic skeletal neuromuscular dysfunction as well as autonomic dysfunction, specifically salivation, fever, tongue and muscle fasciculations, opsoclonus, vomiting, bronchoconstriction, diaphoresis, nystagmus, blurred vision, slurred speech, hypertension, rhabdomyolysis, stridor, wheezing, aspiration, anaphylaxis, and tachycardia, leading to cardiac and respiratory compromise.4,8 Some patients have experienced a decreased sense of smell or hearing and decreased fine motor movements.7 Although pancreatitis may occur with scorpion stings, it is not common for C exilicauda.9 Comorbidities such as cardiac disease and substance use disorders contribute to prolonged length of hospital stay and poor outcome.8

Treatment

Most Centruroides stings can be managed at home, but patients with more serious symptoms and children younger than 2 years should be taken to a hospital for treatment.7 If a patient reports only pain but shows no other signs of neurotoxicity, observation and pain relief with rest, ice, and elevation is appropriate management. Patients with severe manifestations have been treated with various combinations of lorazepam, glycopyrrolate, ipratropium bromide, and ondansetron, but the only treatment definitively shown to decrease time to symptom abatement is antivenin.7 It has been demonstrated that C exilicauda and C sculpturatus antivenin is relatively safe.7 Most patients, especially adults, do not die from C exilicauda and C sculpturatus stings; therefore, antivenin more commonly is symptom abating than it is lifesaving.10 In children, time to symptom resolution was decreased to fewer than 4 hours with antivenin, and there is a lower rate of inpatient admission when antivenin is administered.4,10,11 There is a low incidence of anaphylactic reaction after antivenin, but there have been reported cases of self-limited serum sickness after antivenin use that generally can be managed with antihistamines and corticosteroids.4,7

References
  1. Gonzalez-Santillan E, Possani LD. North American scorpion species of public health importance with reappraisal of historical epidemiology. Acta Tropica. 2018;187:264-274.
  2. Goldsmith LA, Katz SI, Gilchrest BA, et al, eds. Fitzpatrick’s Dermatology in General Medicine. 8th ed. New York, NY: McGraw-Hill; 2012.
  3. Carcamo-Noriega EN, Olamendi-Portugal T, Restano-Cassulini R, et al. Intraspecific variation of Centruroides sculpturatus scorpion venom from two regions of Arizona. Arch Biochem Biophys. 2018;638:52-57.
  4. Kang AM, Brooks DE. Nationwide scorpion exposures reported to US Poison Control centers from 2005 to 2015. J Med Toxicol. 2017;13:158-165.
  5. Valdez-Cruz N, Dávila S, Licea A, et al. Biochemical, genetic and physiological characterization of venom components from two species of scorpions: Centruroides exilicauda Wood and Centruroides sculpturatus Ewing. Biochimie. 2004;86:387-396.
  6. Jiménez-Vargas JM, Quintero-Hernández V, Gonzáles-Morales L, et al. Design and expression of recombinant toxins from Mexican scorpions of the genus Centruroides for production of antivenoms. Toxicon. 2017;128:5-14.
  7. Hurst NB, Lipe DN, Karpen SR, et al. Centruroides sculpturatus envenomation in three adult patients requiring treatment with antivenom. Clin Toxicol (Phila). 2018;56:294-296.
  8. O’Connor A, Padilla-Jones A, Ruha A. Severe bark scorpion envenomation in adults. Clin Toxicol. 2018;56:170-174.
  9. Berg R, Tarantino M. Envenomation by the scorpion Centruroides exilicauda (C sculpturatus): severe and unusual manifestations. Pediatrics. 1991;87:930-933.
  10. LoVecchio F, McBride C. Scorpion envenomations in young children in central Arizona. J Toxicol Clin Toxicol. 2003;41:937-940.
  11. Rodrigo C, Gnanathasan A. Management of scorpion envenoming: a systematic review and meta-analysis of controlled clinical trials. Syst Rev. 2017;6:74.
Article PDF
Author and Disclosure Information

From the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Image is in the public domain.

Correspondence: Dirk M. Elston, MD, Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, 135 Rutledge Ave, MSC 578, Charleston, SC 29425-5780 (elstond@musc.edu).

Issue
Cutis - 105(5)
Publications
Topics
Page Number
239-240
Sections
Author and Disclosure Information

From the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Image is in the public domain.

Correspondence: Dirk M. Elston, MD, Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, 135 Rutledge Ave, MSC 578, Charleston, SC 29425-5780 (elstond@musc.edu).

Author and Disclosure Information

From the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Image is in the public domain.

Correspondence: Dirk M. Elston, MD, Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, 135 Rutledge Ave, MSC 578, Charleston, SC 29425-5780 (elstond@musc.edu).

Article PDF
Article PDF
Related Articles

Epidemiology and Identification

Centruroides is a common genus of bark scorpions in the United States with at least 21 species considered to be medically important, including the closely related Centruroides exilicauda and Centruroides sculpturatus.1 Scorpions can be recognized by a bulbous sac and pointed stinger at the end of a tail-like abdomen. They also have long lobsterlike pedipalps (pincers) for grasping their prey. Identifying characteristics for C exilicauda and C sculpturatus include a small, slender, yellow to light brown or tan body typically measuring 1.3 to 7.6 cm in length with a subaculear tooth or tubercle at the base of the stinger, a characteristic that is common to all Centruroides species (Figure).2 Some variability in size has been shown, with smaller scorpions found in increased elevations and cooler temperatures.1,3 Both C exilicauda and C sculpturatus are found in northern Mexico as well as the southwestern United States (eg, Arizona, New Mexico, Texas, California, Nevada).1 They have a preference for residing in or around trees and often are found on the underside of bark, stones, or tables as well as inside shoes or small cracks and crevices. Scorpions typically sting in self-defense, and stings commonly occur when humans attempt to move tables, put on shoes, or walk barefoot in scorpion-infested areas. Most stings occur from the end of spring through the end summer, but many may go unreported.1,4

Bark scorpion (Centruroides sculpturatus).

The venom of the Centruroides genus includes peptides and proteins that play a fundamental role in toxic activity by impairing potassium, sodium, and calcium ion channels.1,3 Toxins have been shown to be species specific, functioning either in capturing prey or deterring predators. Intraspecies variability in toxins has been demonstrated, which may complicate the production of adequate antivenin.3 Many have thought that C exilicauda Wood and C sculpturatus Ewing are the same species, and the names have been used synonymously in the past; however, genetic and biochemical studies of their venom components have shown that they are distinct species and that C sculpturatus is the more dangerous of the two.5 The median lethal dose 50% of C sculpturatus was found to be 22.7 μg in CD1 mice.6

Envenomation and Clinical Manifestations

Stings from C exilicauda and C sculpturatus have been shown to cause fatality in children more often than in adults.7 In the United States, Arizona has the highest frequency of serious symptoms of envenomation as well as the highest hospital and intensive care unit admission rates.6 Envenomation results in an immediate sharp burning pain followed by numbness.4 Wounds can produce some regional lymph node swelling, ecchymosis, paresthesia, and lymphangitis. More often than not, however, wounds have little to no inflammation and are characterized only by pain.4 The puncture wound is too small to be seen, and C exilicauda and C sculpturatus venom do not cause local tissue destruction, an important factor in distinguishing it from other scorpion envenomations.

More severe complications that may follow are caused by the neurotoxin released by Centruroides stings. The toxin components can increase the duration and amplitude of the neuronal action potential and enhance the release of neurotransmitters such as acetylcholine and norepinephrine.8 Stings can lead to cranial nerve dysfunction and somatic skeletal neuromuscular dysfunction as well as autonomic dysfunction, specifically salivation, fever, tongue and muscle fasciculations, opsoclonus, vomiting, bronchoconstriction, diaphoresis, nystagmus, blurred vision, slurred speech, hypertension, rhabdomyolysis, stridor, wheezing, aspiration, anaphylaxis, and tachycardia, leading to cardiac and respiratory compromise.4,8 Some patients have experienced a decreased sense of smell or hearing and decreased fine motor movements.7 Although pancreatitis may occur with scorpion stings, it is not common for C exilicauda.9 Comorbidities such as cardiac disease and substance use disorders contribute to prolonged length of hospital stay and poor outcome.8

Treatment

Most Centruroides stings can be managed at home, but patients with more serious symptoms and children younger than 2 years should be taken to a hospital for treatment.7 If a patient reports only pain but shows no other signs of neurotoxicity, observation and pain relief with rest, ice, and elevation is appropriate management. Patients with severe manifestations have been treated with various combinations of lorazepam, glycopyrrolate, ipratropium bromide, and ondansetron, but the only treatment definitively shown to decrease time to symptom abatement is antivenin.7 It has been demonstrated that C exilicauda and C sculpturatus antivenin is relatively safe.7 Most patients, especially adults, do not die from C exilicauda and C sculpturatus stings; therefore, antivenin more commonly is symptom abating than it is lifesaving.10 In children, time to symptom resolution was decreased to fewer than 4 hours with antivenin, and there is a lower rate of inpatient admission when antivenin is administered.4,10,11 There is a low incidence of anaphylactic reaction after antivenin, but there have been reported cases of self-limited serum sickness after antivenin use that generally can be managed with antihistamines and corticosteroids.4,7

Epidemiology and Identification

Centruroides is a common genus of bark scorpions in the United States with at least 21 species considered to be medically important, including the closely related Centruroides exilicauda and Centruroides sculpturatus.1 Scorpions can be recognized by a bulbous sac and pointed stinger at the end of a tail-like abdomen. They also have long lobsterlike pedipalps (pincers) for grasping their prey. Identifying characteristics for C exilicauda and C sculpturatus include a small, slender, yellow to light brown or tan body typically measuring 1.3 to 7.6 cm in length with a subaculear tooth or tubercle at the base of the stinger, a characteristic that is common to all Centruroides species (Figure).2 Some variability in size has been shown, with smaller scorpions found in increased elevations and cooler temperatures.1,3 Both C exilicauda and C sculpturatus are found in northern Mexico as well as the southwestern United States (eg, Arizona, New Mexico, Texas, California, Nevada).1 They have a preference for residing in or around trees and often are found on the underside of bark, stones, or tables as well as inside shoes or small cracks and crevices. Scorpions typically sting in self-defense, and stings commonly occur when humans attempt to move tables, put on shoes, or walk barefoot in scorpion-infested areas. Most stings occur from the end of spring through the end summer, but many may go unreported.1,4

Bark scorpion (Centruroides sculpturatus).

The venom of the Centruroides genus includes peptides and proteins that play a fundamental role in toxic activity by impairing potassium, sodium, and calcium ion channels.1,3 Toxins have been shown to be species specific, functioning either in capturing prey or deterring predators. Intraspecies variability in toxins has been demonstrated, which may complicate the production of adequate antivenin.3 Many have thought that C exilicauda Wood and C sculpturatus Ewing are the same species, and the names have been used synonymously in the past; however, genetic and biochemical studies of their venom components have shown that they are distinct species and that C sculpturatus is the more dangerous of the two.5 The median lethal dose 50% of C sculpturatus was found to be 22.7 μg in CD1 mice.6

Envenomation and Clinical Manifestations

Stings from C exilicauda and C sculpturatus have been shown to cause fatality in children more often than in adults.7 In the United States, Arizona has the highest frequency of serious symptoms of envenomation as well as the highest hospital and intensive care unit admission rates.6 Envenomation results in an immediate sharp burning pain followed by numbness.4 Wounds can produce some regional lymph node swelling, ecchymosis, paresthesia, and lymphangitis. More often than not, however, wounds have little to no inflammation and are characterized only by pain.4 The puncture wound is too small to be seen, and C exilicauda and C sculpturatus venom do not cause local tissue destruction, an important factor in distinguishing it from other scorpion envenomations.

More severe complications that may follow are caused by the neurotoxin released by Centruroides stings. The toxin components can increase the duration and amplitude of the neuronal action potential and enhance the release of neurotransmitters such as acetylcholine and norepinephrine.8 Stings can lead to cranial nerve dysfunction and somatic skeletal neuromuscular dysfunction as well as autonomic dysfunction, specifically salivation, fever, tongue and muscle fasciculations, opsoclonus, vomiting, bronchoconstriction, diaphoresis, nystagmus, blurred vision, slurred speech, hypertension, rhabdomyolysis, stridor, wheezing, aspiration, anaphylaxis, and tachycardia, leading to cardiac and respiratory compromise.4,8 Some patients have experienced a decreased sense of smell or hearing and decreased fine motor movements.7 Although pancreatitis may occur with scorpion stings, it is not common for C exilicauda.9 Comorbidities such as cardiac disease and substance use disorders contribute to prolonged length of hospital stay and poor outcome.8

Treatment

Most Centruroides stings can be managed at home, but patients with more serious symptoms and children younger than 2 years should be taken to a hospital for treatment.7 If a patient reports only pain but shows no other signs of neurotoxicity, observation and pain relief with rest, ice, and elevation is appropriate management. Patients with severe manifestations have been treated with various combinations of lorazepam, glycopyrrolate, ipratropium bromide, and ondansetron, but the only treatment definitively shown to decrease time to symptom abatement is antivenin.7 It has been demonstrated that C exilicauda and C sculpturatus antivenin is relatively safe.7 Most patients, especially adults, do not die from C exilicauda and C sculpturatus stings; therefore, antivenin more commonly is symptom abating than it is lifesaving.10 In children, time to symptom resolution was decreased to fewer than 4 hours with antivenin, and there is a lower rate of inpatient admission when antivenin is administered.4,10,11 There is a low incidence of anaphylactic reaction after antivenin, but there have been reported cases of self-limited serum sickness after antivenin use that generally can be managed with antihistamines and corticosteroids.4,7

References
  1. Gonzalez-Santillan E, Possani LD. North American scorpion species of public health importance with reappraisal of historical epidemiology. Acta Tropica. 2018;187:264-274.
  2. Goldsmith LA, Katz SI, Gilchrest BA, et al, eds. Fitzpatrick’s Dermatology in General Medicine. 8th ed. New York, NY: McGraw-Hill; 2012.
  3. Carcamo-Noriega EN, Olamendi-Portugal T, Restano-Cassulini R, et al. Intraspecific variation of Centruroides sculpturatus scorpion venom from two regions of Arizona. Arch Biochem Biophys. 2018;638:52-57.
  4. Kang AM, Brooks DE. Nationwide scorpion exposures reported to US Poison Control centers from 2005 to 2015. J Med Toxicol. 2017;13:158-165.
  5. Valdez-Cruz N, Dávila S, Licea A, et al. Biochemical, genetic and physiological characterization of venom components from two species of scorpions: Centruroides exilicauda Wood and Centruroides sculpturatus Ewing. Biochimie. 2004;86:387-396.
  6. Jiménez-Vargas JM, Quintero-Hernández V, Gonzáles-Morales L, et al. Design and expression of recombinant toxins from Mexican scorpions of the genus Centruroides for production of antivenoms. Toxicon. 2017;128:5-14.
  7. Hurst NB, Lipe DN, Karpen SR, et al. Centruroides sculpturatus envenomation in three adult patients requiring treatment with antivenom. Clin Toxicol (Phila). 2018;56:294-296.
  8. O’Connor A, Padilla-Jones A, Ruha A. Severe bark scorpion envenomation in adults. Clin Toxicol. 2018;56:170-174.
  9. Berg R, Tarantino M. Envenomation by the scorpion Centruroides exilicauda (C sculpturatus): severe and unusual manifestations. Pediatrics. 1991;87:930-933.
  10. LoVecchio F, McBride C. Scorpion envenomations in young children in central Arizona. J Toxicol Clin Toxicol. 2003;41:937-940.
  11. Rodrigo C, Gnanathasan A. Management of scorpion envenoming: a systematic review and meta-analysis of controlled clinical trials. Syst Rev. 2017;6:74.
References
  1. Gonzalez-Santillan E, Possani LD. North American scorpion species of public health importance with reappraisal of historical epidemiology. Acta Tropica. 2018;187:264-274.
  2. Goldsmith LA, Katz SI, Gilchrest BA, et al, eds. Fitzpatrick’s Dermatology in General Medicine. 8th ed. New York, NY: McGraw-Hill; 2012.
  3. Carcamo-Noriega EN, Olamendi-Portugal T, Restano-Cassulini R, et al. Intraspecific variation of Centruroides sculpturatus scorpion venom from two regions of Arizona. Arch Biochem Biophys. 2018;638:52-57.
  4. Kang AM, Brooks DE. Nationwide scorpion exposures reported to US Poison Control centers from 2005 to 2015. J Med Toxicol. 2017;13:158-165.
  5. Valdez-Cruz N, Dávila S, Licea A, et al. Biochemical, genetic and physiological characterization of venom components from two species of scorpions: Centruroides exilicauda Wood and Centruroides sculpturatus Ewing. Biochimie. 2004;86:387-396.
  6. Jiménez-Vargas JM, Quintero-Hernández V, Gonzáles-Morales L, et al. Design and expression of recombinant toxins from Mexican scorpions of the genus Centruroides for production of antivenoms. Toxicon. 2017;128:5-14.
  7. Hurst NB, Lipe DN, Karpen SR, et al. Centruroides sculpturatus envenomation in three adult patients requiring treatment with antivenom. Clin Toxicol (Phila). 2018;56:294-296.
  8. O’Connor A, Padilla-Jones A, Ruha A. Severe bark scorpion envenomation in adults. Clin Toxicol. 2018;56:170-174.
  9. Berg R, Tarantino M. Envenomation by the scorpion Centruroides exilicauda (C sculpturatus): severe and unusual manifestations. Pediatrics. 1991;87:930-933.
  10. LoVecchio F, McBride C. Scorpion envenomations in young children in central Arizona. J Toxicol Clin Toxicol. 2003;41:937-940.
  11. Rodrigo C, Gnanathasan A. Management of scorpion envenoming: a systematic review and meta-analysis of controlled clinical trials. Syst Rev. 2017;6:74.
Issue
Cutis - 105(5)
Issue
Cutis - 105(5)
Page Number
239-240
Page Number
239-240
Publications
Publications
Topics
Article Type
Display Headline
What’s Eating You? Bark Scorpions (Centruroides exilicauda and Centruroides sculpturatus)
Display Headline
What’s Eating You? Bark Scorpions (Centruroides exilicauda and Centruroides sculpturatus)
Sections
Inside the Article

Practice Points

  • Centruroides scorpions can inflict painful stings.
  • Children are at greatest risk for systemic toxicity.
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
Article PDF Media

Tender White Lesions on the Groin

Article Type
Changed
Wed, 04/29/2020 - 16:22
Display Headline
Tender White Lesions on the Groin

The Diagnosis: Candidal Intertrigo  

The biopsy confirmed a diagnosis of severe hyperkeratotic candidal intertrigo with no evidence of Hailey-Hailey disease. Hematoxylin and eosin- stained sections demonstrated irregular acanthosis and variable spongiosis. The stratum corneum was predominantly orthokeratotic with overlying psuedohyphae and yeast fungal elements (Figure 1). 

Figure 1. Irregular acanthosis and variable spongiosis. The stratum corneum was predominantly orthokeratotic. On higher magnification, yeast forms and pseudohyphae diagnostic of Candida albicans were appreciated (H&E, original magnification ×100; inset: H&E oil immersion, original magnification ×1000).

Hyperimmunoglobulinemia E syndrome (HIES), also known as hyper-IgE syndrome or Job syndrome, is a rare immunodeficiency disorder characterized by an eczematous dermatitis-like rash, recurrent skin and lung abscesses, eosinophilia, and elevated serum IgE. Facial asymmetry, prominent forehead, deep-set eyes, broad nose, and roughened facial skin with large pores are characteristic of the sporadic and autosomal-recessive forms. Other common findings include retained primary teeth, hyperextensible joints, and recurrent mucocutaneous candidiasis.

Although autosomal-dominant and autosomal-recessive inheritance patterns exist, sporadic mutations are the most common cause of HIES.2 Several genes have been implicated depending on the inheritance pattern. The majority of autosomal-dominant cases are associated with inactivating STAT3 (signal transducer and activator of transcription 3) mutations, whereas the majority of autosomal-recessive cases are associated with inactivating DOCK8 (dedicator of cytokinesis 8) mutations.1 Ultimately, all of these mutations lead to an impaired helper T cell (TH17) response, which is crucial for clearing fungal and extracellular bacterial infections.3  

Skin eruptions typically are the first manifestation of HIES; they appear within the first week to month of life as papulopustular eruptions on the face and scalp and rapidly generalize to the rest of the body, favoring the shoulders, arms, chest, and buttocks. The pustules then coalesce into crusted plaques that resemble atopic dermatitis, frequently with superimposed Staphylococcus aureus infection. On microscopy, the pustules are folliculocentric and often contain eosinophils, whereas the plaques may contain intraepidermal collections of eosinophils.1 

Mucocutaneous candidiasis is seen in approximately 60% of HIES cases and is closely linked to STAT3 inactivating mutations.3 Histologically, there is marked acanthosis with neutrophil exocytosis and abundant yeast and pseudohyphal forms within the stratum corneum (Figure 2).4 Cutaneous candidal infections typically require both oral and topical antifungal agents to clear the infection.3 Most cases of mucocutaneous candidiasis are caused by Candida albicans; however, other known culprits include Candida glabrata, Candida tropicalis, Candida parapsilosis, and Candida krusei.5,6 Of note, species identification and antifungal susceptibility studies may be useful in refractory cases, especially with C glabrata, which is known to acquire resistance to azoles, such as fluconazole, with emerging resistance to echinocandins.6  

Figure 2. A, The epidermis displayed irregular acanthosis and variable spongiosis. The stratum corneum was predominantly orthokeratotic with overlying fungal elements (H&E, original magnification ×100). B, Closer view of the cornified layer showed pseudohyphae and budding yeast (H&E oil immersion, original magnification ×1000).

The differential diagnosis of this groin eruption included Hailey-Hailey disease; pemphigus vegetans, Hallopeau type; tinea cruris; and inverse psoriasis. Hailey-Hailey disease can be complicated by a superimposed candidal infection with similar clinical features, and biopsy may be required for definitive diagnosis. Hailey-Hailey disease typically presents with macerated fissured plaques that resemble macerated tissue paper with red fissures (Figure 3). Biopsy confirms full-thickness acantholysis resembling a dilapidated brick wall with minimal dyskeratosis.1 Pemphigus vegetans is a localized variant of pemphigus vulgaris with a predilection for flexural surfaces. The lesions progress to vegetating erosive plaques.4 The Hallopeau type often is studded with pustules and typically remains more localized than the Neumann type. Direct immunofluorescence demonstrates intercellular deposition of IgG and C3, and routine sections characteristically show pseudoepitheliomatous hyperplasia with intraepidermal eosinophilic microabscesses.1,4 Tinea cruris is characterized by erythematous annular lesions with raised scaly borders spreading down the inner thighs.7 The epidermis is variably spongiotic with parakeratosis, and neutrophils often present in a layered stratum corneum with basketweave keratin above a layer of more compact and eosinophilic keratin. Fungal stains, such as periodic acid-Schiff, will highlight the fungal hyphae within the stratum corneum. The inguinal folds are a typical location for inverse psoriasis, which generally appears as thin, sharply demarcated, shiny red plaques with less scale than plaque psoriasis.1 Psoriasiform hyperplasia with a diminished granular layer and tortuous papillary dermal vessels would be expected histologically.1 

Figure 3. Hailey-Hailey disease with superimposed candidal infection. White macerated scale was seen overlying a large pink plaque and a smaller satellite pink scaly plaque in the groin.

References
  1. James WD, Berger TG, Elston DM. Andrews' Diseases of the Skin. 12th ed. Philadelphia, PA: Elsevier; 2016. 
  2. Schwartz RA, Tarlow MM. Dermatologic manifestations of Job syndrome. Medscape website. https://emedicine.medscape.com/article/1050852-overview. Updated April 22, 2019. Accessed March 28, 2020. 
  3. Minegishi Y, Saito M. Cutaneous manifestations of hyper IgE syndrome. Allergol Int. 2012;61:191-196. 
  4. Patterson JW. Weedon's Skin Pathology. 4th ed. China: Churchill Livingstone Elsevier; 2016. 
  5. Pappas PG, Kauffman CA, Andes DR, et al. Executive summary: clinical practice guideline for the management of candidiasis: 2016 update by the Infectious Diseases Society of America. Clin Infect Dis. 2016;62:409-417. 
  6. Center for Disease Control and Prevention. Antifungal resistance. https://www.cdc.gov/fungal/antifungal-resistance.html. Updated March 17, 2020. Accessed April 20, 2020.  
  7. Tinea cruris. DermNet NZ website. https://www.dermnetnz.org/topics/tinea-cruris/. Published 2003. Accessed March 28, 2020. 
Article PDF
Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Drs. O’Connor and Forcucci are from the Department of Pathology and Laboratory Medicine, and Drs. Zimmer and Elston are from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Heather M. O’Connor, DO, Medical University of South Carolina, Department of Pathology and Laboratory Medicine, 171 Ashley Ave, MSC908, Charleston, SC 29425 (oaconnor@musc.edu).

Issue
Cutis - 105(4)
Publications
Topics
Page Number
E28-E30
Sections
Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Drs. O’Connor and Forcucci are from the Department of Pathology and Laboratory Medicine, and Drs. Zimmer and Elston are from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Heather M. O’Connor, DO, Medical University of South Carolina, Department of Pathology and Laboratory Medicine, 171 Ashley Ave, MSC908, Charleston, SC 29425 (oaconnor@musc.edu).

Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Drs. O’Connor and Forcucci are from the Department of Pathology and Laboratory Medicine, and Drs. Zimmer and Elston are from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Heather M. O’Connor, DO, Medical University of South Carolina, Department of Pathology and Laboratory Medicine, 171 Ashley Ave, MSC908, Charleston, SC 29425 (oaconnor@musc.edu).

Article PDF
Article PDF
Related Articles

The Diagnosis: Candidal Intertrigo  

The biopsy confirmed a diagnosis of severe hyperkeratotic candidal intertrigo with no evidence of Hailey-Hailey disease. Hematoxylin and eosin- stained sections demonstrated irregular acanthosis and variable spongiosis. The stratum corneum was predominantly orthokeratotic with overlying psuedohyphae and yeast fungal elements (Figure 1). 

Figure 1. Irregular acanthosis and variable spongiosis. The stratum corneum was predominantly orthokeratotic. On higher magnification, yeast forms and pseudohyphae diagnostic of Candida albicans were appreciated (H&E, original magnification ×100; inset: H&E oil immersion, original magnification ×1000).

Hyperimmunoglobulinemia E syndrome (HIES), also known as hyper-IgE syndrome or Job syndrome, is a rare immunodeficiency disorder characterized by an eczematous dermatitis-like rash, recurrent skin and lung abscesses, eosinophilia, and elevated serum IgE. Facial asymmetry, prominent forehead, deep-set eyes, broad nose, and roughened facial skin with large pores are characteristic of the sporadic and autosomal-recessive forms. Other common findings include retained primary teeth, hyperextensible joints, and recurrent mucocutaneous candidiasis.

Although autosomal-dominant and autosomal-recessive inheritance patterns exist, sporadic mutations are the most common cause of HIES.2 Several genes have been implicated depending on the inheritance pattern. The majority of autosomal-dominant cases are associated with inactivating STAT3 (signal transducer and activator of transcription 3) mutations, whereas the majority of autosomal-recessive cases are associated with inactivating DOCK8 (dedicator of cytokinesis 8) mutations.1 Ultimately, all of these mutations lead to an impaired helper T cell (TH17) response, which is crucial for clearing fungal and extracellular bacterial infections.3  

Skin eruptions typically are the first manifestation of HIES; they appear within the first week to month of life as papulopustular eruptions on the face and scalp and rapidly generalize to the rest of the body, favoring the shoulders, arms, chest, and buttocks. The pustules then coalesce into crusted plaques that resemble atopic dermatitis, frequently with superimposed Staphylococcus aureus infection. On microscopy, the pustules are folliculocentric and often contain eosinophils, whereas the plaques may contain intraepidermal collections of eosinophils.1 

Mucocutaneous candidiasis is seen in approximately 60% of HIES cases and is closely linked to STAT3 inactivating mutations.3 Histologically, there is marked acanthosis with neutrophil exocytosis and abundant yeast and pseudohyphal forms within the stratum corneum (Figure 2).4 Cutaneous candidal infections typically require both oral and topical antifungal agents to clear the infection.3 Most cases of mucocutaneous candidiasis are caused by Candida albicans; however, other known culprits include Candida glabrata, Candida tropicalis, Candida parapsilosis, and Candida krusei.5,6 Of note, species identification and antifungal susceptibility studies may be useful in refractory cases, especially with C glabrata, which is known to acquire resistance to azoles, such as fluconazole, with emerging resistance to echinocandins.6  

Figure 2. A, The epidermis displayed irregular acanthosis and variable spongiosis. The stratum corneum was predominantly orthokeratotic with overlying fungal elements (H&E, original magnification ×100). B, Closer view of the cornified layer showed pseudohyphae and budding yeast (H&E oil immersion, original magnification ×1000).

The differential diagnosis of this groin eruption included Hailey-Hailey disease; pemphigus vegetans, Hallopeau type; tinea cruris; and inverse psoriasis. Hailey-Hailey disease can be complicated by a superimposed candidal infection with similar clinical features, and biopsy may be required for definitive diagnosis. Hailey-Hailey disease typically presents with macerated fissured plaques that resemble macerated tissue paper with red fissures (Figure 3). Biopsy confirms full-thickness acantholysis resembling a dilapidated brick wall with minimal dyskeratosis.1 Pemphigus vegetans is a localized variant of pemphigus vulgaris with a predilection for flexural surfaces. The lesions progress to vegetating erosive plaques.4 The Hallopeau type often is studded with pustules and typically remains more localized than the Neumann type. Direct immunofluorescence demonstrates intercellular deposition of IgG and C3, and routine sections characteristically show pseudoepitheliomatous hyperplasia with intraepidermal eosinophilic microabscesses.1,4 Tinea cruris is characterized by erythematous annular lesions with raised scaly borders spreading down the inner thighs.7 The epidermis is variably spongiotic with parakeratosis, and neutrophils often present in a layered stratum corneum with basketweave keratin above a layer of more compact and eosinophilic keratin. Fungal stains, such as periodic acid-Schiff, will highlight the fungal hyphae within the stratum corneum. The inguinal folds are a typical location for inverse psoriasis, which generally appears as thin, sharply demarcated, shiny red plaques with less scale than plaque psoriasis.1 Psoriasiform hyperplasia with a diminished granular layer and tortuous papillary dermal vessels would be expected histologically.1 

Figure 3. Hailey-Hailey disease with superimposed candidal infection. White macerated scale was seen overlying a large pink plaque and a smaller satellite pink scaly plaque in the groin.

The Diagnosis: Candidal Intertrigo  

The biopsy confirmed a diagnosis of severe hyperkeratotic candidal intertrigo with no evidence of Hailey-Hailey disease. Hematoxylin and eosin- stained sections demonstrated irregular acanthosis and variable spongiosis. The stratum corneum was predominantly orthokeratotic with overlying psuedohyphae and yeast fungal elements (Figure 1). 

Figure 1. Irregular acanthosis and variable spongiosis. The stratum corneum was predominantly orthokeratotic. On higher magnification, yeast forms and pseudohyphae diagnostic of Candida albicans were appreciated (H&E, original magnification ×100; inset: H&E oil immersion, original magnification ×1000).

Hyperimmunoglobulinemia E syndrome (HIES), also known as hyper-IgE syndrome or Job syndrome, is a rare immunodeficiency disorder characterized by an eczematous dermatitis-like rash, recurrent skin and lung abscesses, eosinophilia, and elevated serum IgE. Facial asymmetry, prominent forehead, deep-set eyes, broad nose, and roughened facial skin with large pores are characteristic of the sporadic and autosomal-recessive forms. Other common findings include retained primary teeth, hyperextensible joints, and recurrent mucocutaneous candidiasis.

Although autosomal-dominant and autosomal-recessive inheritance patterns exist, sporadic mutations are the most common cause of HIES.2 Several genes have been implicated depending on the inheritance pattern. The majority of autosomal-dominant cases are associated with inactivating STAT3 (signal transducer and activator of transcription 3) mutations, whereas the majority of autosomal-recessive cases are associated with inactivating DOCK8 (dedicator of cytokinesis 8) mutations.1 Ultimately, all of these mutations lead to an impaired helper T cell (TH17) response, which is crucial for clearing fungal and extracellular bacterial infections.3  

Skin eruptions typically are the first manifestation of HIES; they appear within the first week to month of life as papulopustular eruptions on the face and scalp and rapidly generalize to the rest of the body, favoring the shoulders, arms, chest, and buttocks. The pustules then coalesce into crusted plaques that resemble atopic dermatitis, frequently with superimposed Staphylococcus aureus infection. On microscopy, the pustules are folliculocentric and often contain eosinophils, whereas the plaques may contain intraepidermal collections of eosinophils.1 

Mucocutaneous candidiasis is seen in approximately 60% of HIES cases and is closely linked to STAT3 inactivating mutations.3 Histologically, there is marked acanthosis with neutrophil exocytosis and abundant yeast and pseudohyphal forms within the stratum corneum (Figure 2).4 Cutaneous candidal infections typically require both oral and topical antifungal agents to clear the infection.3 Most cases of mucocutaneous candidiasis are caused by Candida albicans; however, other known culprits include Candida glabrata, Candida tropicalis, Candida parapsilosis, and Candida krusei.5,6 Of note, species identification and antifungal susceptibility studies may be useful in refractory cases, especially with C glabrata, which is known to acquire resistance to azoles, such as fluconazole, with emerging resistance to echinocandins.6  

Figure 2. A, The epidermis displayed irregular acanthosis and variable spongiosis. The stratum corneum was predominantly orthokeratotic with overlying fungal elements (H&E, original magnification ×100). B, Closer view of the cornified layer showed pseudohyphae and budding yeast (H&E oil immersion, original magnification ×1000).

The differential diagnosis of this groin eruption included Hailey-Hailey disease; pemphigus vegetans, Hallopeau type; tinea cruris; and inverse psoriasis. Hailey-Hailey disease can be complicated by a superimposed candidal infection with similar clinical features, and biopsy may be required for definitive diagnosis. Hailey-Hailey disease typically presents with macerated fissured plaques that resemble macerated tissue paper with red fissures (Figure 3). Biopsy confirms full-thickness acantholysis resembling a dilapidated brick wall with minimal dyskeratosis.1 Pemphigus vegetans is a localized variant of pemphigus vulgaris with a predilection for flexural surfaces. The lesions progress to vegetating erosive plaques.4 The Hallopeau type often is studded with pustules and typically remains more localized than the Neumann type. Direct immunofluorescence demonstrates intercellular deposition of IgG and C3, and routine sections characteristically show pseudoepitheliomatous hyperplasia with intraepidermal eosinophilic microabscesses.1,4 Tinea cruris is characterized by erythematous annular lesions with raised scaly borders spreading down the inner thighs.7 The epidermis is variably spongiotic with parakeratosis, and neutrophils often present in a layered stratum corneum with basketweave keratin above a layer of more compact and eosinophilic keratin. Fungal stains, such as periodic acid-Schiff, will highlight the fungal hyphae within the stratum corneum. The inguinal folds are a typical location for inverse psoriasis, which generally appears as thin, sharply demarcated, shiny red plaques with less scale than plaque psoriasis.1 Psoriasiform hyperplasia with a diminished granular layer and tortuous papillary dermal vessels would be expected histologically.1 

Figure 3. Hailey-Hailey disease with superimposed candidal infection. White macerated scale was seen overlying a large pink plaque and a smaller satellite pink scaly plaque in the groin.

References
  1. James WD, Berger TG, Elston DM. Andrews' Diseases of the Skin. 12th ed. Philadelphia, PA: Elsevier; 2016. 
  2. Schwartz RA, Tarlow MM. Dermatologic manifestations of Job syndrome. Medscape website. https://emedicine.medscape.com/article/1050852-overview. Updated April 22, 2019. Accessed March 28, 2020. 
  3. Minegishi Y, Saito M. Cutaneous manifestations of hyper IgE syndrome. Allergol Int. 2012;61:191-196. 
  4. Patterson JW. Weedon's Skin Pathology. 4th ed. China: Churchill Livingstone Elsevier; 2016. 
  5. Pappas PG, Kauffman CA, Andes DR, et al. Executive summary: clinical practice guideline for the management of candidiasis: 2016 update by the Infectious Diseases Society of America. Clin Infect Dis. 2016;62:409-417. 
  6. Center for Disease Control and Prevention. Antifungal resistance. https://www.cdc.gov/fungal/antifungal-resistance.html. Updated March 17, 2020. Accessed April 20, 2020.  
  7. Tinea cruris. DermNet NZ website. https://www.dermnetnz.org/topics/tinea-cruris/. Published 2003. Accessed March 28, 2020. 
References
  1. James WD, Berger TG, Elston DM. Andrews' Diseases of the Skin. 12th ed. Philadelphia, PA: Elsevier; 2016. 
  2. Schwartz RA, Tarlow MM. Dermatologic manifestations of Job syndrome. Medscape website. https://emedicine.medscape.com/article/1050852-overview. Updated April 22, 2019. Accessed March 28, 2020. 
  3. Minegishi Y, Saito M. Cutaneous manifestations of hyper IgE syndrome. Allergol Int. 2012;61:191-196. 
  4. Patterson JW. Weedon's Skin Pathology. 4th ed. China: Churchill Livingstone Elsevier; 2016. 
  5. Pappas PG, Kauffman CA, Andes DR, et al. Executive summary: clinical practice guideline for the management of candidiasis: 2016 update by the Infectious Diseases Society of America. Clin Infect Dis. 2016;62:409-417. 
  6. Center for Disease Control and Prevention. Antifungal resistance. https://www.cdc.gov/fungal/antifungal-resistance.html. Updated March 17, 2020. Accessed April 20, 2020.  
  7. Tinea cruris. DermNet NZ website. https://www.dermnetnz.org/topics/tinea-cruris/. Published 2003. Accessed March 28, 2020. 
Issue
Cutis - 105(4)
Issue
Cutis - 105(4)
Page Number
E28-E30
Page Number
E28-E30
Publications
Publications
Topics
Article Type
Display Headline
Tender White Lesions on the Groin
Display Headline
Tender White Lesions on the Groin
Sections
Questionnaire Body

A 28-year-old man with a history of hyperimmunoglobulinemia E syndrome (previously known as Job syndrome), coarse facial features, and multiple skin and soft tissue infections presented to the university dermatology clinic with persistent white, macerated, fissured groin plaques that were present for months. The lesions were tender and pruritic with a burning sensation. Treatment with topical terbinafine and oral fluconazole was attempted without resolution of the eruption. A biopsy of the groin lesion was performed.  

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 04/29/2020 - 10:00
Un-Gate On Date
Wed, 04/29/2020 - 10:00
Use ProPublica
CFC Schedule Remove Status
Wed, 04/29/2020 - 10:00
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Climate Change and Expansion of Tick Geography

Article Type
Changed
Mon, 04/13/2020 - 11:15
Display Headline
Climate Change and Expansion of Tick Geography

 

The expanding range of tick-borne diseases is a growing problem worldwide. Climate change plays a preeminent role in the expansion of tick species, especially for southern ticks in the United States such as Amblyomma species, which have introduced new pathogens to northern states.1-5 In addition to well-known tick-borne diseases, Amblyomma ticks have been implicated in the spread of emerging severe and potentially fatal viral illnesses, including Bourbon virus and Heartland virus.6 The increasing range of Amblyomma ticks also exposes new populations to tick-induced meat allergy (alpha-gal) syndrome, whereby development of specific IgE antibodies to the oligosaccharide galactose-alpha-1,3-galactose (alpha-gal) following tick bites results in severe allergic responses to consumption of beef, pork, and lamb.7

Amblyomma ticks have now been identified close to the Canadian border in Michigan and New York, and predictions of continued climate change raise the possibility of northward range expansion into all provinces of Canada from Alberta to Newfoundland and Labrador during the coming decades.8,9 Additional factors that contribute to the expanding range of many tick species include international travel, migratory patterns of birds, competition, and natural predators such as fire ants that feed on tick eggs and influence the feeding behavior of adults.10

Traditional methods of tick identification rely on gross morphology, including the presence of festoons, shape of the coxae where the legs attach, and markings on the hard overlying scutum. More recently, molecular identification has improved tick identification, leading to more accurate assessment of tick prevalence. These modern identification studies include analysis of 16S ribosomal DNA (rDNA), 12S rDNA, and ITS1 rDNA, and ITS2 rDNA genes.11

The spread of tick vectors has huge public health implications, and better methods to control tick populations are needed.12 New acaricides and growth regulators are being developed,13 and early spring applications of acaricides such as bifenthrin can suppress nymphs prior to the initiation of host-seeking activity.14 Controlled burns within tick habitats have proved helpful in reducing the risk for vector-borne disease.15,16 Personal protection is best accomplished with the use of a repellent together with clothing impregnated with an acaricide such as permethrin.17 Efforts to slow climate change and continued surveillance for the spread of tick vectors is urgently needed.

References
  1. Sanchez-Vicente S, Tagliafierro T, Coleman JL, et al. Polymicrobial nature of tick-borne diseases [published online September 10, 2019]. MBio. doi:10.1128/mBio.02055-19.
  2. Raghavan RK, Peterson AT, Cobos ME, et al. Current and future distribution of the Lone Star tick, Amblyomma americanum (L.) (Acari: Ixodidae) in North America. PLoS One. 2019;14:e0209082.
  3. Stafford KC 3rd, Molaei G, Little EAH, et al. Distribution and establishment of the Lone Star tick in Connecticut and implications for range expansion and public health. J Med Entomol. 2018;25:1561-1568.
  4. Gilliam ME, Rechkemmer WT, McCravy KW, et al. The influence of prescribed fire, habitat, and weather on Amblyomma americanum (Ixodida: Ixodidae) in West-Central Illinois, USA [published online March 22, 2018]. Insects. doi:10.3390/insects9020036.
  5. Sonenshine DE. Range expansion of tick disease vectors in North America: implications for spread of tick-borne disease [published online March 9, 2018]. Int J Environ Res Public Health. doi:10.3390/ijerph15030478.
  6. Savage HM, Godsey MS Jr, Panella NA, et al. Surveillance for tick-borne viruses near the location of a fatal human case of Bourbon virus (family Orthomyxoviridae: genus Thogotovirus) in eastern Kansas, 2015. J Med Entomol. 2018;55:701-705.
  7. Crispell G, Commins SP, Archer-Hartman SA, et al. Discovery of alpha-gal-containing antigens in North American tick species believed to induce red meat allergy. Front Immunol. 2019;10:1056.
  8. Gasmi S, Bouchard C, Ogden NH, et al. Evidence for increasing densities and geographic ranges of tick species of public health significance other than Ixodes scapularis in Québec, Canada. PLoS One. 2018;13:e0201924.
  9. Sagurova I, Ludwig A, Ogden NH, et al. Predicted northward expansion of the geographic range of the tick vector Amblyomma americanum in North America under future climate conditions. Environ Health Perspect. 2019;127:107014.
  10. Kjeldgaard MK, Takano OM, Bockoven AA, et al. Red imported fire ant (Solenopsis invicta) aggression influences the behavior of three hard tick species. Exp Appl Acarol. 2019;79:87-97.
  11. Abouelhassan EM, El-Gawady HM, Abdel-Aal AA, et al. Comparison of some molecular markers for tick species identification. J Arthropod Borne Dis. 2019;13:153-164.
  12. Jordan RA, Egizi A. The growing importance of lone star ticks in a Lyme disease endemic county: passive tick surveillance in Monmouth County, NJ, 2006–2016. PLoS One. 2019;14:e0211778.
  13. Showler AT, Donahue WA, Harlien JL, et al. Efficacy of novaluron + pyriproxyfen (Tekko Pro) insect growth regulators against Amblyomma americanum (Acari: Ixodidae), Rhipicephalus (Boophilus) annulatus, Rhipicephalus (Boophilus) microplus, and Rhipicephalus sanguineus. J Med Entomol. 2019;56:1338-1345.
  14. Schulze TL, Jordan RA. Early season applications of bifenthrin suppress host-seeking Ixodes scapularis and Amblyomma americanum (Acari: Ixodidae) nymphs [published online November 26, 2019]. J Med Entomol. doi:10.1093/jme/tjz202.
  15. Hodo CL, Forgacs D, Auckland LD, et al. Presence of diverse Rickettsia spp. and absence of Borrelia burgdorferi sensu lato in ticks in an East Texas forest with reduced tick density associated with controlled burns. Ticks Tick Borne Dis. 2020;11:101310.
  16. Gleim ER, Zemtsova GE, Berghaus RD, et al. Frequent prescribed fires can reduce risk of tick-borne diseases. Sci Rep. 2019;9:9974.
  17. Prose R, Breuner NE, Johnson TL, et al. Contact irritancy and toxicity of permethrin-treated clothing for Ixodes scapularis, Amblyomma americanum, and Dermacentor variabilis ticks (Acari: Ixodidae). J Med Entomol. 2018;55:1217-1224.
Article PDF
Author and Disclosure Information

 

From the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The author reports no conflict of interest.

Correspondence: Dirk M. Elston, MD, 135 Rutledge Ave, MSC 578, Charleston, SC 29425-5780 (elstond@musc.edu).

Issue
Cutis - 105(4)
Publications
Topics
Page Number
161-162
Sections
Author and Disclosure Information

 

From the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The author reports no conflict of interest.

Correspondence: Dirk M. Elston, MD, 135 Rutledge Ave, MSC 578, Charleston, SC 29425-5780 (elstond@musc.edu).

Author and Disclosure Information

 

From the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The author reports no conflict of interest.

Correspondence: Dirk M. Elston, MD, 135 Rutledge Ave, MSC 578, Charleston, SC 29425-5780 (elstond@musc.edu).

Article PDF
Article PDF
Related Articles

 

The expanding range of tick-borne diseases is a growing problem worldwide. Climate change plays a preeminent role in the expansion of tick species, especially for southern ticks in the United States such as Amblyomma species, which have introduced new pathogens to northern states.1-5 In addition to well-known tick-borne diseases, Amblyomma ticks have been implicated in the spread of emerging severe and potentially fatal viral illnesses, including Bourbon virus and Heartland virus.6 The increasing range of Amblyomma ticks also exposes new populations to tick-induced meat allergy (alpha-gal) syndrome, whereby development of specific IgE antibodies to the oligosaccharide galactose-alpha-1,3-galactose (alpha-gal) following tick bites results in severe allergic responses to consumption of beef, pork, and lamb.7

Amblyomma ticks have now been identified close to the Canadian border in Michigan and New York, and predictions of continued climate change raise the possibility of northward range expansion into all provinces of Canada from Alberta to Newfoundland and Labrador during the coming decades.8,9 Additional factors that contribute to the expanding range of many tick species include international travel, migratory patterns of birds, competition, and natural predators such as fire ants that feed on tick eggs and influence the feeding behavior of adults.10

Traditional methods of tick identification rely on gross morphology, including the presence of festoons, shape of the coxae where the legs attach, and markings on the hard overlying scutum. More recently, molecular identification has improved tick identification, leading to more accurate assessment of tick prevalence. These modern identification studies include analysis of 16S ribosomal DNA (rDNA), 12S rDNA, and ITS1 rDNA, and ITS2 rDNA genes.11

The spread of tick vectors has huge public health implications, and better methods to control tick populations are needed.12 New acaricides and growth regulators are being developed,13 and early spring applications of acaricides such as bifenthrin can suppress nymphs prior to the initiation of host-seeking activity.14 Controlled burns within tick habitats have proved helpful in reducing the risk for vector-borne disease.15,16 Personal protection is best accomplished with the use of a repellent together with clothing impregnated with an acaricide such as permethrin.17 Efforts to slow climate change and continued surveillance for the spread of tick vectors is urgently needed.

 

The expanding range of tick-borne diseases is a growing problem worldwide. Climate change plays a preeminent role in the expansion of tick species, especially for southern ticks in the United States such as Amblyomma species, which have introduced new pathogens to northern states.1-5 In addition to well-known tick-borne diseases, Amblyomma ticks have been implicated in the spread of emerging severe and potentially fatal viral illnesses, including Bourbon virus and Heartland virus.6 The increasing range of Amblyomma ticks also exposes new populations to tick-induced meat allergy (alpha-gal) syndrome, whereby development of specific IgE antibodies to the oligosaccharide galactose-alpha-1,3-galactose (alpha-gal) following tick bites results in severe allergic responses to consumption of beef, pork, and lamb.7

Amblyomma ticks have now been identified close to the Canadian border in Michigan and New York, and predictions of continued climate change raise the possibility of northward range expansion into all provinces of Canada from Alberta to Newfoundland and Labrador during the coming decades.8,9 Additional factors that contribute to the expanding range of many tick species include international travel, migratory patterns of birds, competition, and natural predators such as fire ants that feed on tick eggs and influence the feeding behavior of adults.10

Traditional methods of tick identification rely on gross morphology, including the presence of festoons, shape of the coxae where the legs attach, and markings on the hard overlying scutum. More recently, molecular identification has improved tick identification, leading to more accurate assessment of tick prevalence. These modern identification studies include analysis of 16S ribosomal DNA (rDNA), 12S rDNA, and ITS1 rDNA, and ITS2 rDNA genes.11

The spread of tick vectors has huge public health implications, and better methods to control tick populations are needed.12 New acaricides and growth regulators are being developed,13 and early spring applications of acaricides such as bifenthrin can suppress nymphs prior to the initiation of host-seeking activity.14 Controlled burns within tick habitats have proved helpful in reducing the risk for vector-borne disease.15,16 Personal protection is best accomplished with the use of a repellent together with clothing impregnated with an acaricide such as permethrin.17 Efforts to slow climate change and continued surveillance for the spread of tick vectors is urgently needed.

References
  1. Sanchez-Vicente S, Tagliafierro T, Coleman JL, et al. Polymicrobial nature of tick-borne diseases [published online September 10, 2019]. MBio. doi:10.1128/mBio.02055-19.
  2. Raghavan RK, Peterson AT, Cobos ME, et al. Current and future distribution of the Lone Star tick, Amblyomma americanum (L.) (Acari: Ixodidae) in North America. PLoS One. 2019;14:e0209082.
  3. Stafford KC 3rd, Molaei G, Little EAH, et al. Distribution and establishment of the Lone Star tick in Connecticut and implications for range expansion and public health. J Med Entomol. 2018;25:1561-1568.
  4. Gilliam ME, Rechkemmer WT, McCravy KW, et al. The influence of prescribed fire, habitat, and weather on Amblyomma americanum (Ixodida: Ixodidae) in West-Central Illinois, USA [published online March 22, 2018]. Insects. doi:10.3390/insects9020036.
  5. Sonenshine DE. Range expansion of tick disease vectors in North America: implications for spread of tick-borne disease [published online March 9, 2018]. Int J Environ Res Public Health. doi:10.3390/ijerph15030478.
  6. Savage HM, Godsey MS Jr, Panella NA, et al. Surveillance for tick-borne viruses near the location of a fatal human case of Bourbon virus (family Orthomyxoviridae: genus Thogotovirus) in eastern Kansas, 2015. J Med Entomol. 2018;55:701-705.
  7. Crispell G, Commins SP, Archer-Hartman SA, et al. Discovery of alpha-gal-containing antigens in North American tick species believed to induce red meat allergy. Front Immunol. 2019;10:1056.
  8. Gasmi S, Bouchard C, Ogden NH, et al. Evidence for increasing densities and geographic ranges of tick species of public health significance other than Ixodes scapularis in Québec, Canada. PLoS One. 2018;13:e0201924.
  9. Sagurova I, Ludwig A, Ogden NH, et al. Predicted northward expansion of the geographic range of the tick vector Amblyomma americanum in North America under future climate conditions. Environ Health Perspect. 2019;127:107014.
  10. Kjeldgaard MK, Takano OM, Bockoven AA, et al. Red imported fire ant (Solenopsis invicta) aggression influences the behavior of three hard tick species. Exp Appl Acarol. 2019;79:87-97.
  11. Abouelhassan EM, El-Gawady HM, Abdel-Aal AA, et al. Comparison of some molecular markers for tick species identification. J Arthropod Borne Dis. 2019;13:153-164.
  12. Jordan RA, Egizi A. The growing importance of lone star ticks in a Lyme disease endemic county: passive tick surveillance in Monmouth County, NJ, 2006–2016. PLoS One. 2019;14:e0211778.
  13. Showler AT, Donahue WA, Harlien JL, et al. Efficacy of novaluron + pyriproxyfen (Tekko Pro) insect growth regulators against Amblyomma americanum (Acari: Ixodidae), Rhipicephalus (Boophilus) annulatus, Rhipicephalus (Boophilus) microplus, and Rhipicephalus sanguineus. J Med Entomol. 2019;56:1338-1345.
  14. Schulze TL, Jordan RA. Early season applications of bifenthrin suppress host-seeking Ixodes scapularis and Amblyomma americanum (Acari: Ixodidae) nymphs [published online November 26, 2019]. J Med Entomol. doi:10.1093/jme/tjz202.
  15. Hodo CL, Forgacs D, Auckland LD, et al. Presence of diverse Rickettsia spp. and absence of Borrelia burgdorferi sensu lato in ticks in an East Texas forest with reduced tick density associated with controlled burns. Ticks Tick Borne Dis. 2020;11:101310.
  16. Gleim ER, Zemtsova GE, Berghaus RD, et al. Frequent prescribed fires can reduce risk of tick-borne diseases. Sci Rep. 2019;9:9974.
  17. Prose R, Breuner NE, Johnson TL, et al. Contact irritancy and toxicity of permethrin-treated clothing for Ixodes scapularis, Amblyomma americanum, and Dermacentor variabilis ticks (Acari: Ixodidae). J Med Entomol. 2018;55:1217-1224.
References
  1. Sanchez-Vicente S, Tagliafierro T, Coleman JL, et al. Polymicrobial nature of tick-borne diseases [published online September 10, 2019]. MBio. doi:10.1128/mBio.02055-19.
  2. Raghavan RK, Peterson AT, Cobos ME, et al. Current and future distribution of the Lone Star tick, Amblyomma americanum (L.) (Acari: Ixodidae) in North America. PLoS One. 2019;14:e0209082.
  3. Stafford KC 3rd, Molaei G, Little EAH, et al. Distribution and establishment of the Lone Star tick in Connecticut and implications for range expansion and public health. J Med Entomol. 2018;25:1561-1568.
  4. Gilliam ME, Rechkemmer WT, McCravy KW, et al. The influence of prescribed fire, habitat, and weather on Amblyomma americanum (Ixodida: Ixodidae) in West-Central Illinois, USA [published online March 22, 2018]. Insects. doi:10.3390/insects9020036.
  5. Sonenshine DE. Range expansion of tick disease vectors in North America: implications for spread of tick-borne disease [published online March 9, 2018]. Int J Environ Res Public Health. doi:10.3390/ijerph15030478.
  6. Savage HM, Godsey MS Jr, Panella NA, et al. Surveillance for tick-borne viruses near the location of a fatal human case of Bourbon virus (family Orthomyxoviridae: genus Thogotovirus) in eastern Kansas, 2015. J Med Entomol. 2018;55:701-705.
  7. Crispell G, Commins SP, Archer-Hartman SA, et al. Discovery of alpha-gal-containing antigens in North American tick species believed to induce red meat allergy. Front Immunol. 2019;10:1056.
  8. Gasmi S, Bouchard C, Ogden NH, et al. Evidence for increasing densities and geographic ranges of tick species of public health significance other than Ixodes scapularis in Québec, Canada. PLoS One. 2018;13:e0201924.
  9. Sagurova I, Ludwig A, Ogden NH, et al. Predicted northward expansion of the geographic range of the tick vector Amblyomma americanum in North America under future climate conditions. Environ Health Perspect. 2019;127:107014.
  10. Kjeldgaard MK, Takano OM, Bockoven AA, et al. Red imported fire ant (Solenopsis invicta) aggression influences the behavior of three hard tick species. Exp Appl Acarol. 2019;79:87-97.
  11. Abouelhassan EM, El-Gawady HM, Abdel-Aal AA, et al. Comparison of some molecular markers for tick species identification. J Arthropod Borne Dis. 2019;13:153-164.
  12. Jordan RA, Egizi A. The growing importance of lone star ticks in a Lyme disease endemic county: passive tick surveillance in Monmouth County, NJ, 2006–2016. PLoS One. 2019;14:e0211778.
  13. Showler AT, Donahue WA, Harlien JL, et al. Efficacy of novaluron + pyriproxyfen (Tekko Pro) insect growth regulators against Amblyomma americanum (Acari: Ixodidae), Rhipicephalus (Boophilus) annulatus, Rhipicephalus (Boophilus) microplus, and Rhipicephalus sanguineus. J Med Entomol. 2019;56:1338-1345.
  14. Schulze TL, Jordan RA. Early season applications of bifenthrin suppress host-seeking Ixodes scapularis and Amblyomma americanum (Acari: Ixodidae) nymphs [published online November 26, 2019]. J Med Entomol. doi:10.1093/jme/tjz202.
  15. Hodo CL, Forgacs D, Auckland LD, et al. Presence of diverse Rickettsia spp. and absence of Borrelia burgdorferi sensu lato in ticks in an East Texas forest with reduced tick density associated with controlled burns. Ticks Tick Borne Dis. 2020;11:101310.
  16. Gleim ER, Zemtsova GE, Berghaus RD, et al. Frequent prescribed fires can reduce risk of tick-borne diseases. Sci Rep. 2019;9:9974.
  17. Prose R, Breuner NE, Johnson TL, et al. Contact irritancy and toxicity of permethrin-treated clothing for Ixodes scapularis, Amblyomma americanum, and Dermacentor variabilis ticks (Acari: Ixodidae). J Med Entomol. 2018;55:1217-1224.
Issue
Cutis - 105(4)
Issue
Cutis - 105(4)
Page Number
161-162
Page Number
161-162
Publications
Publications
Topics
Article Type
Display Headline
Climate Change and Expansion of Tick Geography
Display Headline
Climate Change and Expansion of Tick Geography
Sections
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.
Article PDF Media

Multinodular Plaque on the Penis

Article Type
Changed
Wed, 03/25/2020 - 13:55
Display Headline
Multinodular Plaque on the Penis

The Diagnosis: Tophaceous Gout  

Biopsy revealed amorphous pink material within the center of palisading granulomas lined by histiocytes and giant cells. Scattered crystal remnants also were identified within the center of the granulomas; however, the majority of the crystals were dissolved during the formalin processing of the tissue to become the amorphous material. A perivascular mixed inflammatory infiltrate composed of lymphocytes, histiocytes, and plasma cells surrounded the tophi nodules. A biopsy confirmed the diagnosis of tophaceous gout (Figure). 

At scanning magnification, palisaded granulomas surrounding feathery gray amorphous material were seen (H&E, original magnification ×40).

Gout is a systemic metabolic disease characterized by the supersaturation of monosodium urate (MSU) crystals in joints and bursae. Peripheral joints most commonly are affected due to the poor solubility of MSU crystals at low temperatures.1 It is one of the most common forms of inflammatory arthritis, with an estimated prevalence of 4% of adults in the United States.2 An estimated $1 billion is spent each year on ambulatory care for gout.3 Gout occurs most commonly in men and usually manifests in the fifth or sixth decades of life.4 Risk factors for the development of gout include obesity, hypertension, poor dietary habits and kidney function, excessive alcohol intake, and diuretic use.3 

Disease manifestations range from asymptomatic hyperuricemia to acute gouty arthritis and chronic tophaceous gout. Patients may present with chronic tophaceous gout without a prior clinically apparent acute gout episode.5,6 Uncontrolled gout may result in large accumulations of MSU crystals, leading to well-circumscribed masses (known as tophi), as demonstrated in our patient.1 Tophi are pathognomonic features of gout and are the sine qua non of advanced gout (also known as chronic tophaceous gout).2 Clinically, these tophi appear as subcutaneous, yellowish white, firm and smooth nodules that are highlighted on the skin.4 Tophi most commonly are found on the helix, articular and periarticular tissue, and the tissue of the hands and feet. They usually are visible on physical examination but also may be detected on imaging studies.2,4 

Gouty tophi have been reported in extraordinary locations, such as in sclerae; vocal cords; heart valves; abdominal striae; nerves; axial skeleton4,7; and the penis, as in our patient and one other case.2 These gouty deposits can appear similarly to lipomas, rheumatoid and osteoarthritic nodules, and infectious and malignant processes.1,5 When tophi present in unusual locations, tissue biopsy often is necessary to confirm the diagnosis. Tissue preservation in alcohol is required to preserve the urate crystals. Microscopically, urate crystals appear as tightly packed, brown, needle-shaped crystals surrounded by granulomatous inflammation with foreign body giant cells, macrophages, and possibly some fibrosis. When examined under polarized light, the MSU crystals are negatively birefringent. However, when clinical suspicion for gout is low and the tissue is instead formalin fixed, as was performed in our case, the crystals dissolve into fibrillary amorphous deposits within the center of the granulomatous inflammation, which is another characteristic histologic finding in tophaceous gout.8 

Management of gout focuses on urate-lowering therapy including lifestyle changes. Lower serum urate levels are associated with a decreased incidence of acute gout attacks and chronic tophaceous gout.2 Urate-lowering drugs often are combined with anti-inflammatory drugs during acute attacks. Lifestyle changes, such as weight loss, exercise, reduced alcohol consumption, high fluid intake, and a low-purine diet also are beneficial.3,4 Although gout cannot be cured, it can be effectively managed, and appropriate treatment can improve quality of life and reduce the risk for permanent joint damage and structural deformities. If medical treatment and lifestyle changes fail to adequately control tophaceous gout or if tophi become symptomatic, surgical removal of tophi is appropriate.4 

At follow-up, our patient opted for surgical removal of the penile tophi. Using local anesthesia, surgical debulking via curettage was performed. Open defects were closed with fine absorbable sutures, and prophylactic antibiotics were given. Allopurinol also was started. Six weeks following extraction, the patient reported no complications and the area was continuing to heal.

Tophaceous gout would be distinguished from conditions in the differential diagnosis based on histologic findings from hematoxylin and eosin (H&E)-stained sections. Actinomycotic mycetoma is rare in the United States and is characterized by a seropurulent or stringy exudate with grains, ulcerations, melicerous scabs, and retractable scarring.9 On H&E-stained sections, actinomyces appear filamentous with deeply basophilic staining and radially oriented acidophilic projections.10 Calcinosis cutis of the penis has been reported to appear as asymptomatic papules; however, microscopic sections reveal deeply basophilic calcium deposits within the tissue.11 Multinodular syphilis shows characteristic histology with lichenoid or vacuolar interface dermatitis, slender acanthosis, plasma cells, and endothelial swelling of the small vessels. A Treponema pallidum immunoperoxidase stain shows numerous organisms. Planar xanthoma shows xanthomatous or foamy histiocytes throughout the dermis on H&E-stained sections.12  

References
  1. Ragab G, Elshahaly M, Bardin T. Gout: an old disease in new perspective--a review. J Adv Res. 2007;8:495-511.  
  2. Flores Martín JF, Vázquez Alonso F, Puche Sanz I, et al. Gouty tophi in the penis: a case report and review of the literature. Case Rep Urol. 2012;2012:594905.  
  3. Qaseem A, Harris RP, Forciea MA; Clinical Guidelines Committee of the American College of Physicians. Management of acute and recurrent gout: a clinical practice guideline from the American College of Physicians. Ann Intern Med. 2017;166:58-68.  
  4. Forbess LJ, Fields TR. The broad spectrum of urate crystal deposition: unusual presentations of gouty tophi. Semin Arthritis Rheum. 2012;42:146-154.  
  5. Khanna D, Fitzgerald JD, Khanna PP, et al. 2012 American College of Rheumatology guidelines for management of gout. part 1: systematic nonpharmacologic and pharmacologic therapeutic approaches to hyperuricemia. Arthritis Care Res. 2012;64:1431-1446.  
  6. Khanna D, Khanna PP, Fitzgerald JD, et al. 2012 American College of Rheumatology guidelines for management of gout. part 2: therapy and anti-inflammatory prophylaxis of acute gouty arthritis. Arthritis Care Res. 2012;64:1447-1461.  
  7. Gaviria JL, Ortega VG, Gaona J. Unusual dermatological manifestations of gout: review of literature and a case report. Plast Reconstr Surg Glob Open. 2015;3:E445.  
  8. Patterson JW, Hosler GA, Weedon D. Weedon's Skin Pathology. Edinburgh, Scotland: Churchill Livingstone/Elsevier; 2016. 
  9. Guerra-Leal JD, Medrano-Danés LA, Montemayor-Martinez A, et al. The importance of diagnostic imaging of mycetoma in the foot [published online December 18, 2018]. Int J Dermatol. 2019;58:600-604.  
  10. Fazeli MS, Bateni H. Actinomycosis: a rare soft tissue infection. Dermatol Online J. 2005;11:18.  
  11. Cohen PR, Tschen JA. Idiopathic calcinosis cutis of the penis. J Clin Aesthet Dermatol. 2012;5:23-30.  
  12. Ko C, Elston DM, Ferringer T. Dermatopathology. 3rd ed. Philadelphia, PA: Elsevier; 2019. 
Article PDF
Author and Disclosure Information

Drs. Nickerson, O’Connor, Lopez, and Elston are from the Medical University of South Carolina, Charleston. Drs. Nickerson and Elston are from the Department of Dermatology and Dermatologic Surgery, and Drs. O’Connor and Lopez are from the Department of Pathology. Dr. Dowling is from the Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton.

The authors report no conflict of interest.

Correspondence: Derek W. Nickerson, MD, Medical University of South Carolina, Department of Dermatology and Dermatologic Surgery, 135 Rutledge Ave, MSC 578, Charleston, SC 29464 (nickersd@musc.edu).

Issue
Cutis - 105(3)
Publications
Topics
Page Number
E19-E21
Sections
Author and Disclosure Information

Drs. Nickerson, O’Connor, Lopez, and Elston are from the Medical University of South Carolina, Charleston. Drs. Nickerson and Elston are from the Department of Dermatology and Dermatologic Surgery, and Drs. O’Connor and Lopez are from the Department of Pathology. Dr. Dowling is from the Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton.

The authors report no conflict of interest.

Correspondence: Derek W. Nickerson, MD, Medical University of South Carolina, Department of Dermatology and Dermatologic Surgery, 135 Rutledge Ave, MSC 578, Charleston, SC 29464 (nickersd@musc.edu).

Author and Disclosure Information

Drs. Nickerson, O’Connor, Lopez, and Elston are from the Medical University of South Carolina, Charleston. Drs. Nickerson and Elston are from the Department of Dermatology and Dermatologic Surgery, and Drs. O’Connor and Lopez are from the Department of Pathology. Dr. Dowling is from the Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton.

The authors report no conflict of interest.

Correspondence: Derek W. Nickerson, MD, Medical University of South Carolina, Department of Dermatology and Dermatologic Surgery, 135 Rutledge Ave, MSC 578, Charleston, SC 29464 (nickersd@musc.edu).

Article PDF
Article PDF
Related Articles

The Diagnosis: Tophaceous Gout  

Biopsy revealed amorphous pink material within the center of palisading granulomas lined by histiocytes and giant cells. Scattered crystal remnants also were identified within the center of the granulomas; however, the majority of the crystals were dissolved during the formalin processing of the tissue to become the amorphous material. A perivascular mixed inflammatory infiltrate composed of lymphocytes, histiocytes, and plasma cells surrounded the tophi nodules. A biopsy confirmed the diagnosis of tophaceous gout (Figure). 

At scanning magnification, palisaded granulomas surrounding feathery gray amorphous material were seen (H&E, original magnification ×40).

Gout is a systemic metabolic disease characterized by the supersaturation of monosodium urate (MSU) crystals in joints and bursae. Peripheral joints most commonly are affected due to the poor solubility of MSU crystals at low temperatures.1 It is one of the most common forms of inflammatory arthritis, with an estimated prevalence of 4% of adults in the United States.2 An estimated $1 billion is spent each year on ambulatory care for gout.3 Gout occurs most commonly in men and usually manifests in the fifth or sixth decades of life.4 Risk factors for the development of gout include obesity, hypertension, poor dietary habits and kidney function, excessive alcohol intake, and diuretic use.3 

Disease manifestations range from asymptomatic hyperuricemia to acute gouty arthritis and chronic tophaceous gout. Patients may present with chronic tophaceous gout without a prior clinically apparent acute gout episode.5,6 Uncontrolled gout may result in large accumulations of MSU crystals, leading to well-circumscribed masses (known as tophi), as demonstrated in our patient.1 Tophi are pathognomonic features of gout and are the sine qua non of advanced gout (also known as chronic tophaceous gout).2 Clinically, these tophi appear as subcutaneous, yellowish white, firm and smooth nodules that are highlighted on the skin.4 Tophi most commonly are found on the helix, articular and periarticular tissue, and the tissue of the hands and feet. They usually are visible on physical examination but also may be detected on imaging studies.2,4 

Gouty tophi have been reported in extraordinary locations, such as in sclerae; vocal cords; heart valves; abdominal striae; nerves; axial skeleton4,7; and the penis, as in our patient and one other case.2 These gouty deposits can appear similarly to lipomas, rheumatoid and osteoarthritic nodules, and infectious and malignant processes.1,5 When tophi present in unusual locations, tissue biopsy often is necessary to confirm the diagnosis. Tissue preservation in alcohol is required to preserve the urate crystals. Microscopically, urate crystals appear as tightly packed, brown, needle-shaped crystals surrounded by granulomatous inflammation with foreign body giant cells, macrophages, and possibly some fibrosis. When examined under polarized light, the MSU crystals are negatively birefringent. However, when clinical suspicion for gout is low and the tissue is instead formalin fixed, as was performed in our case, the crystals dissolve into fibrillary amorphous deposits within the center of the granulomatous inflammation, which is another characteristic histologic finding in tophaceous gout.8 

Management of gout focuses on urate-lowering therapy including lifestyle changes. Lower serum urate levels are associated with a decreased incidence of acute gout attacks and chronic tophaceous gout.2 Urate-lowering drugs often are combined with anti-inflammatory drugs during acute attacks. Lifestyle changes, such as weight loss, exercise, reduced alcohol consumption, high fluid intake, and a low-purine diet also are beneficial.3,4 Although gout cannot be cured, it can be effectively managed, and appropriate treatment can improve quality of life and reduce the risk for permanent joint damage and structural deformities. If medical treatment and lifestyle changes fail to adequately control tophaceous gout or if tophi become symptomatic, surgical removal of tophi is appropriate.4 

At follow-up, our patient opted for surgical removal of the penile tophi. Using local anesthesia, surgical debulking via curettage was performed. Open defects were closed with fine absorbable sutures, and prophylactic antibiotics were given. Allopurinol also was started. Six weeks following extraction, the patient reported no complications and the area was continuing to heal.

Tophaceous gout would be distinguished from conditions in the differential diagnosis based on histologic findings from hematoxylin and eosin (H&E)-stained sections. Actinomycotic mycetoma is rare in the United States and is characterized by a seropurulent or stringy exudate with grains, ulcerations, melicerous scabs, and retractable scarring.9 On H&E-stained sections, actinomyces appear filamentous with deeply basophilic staining and radially oriented acidophilic projections.10 Calcinosis cutis of the penis has been reported to appear as asymptomatic papules; however, microscopic sections reveal deeply basophilic calcium deposits within the tissue.11 Multinodular syphilis shows characteristic histology with lichenoid or vacuolar interface dermatitis, slender acanthosis, plasma cells, and endothelial swelling of the small vessels. A Treponema pallidum immunoperoxidase stain shows numerous organisms. Planar xanthoma shows xanthomatous or foamy histiocytes throughout the dermis on H&E-stained sections.12  

The Diagnosis: Tophaceous Gout  

Biopsy revealed amorphous pink material within the center of palisading granulomas lined by histiocytes and giant cells. Scattered crystal remnants also were identified within the center of the granulomas; however, the majority of the crystals were dissolved during the formalin processing of the tissue to become the amorphous material. A perivascular mixed inflammatory infiltrate composed of lymphocytes, histiocytes, and plasma cells surrounded the tophi nodules. A biopsy confirmed the diagnosis of tophaceous gout (Figure). 

At scanning magnification, palisaded granulomas surrounding feathery gray amorphous material were seen (H&E, original magnification ×40).

Gout is a systemic metabolic disease characterized by the supersaturation of monosodium urate (MSU) crystals in joints and bursae. Peripheral joints most commonly are affected due to the poor solubility of MSU crystals at low temperatures.1 It is one of the most common forms of inflammatory arthritis, with an estimated prevalence of 4% of adults in the United States.2 An estimated $1 billion is spent each year on ambulatory care for gout.3 Gout occurs most commonly in men and usually manifests in the fifth or sixth decades of life.4 Risk factors for the development of gout include obesity, hypertension, poor dietary habits and kidney function, excessive alcohol intake, and diuretic use.3 

Disease manifestations range from asymptomatic hyperuricemia to acute gouty arthritis and chronic tophaceous gout. Patients may present with chronic tophaceous gout without a prior clinically apparent acute gout episode.5,6 Uncontrolled gout may result in large accumulations of MSU crystals, leading to well-circumscribed masses (known as tophi), as demonstrated in our patient.1 Tophi are pathognomonic features of gout and are the sine qua non of advanced gout (also known as chronic tophaceous gout).2 Clinically, these tophi appear as subcutaneous, yellowish white, firm and smooth nodules that are highlighted on the skin.4 Tophi most commonly are found on the helix, articular and periarticular tissue, and the tissue of the hands and feet. They usually are visible on physical examination but also may be detected on imaging studies.2,4 

Gouty tophi have been reported in extraordinary locations, such as in sclerae; vocal cords; heart valves; abdominal striae; nerves; axial skeleton4,7; and the penis, as in our patient and one other case.2 These gouty deposits can appear similarly to lipomas, rheumatoid and osteoarthritic nodules, and infectious and malignant processes.1,5 When tophi present in unusual locations, tissue biopsy often is necessary to confirm the diagnosis. Tissue preservation in alcohol is required to preserve the urate crystals. Microscopically, urate crystals appear as tightly packed, brown, needle-shaped crystals surrounded by granulomatous inflammation with foreign body giant cells, macrophages, and possibly some fibrosis. When examined under polarized light, the MSU crystals are negatively birefringent. However, when clinical suspicion for gout is low and the tissue is instead formalin fixed, as was performed in our case, the crystals dissolve into fibrillary amorphous deposits within the center of the granulomatous inflammation, which is another characteristic histologic finding in tophaceous gout.8 

Management of gout focuses on urate-lowering therapy including lifestyle changes. Lower serum urate levels are associated with a decreased incidence of acute gout attacks and chronic tophaceous gout.2 Urate-lowering drugs often are combined with anti-inflammatory drugs during acute attacks. Lifestyle changes, such as weight loss, exercise, reduced alcohol consumption, high fluid intake, and a low-purine diet also are beneficial.3,4 Although gout cannot be cured, it can be effectively managed, and appropriate treatment can improve quality of life and reduce the risk for permanent joint damage and structural deformities. If medical treatment and lifestyle changes fail to adequately control tophaceous gout or if tophi become symptomatic, surgical removal of tophi is appropriate.4 

At follow-up, our patient opted for surgical removal of the penile tophi. Using local anesthesia, surgical debulking via curettage was performed. Open defects were closed with fine absorbable sutures, and prophylactic antibiotics were given. Allopurinol also was started. Six weeks following extraction, the patient reported no complications and the area was continuing to heal.

Tophaceous gout would be distinguished from conditions in the differential diagnosis based on histologic findings from hematoxylin and eosin (H&E)-stained sections. Actinomycotic mycetoma is rare in the United States and is characterized by a seropurulent or stringy exudate with grains, ulcerations, melicerous scabs, and retractable scarring.9 On H&E-stained sections, actinomyces appear filamentous with deeply basophilic staining and radially oriented acidophilic projections.10 Calcinosis cutis of the penis has been reported to appear as asymptomatic papules; however, microscopic sections reveal deeply basophilic calcium deposits within the tissue.11 Multinodular syphilis shows characteristic histology with lichenoid or vacuolar interface dermatitis, slender acanthosis, plasma cells, and endothelial swelling of the small vessels. A Treponema pallidum immunoperoxidase stain shows numerous organisms. Planar xanthoma shows xanthomatous or foamy histiocytes throughout the dermis on H&E-stained sections.12  

References
  1. Ragab G, Elshahaly M, Bardin T. Gout: an old disease in new perspective--a review. J Adv Res. 2007;8:495-511.  
  2. Flores Martín JF, Vázquez Alonso F, Puche Sanz I, et al. Gouty tophi in the penis: a case report and review of the literature. Case Rep Urol. 2012;2012:594905.  
  3. Qaseem A, Harris RP, Forciea MA; Clinical Guidelines Committee of the American College of Physicians. Management of acute and recurrent gout: a clinical practice guideline from the American College of Physicians. Ann Intern Med. 2017;166:58-68.  
  4. Forbess LJ, Fields TR. The broad spectrum of urate crystal deposition: unusual presentations of gouty tophi. Semin Arthritis Rheum. 2012;42:146-154.  
  5. Khanna D, Fitzgerald JD, Khanna PP, et al. 2012 American College of Rheumatology guidelines for management of gout. part 1: systematic nonpharmacologic and pharmacologic therapeutic approaches to hyperuricemia. Arthritis Care Res. 2012;64:1431-1446.  
  6. Khanna D, Khanna PP, Fitzgerald JD, et al. 2012 American College of Rheumatology guidelines for management of gout. part 2: therapy and anti-inflammatory prophylaxis of acute gouty arthritis. Arthritis Care Res. 2012;64:1447-1461.  
  7. Gaviria JL, Ortega VG, Gaona J. Unusual dermatological manifestations of gout: review of literature and a case report. Plast Reconstr Surg Glob Open. 2015;3:E445.  
  8. Patterson JW, Hosler GA, Weedon D. Weedon's Skin Pathology. Edinburgh, Scotland: Churchill Livingstone/Elsevier; 2016. 
  9. Guerra-Leal JD, Medrano-Danés LA, Montemayor-Martinez A, et al. The importance of diagnostic imaging of mycetoma in the foot [published online December 18, 2018]. Int J Dermatol. 2019;58:600-604.  
  10. Fazeli MS, Bateni H. Actinomycosis: a rare soft tissue infection. Dermatol Online J. 2005;11:18.  
  11. Cohen PR, Tschen JA. Idiopathic calcinosis cutis of the penis. J Clin Aesthet Dermatol. 2012;5:23-30.  
  12. Ko C, Elston DM, Ferringer T. Dermatopathology. 3rd ed. Philadelphia, PA: Elsevier; 2019. 
References
  1. Ragab G, Elshahaly M, Bardin T. Gout: an old disease in new perspective--a review. J Adv Res. 2007;8:495-511.  
  2. Flores Martín JF, Vázquez Alonso F, Puche Sanz I, et al. Gouty tophi in the penis: a case report and review of the literature. Case Rep Urol. 2012;2012:594905.  
  3. Qaseem A, Harris RP, Forciea MA; Clinical Guidelines Committee of the American College of Physicians. Management of acute and recurrent gout: a clinical practice guideline from the American College of Physicians. Ann Intern Med. 2017;166:58-68.  
  4. Forbess LJ, Fields TR. The broad spectrum of urate crystal deposition: unusual presentations of gouty tophi. Semin Arthritis Rheum. 2012;42:146-154.  
  5. Khanna D, Fitzgerald JD, Khanna PP, et al. 2012 American College of Rheumatology guidelines for management of gout. part 1: systematic nonpharmacologic and pharmacologic therapeutic approaches to hyperuricemia. Arthritis Care Res. 2012;64:1431-1446.  
  6. Khanna D, Khanna PP, Fitzgerald JD, et al. 2012 American College of Rheumatology guidelines for management of gout. part 2: therapy and anti-inflammatory prophylaxis of acute gouty arthritis. Arthritis Care Res. 2012;64:1447-1461.  
  7. Gaviria JL, Ortega VG, Gaona J. Unusual dermatological manifestations of gout: review of literature and a case report. Plast Reconstr Surg Glob Open. 2015;3:E445.  
  8. Patterson JW, Hosler GA, Weedon D. Weedon's Skin Pathology. Edinburgh, Scotland: Churchill Livingstone/Elsevier; 2016. 
  9. Guerra-Leal JD, Medrano-Danés LA, Montemayor-Martinez A, et al. The importance of diagnostic imaging of mycetoma in the foot [published online December 18, 2018]. Int J Dermatol. 2019;58:600-604.  
  10. Fazeli MS, Bateni H. Actinomycosis: a rare soft tissue infection. Dermatol Online J. 2005;11:18.  
  11. Cohen PR, Tschen JA. Idiopathic calcinosis cutis of the penis. J Clin Aesthet Dermatol. 2012;5:23-30.  
  12. Ko C, Elston DM, Ferringer T. Dermatopathology. 3rd ed. Philadelphia, PA: Elsevier; 2019. 
Issue
Cutis - 105(3)
Issue
Cutis - 105(3)
Page Number
E19-E21
Page Number
E19-E21
Publications
Publications
Topics
Article Type
Display Headline
Multinodular Plaque on the Penis
Display Headline
Multinodular Plaque on the Penis
Sections
Questionnaire Body

A 34-year-old man presented for evaluation of a slowly growing group of firm white bumps on the penis. The lesions were nontender and asymptomatic. Medical and family history was notable for gout, though he was not being treated. Physical examination revealed a 3-cm, firm, multinodular, chalky white plaque on the dorsal aspect of the penile shaft. A tangential biopsy was performed and sent for hematoxylin and eosin staining.  

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 03/25/2020 - 12:00
Un-Gate On Date
Wed, 03/25/2020 - 12:00
Use ProPublica
CFC Schedule Remove Status
Wed, 03/25/2020 - 12:00
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

What’s Eating You? Human Body Lice (Pediculus humanus corporis)

Article Type
Changed
Wed, 05/13/2020 - 11:59
Display Headline
What’s Eating You? Human Body Lice (Pediculus humanus corporis)

Epidemiology and Transmission

Pediculus humanus corporis, commonly known as the human body louse, is one in a family of 3 ectoparasites of the same suborder that also encompasses pubic lice (Phthirus pubis) and head lice (Pediculus humanus capitis). Adults are approximately 2 mm in size, with the same life cycle as head lice (Figure 1). They require blood meals roughly 5 times per day and cannot survive longer than 2 days without feeding.1 Although similar in structure to head lice, body lice differ behaviorally in that they do not reside on their human host’s body; instead, they infest the host’s clothing, localizing to seams (Figure 2), and migrate to the host for blood meals. In fact, based on this behavior, genetic analysis of early human body lice has been used to postulate when clothing was first used by humans as well as to determine early human migration patterns.2,3

Figure 1. Adult body louse (Pediculus humanus corporis).

Figure 2. Body lice nits localized in clothing seams.

Although clinicians in developed countries may be less familiar with body lice compared to their counterparts, body lice nevertheless remain a global health concern in impoverished, densely populated areas, as well as in homeless populations due to poor hygiene. Transmission frequently occurs via physical contact with an affected individual and his/her personal items (eg, linens) via fomites.4,5 Body louse infestation is more prevalent in homeless individuals who sleep outside vs in shelters; a history of pubic lice and lack of regular bathing have been reported as additional risk factors.6 Outbreaks have been noted in the wake of natural disasters, in the setting of political upheavals, and in refugee camps, as well as in individuals seeking political asylum.7 Unlike head and pubic lice, body lice can serve as vectors for infectious diseases including Rickettsia prowazekii (epidemic typhus), Borrelia recurrentis (louse-borne relapsing fever), Bartonella quintana (trench fever), and Yersinia pestis (plague).5,8,9 Several Acinetobacter species were isolated from nearly one-third of collected body louse specimens in a French study.10 Additionally, serology for B quintana was found to be positive in up to 30% of cases in one United States urban homeless population.4

Clinical Manifestations

Patients often present with generalized pruritus, usually considerably more severe than with P humanus capitis, with lesions concentrated on the trunk.11 In addition to often impetiginized, self-inflicted excoriations, feeding sites may present as erythematous macules (Figure 3), papules, or papular urticaria with a central hemorrhagic punctum. Extensive infestation also can manifest as the colloquial vagabond disease, characterized by postinflammatory hyperpigmentation and thickening of the involved skin. Remarkably, patients also may present with considerable iron-deficiency anemia secondary to high parasite load and large volume blood feeding. Multiple case reports have demonstrated associated morbidity.12-14 The differential diagnosis for pediculosis may include scabies, lichen simplex chronicus, and eczematous dermatitis, though the clinician should prudently consider whether both scabies and pediculosis may be present, as coexistence is possible.4,15

Figure 3. Erythematous papules secondary to body lice infestation.

 

 

Diagnosis

Diagnosis can be reached by visualizing adult lice, nymphs, or viable nits on the body or more commonly within inner clothing seams; nits also fluoresce under Wood light.15 Although dermoscopy has proven useful for increased sensitivity and differentiation between viable and hatched nits, the insects also can be viewed with the unaided eye.16

Treatment: New Concerns and Strategies

The mainstay of treatment for body lice has long consisted of thorough washing and drying of all clothing and linens in a hot dryer. Treatment can be augmented with the addition of pharmacotherapy, plus antibiotics as warranted for louse-borne disease. Pharmacologic intervention often is used in cases of mass infestation and is similar to head lice.

Options for head lice include topical permethrin, malathion, lindane, spinosad, benzyl alcohol, and ivermectin. Pyrethroids, derived from the chrysanthemum, generally are considered safe for human use with a side-effect profile limited to irritation and allergy17; however, neurotoxicity and leukemia are clinical concerns, with an association more recently shown between large-volume use of pyrethroids and acute lymphoblastic leukemia.18,19 Use of lindane is not recommended due to a greater potential for central nervous system neurotoxicity, manifested by seizures, with repeated large surface application. Malathion is problematic due to the risk for mucosal irritation, flammability of some formulations, and theoretical organophosphate poisoning, as its mechanism of action involves inhibition of acetylcholinesterase.15 However, in the context of head lice treatment, a randomized controlled trial reported no incidence of acetylcholinesterase inhibition.20 Spinosad, manufactured from the soil bacterium Saccharopolyspora spinosa, functions similarly by interfering with the nicotinic acetylcholine receptor and also carries a risk for skin irritation.21 Among all the treatment options, we prefer benzyl alcohol, particularly in the context of resistance, as it is effective via a physical mechanism of action and lacks notable neurotoxic effects to the host. Use of benzyl alcohol is approved for patients as young as 6 months; it functions by asphyxiating the lice via paralysis of the respiratory spiracle with occlusion by inert ingredients. Itching, episodic numbness, and scalp or mucosal irritation are possible complications of treatment.22

Treatment resistance of body lice has increased in recent years, warranting exploration of additional management strategies. Moreover, developing resistance to lindane and malathion has been reported.23 Resistance to pyrethroids has been attributed to mutations in a voltage-gated sodium channel, one of which was universally present in the sampling of a single population.24 A randomized controlled trial showed that off-label oral ivermectin 400 μg/kg was superior to malathion lotion 0.5% in difficult-to-treat cases of head lice25; utility of oral ivermectin also has been reported in body lice.26 In vitro studies also have shown promise for pursuing synergistic treatment of body lice with both ivermectin and antibiotics.27



A novel primary prophylaxis approach for at-risk homeless individuals recently utilized permethrin-impregnated underwear. Although the intervention provided short-term infestation improvement, longer-term use did not show improvement from placebo and also increased prevalence of permethrin-resistant haplotypes.2

References
  1. Veracx A, Raoult D. Biology and genetics of human head and body lice. Trends Parasitol. 2012;28:563-571.
  2. Kittler R, Kayser M, Stoneking M. Molecular evolution of Pediculus humanus and the origin of clothing. Curr Biol. 2003;13:1414-1417.
  3. Drali R, Mumcuoglu KY, Yesilyurt G, et al. Studies of ancient lice reveal unsuspected past migrations of vectors. Am J Trop Med Hyg. 2015;93:623-625.
  4. Chosidow O. Scabies and pediculosis. Lancet. 2000;355:819-826.
  5. Feldmeier H, Heukelbach J. Epidermal parasitic skin diseases: a neglected category of poverty-associated plagues. Bull World Health Organ. 2009;87:152-159.
  6. Arnaud A, Chosidow O, Detrez MA, et al. Prevalence 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.
  7. Hytonen J, Khawaja T, Gronroos JO, et al. Louse-borne relapsing fever in Finland in two asylum seekers from Somalia. APMIS. 2017;125:59-62.
  8. Nordmann T, Feldt T, Bosselmann M, et al. Outbreak of louse-borne relapsing fever among urban dwellers in Arsi Zone, Central Ethiopia, from July to November 2016. Am J Trop Med Hyg. 2018;98:1599-1602.
  9. Louni M, Mana N, Bitam I, et al. Body lice of homeless people reveal the presence of several emerging bacterial pathogens in northern Algeria. PLoS Negl Trop Dis. 2018;12:E0006397.
  10. Candy K, Amanzougaghene N, Izri A, et al. Molecular survey of head and body lice, Pediculus humanus, in France. Vector Borne Zoonotic Dis. 2018;18:243-251.
  11. Bolognia JL, Schaffer JV, Cerroni L. Dermatology. 4th ed. Elsevier Limited; 2018.
  12. Nara A, Nagai H, Yamaguchi R, et al. An unusual autopsy case of lethal hypothermia exacerbated by body lice-induced severe anemia. Int J Legal Med. 2016;130:765-769.
  13. Althomali SA, Alzubaidi LM, Alkhaldi DM. Severe iron deficiency anaemia associated with heavy lice infestation in a young woman [published online November 5, 2015]. BMJ Case Rep. doi:10.1136/bcr-2015-212207.
  14. Hau V, Muhi-Iddin N. A ghost covered in lice: a case of severe blood loss with long-standing heavy pediculosis capitis infestation [published online December 19, 2014]. BMJ Case Rep. doi:10.1136/bcr-2014-206623.
  15. Diaz JH. Lice (Pediculosis). In: Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 9th ed. New York, NY: Elsevier; 2020:3482-3486.
  16. Martins LG, Bernardes Filho F, Quaresma MV, et al. Dermoscopy applied to pediculosis corporis diagnosis. An Bras Dermatol. 2014;89:513-514.
  17. Devore CD, Schutze GE; Council on School Health and Committee on Infectious Diseases, American Academy of Pediatrics. Head lice. Pediatrics. 2015;135:E1355-E1365.
  18. Shafer TJ, Meyer DA, Crofton KM. Developmental neurotoxicity of pyrethroid insecticides: critical review and future research needs. Environ Health Perspect. 2005;113:123-136.
  19. Ding G, Shi R, Gao Y, et al. Pyrethroid pesticide exposure and risk of childhood acute lymphocytic leukemia in Shanghai. Environ Sci Technol. 2012;46:13480-13487.
  20. Meinking TL, Vicaria M, Eyerdam DH, et al. A randomized, investigator-blinded, time-ranging study of the comparative efficacy of 0.5% malathion gel versus Ovide Lotion (0.5% malathion) or Nix Crème Rinse (1% permethrin) used as labeled, for the treatment of head lice. Pediatr Dermatol. 2007;24:405-411.
  21. McCormack PL. Spinosad: in pediculosis capitis. Am J Clin Dermatol. 2011;12:349-353.
  22. Meinking TL, Villar ME, Vicaria M, et al. The clinical trials supporting benzyl alcohol lotion 5% (Ulesfia): a safe and effective topical treatment for head lice (pediculosis humanus capitis). Pediatr Dermatol. 2010;27:19-24.
  23. Lebwohl M, Clark L, Levitt J. Therapy for head lice based on life cycle, resistance, and safety considerations. Pediatrics. 2007;119:965-974
  24. Drali R, Benkouiten S, Badiaga S, et al. Detection of a knockdown resistance mutation associated with permethrin resistance in the body louse Pediculus humanus corporis by use of melting curve analysis genotyping. J Clin Microbiol. 2012;50:2229-2233.
  25. Chosidow O, Giraudeau B, Cottrell J, et al. Oral ivermectin versus malathion lotion for difficult-to-treat head lice. N Engl J Med. 2010;362:896-905.
  26. Foucault C, Ranque S, Badiaga S, et al. Oral ivermectin in the treatment of body lice. J Infect Dis. 2006;193:474-476.
  27. Sangaré AK, Doumbo OK, Raoult D. Management and treatment of human lice [published online July 27, 2016]. Biomed Res Int. doi:10.1155/2016/8962685.
  28. 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.
Article PDF
Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Dr. Nyers 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.

Images are in the public domain.

Correspondence: Emily S. Nyers, MD, 135 Rutledge Ave, MSC 578, Charleston, SC 29425 (nyers@musc.edu).

Issue
Cutis - 105(3)
Publications
Topics
Page Number
118-120
Sections
Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Dr. Nyers 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.

Images are in the public domain.

Correspondence: Emily S. Nyers, MD, 135 Rutledge Ave, MSC 578, Charleston, SC 29425 (nyers@musc.edu).

Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Dr. Nyers 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.

Images are in the public domain.

Correspondence: Emily S. Nyers, MD, 135 Rutledge Ave, MSC 578, Charleston, SC 29425 (nyers@musc.edu).

Article PDF
Article PDF

Epidemiology and Transmission

Pediculus humanus corporis, commonly known as the human body louse, is one in a family of 3 ectoparasites of the same suborder that also encompasses pubic lice (Phthirus pubis) and head lice (Pediculus humanus capitis). Adults are approximately 2 mm in size, with the same life cycle as head lice (Figure 1). They require blood meals roughly 5 times per day and cannot survive longer than 2 days without feeding.1 Although similar in structure to head lice, body lice differ behaviorally in that they do not reside on their human host’s body; instead, they infest the host’s clothing, localizing to seams (Figure 2), and migrate to the host for blood meals. In fact, based on this behavior, genetic analysis of early human body lice has been used to postulate when clothing was first used by humans as well as to determine early human migration patterns.2,3

Figure 1. Adult body louse (Pediculus humanus corporis).

Figure 2. Body lice nits localized in clothing seams.

Although clinicians in developed countries may be less familiar with body lice compared to their counterparts, body lice nevertheless remain a global health concern in impoverished, densely populated areas, as well as in homeless populations due to poor hygiene. Transmission frequently occurs via physical contact with an affected individual and his/her personal items (eg, linens) via fomites.4,5 Body louse infestation is more prevalent in homeless individuals who sleep outside vs in shelters; a history of pubic lice and lack of regular bathing have been reported as additional risk factors.6 Outbreaks have been noted in the wake of natural disasters, in the setting of political upheavals, and in refugee camps, as well as in individuals seeking political asylum.7 Unlike head and pubic lice, body lice can serve as vectors for infectious diseases including Rickettsia prowazekii (epidemic typhus), Borrelia recurrentis (louse-borne relapsing fever), Bartonella quintana (trench fever), and Yersinia pestis (plague).5,8,9 Several Acinetobacter species were isolated from nearly one-third of collected body louse specimens in a French study.10 Additionally, serology for B quintana was found to be positive in up to 30% of cases in one United States urban homeless population.4

Clinical Manifestations

Patients often present with generalized pruritus, usually considerably more severe than with P humanus capitis, with lesions concentrated on the trunk.11 In addition to often impetiginized, self-inflicted excoriations, feeding sites may present as erythematous macules (Figure 3), papules, or papular urticaria with a central hemorrhagic punctum. Extensive infestation also can manifest as the colloquial vagabond disease, characterized by postinflammatory hyperpigmentation and thickening of the involved skin. Remarkably, patients also may present with considerable iron-deficiency anemia secondary to high parasite load and large volume blood feeding. Multiple case reports have demonstrated associated morbidity.12-14 The differential diagnosis for pediculosis may include scabies, lichen simplex chronicus, and eczematous dermatitis, though the clinician should prudently consider whether both scabies and pediculosis may be present, as coexistence is possible.4,15

Figure 3. Erythematous papules secondary to body lice infestation.

 

 

Diagnosis

Diagnosis can be reached by visualizing adult lice, nymphs, or viable nits on the body or more commonly within inner clothing seams; nits also fluoresce under Wood light.15 Although dermoscopy has proven useful for increased sensitivity and differentiation between viable and hatched nits, the insects also can be viewed with the unaided eye.16

Treatment: New Concerns and Strategies

The mainstay of treatment for body lice has long consisted of thorough washing and drying of all clothing and linens in a hot dryer. Treatment can be augmented with the addition of pharmacotherapy, plus antibiotics as warranted for louse-borne disease. Pharmacologic intervention often is used in cases of mass infestation and is similar to head lice.

Options for head lice include topical permethrin, malathion, lindane, spinosad, benzyl alcohol, and ivermectin. Pyrethroids, derived from the chrysanthemum, generally are considered safe for human use with a side-effect profile limited to irritation and allergy17; however, neurotoxicity and leukemia are clinical concerns, with an association more recently shown between large-volume use of pyrethroids and acute lymphoblastic leukemia.18,19 Use of lindane is not recommended due to a greater potential for central nervous system neurotoxicity, manifested by seizures, with repeated large surface application. Malathion is problematic due to the risk for mucosal irritation, flammability of some formulations, and theoretical organophosphate poisoning, as its mechanism of action involves inhibition of acetylcholinesterase.15 However, in the context of head lice treatment, a randomized controlled trial reported no incidence of acetylcholinesterase inhibition.20 Spinosad, manufactured from the soil bacterium Saccharopolyspora spinosa, functions similarly by interfering with the nicotinic acetylcholine receptor and also carries a risk for skin irritation.21 Among all the treatment options, we prefer benzyl alcohol, particularly in the context of resistance, as it is effective via a physical mechanism of action and lacks notable neurotoxic effects to the host. Use of benzyl alcohol is approved for patients as young as 6 months; it functions by asphyxiating the lice via paralysis of the respiratory spiracle with occlusion by inert ingredients. Itching, episodic numbness, and scalp or mucosal irritation are possible complications of treatment.22

Treatment resistance of body lice has increased in recent years, warranting exploration of additional management strategies. Moreover, developing resistance to lindane and malathion has been reported.23 Resistance to pyrethroids has been attributed to mutations in a voltage-gated sodium channel, one of which was universally present in the sampling of a single population.24 A randomized controlled trial showed that off-label oral ivermectin 400 μg/kg was superior to malathion lotion 0.5% in difficult-to-treat cases of head lice25; utility of oral ivermectin also has been reported in body lice.26 In vitro studies also have shown promise for pursuing synergistic treatment of body lice with both ivermectin and antibiotics.27



A novel primary prophylaxis approach for at-risk homeless individuals recently utilized permethrin-impregnated underwear. Although the intervention provided short-term infestation improvement, longer-term use did not show improvement from placebo and also increased prevalence of permethrin-resistant haplotypes.2

Epidemiology and Transmission

Pediculus humanus corporis, commonly known as the human body louse, is one in a family of 3 ectoparasites of the same suborder that also encompasses pubic lice (Phthirus pubis) and head lice (Pediculus humanus capitis). Adults are approximately 2 mm in size, with the same life cycle as head lice (Figure 1). They require blood meals roughly 5 times per day and cannot survive longer than 2 days without feeding.1 Although similar in structure to head lice, body lice differ behaviorally in that they do not reside on their human host’s body; instead, they infest the host’s clothing, localizing to seams (Figure 2), and migrate to the host for blood meals. In fact, based on this behavior, genetic analysis of early human body lice has been used to postulate when clothing was first used by humans as well as to determine early human migration patterns.2,3

Figure 1. Adult body louse (Pediculus humanus corporis).

Figure 2. Body lice nits localized in clothing seams.

Although clinicians in developed countries may be less familiar with body lice compared to their counterparts, body lice nevertheless remain a global health concern in impoverished, densely populated areas, as well as in homeless populations due to poor hygiene. Transmission frequently occurs via physical contact with an affected individual and his/her personal items (eg, linens) via fomites.4,5 Body louse infestation is more prevalent in homeless individuals who sleep outside vs in shelters; a history of pubic lice and lack of regular bathing have been reported as additional risk factors.6 Outbreaks have been noted in the wake of natural disasters, in the setting of political upheavals, and in refugee camps, as well as in individuals seeking political asylum.7 Unlike head and pubic lice, body lice can serve as vectors for infectious diseases including Rickettsia prowazekii (epidemic typhus), Borrelia recurrentis (louse-borne relapsing fever), Bartonella quintana (trench fever), and Yersinia pestis (plague).5,8,9 Several Acinetobacter species were isolated from nearly one-third of collected body louse specimens in a French study.10 Additionally, serology for B quintana was found to be positive in up to 30% of cases in one United States urban homeless population.4

Clinical Manifestations

Patients often present with generalized pruritus, usually considerably more severe than with P humanus capitis, with lesions concentrated on the trunk.11 In addition to often impetiginized, self-inflicted excoriations, feeding sites may present as erythematous macules (Figure 3), papules, or papular urticaria with a central hemorrhagic punctum. Extensive infestation also can manifest as the colloquial vagabond disease, characterized by postinflammatory hyperpigmentation and thickening of the involved skin. Remarkably, patients also may present with considerable iron-deficiency anemia secondary to high parasite load and large volume blood feeding. Multiple case reports have demonstrated associated morbidity.12-14 The differential diagnosis for pediculosis may include scabies, lichen simplex chronicus, and eczematous dermatitis, though the clinician should prudently consider whether both scabies and pediculosis may be present, as coexistence is possible.4,15

Figure 3. Erythematous papules secondary to body lice infestation.

 

 

Diagnosis

Diagnosis can be reached by visualizing adult lice, nymphs, or viable nits on the body or more commonly within inner clothing seams; nits also fluoresce under Wood light.15 Although dermoscopy has proven useful for increased sensitivity and differentiation between viable and hatched nits, the insects also can be viewed with the unaided eye.16

Treatment: New Concerns and Strategies

The mainstay of treatment for body lice has long consisted of thorough washing and drying of all clothing and linens in a hot dryer. Treatment can be augmented with the addition of pharmacotherapy, plus antibiotics as warranted for louse-borne disease. Pharmacologic intervention often is used in cases of mass infestation and is similar to head lice.

Options for head lice include topical permethrin, malathion, lindane, spinosad, benzyl alcohol, and ivermectin. Pyrethroids, derived from the chrysanthemum, generally are considered safe for human use with a side-effect profile limited to irritation and allergy17; however, neurotoxicity and leukemia are clinical concerns, with an association more recently shown between large-volume use of pyrethroids and acute lymphoblastic leukemia.18,19 Use of lindane is not recommended due to a greater potential for central nervous system neurotoxicity, manifested by seizures, with repeated large surface application. Malathion is problematic due to the risk for mucosal irritation, flammability of some formulations, and theoretical organophosphate poisoning, as its mechanism of action involves inhibition of acetylcholinesterase.15 However, in the context of head lice treatment, a randomized controlled trial reported no incidence of acetylcholinesterase inhibition.20 Spinosad, manufactured from the soil bacterium Saccharopolyspora spinosa, functions similarly by interfering with the nicotinic acetylcholine receptor and also carries a risk for skin irritation.21 Among all the treatment options, we prefer benzyl alcohol, particularly in the context of resistance, as it is effective via a physical mechanism of action and lacks notable neurotoxic effects to the host. Use of benzyl alcohol is approved for patients as young as 6 months; it functions by asphyxiating the lice via paralysis of the respiratory spiracle with occlusion by inert ingredients. Itching, episodic numbness, and scalp or mucosal irritation are possible complications of treatment.22

Treatment resistance of body lice has increased in recent years, warranting exploration of additional management strategies. Moreover, developing resistance to lindane and malathion has been reported.23 Resistance to pyrethroids has been attributed to mutations in a voltage-gated sodium channel, one of which was universally present in the sampling of a single population.24 A randomized controlled trial showed that off-label oral ivermectin 400 μg/kg was superior to malathion lotion 0.5% in difficult-to-treat cases of head lice25; utility of oral ivermectin also has been reported in body lice.26 In vitro studies also have shown promise for pursuing synergistic treatment of body lice with both ivermectin and antibiotics.27



A novel primary prophylaxis approach for at-risk homeless individuals recently utilized permethrin-impregnated underwear. Although the intervention provided short-term infestation improvement, longer-term use did not show improvement from placebo and also increased prevalence of permethrin-resistant haplotypes.2

References
  1. Veracx A, Raoult D. Biology and genetics of human head and body lice. Trends Parasitol. 2012;28:563-571.
  2. Kittler R, Kayser M, Stoneking M. Molecular evolution of Pediculus humanus and the origin of clothing. Curr Biol. 2003;13:1414-1417.
  3. Drali R, Mumcuoglu KY, Yesilyurt G, et al. Studies of ancient lice reveal unsuspected past migrations of vectors. Am J Trop Med Hyg. 2015;93:623-625.
  4. Chosidow O. Scabies and pediculosis. Lancet. 2000;355:819-826.
  5. Feldmeier H, Heukelbach J. Epidermal parasitic skin diseases: a neglected category of poverty-associated plagues. Bull World Health Organ. 2009;87:152-159.
  6. Arnaud A, Chosidow O, Detrez MA, et al. Prevalence 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.
  7. Hytonen J, Khawaja T, Gronroos JO, et al. Louse-borne relapsing fever in Finland in two asylum seekers from Somalia. APMIS. 2017;125:59-62.
  8. Nordmann T, Feldt T, Bosselmann M, et al. Outbreak of louse-borne relapsing fever among urban dwellers in Arsi Zone, Central Ethiopia, from July to November 2016. Am J Trop Med Hyg. 2018;98:1599-1602.
  9. Louni M, Mana N, Bitam I, et al. Body lice of homeless people reveal the presence of several emerging bacterial pathogens in northern Algeria. PLoS Negl Trop Dis. 2018;12:E0006397.
  10. Candy K, Amanzougaghene N, Izri A, et al. Molecular survey of head and body lice, Pediculus humanus, in France. Vector Borne Zoonotic Dis. 2018;18:243-251.
  11. Bolognia JL, Schaffer JV, Cerroni L. Dermatology. 4th ed. Elsevier Limited; 2018.
  12. Nara A, Nagai H, Yamaguchi R, et al. An unusual autopsy case of lethal hypothermia exacerbated by body lice-induced severe anemia. Int J Legal Med. 2016;130:765-769.
  13. Althomali SA, Alzubaidi LM, Alkhaldi DM. Severe iron deficiency anaemia associated with heavy lice infestation in a young woman [published online November 5, 2015]. BMJ Case Rep. doi:10.1136/bcr-2015-212207.
  14. Hau V, Muhi-Iddin N. A ghost covered in lice: a case of severe blood loss with long-standing heavy pediculosis capitis infestation [published online December 19, 2014]. BMJ Case Rep. doi:10.1136/bcr-2014-206623.
  15. Diaz JH. Lice (Pediculosis). In: Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 9th ed. New York, NY: Elsevier; 2020:3482-3486.
  16. Martins LG, Bernardes Filho F, Quaresma MV, et al. Dermoscopy applied to pediculosis corporis diagnosis. An Bras Dermatol. 2014;89:513-514.
  17. Devore CD, Schutze GE; Council on School Health and Committee on Infectious Diseases, American Academy of Pediatrics. Head lice. Pediatrics. 2015;135:E1355-E1365.
  18. Shafer TJ, Meyer DA, Crofton KM. Developmental neurotoxicity of pyrethroid insecticides: critical review and future research needs. Environ Health Perspect. 2005;113:123-136.
  19. Ding G, Shi R, Gao Y, et al. Pyrethroid pesticide exposure and risk of childhood acute lymphocytic leukemia in Shanghai. Environ Sci Technol. 2012;46:13480-13487.
  20. Meinking TL, Vicaria M, Eyerdam DH, et al. A randomized, investigator-blinded, time-ranging study of the comparative efficacy of 0.5% malathion gel versus Ovide Lotion (0.5% malathion) or Nix Crème Rinse (1% permethrin) used as labeled, for the treatment of head lice. Pediatr Dermatol. 2007;24:405-411.
  21. McCormack PL. Spinosad: in pediculosis capitis. Am J Clin Dermatol. 2011;12:349-353.
  22. Meinking TL, Villar ME, Vicaria M, et al. The clinical trials supporting benzyl alcohol lotion 5% (Ulesfia): a safe and effective topical treatment for head lice (pediculosis humanus capitis). Pediatr Dermatol. 2010;27:19-24.
  23. Lebwohl M, Clark L, Levitt J. Therapy for head lice based on life cycle, resistance, and safety considerations. Pediatrics. 2007;119:965-974
  24. Drali R, Benkouiten S, Badiaga S, et al. Detection of a knockdown resistance mutation associated with permethrin resistance in the body louse Pediculus humanus corporis by use of melting curve analysis genotyping. J Clin Microbiol. 2012;50:2229-2233.
  25. Chosidow O, Giraudeau B, Cottrell J, et al. Oral ivermectin versus malathion lotion for difficult-to-treat head lice. N Engl J Med. 2010;362:896-905.
  26. Foucault C, Ranque S, Badiaga S, et al. Oral ivermectin in the treatment of body lice. J Infect Dis. 2006;193:474-476.
  27. Sangaré AK, Doumbo OK, Raoult D. Management and treatment of human lice [published online July 27, 2016]. Biomed Res Int. doi:10.1155/2016/8962685.
  28. 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.
References
  1. Veracx A, Raoult D. Biology and genetics of human head and body lice. Trends Parasitol. 2012;28:563-571.
  2. Kittler R, Kayser M, Stoneking M. Molecular evolution of Pediculus humanus and the origin of clothing. Curr Biol. 2003;13:1414-1417.
  3. Drali R, Mumcuoglu KY, Yesilyurt G, et al. Studies of ancient lice reveal unsuspected past migrations of vectors. Am J Trop Med Hyg. 2015;93:623-625.
  4. Chosidow O. Scabies and pediculosis. Lancet. 2000;355:819-826.
  5. Feldmeier H, Heukelbach J. Epidermal parasitic skin diseases: a neglected category of poverty-associated plagues. Bull World Health Organ. 2009;87:152-159.
  6. Arnaud A, Chosidow O, Detrez MA, et al. Prevalence 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.
  7. Hytonen J, Khawaja T, Gronroos JO, et al. Louse-borne relapsing fever in Finland in two asylum seekers from Somalia. APMIS. 2017;125:59-62.
  8. Nordmann T, Feldt T, Bosselmann M, et al. Outbreak of louse-borne relapsing fever among urban dwellers in Arsi Zone, Central Ethiopia, from July to November 2016. Am J Trop Med Hyg. 2018;98:1599-1602.
  9. Louni M, Mana N, Bitam I, et al. Body lice of homeless people reveal the presence of several emerging bacterial pathogens in northern Algeria. PLoS Negl Trop Dis. 2018;12:E0006397.
  10. Candy K, Amanzougaghene N, Izri A, et al. Molecular survey of head and body lice, Pediculus humanus, in France. Vector Borne Zoonotic Dis. 2018;18:243-251.
  11. Bolognia JL, Schaffer JV, Cerroni L. Dermatology. 4th ed. Elsevier Limited; 2018.
  12. Nara A, Nagai H, Yamaguchi R, et al. An unusual autopsy case of lethal hypothermia exacerbated by body lice-induced severe anemia. Int J Legal Med. 2016;130:765-769.
  13. Althomali SA, Alzubaidi LM, Alkhaldi DM. Severe iron deficiency anaemia associated with heavy lice infestation in a young woman [published online November 5, 2015]. BMJ Case Rep. doi:10.1136/bcr-2015-212207.
  14. Hau V, Muhi-Iddin N. A ghost covered in lice: a case of severe blood loss with long-standing heavy pediculosis capitis infestation [published online December 19, 2014]. BMJ Case Rep. doi:10.1136/bcr-2014-206623.
  15. Diaz JH. Lice (Pediculosis). In: Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 9th ed. New York, NY: Elsevier; 2020:3482-3486.
  16. Martins LG, Bernardes Filho F, Quaresma MV, et al. Dermoscopy applied to pediculosis corporis diagnosis. An Bras Dermatol. 2014;89:513-514.
  17. Devore CD, Schutze GE; Council on School Health and Committee on Infectious Diseases, American Academy of Pediatrics. Head lice. Pediatrics. 2015;135:E1355-E1365.
  18. Shafer TJ, Meyer DA, Crofton KM. Developmental neurotoxicity of pyrethroid insecticides: critical review and future research needs. Environ Health Perspect. 2005;113:123-136.
  19. Ding G, Shi R, Gao Y, et al. Pyrethroid pesticide exposure and risk of childhood acute lymphocytic leukemia in Shanghai. Environ Sci Technol. 2012;46:13480-13487.
  20. Meinking TL, Vicaria M, Eyerdam DH, et al. A randomized, investigator-blinded, time-ranging study of the comparative efficacy of 0.5% malathion gel versus Ovide Lotion (0.5% malathion) or Nix Crème Rinse (1% permethrin) used as labeled, for the treatment of head lice. Pediatr Dermatol. 2007;24:405-411.
  21. McCormack PL. Spinosad: in pediculosis capitis. Am J Clin Dermatol. 2011;12:349-353.
  22. Meinking TL, Villar ME, Vicaria M, et al. The clinical trials supporting benzyl alcohol lotion 5% (Ulesfia): a safe and effective topical treatment for head lice (pediculosis humanus capitis). Pediatr Dermatol. 2010;27:19-24.
  23. Lebwohl M, Clark L, Levitt J. Therapy for head lice based on life cycle, resistance, and safety considerations. Pediatrics. 2007;119:965-974
  24. Drali R, Benkouiten S, Badiaga S, et al. Detection of a knockdown resistance mutation associated with permethrin resistance in the body louse Pediculus humanus corporis by use of melting curve analysis genotyping. J Clin Microbiol. 2012;50:2229-2233.
  25. Chosidow O, Giraudeau B, Cottrell J, et al. Oral ivermectin versus malathion lotion for difficult-to-treat head lice. N Engl J Med. 2010;362:896-905.
  26. Foucault C, Ranque S, Badiaga S, et al. Oral ivermectin in the treatment of body lice. J Infect Dis. 2006;193:474-476.
  27. Sangaré AK, Doumbo OK, Raoult D. Management and treatment of human lice [published online July 27, 2016]. Biomed Res Int. doi:10.1155/2016/8962685.
  28. 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.
Issue
Cutis - 105(3)
Issue
Cutis - 105(3)
Page Number
118-120
Page Number
118-120
Publications
Publications
Topics
Article Type
Display Headline
What’s Eating You? Human Body Lice (Pediculus humanus corporis)
Display Headline
What’s Eating You? Human Body Lice (Pediculus humanus corporis)
Sections
Inside the Article

Practice Points

  • Body lice reside in clothing, particularly folds and seams, and migrate to the host for blood meals. To evaluate for infestation, the clinician should not only look at the skin but also closely examine the patient’s clothing. Clothes also are a target for treatment via washing in hot water.
  • Due to observed and theoretical adverse effects of other chemical treatments, benzyl alcohol is the authors’ choice for treatment of head lice.
  • Oral ivermectin is a promising future treatment for body lice.
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
Article PDF Media

What’s Eating You? Vespids Revisited

Article Type
Changed
Mon, 01/13/2020 - 10:02

Identification

The Hymenoptera order of insects includes Apidae (bees), Vespidae (wasps, yellow jackets, hornets), and Formicidae (fire ants). All 3 of these families of insects inject venom into their prey or as a defense mechanism via ovipositors in their abdomen. Vespids are the most aggressive and are found in each of the United States.1 They have membranous wings, broad antennae, and a nonbarbed stinger (Figure 1).2 The nonbarbed stinger of Vespidae differentiates them from Apidae and allows these insects to sting their prey multiple times. Vespids can build nests in the ground (yellow jackets), trees (hornets), or areas of cover such as window shutters (mud wasps). Because only the queens survive winter, larger populations do not develop until late summer when the most stings take place. Stings most often take place near the nest of the vespid or while the victim is eating outdoors.3

Figure 1. Vespids. A, The anatomy of a hornet. B, A brilliant image of a yellow jacket. C, A detailed image of a wasp.

Envenomation

When vespids sting their prey they inject venom via their ovipositors.1 The venom is composed of a mixture of low-molecular-weight proteins, kinins, proteolytic enzymes, lipids, carbohydrates, and high-molecular-weight proteins that act as allergens.1,4,5 The proteolytic enzymes degrade the surrounding tissue, basophils become activated, and histamine is released secondary to mast cell degranulation, which results in vasodilation and an inflammatory response characterized by edema, erythema, warmth, and pain.1 The pain of the sting is immediate and can be intense; almost all victims are acutely aware of the discomforting sensation.4

Management of Reactions

Three types of reactions can be seen after a vespid sting: uncomplicated local reactions, large local reactions, and systemic reactions (SRs). The most common reaction is the self-limiting uncomplicated local reaction that includes a focal area of warmth, edema, erythema, induration, and tenderness.1 Treatment of this kind of reaction is supportive, with ice, nonsteroidal anti-inflammatory drugs, and H1 and H2 blockers being commonly used methods. Large local reactions (Figure 2) are similar to uncomplicated local reactions but are greater than 10 cm in diameter and last longer. The same symptomatic treatment may be used along with possible short (3–5 days) oral glucocorticoid (40–60 mg prednisone) or potent topical steroid administration if symptoms persist. Systemic reactions involve IgE-mediated generalized urticaria, angioedema, face swelling, stridor, bronchospasm, nausea, vomiting, flushing, and respiratory distress.1 Emergency management includes maintenance of airway, breathing, and circulation. Epinephrine injection commonly is employed and should be given via intramuscular injection into the anterolateral thigh; a dose of 0.3 to 0.5 mg can be repeatedly injected every 5 to 15 minutes, as needed.1

Figure 2. A large local reaction after a wasp sting.

If an individual has an SR, it is recommended to go to an emergency department after stabilization for monitoring. Referral to an allergist for desensitization is appropriate. A radioallergosorbent test to measure allergen-specific IgE can be helpful to confirm an allergy.4 This test also should be done weeks after the incident because during the first few days IgE may be too low to measure. Once the allergy is confirmed, the desensitization with venom immunotherapy (VIT) can begin. Venom immunotherapy is effective and reduces a patient’s risk for recurrent SRs to less than 5% to 20%.6 A 2015 study recommended longer duration of VIT therapy due to risk for repeat SRs after discontinuing therapy. This study concluded that VIT is to be administered for 5 years, unless the patient is at high risk for SRs after VIT therapy—risk factors include older age, cardiopulmonary disease, SR during VIT treatment, mast cell disorders, and elevated serum tryptase—in which case VIT may have to be continued indefinitely. It is recommended that all patients with history of SR carry an epinephrine autoinjector in case of emergency.6



Epidemiologic data show a prevalence of 0.3% to 7.5% for self-reported SRs due to stings, with lower prevalence in children (0.15%–0.3%).4,7 An additional study looking at data from an allergy practice determined 24% of all cases of anaphylaxis were due to insect stings.5

Conclusion

Although many vespid stings can be managed symptomatically, it is imperative for patients and providers to be aware of the possible severe reactions that can take place. It is essential for providers to be aware of how to care for and treat large local reactions and SRs, as symptom recognition and timely treatment can improve patient safety and result in better outcomes.

References
  1. Arif F, Williams M. Hymenoptera Stings (Bee, Vespids and Ants). Treasure Island, FL: StatPearls Publishing LLC; 2019.  https://www.ncbi.nlm.nih.gov/books/NBK518972/. Updated April 20, 2019. Accessed December 11, 2019.
  2. Elston, DM. Life-threatening stings, bites, infestations, and parasitic diseases. Clin Dermatol. 2005;23:164-170.
  3. Ulrich RM, Gabrielle H, Arthur H. Allergic reactions to stinging and biting insects. In: Rich RR, Fleisher T, Shearer W, et al, eds. Clinical Immunology: Principles and Practice. 3rd ed. St. Louis, MO: Mosby/Elsevier; 2008:657-666.
  4. Biló BM, Rueff F, Mosbech H, et al. Diagnosis of hymenoptera venom allergy. Allergy. 2005;60:1339-1349.
  5. Schafer T, Przybilla B. IgE antibodies to hymenoptera venoms in the serum are common in the general population and are related to indication of atopy. Allergy. 1996;51:372-377.
  6. Ulrich MR, Johannes R. When can immunotherapy for insect sting allergy be stopped? J Allergy Clin Immunol. 2015;3:324-328.
  7. Abrishami MH, Boyd GK, Settipane GA. Prevalence of bee sting allergy in 2010 girl scouts. Acta Allergol. 1971;26:117-120.
Article PDF
Author and Disclosure Information

Dr. Evans is from the University of Nebraska Medical Center, Omaha. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Images are in the public domain.

Correspondence: Dirk M. Elston, MD, Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, 135 Rutledge Ave, MSC 578, Charleston, SC 29425 (elstond@musc.edu).

Issue
Cutis - 105(1)
Publications
Topics
Page Number
17-18
Sections
Author and Disclosure Information

Dr. Evans is from the University of Nebraska Medical Center, Omaha. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Images are in the public domain.

Correspondence: Dirk M. Elston, MD, Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, 135 Rutledge Ave, MSC 578, Charleston, SC 29425 (elstond@musc.edu).

Author and Disclosure Information

Dr. Evans is from the University of Nebraska Medical Center, Omaha. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Images are in the public domain.

Correspondence: Dirk M. Elston, MD, Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, 135 Rutledge Ave, MSC 578, Charleston, SC 29425 (elstond@musc.edu).

Article PDF
Article PDF

Identification

The Hymenoptera order of insects includes Apidae (bees), Vespidae (wasps, yellow jackets, hornets), and Formicidae (fire ants). All 3 of these families of insects inject venom into their prey or as a defense mechanism via ovipositors in their abdomen. Vespids are the most aggressive and are found in each of the United States.1 They have membranous wings, broad antennae, and a nonbarbed stinger (Figure 1).2 The nonbarbed stinger of Vespidae differentiates them from Apidae and allows these insects to sting their prey multiple times. Vespids can build nests in the ground (yellow jackets), trees (hornets), or areas of cover such as window shutters (mud wasps). Because only the queens survive winter, larger populations do not develop until late summer when the most stings take place. Stings most often take place near the nest of the vespid or while the victim is eating outdoors.3

Figure 1. Vespids. A, The anatomy of a hornet. B, A brilliant image of a yellow jacket. C, A detailed image of a wasp.

Envenomation

When vespids sting their prey they inject venom via their ovipositors.1 The venom is composed of a mixture of low-molecular-weight proteins, kinins, proteolytic enzymes, lipids, carbohydrates, and high-molecular-weight proteins that act as allergens.1,4,5 The proteolytic enzymes degrade the surrounding tissue, basophils become activated, and histamine is released secondary to mast cell degranulation, which results in vasodilation and an inflammatory response characterized by edema, erythema, warmth, and pain.1 The pain of the sting is immediate and can be intense; almost all victims are acutely aware of the discomforting sensation.4

Management of Reactions

Three types of reactions can be seen after a vespid sting: uncomplicated local reactions, large local reactions, and systemic reactions (SRs). The most common reaction is the self-limiting uncomplicated local reaction that includes a focal area of warmth, edema, erythema, induration, and tenderness.1 Treatment of this kind of reaction is supportive, with ice, nonsteroidal anti-inflammatory drugs, and H1 and H2 blockers being commonly used methods. Large local reactions (Figure 2) are similar to uncomplicated local reactions but are greater than 10 cm in diameter and last longer. The same symptomatic treatment may be used along with possible short (3–5 days) oral glucocorticoid (40–60 mg prednisone) or potent topical steroid administration if symptoms persist. Systemic reactions involve IgE-mediated generalized urticaria, angioedema, face swelling, stridor, bronchospasm, nausea, vomiting, flushing, and respiratory distress.1 Emergency management includes maintenance of airway, breathing, and circulation. Epinephrine injection commonly is employed and should be given via intramuscular injection into the anterolateral thigh; a dose of 0.3 to 0.5 mg can be repeatedly injected every 5 to 15 minutes, as needed.1

Figure 2. A large local reaction after a wasp sting.

If an individual has an SR, it is recommended to go to an emergency department after stabilization for monitoring. Referral to an allergist for desensitization is appropriate. A radioallergosorbent test to measure allergen-specific IgE can be helpful to confirm an allergy.4 This test also should be done weeks after the incident because during the first few days IgE may be too low to measure. Once the allergy is confirmed, the desensitization with venom immunotherapy (VIT) can begin. Venom immunotherapy is effective and reduces a patient’s risk for recurrent SRs to less than 5% to 20%.6 A 2015 study recommended longer duration of VIT therapy due to risk for repeat SRs after discontinuing therapy. This study concluded that VIT is to be administered for 5 years, unless the patient is at high risk for SRs after VIT therapy—risk factors include older age, cardiopulmonary disease, SR during VIT treatment, mast cell disorders, and elevated serum tryptase—in which case VIT may have to be continued indefinitely. It is recommended that all patients with history of SR carry an epinephrine autoinjector in case of emergency.6



Epidemiologic data show a prevalence of 0.3% to 7.5% for self-reported SRs due to stings, with lower prevalence in children (0.15%–0.3%).4,7 An additional study looking at data from an allergy practice determined 24% of all cases of anaphylaxis were due to insect stings.5

Conclusion

Although many vespid stings can be managed symptomatically, it is imperative for patients and providers to be aware of the possible severe reactions that can take place. It is essential for providers to be aware of how to care for and treat large local reactions and SRs, as symptom recognition and timely treatment can improve patient safety and result in better outcomes.

Identification

The Hymenoptera order of insects includes Apidae (bees), Vespidae (wasps, yellow jackets, hornets), and Formicidae (fire ants). All 3 of these families of insects inject venom into their prey or as a defense mechanism via ovipositors in their abdomen. Vespids are the most aggressive and are found in each of the United States.1 They have membranous wings, broad antennae, and a nonbarbed stinger (Figure 1).2 The nonbarbed stinger of Vespidae differentiates them from Apidae and allows these insects to sting their prey multiple times. Vespids can build nests in the ground (yellow jackets), trees (hornets), or areas of cover such as window shutters (mud wasps). Because only the queens survive winter, larger populations do not develop until late summer when the most stings take place. Stings most often take place near the nest of the vespid or while the victim is eating outdoors.3

Figure 1. Vespids. A, The anatomy of a hornet. B, A brilliant image of a yellow jacket. C, A detailed image of a wasp.

Envenomation

When vespids sting their prey they inject venom via their ovipositors.1 The venom is composed of a mixture of low-molecular-weight proteins, kinins, proteolytic enzymes, lipids, carbohydrates, and high-molecular-weight proteins that act as allergens.1,4,5 The proteolytic enzymes degrade the surrounding tissue, basophils become activated, and histamine is released secondary to mast cell degranulation, which results in vasodilation and an inflammatory response characterized by edema, erythema, warmth, and pain.1 The pain of the sting is immediate and can be intense; almost all victims are acutely aware of the discomforting sensation.4

Management of Reactions

Three types of reactions can be seen after a vespid sting: uncomplicated local reactions, large local reactions, and systemic reactions (SRs). The most common reaction is the self-limiting uncomplicated local reaction that includes a focal area of warmth, edema, erythema, induration, and tenderness.1 Treatment of this kind of reaction is supportive, with ice, nonsteroidal anti-inflammatory drugs, and H1 and H2 blockers being commonly used methods. Large local reactions (Figure 2) are similar to uncomplicated local reactions but are greater than 10 cm in diameter and last longer. The same symptomatic treatment may be used along with possible short (3–5 days) oral glucocorticoid (40–60 mg prednisone) or potent topical steroid administration if symptoms persist. Systemic reactions involve IgE-mediated generalized urticaria, angioedema, face swelling, stridor, bronchospasm, nausea, vomiting, flushing, and respiratory distress.1 Emergency management includes maintenance of airway, breathing, and circulation. Epinephrine injection commonly is employed and should be given via intramuscular injection into the anterolateral thigh; a dose of 0.3 to 0.5 mg can be repeatedly injected every 5 to 15 minutes, as needed.1

Figure 2. A large local reaction after a wasp sting.

If an individual has an SR, it is recommended to go to an emergency department after stabilization for monitoring. Referral to an allergist for desensitization is appropriate. A radioallergosorbent test to measure allergen-specific IgE can be helpful to confirm an allergy.4 This test also should be done weeks after the incident because during the first few days IgE may be too low to measure. Once the allergy is confirmed, the desensitization with venom immunotherapy (VIT) can begin. Venom immunotherapy is effective and reduces a patient’s risk for recurrent SRs to less than 5% to 20%.6 A 2015 study recommended longer duration of VIT therapy due to risk for repeat SRs after discontinuing therapy. This study concluded that VIT is to be administered for 5 years, unless the patient is at high risk for SRs after VIT therapy—risk factors include older age, cardiopulmonary disease, SR during VIT treatment, mast cell disorders, and elevated serum tryptase—in which case VIT may have to be continued indefinitely. It is recommended that all patients with history of SR carry an epinephrine autoinjector in case of emergency.6



Epidemiologic data show a prevalence of 0.3% to 7.5% for self-reported SRs due to stings, with lower prevalence in children (0.15%–0.3%).4,7 An additional study looking at data from an allergy practice determined 24% of all cases of anaphylaxis were due to insect stings.5

Conclusion

Although many vespid stings can be managed symptomatically, it is imperative for patients and providers to be aware of the possible severe reactions that can take place. It is essential for providers to be aware of how to care for and treat large local reactions and SRs, as symptom recognition and timely treatment can improve patient safety and result in better outcomes.

References
  1. Arif F, Williams M. Hymenoptera Stings (Bee, Vespids and Ants). Treasure Island, FL: StatPearls Publishing LLC; 2019.  https://www.ncbi.nlm.nih.gov/books/NBK518972/. Updated April 20, 2019. Accessed December 11, 2019.
  2. Elston, DM. Life-threatening stings, bites, infestations, and parasitic diseases. Clin Dermatol. 2005;23:164-170.
  3. Ulrich RM, Gabrielle H, Arthur H. Allergic reactions to stinging and biting insects. In: Rich RR, Fleisher T, Shearer W, et al, eds. Clinical Immunology: Principles and Practice. 3rd ed. St. Louis, MO: Mosby/Elsevier; 2008:657-666.
  4. Biló BM, Rueff F, Mosbech H, et al. Diagnosis of hymenoptera venom allergy. Allergy. 2005;60:1339-1349.
  5. Schafer T, Przybilla B. IgE antibodies to hymenoptera venoms in the serum are common in the general population and are related to indication of atopy. Allergy. 1996;51:372-377.
  6. Ulrich MR, Johannes R. When can immunotherapy for insect sting allergy be stopped? J Allergy Clin Immunol. 2015;3:324-328.
  7. Abrishami MH, Boyd GK, Settipane GA. Prevalence of bee sting allergy in 2010 girl scouts. Acta Allergol. 1971;26:117-120.
References
  1. Arif F, Williams M. Hymenoptera Stings (Bee, Vespids and Ants). Treasure Island, FL: StatPearls Publishing LLC; 2019.  https://www.ncbi.nlm.nih.gov/books/NBK518972/. Updated April 20, 2019. Accessed December 11, 2019.
  2. Elston, DM. Life-threatening stings, bites, infestations, and parasitic diseases. Clin Dermatol. 2005;23:164-170.
  3. Ulrich RM, Gabrielle H, Arthur H. Allergic reactions to stinging and biting insects. In: Rich RR, Fleisher T, Shearer W, et al, eds. Clinical Immunology: Principles and Practice. 3rd ed. St. Louis, MO: Mosby/Elsevier; 2008:657-666.
  4. Biló BM, Rueff F, Mosbech H, et al. Diagnosis of hymenoptera venom allergy. Allergy. 2005;60:1339-1349.
  5. Schafer T, Przybilla B. IgE antibodies to hymenoptera venoms in the serum are common in the general population and are related to indication of atopy. Allergy. 1996;51:372-377.
  6. Ulrich MR, Johannes R. When can immunotherapy for insect sting allergy be stopped? J Allergy Clin Immunol. 2015;3:324-328.
  7. Abrishami MH, Boyd GK, Settipane GA. Prevalence of bee sting allergy in 2010 girl scouts. Acta Allergol. 1971;26:117-120.
Issue
Cutis - 105(1)
Issue
Cutis - 105(1)
Page Number
17-18
Page Number
17-18
Publications
Publications
Topics
Article Type
Sections
Inside the Article

Practice Points

  • Most vespid stings can be managed with nonsteroidal anti-inflammatory drugs, ice, and antihistamines.
  • For systemic reactions, prompt recognition and initiation of intramuscular epinephrine is recommended.
  • In patients with confirmed allergy, recent data now suggest at least 5 years of venom immunotherapy and potentially lifelong for specific patients.
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.
Article PDF Media