User login
Botanical Briefs: Bloodroot (Sanguinaria canadensis)
Bloodroot (Sanguinaria canadensis) is a member of the family Papaveraceae.1 This North American plant commonly is found in widespread distribution from Nova Scotia, Canada, to Florida and from the Great Lakes to Mississippi.2 Historically, Native Americans used bloodroot as a skin dye and as a medicine for many ailments.3
Bloodroot blooms for only a few days, starting in March, and fruits in June. The flowers comprise 8 to 10 white petals, surrounding a bed of yellow stamens (Figure). The plant thrives in wooded areas and grows to 12 inches tall. In its off-season, the plant remains dormant and can survive below-freezing temperatures.4
Chemical Constituents
Bloodroot gets its colloquial name from its red sap, which is released when the plant’s rhizome is cut. This sap contains a high concentration of alkaloids that are used for protection against predators. The rhizome itself has a rusty, red-brown color; the roots are a brighter red-orange.4
The rhizome of S canadensis contains the highest concentration of active alkaloids; the roots also contain these chemicals, though to a lesser degree; and the leaves, flowers, and fruits harvest approximately 1% of the alkaloids found in the roots.4 The concentration of alkaloids can vary from one plant to the next, depending on environmental conditions.5,6
The major alkaloids in S canadensis include both quaternary benzophenanthridine alkaloids (eg, sanguinarine, chelerythrine, sanguilutine, chelilutine, sanguirubine, chelirubine) and protopin alkaloids (eg, protopine, allocryptopine).3,7 Of these, sanguinarine and chelerythrine typically are the most potent.1 Oral ingestion or topical application of these molecules can have therapeutic and toxic effects.8
Biophysiological Effects
Bloodroot has been shown to have remarkable antimicrobial effects.9 The plant produces hydrogen peroxide and superoxide anion.10 These mediators cause oxidative stress, thus inducing destruction of cellular DNA and the cell membrane.11 Although these effects can be helpful when fighting infection, they are not necessarily selective against healthy cells.12
Alkaloids of bloodroot also have cardiovascular therapeutic effects. Sanguinarine blocks angiotensin II and causes vasodilation, thus helping treat hypertension.13 It also acts as an inotrope by blocking the Na+/K+ ATPase pump. These effects in a patient who is already taking digoxin can cause notable cardiotoxicity because the 2 drugs share a mechanism of action.14
Chelerythrine blocks production of cyclooxygenase 2 and prostaglandin E2.15 This pathway modification results in anti-inflammatory effects that can help treat arthritis, edema, and other inflammatory conditions.16 Moreover, sanguinarine has demonstrated efficacy in numerous anticancer pathways,17 including downregulation of intercellular adhesion molecules, vascular cell adhesion molecules, and vascular endothelial growth factor (VEGF).18-20 Blocking VEGF is one way to inhibit angiogenesis,21 which is upregulated in tumor formation, thus sanguinarine can have an antiproliferative anticancer effect.22 Sanguinarine also upregulates molecules such as nuclear factor–κB and the protease enzymes known as caspases to cause proapoptotic effects, furthering its antitumor potential.23,24
Treatment of Dermatologic Conditions
The initial technique of Mohs micrographic surgery employed a chemopaste that utilized an extract of S canadensis to preserve tissue.25 Outside the dermatologist’s office, bloodroot is used as a topical home remedy for a variety of cutaneous conditions, including cancer, skin tags, and warts.26 Bloodroot is advertised as black salve, an alternative anticancer treatment.27,28
As useful as this natural agent sounds, it has a pitfall: The alkaloids of S canadensis are nonspecific in their cytotoxicity, damaging neoplastic and healthy tissue.29 This cytotoxic effect can cause escharification through diffuse tissue destruction and has been observed to result in formation of a keloid scar.30 The alkaloids in black salve also have been shown to cause skin erosions and cellular atypia.28,31 Therefore, the utility of this escharotic in medical treatment is limited.32 Fortuitously, oral antibiotics and wound care can help address this adverse effect.28
Bloodroot was once used as a mouth rinse and toothpaste to treat gingivitis, but this application was later associated with oral leukoplakia, a premalignant condition.33 Leukoplakia associated with S canadensis extract often is unremitting. Immediate discontinuation of the offending agent produces little regression, suggesting that cellular damage is irreversible.34
Final Thoughts
Although bloodroot demonstrates efficacy as a phytotherapeutic, it does come with notable toxicity. Physicians should warn patients of the unwanted cosmetic effects of black salve, especially oral products that incorporate sanguinarine. Adverse effects on the oropharynx can be irreversible, though the eschar associated with black salve can be treated with a topical or oral corticosteroid.29
- Vogel M, Lawson M, Sippl W, et al. Structure and mechanism of sanguinarine reductase, an enzyme of alkaloid detoxification. J Biol Chem. 2010;285:18397-18406. doi:10.1074/jbc.M109.088989
- Maranda EL, Wang MX, Cortizo J, et al. Flower power—the versatility of bloodroot. JAMA Dermatol. 2016;152:824. doi:10.1001/jamadermatol.2015.5522
- Setzer WN. The phytochemistry of Cherokee aromatic medicinal plants. Medicines (Basel). 2018;5:121. doi:10.3390/medicines5040121
- Croaker A, King GJ, Pyne JH, et al. Sanguinaria canadensis: traditional medicine, phytochemical composition, biological activities and current uses. Int J Mol Sci. 2016;17:1414. doi:10.3390/ijms17091414
- Graf TN, Levine KE, Andrews ME, et al. Variability in the yield of benzophenanthridine alkaloids in wildcrafted vs cultivated bloodroot (Sanguinaria canadensis L.) J Agric Food Chem. 2007; 55:1205-1211. doi:10.1021/jf062498f
- Bennett BC, Bell CR, Boulware RT. Geographic variation in alkaloid content of Sanguinaria canadensis (Papaveraceae). Rhodora. 1990;92:57-69.
- Leaver CA, Yuan H, Wallen GR. Apoptotic activities of Sanguinaria canadensis: primary human keratinocytes, C-33A, and human papillomavirus HeLa cervical cancer lines. Integr Med (Encinitas). 2018;17:32-37.
- Kutchan TM. Molecular genetics of plant alkaloid biosynthesis. In: Cordell GA, ed. The Alkaloids. Vol 50. Elsevier Science Publishing Co, Inc; 1997:257-316.
- Obiang-Obounou BW, Kang O-H, Choi J-G, et al. The mechanism of action of sanguinarine against methicillin-resistant Staphylococcus aureus. J Toxicol Sci. 2011;36:277-283. doi:10.2131/jts.36.277
- Z˙abka A, Winnicki K, Polit JT, et al. Sanguinarine-induced oxidative stress and apoptosis-like programmed cell death (AL-PCD) in root meristem cells of Allium cepa. Plant Physiol Biochem. 2017;112:193-206. doi:10.1016/j.plaphy.2017.01.004
- Kumar GS, Hazra S. Sanguinarine, a promising anticancer therapeutic: photochemical and nucleic acid binding properties. RSC Advances. 2014;4:56518-56531.
- Ping G, Wang Y, Shen L, et al. Highly efficient complexation of sanguinarine alkaloid by carboxylatopillar[6]arene: pKa shift, increased solubility and enhanced antibacterial activity. Chemical Commun (Camb). 2017;53:7381-7384. doi:10.1039/c7cc02799k
- Caballero-George C, Vanderheyden PM, Solis PN, et al. Biological screening of selected medicinal Panamanian plants by radioligand-binding techniques. Phytomedicine. 2001;8:59-70. doi:10.1078/0944-7113-00011
- Seifen E, Adams RJ, Riemer RK. Sanguinarine: a positive inotropic alkaloid which inhibits cardiac Na+, K+-ATPase. Eur J Pharmacol. 1979;60:373-377. doi:10.1016/0014-2999(79)90245-0
- Debprasad C, Hemanta M, Paromita B, et al. Inhibition of NO2, PGE2, TNF-α, and iNOS EXpression by Shorea robusta L.: an ethnomedicine used for anti-inflammatory and analgesic activity. Evid Based Complement Alternat Med. 2012; 2012:254849. doi:10.1155/2012/254849
- Melov S, Ravenscroft J, Malik S, et al. Extension of life-span with superoxide dismutase/catalase mimetics. Science. 2000;289:1567-1569. doi:10.1126/science.289.5484.1567
- Basu P, Kumar GS. Sanguinarine and its role in chronic diseases. In: Gupta SC, Prasad S, Aggarwal BB, eds. Advances in Experimental Medicine and Biology: Anti-inflammatory Nutraceuticals and Chronic Diseases. Vol 928. Springer International Publishing; 2016:155-172.
- Alasvand M, Assadollahi V, Ambra R, et al. Antiangiogenic effect of alkaloids. Oxid Med Cell Longev. 2019;2019:9475908. doi:10.1155/2019/9475908
- Basini G, Santini SE, Bussolati S, et al. The plant alkaloid sanguinarine is a potential inhibitor of follicular angiogenesis. J Reprod Dev. 2007;53:573-579. doi:10.1262/jrd.18126
- Xu J-Y, Meng Q-H, Chong Y, et al. Sanguinarine is a novel VEGF inhibitor involved in the suppression of angiogenesis and cell migration. Mol Clin Oncol. 2013;1:331-336. doi:10.3892/mco.2012.41
- Lu K, Bhat M, Basu S. Plants and their active compounds: natural molecules to target angiogenesis. Angiogenesis. 2016;19:287-295. doi:10.1007/s10456-016-9512-y
- Achkar IW, Mraiche F, Mohammad RM, et al. Anticancer potential of sanguinarine for various human malignancies. Future Med Chem. 2017;9:933-950. doi:10.4155/fmc-2017-0041
- Lee TK, Park C, Jeong S-J, et al. Sanguinarine induces apoptosis of human oral squamous cell carcinoma KB cells via inactivation of the PI3K/Akt signaling pathway. Drug Dev Res. 2016;77:227-240. doi:10.1002/ddr.21315
- Gaziano R, Moroni G, Buè C, et al. Antitumor effects of the benzophenanthridine alkaloid sanguinarine: evidence and perspectives. World J Gastrointest Oncol. 2016;8:30-39. doi:10.4251/wjgo.v8.i1.30
- Mohs FE. Chemosurgery for skin cancer: fixed tissue and fresh tissue techniques. Arch Dermatol. 1976;112:211-215.
- Affleck AG, Varma S. A case of do-it-yourself Mohs’ surgery using bloodroot obtained from the internet. Br J Dermatol. 2007;157:1078-1079. doi:10.1111/j.1365-2133.2007.08180.x
- Eastman KL, McFarland LV, Raugi GJ. Buyer beware: a black salve caution. J Am Acad Dermatol. 2011;65:E154-E155. doi:10.1016/j.jaad.2011.07.031
- Osswald SS, Elston DM, Farley MF, et al. Self-treatment of a basal cell carcinoma with “black and yellow salve.” J Am Acad Dermatol. 2005;53:508-510. doi:10.1016/j.jaad.2005.04.007
- Schlichte MJ, Downing CP, Ramirez-Fort M, et al. Bloodroot associated eschar. Dermatol Online J. 2015;20:13030/qt05r0r2wr.
- Wang MZ, Warshaw EM. Bloodroot. Dermatitis. 2012;23:281-283. doi:10.1097/DER.0b013e318273a4dd
- Tan JM, Peters P, Ong N, et al. Histopathological features after topical black salve application. Australas J Dermatol. 2015;56:75-76.
- Hou JL, Brewer JD. Black salve and bloodroot extract in dermatologic conditions. Cutis. 2015;95:309-311.
- Eversole LR, Eversole GM, Kopcik J. Sanguinaria-associated oral leukoplakia: comparison with other benign and dysplastic leukoplakic lesions. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2000;89:455-464. doi:10.1016/s1079-2104(00)70125-9
- Mascarenhas AK, Allen CM, Moeschberger ML. The association between Viadent® use and oral leukoplakia—results of a matched case-control study. J Public Health Dent. 2002;62:158-162. doi:10.1111/j.1752-7325.2002.tb03437.x
Bloodroot (Sanguinaria canadensis) is a member of the family Papaveraceae.1 This North American plant commonly is found in widespread distribution from Nova Scotia, Canada, to Florida and from the Great Lakes to Mississippi.2 Historically, Native Americans used bloodroot as a skin dye and as a medicine for many ailments.3
Bloodroot blooms for only a few days, starting in March, and fruits in June. The flowers comprise 8 to 10 white petals, surrounding a bed of yellow stamens (Figure). The plant thrives in wooded areas and grows to 12 inches tall. In its off-season, the plant remains dormant and can survive below-freezing temperatures.4
Chemical Constituents
Bloodroot gets its colloquial name from its red sap, which is released when the plant’s rhizome is cut. This sap contains a high concentration of alkaloids that are used for protection against predators. The rhizome itself has a rusty, red-brown color; the roots are a brighter red-orange.4
The rhizome of S canadensis contains the highest concentration of active alkaloids; the roots also contain these chemicals, though to a lesser degree; and the leaves, flowers, and fruits harvest approximately 1% of the alkaloids found in the roots.4 The concentration of alkaloids can vary from one plant to the next, depending on environmental conditions.5,6
The major alkaloids in S canadensis include both quaternary benzophenanthridine alkaloids (eg, sanguinarine, chelerythrine, sanguilutine, chelilutine, sanguirubine, chelirubine) and protopin alkaloids (eg, protopine, allocryptopine).3,7 Of these, sanguinarine and chelerythrine typically are the most potent.1 Oral ingestion or topical application of these molecules can have therapeutic and toxic effects.8
Biophysiological Effects
Bloodroot has been shown to have remarkable antimicrobial effects.9 The plant produces hydrogen peroxide and superoxide anion.10 These mediators cause oxidative stress, thus inducing destruction of cellular DNA and the cell membrane.11 Although these effects can be helpful when fighting infection, they are not necessarily selective against healthy cells.12
Alkaloids of bloodroot also have cardiovascular therapeutic effects. Sanguinarine blocks angiotensin II and causes vasodilation, thus helping treat hypertension.13 It also acts as an inotrope by blocking the Na+/K+ ATPase pump. These effects in a patient who is already taking digoxin can cause notable cardiotoxicity because the 2 drugs share a mechanism of action.14
Chelerythrine blocks production of cyclooxygenase 2 and prostaglandin E2.15 This pathway modification results in anti-inflammatory effects that can help treat arthritis, edema, and other inflammatory conditions.16 Moreover, sanguinarine has demonstrated efficacy in numerous anticancer pathways,17 including downregulation of intercellular adhesion molecules, vascular cell adhesion molecules, and vascular endothelial growth factor (VEGF).18-20 Blocking VEGF is one way to inhibit angiogenesis,21 which is upregulated in tumor formation, thus sanguinarine can have an antiproliferative anticancer effect.22 Sanguinarine also upregulates molecules such as nuclear factor–κB and the protease enzymes known as caspases to cause proapoptotic effects, furthering its antitumor potential.23,24
Treatment of Dermatologic Conditions
The initial technique of Mohs micrographic surgery employed a chemopaste that utilized an extract of S canadensis to preserve tissue.25 Outside the dermatologist’s office, bloodroot is used as a topical home remedy for a variety of cutaneous conditions, including cancer, skin tags, and warts.26 Bloodroot is advertised as black salve, an alternative anticancer treatment.27,28
As useful as this natural agent sounds, it has a pitfall: The alkaloids of S canadensis are nonspecific in their cytotoxicity, damaging neoplastic and healthy tissue.29 This cytotoxic effect can cause escharification through diffuse tissue destruction and has been observed to result in formation of a keloid scar.30 The alkaloids in black salve also have been shown to cause skin erosions and cellular atypia.28,31 Therefore, the utility of this escharotic in medical treatment is limited.32 Fortuitously, oral antibiotics and wound care can help address this adverse effect.28
Bloodroot was once used as a mouth rinse and toothpaste to treat gingivitis, but this application was later associated with oral leukoplakia, a premalignant condition.33 Leukoplakia associated with S canadensis extract often is unremitting. Immediate discontinuation of the offending agent produces little regression, suggesting that cellular damage is irreversible.34
Final Thoughts
Although bloodroot demonstrates efficacy as a phytotherapeutic, it does come with notable toxicity. Physicians should warn patients of the unwanted cosmetic effects of black salve, especially oral products that incorporate sanguinarine. Adverse effects on the oropharynx can be irreversible, though the eschar associated with black salve can be treated with a topical or oral corticosteroid.29
Bloodroot (Sanguinaria canadensis) is a member of the family Papaveraceae.1 This North American plant commonly is found in widespread distribution from Nova Scotia, Canada, to Florida and from the Great Lakes to Mississippi.2 Historically, Native Americans used bloodroot as a skin dye and as a medicine for many ailments.3
Bloodroot blooms for only a few days, starting in March, and fruits in June. The flowers comprise 8 to 10 white petals, surrounding a bed of yellow stamens (Figure). The plant thrives in wooded areas and grows to 12 inches tall. In its off-season, the plant remains dormant and can survive below-freezing temperatures.4
Chemical Constituents
Bloodroot gets its colloquial name from its red sap, which is released when the plant’s rhizome is cut. This sap contains a high concentration of alkaloids that are used for protection against predators. The rhizome itself has a rusty, red-brown color; the roots are a brighter red-orange.4
The rhizome of S canadensis contains the highest concentration of active alkaloids; the roots also contain these chemicals, though to a lesser degree; and the leaves, flowers, and fruits harvest approximately 1% of the alkaloids found in the roots.4 The concentration of alkaloids can vary from one plant to the next, depending on environmental conditions.5,6
The major alkaloids in S canadensis include both quaternary benzophenanthridine alkaloids (eg, sanguinarine, chelerythrine, sanguilutine, chelilutine, sanguirubine, chelirubine) and protopin alkaloids (eg, protopine, allocryptopine).3,7 Of these, sanguinarine and chelerythrine typically are the most potent.1 Oral ingestion or topical application of these molecules can have therapeutic and toxic effects.8
Biophysiological Effects
Bloodroot has been shown to have remarkable antimicrobial effects.9 The plant produces hydrogen peroxide and superoxide anion.10 These mediators cause oxidative stress, thus inducing destruction of cellular DNA and the cell membrane.11 Although these effects can be helpful when fighting infection, they are not necessarily selective against healthy cells.12
Alkaloids of bloodroot also have cardiovascular therapeutic effects. Sanguinarine blocks angiotensin II and causes vasodilation, thus helping treat hypertension.13 It also acts as an inotrope by blocking the Na+/K+ ATPase pump. These effects in a patient who is already taking digoxin can cause notable cardiotoxicity because the 2 drugs share a mechanism of action.14
Chelerythrine blocks production of cyclooxygenase 2 and prostaglandin E2.15 This pathway modification results in anti-inflammatory effects that can help treat arthritis, edema, and other inflammatory conditions.16 Moreover, sanguinarine has demonstrated efficacy in numerous anticancer pathways,17 including downregulation of intercellular adhesion molecules, vascular cell adhesion molecules, and vascular endothelial growth factor (VEGF).18-20 Blocking VEGF is one way to inhibit angiogenesis,21 which is upregulated in tumor formation, thus sanguinarine can have an antiproliferative anticancer effect.22 Sanguinarine also upregulates molecules such as nuclear factor–κB and the protease enzymes known as caspases to cause proapoptotic effects, furthering its antitumor potential.23,24
Treatment of Dermatologic Conditions
The initial technique of Mohs micrographic surgery employed a chemopaste that utilized an extract of S canadensis to preserve tissue.25 Outside the dermatologist’s office, bloodroot is used as a topical home remedy for a variety of cutaneous conditions, including cancer, skin tags, and warts.26 Bloodroot is advertised as black salve, an alternative anticancer treatment.27,28
As useful as this natural agent sounds, it has a pitfall: The alkaloids of S canadensis are nonspecific in their cytotoxicity, damaging neoplastic and healthy tissue.29 This cytotoxic effect can cause escharification through diffuse tissue destruction and has been observed to result in formation of a keloid scar.30 The alkaloids in black salve also have been shown to cause skin erosions and cellular atypia.28,31 Therefore, the utility of this escharotic in medical treatment is limited.32 Fortuitously, oral antibiotics and wound care can help address this adverse effect.28
Bloodroot was once used as a mouth rinse and toothpaste to treat gingivitis, but this application was later associated with oral leukoplakia, a premalignant condition.33 Leukoplakia associated with S canadensis extract often is unremitting. Immediate discontinuation of the offending agent produces little regression, suggesting that cellular damage is irreversible.34
Final Thoughts
Although bloodroot demonstrates efficacy as a phytotherapeutic, it does come with notable toxicity. Physicians should warn patients of the unwanted cosmetic effects of black salve, especially oral products that incorporate sanguinarine. Adverse effects on the oropharynx can be irreversible, though the eschar associated with black salve can be treated with a topical or oral corticosteroid.29
- Vogel M, Lawson M, Sippl W, et al. Structure and mechanism of sanguinarine reductase, an enzyme of alkaloid detoxification. J Biol Chem. 2010;285:18397-18406. doi:10.1074/jbc.M109.088989
- Maranda EL, Wang MX, Cortizo J, et al. Flower power—the versatility of bloodroot. JAMA Dermatol. 2016;152:824. doi:10.1001/jamadermatol.2015.5522
- Setzer WN. The phytochemistry of Cherokee aromatic medicinal plants. Medicines (Basel). 2018;5:121. doi:10.3390/medicines5040121
- Croaker A, King GJ, Pyne JH, et al. Sanguinaria canadensis: traditional medicine, phytochemical composition, biological activities and current uses. Int J Mol Sci. 2016;17:1414. doi:10.3390/ijms17091414
- Graf TN, Levine KE, Andrews ME, et al. Variability in the yield of benzophenanthridine alkaloids in wildcrafted vs cultivated bloodroot (Sanguinaria canadensis L.) J Agric Food Chem. 2007; 55:1205-1211. doi:10.1021/jf062498f
- Bennett BC, Bell CR, Boulware RT. Geographic variation in alkaloid content of Sanguinaria canadensis (Papaveraceae). Rhodora. 1990;92:57-69.
- Leaver CA, Yuan H, Wallen GR. Apoptotic activities of Sanguinaria canadensis: primary human keratinocytes, C-33A, and human papillomavirus HeLa cervical cancer lines. Integr Med (Encinitas). 2018;17:32-37.
- Kutchan TM. Molecular genetics of plant alkaloid biosynthesis. In: Cordell GA, ed. The Alkaloids. Vol 50. Elsevier Science Publishing Co, Inc; 1997:257-316.
- Obiang-Obounou BW, Kang O-H, Choi J-G, et al. The mechanism of action of sanguinarine against methicillin-resistant Staphylococcus aureus. J Toxicol Sci. 2011;36:277-283. doi:10.2131/jts.36.277
- Z˙abka A, Winnicki K, Polit JT, et al. Sanguinarine-induced oxidative stress and apoptosis-like programmed cell death (AL-PCD) in root meristem cells of Allium cepa. Plant Physiol Biochem. 2017;112:193-206. doi:10.1016/j.plaphy.2017.01.004
- Kumar GS, Hazra S. Sanguinarine, a promising anticancer therapeutic: photochemical and nucleic acid binding properties. RSC Advances. 2014;4:56518-56531.
- Ping G, Wang Y, Shen L, et al. Highly efficient complexation of sanguinarine alkaloid by carboxylatopillar[6]arene: pKa shift, increased solubility and enhanced antibacterial activity. Chemical Commun (Camb). 2017;53:7381-7384. doi:10.1039/c7cc02799k
- Caballero-George C, Vanderheyden PM, Solis PN, et al. Biological screening of selected medicinal Panamanian plants by radioligand-binding techniques. Phytomedicine. 2001;8:59-70. doi:10.1078/0944-7113-00011
- Seifen E, Adams RJ, Riemer RK. Sanguinarine: a positive inotropic alkaloid which inhibits cardiac Na+, K+-ATPase. Eur J Pharmacol. 1979;60:373-377. doi:10.1016/0014-2999(79)90245-0
- Debprasad C, Hemanta M, Paromita B, et al. Inhibition of NO2, PGE2, TNF-α, and iNOS EXpression by Shorea robusta L.: an ethnomedicine used for anti-inflammatory and analgesic activity. Evid Based Complement Alternat Med. 2012; 2012:254849. doi:10.1155/2012/254849
- Melov S, Ravenscroft J, Malik S, et al. Extension of life-span with superoxide dismutase/catalase mimetics. Science. 2000;289:1567-1569. doi:10.1126/science.289.5484.1567
- Basu P, Kumar GS. Sanguinarine and its role in chronic diseases. In: Gupta SC, Prasad S, Aggarwal BB, eds. Advances in Experimental Medicine and Biology: Anti-inflammatory Nutraceuticals and Chronic Diseases. Vol 928. Springer International Publishing; 2016:155-172.
- Alasvand M, Assadollahi V, Ambra R, et al. Antiangiogenic effect of alkaloids. Oxid Med Cell Longev. 2019;2019:9475908. doi:10.1155/2019/9475908
- Basini G, Santini SE, Bussolati S, et al. The plant alkaloid sanguinarine is a potential inhibitor of follicular angiogenesis. J Reprod Dev. 2007;53:573-579. doi:10.1262/jrd.18126
- Xu J-Y, Meng Q-H, Chong Y, et al. Sanguinarine is a novel VEGF inhibitor involved in the suppression of angiogenesis and cell migration. Mol Clin Oncol. 2013;1:331-336. doi:10.3892/mco.2012.41
- Lu K, Bhat M, Basu S. Plants and their active compounds: natural molecules to target angiogenesis. Angiogenesis. 2016;19:287-295. doi:10.1007/s10456-016-9512-y
- Achkar IW, Mraiche F, Mohammad RM, et al. Anticancer potential of sanguinarine for various human malignancies. Future Med Chem. 2017;9:933-950. doi:10.4155/fmc-2017-0041
- Lee TK, Park C, Jeong S-J, et al. Sanguinarine induces apoptosis of human oral squamous cell carcinoma KB cells via inactivation of the PI3K/Akt signaling pathway. Drug Dev Res. 2016;77:227-240. doi:10.1002/ddr.21315
- Gaziano R, Moroni G, Buè C, et al. Antitumor effects of the benzophenanthridine alkaloid sanguinarine: evidence and perspectives. World J Gastrointest Oncol. 2016;8:30-39. doi:10.4251/wjgo.v8.i1.30
- Mohs FE. Chemosurgery for skin cancer: fixed tissue and fresh tissue techniques. Arch Dermatol. 1976;112:211-215.
- Affleck AG, Varma S. A case of do-it-yourself Mohs’ surgery using bloodroot obtained from the internet. Br J Dermatol. 2007;157:1078-1079. doi:10.1111/j.1365-2133.2007.08180.x
- Eastman KL, McFarland LV, Raugi GJ. Buyer beware: a black salve caution. J Am Acad Dermatol. 2011;65:E154-E155. doi:10.1016/j.jaad.2011.07.031
- Osswald SS, Elston DM, Farley MF, et al. Self-treatment of a basal cell carcinoma with “black and yellow salve.” J Am Acad Dermatol. 2005;53:508-510. doi:10.1016/j.jaad.2005.04.007
- Schlichte MJ, Downing CP, Ramirez-Fort M, et al. Bloodroot associated eschar. Dermatol Online J. 2015;20:13030/qt05r0r2wr.
- Wang MZ, Warshaw EM. Bloodroot. Dermatitis. 2012;23:281-283. doi:10.1097/DER.0b013e318273a4dd
- Tan JM, Peters P, Ong N, et al. Histopathological features after topical black salve application. Australas J Dermatol. 2015;56:75-76.
- Hou JL, Brewer JD. Black salve and bloodroot extract in dermatologic conditions. Cutis. 2015;95:309-311.
- Eversole LR, Eversole GM, Kopcik J. Sanguinaria-associated oral leukoplakia: comparison with other benign and dysplastic leukoplakic lesions. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2000;89:455-464. doi:10.1016/s1079-2104(00)70125-9
- Mascarenhas AK, Allen CM, Moeschberger ML. The association between Viadent® use and oral leukoplakia—results of a matched case-control study. J Public Health Dent. 2002;62:158-162. doi:10.1111/j.1752-7325.2002.tb03437.x
- Vogel M, Lawson M, Sippl W, et al. Structure and mechanism of sanguinarine reductase, an enzyme of alkaloid detoxification. J Biol Chem. 2010;285:18397-18406. doi:10.1074/jbc.M109.088989
- Maranda EL, Wang MX, Cortizo J, et al. Flower power—the versatility of bloodroot. JAMA Dermatol. 2016;152:824. doi:10.1001/jamadermatol.2015.5522
- Setzer WN. The phytochemistry of Cherokee aromatic medicinal plants. Medicines (Basel). 2018;5:121. doi:10.3390/medicines5040121
- Croaker A, King GJ, Pyne JH, et al. Sanguinaria canadensis: traditional medicine, phytochemical composition, biological activities and current uses. Int J Mol Sci. 2016;17:1414. doi:10.3390/ijms17091414
- Graf TN, Levine KE, Andrews ME, et al. Variability in the yield of benzophenanthridine alkaloids in wildcrafted vs cultivated bloodroot (Sanguinaria canadensis L.) J Agric Food Chem. 2007; 55:1205-1211. doi:10.1021/jf062498f
- Bennett BC, Bell CR, Boulware RT. Geographic variation in alkaloid content of Sanguinaria canadensis (Papaveraceae). Rhodora. 1990;92:57-69.
- Leaver CA, Yuan H, Wallen GR. Apoptotic activities of Sanguinaria canadensis: primary human keratinocytes, C-33A, and human papillomavirus HeLa cervical cancer lines. Integr Med (Encinitas). 2018;17:32-37.
- Kutchan TM. Molecular genetics of plant alkaloid biosynthesis. In: Cordell GA, ed. The Alkaloids. Vol 50. Elsevier Science Publishing Co, Inc; 1997:257-316.
- Obiang-Obounou BW, Kang O-H, Choi J-G, et al. The mechanism of action of sanguinarine against methicillin-resistant Staphylococcus aureus. J Toxicol Sci. 2011;36:277-283. doi:10.2131/jts.36.277
- Z˙abka A, Winnicki K, Polit JT, et al. Sanguinarine-induced oxidative stress and apoptosis-like programmed cell death (AL-PCD) in root meristem cells of Allium cepa. Plant Physiol Biochem. 2017;112:193-206. doi:10.1016/j.plaphy.2017.01.004
- Kumar GS, Hazra S. Sanguinarine, a promising anticancer therapeutic: photochemical and nucleic acid binding properties. RSC Advances. 2014;4:56518-56531.
- Ping G, Wang Y, Shen L, et al. Highly efficient complexation of sanguinarine alkaloid by carboxylatopillar[6]arene: pKa shift, increased solubility and enhanced antibacterial activity. Chemical Commun (Camb). 2017;53:7381-7384. doi:10.1039/c7cc02799k
- Caballero-George C, Vanderheyden PM, Solis PN, et al. Biological screening of selected medicinal Panamanian plants by radioligand-binding techniques. Phytomedicine. 2001;8:59-70. doi:10.1078/0944-7113-00011
- Seifen E, Adams RJ, Riemer RK. Sanguinarine: a positive inotropic alkaloid which inhibits cardiac Na+, K+-ATPase. Eur J Pharmacol. 1979;60:373-377. doi:10.1016/0014-2999(79)90245-0
- Debprasad C, Hemanta M, Paromita B, et al. Inhibition of NO2, PGE2, TNF-α, and iNOS EXpression by Shorea robusta L.: an ethnomedicine used for anti-inflammatory and analgesic activity. Evid Based Complement Alternat Med. 2012; 2012:254849. doi:10.1155/2012/254849
- Melov S, Ravenscroft J, Malik S, et al. Extension of life-span with superoxide dismutase/catalase mimetics. Science. 2000;289:1567-1569. doi:10.1126/science.289.5484.1567
- Basu P, Kumar GS. Sanguinarine and its role in chronic diseases. In: Gupta SC, Prasad S, Aggarwal BB, eds. Advances in Experimental Medicine and Biology: Anti-inflammatory Nutraceuticals and Chronic Diseases. Vol 928. Springer International Publishing; 2016:155-172.
- Alasvand M, Assadollahi V, Ambra R, et al. Antiangiogenic effect of alkaloids. Oxid Med Cell Longev. 2019;2019:9475908. doi:10.1155/2019/9475908
- Basini G, Santini SE, Bussolati S, et al. The plant alkaloid sanguinarine is a potential inhibitor of follicular angiogenesis. J Reprod Dev. 2007;53:573-579. doi:10.1262/jrd.18126
- Xu J-Y, Meng Q-H, Chong Y, et al. Sanguinarine is a novel VEGF inhibitor involved in the suppression of angiogenesis and cell migration. Mol Clin Oncol. 2013;1:331-336. doi:10.3892/mco.2012.41
- Lu K, Bhat M, Basu S. Plants and their active compounds: natural molecules to target angiogenesis. Angiogenesis. 2016;19:287-295. doi:10.1007/s10456-016-9512-y
- Achkar IW, Mraiche F, Mohammad RM, et al. Anticancer potential of sanguinarine for various human malignancies. Future Med Chem. 2017;9:933-950. doi:10.4155/fmc-2017-0041
- Lee TK, Park C, Jeong S-J, et al. Sanguinarine induces apoptosis of human oral squamous cell carcinoma KB cells via inactivation of the PI3K/Akt signaling pathway. Drug Dev Res. 2016;77:227-240. doi:10.1002/ddr.21315
- Gaziano R, Moroni G, Buè C, et al. Antitumor effects of the benzophenanthridine alkaloid sanguinarine: evidence and perspectives. World J Gastrointest Oncol. 2016;8:30-39. doi:10.4251/wjgo.v8.i1.30
- Mohs FE. Chemosurgery for skin cancer: fixed tissue and fresh tissue techniques. Arch Dermatol. 1976;112:211-215.
- Affleck AG, Varma S. A case of do-it-yourself Mohs’ surgery using bloodroot obtained from the internet. Br J Dermatol. 2007;157:1078-1079. doi:10.1111/j.1365-2133.2007.08180.x
- Eastman KL, McFarland LV, Raugi GJ. Buyer beware: a black salve caution. J Am Acad Dermatol. 2011;65:E154-E155. doi:10.1016/j.jaad.2011.07.031
- Osswald SS, Elston DM, Farley MF, et al. Self-treatment of a basal cell carcinoma with “black and yellow salve.” J Am Acad Dermatol. 2005;53:508-510. doi:10.1016/j.jaad.2005.04.007
- Schlichte MJ, Downing CP, Ramirez-Fort M, et al. Bloodroot associated eschar. Dermatol Online J. 2015;20:13030/qt05r0r2wr.
- Wang MZ, Warshaw EM. Bloodroot. Dermatitis. 2012;23:281-283. doi:10.1097/DER.0b013e318273a4dd
- Tan JM, Peters P, Ong N, et al. Histopathological features after topical black salve application. Australas J Dermatol. 2015;56:75-76.
- Hou JL, Brewer JD. Black salve and bloodroot extract in dermatologic conditions. Cutis. 2015;95:309-311.
- Eversole LR, Eversole GM, Kopcik J. Sanguinaria-associated oral leukoplakia: comparison with other benign and dysplastic leukoplakic lesions. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2000;89:455-464. doi:10.1016/s1079-2104(00)70125-9
- Mascarenhas AK, Allen CM, Moeschberger ML. The association between Viadent® use and oral leukoplakia—results of a matched case-control study. J Public Health Dent. 2002;62:158-162. doi:10.1111/j.1752-7325.2002.tb03437.x
Practice Points
- Bloodroot (Sanguinaria canadensis) is a plant historically used in Mohs micrographic surgery as chemopaste.
- Bloodroot has been shown to have remarkable antimicrobial effects.
- The alkaloids of S canadensis are nonspecific in their cytotoxicity, damaging both neoplastic and healthy tissue. They have been shown to cause skin erosions and cellular atypia.
Aquatic Antagonists: Sea Cucumbers (Holothuroidea)
Sea cucumbers—commonly known as trepang in Indonesia, namako in Japan, and hai shen in China, where they are treasured as a food delicacy—are sea creatures belonging to the phylum Echinodermata, class Holothuridea, and family Cucumariidae . 1,2 They are an integral part of a variety of marine habitats, serving as cleaners as they filter through sediment for nutrients. They can be found on the ocean floor under hundreds of feet of water or in shallow sandy waters along the coast, but they most commonly are found living among coral reefs. Sea cucumbers look just as they sound—shaped like cucumbers or sausages, ranging from under 1 inch to upwards of 6 feet in length depending on the specific species (Figure 1). They have a group of tentacles around the mouth used for filtering sediment, and they move about the ocean floor on tubular feet protruding through the body wall, similar to a sea star.
Beneficial Properties and Cultural Relevance
Although more than 1200 species of sea cucumbers have been identified thus far, only about 20 of these are edible.2 The most common of the edible species is Stichopus japonicus, which can be found off the coasts of Korea, China, Japan, and Russia. This particular species most commonly is used in traditional dishes and is divided into 3 groups based on the color: red, green, or black. The price and taste of sea cucumbers varies based on the color, with red being the most expensive.2 The body wall of the sea cucumber is cleaned, repeatedly boiled, and dried until edible. It is considered a delicacy, not only in food but also in pharmaceutical forms, as it is comprised of a variety of vitamins, minerals, and other nutrients that are thought to provide anticancer, anticoagulant, antioxidant, antifungal, and anti-inflammatory properties. Components of the body wall include collagen, mucopolysaccharides, peptides, gelatin, glycosaminoglycans, glycosides (including various holotoxins), hydroxylates, saponins, and fatty acids.2 The regenerative properties of the sea cucumber also are important in future biomedical developments.
Toxic Properties
Although sea cucumbers have proven to have many beneficial properties, at least 30 species also produce potent toxins that pose a danger to both humans and other wildlife.3 The toxins are collectively referred to as holothurin; however, specific species actually produce a variety of holothurin toxins with unique chemical structures. Each toxin is a variation of a specific triterpene glycoside called saponins, which are common glycosides in the plant world. Holothurin was the first saponin to be found in animals. The only animals known to contain holothurin are the echinoderms, including sea cucumbers and sea stars.1 Holothurins A and B are the 2 groups of holothurin toxins produced specifically by sea cucumbers. The toxins are composed of roughly 60% glycosides and pigment; 30% free amino acids (alanine, arginine, cysteine, glycine, glutamic acid, histidine, serine, and valine); 5% to 10% insoluble proteins; and 1% cholesterol, salts, and polypeptides.3
Holothurins are concentrated in granules within specialized structures of the sea cucumber called Cuvierian tubules, which freely float in the posterior coelomic cavity of the sea cucumber and are attached at the base of the respiratory tree. It is with these tubules that sea cucumbers utilize a unique defensive mechanism. Upon disturbance, the sea cucumber will turn its posterior end to the threat and squeeze its body in a series of violent contractions, inducing a tear in the cloacal wall.4 The tubules pass through this tear, are autotomized from the attachment point at the respiratory tree, and are finally expelled through the anus onto the predator and into the surrounding waters. The tubules are both sticky on contact and poisonous due to the holothurin, allowing the sea cucumber to crawl away from the threat unscathed. Over time, the tubules will regenerate, allowing the sea cucumber to protect itself again in the face of future danger.
Aside from direct disturbance by a threat, sea cucumbers also are known to undergo evisceration due to high temperatures and oxygen deficiency.3 Species that lack Cuvierian tubules can still produce holothurin toxins, though the toxins are secreted onto the outer surface of the body wall and mainly pose a risk with direct contact undiluted by seawater.5 The toxin induces a neural blockade in other sea creatures through its interaction with ion channels. On Asian islands, sea cucumbers have been exploited for this ability and commonly are thrown into tidal pools by fishermen to paralyze fish for easier capture.1
Effects on Human Skin
In humans, the holothurin toxins of sea cucumbers cause an acute irritant dermatitis upon contact with the skin.6 Fishermen or divers handling sea cucumbers without gloves may present with an irritant contact dermatitis characterized by marked erythema and swelling (Figure 2).6-8 Additionally, holothurin toxins can cause irritation of the mucous membranes of the eyes and mouth. Contact with the mucous membranes of the eyes can induce a painful conjunctivitis that may result in blindness.6,8 Ingestion of large quantities of sea cucumber can produce an anticoagulant effect, and toxins in some species act similar to cardiac glycosides.3,9
In addition to their own toxins, sea cucumbers also can secrete undigested nematocysts of previously consumed cnidarians through the integument.7,10 In this case, the result of direct contact with the body wall is similar to a jellyfish sting in addition to the irritant contact dermatitis caused by the holothurin toxin.
Treatment and Prevention
Irritant dermatitis resulting from contact with a holothurin toxin is first treated with cleansing of the affected area at the time of exposure with generous amounts of seawater or preferably hot seawater and soap. Most marine toxins are inactivated by heat, but holothurin is partially heat stable. Vinegar or isopropyl alcohol also have been used.9 The result is removal of the slime containing the holothurin toxin rather than deactivation of the toxin. Although this alone may relieve symptoms, dermatitis also may be addressed with topical anesthetics, corticosteroids, or, if a severe reaction has occurred, systemic steroids.9
Conjunctivitis should be addressed with copious irrigation with tap water and topical anesthesia. Following proper irrigation, providers may choose to follow up with fluorescein staining to rule out corneal injury.10
The dermatologic effects of holothurin toxins can be prevented with the use of gloves and diving masks or goggles. Proper protective wear should be utilized not only when directly handling sea cucumbers but also when swimming in water where sea cucumbers may be present. Systemic toxicity can be prevented by proper cooking, as holothurin toxins are only partially heat resistant and also are hydrolyzed into nontoxic products by gastric acid. Additionally, the species of the sea cucumber should be confirmed prior to consumption, as edible species are known to contain less toxin.1
Conclusion
Although sea cucumbers have ecologic, culinary, and pharmaceutical value, they also can pose a threat to both humans and wildlife. The holothurin toxins produced by sea cucumbers cause a painful contact dermatitis and can lead to conjunctivitis and even blindness following eye exposure. Although the toxin is broken down into nontoxic metabolites by gastric acid, large amounts of potent variants can induce systemic effects. Individuals who come in contact with sea cucumbers, such as fishermen and divers, should utilize proper protection including gloves and protective eyewear.
- Burnett K, Fenner P, Williamson J. Venomous and Poisonous Marine Animals: A Medical and Biological Handbook. University of New South Wales Press; 1996.
- Oh GW, Ko SC, Lee DH, et al. Biological activities and biomedical potential of sea cucumber (Stichopus japonicus): a review. Fisheries Aquatic Sci. 2017;20:28.
- Nigrelli RF, Jakowska S. Effects of holothurian, a steroid saponin from the Bahamian sea cucumber (Actinopyga agassizi), on various biological systems. Ann NY Acad Sci. 1960;90:884-892.
- Demeuldre M, Hennebert E, Bonneel M, et al. Mechanical adaptability of sea cucumber Cuvierian tubules involves a mutable collagenous tissue. J Exp Biol. 2017;220:2108-2119.
- Matranga V, ed. Echinodermata: Progress in Molecular and Subcellular Biology. Springer; 2005.
- Tlougan, BE, Podjasek, JO, Adams BB. Aquatic sports dermatoses. part 2—in the water: saltwater dermatoses. Int J Dermatol. 2010;49:994-1002.
- Bonamonte D, Verni P, Filoni A, et al. Dermatitis caused by echinoderms. In: Bonamonte D, Angelini G, eds. Springer; 2016:59-72.
- Haddad V Jr. Medical Emergencies Caused by Aquatic Animals: A Zoological and Clinical Guide. Springer International Publishing; 2016.
- French LK, Horowitz BZ. Marine vertebrates, cnidarians, and mollusks. In: Brent J, Burkhart K, Dargan P, et al, eds. Critical Care Toxicology. Springer; 2017:1-30.
- Smith ML. Skin problems from marine echinoderms. Dermatol Ther. 2002;15:30-33.
Sea cucumbers—commonly known as trepang in Indonesia, namako in Japan, and hai shen in China, where they are treasured as a food delicacy—are sea creatures belonging to the phylum Echinodermata, class Holothuridea, and family Cucumariidae . 1,2 They are an integral part of a variety of marine habitats, serving as cleaners as they filter through sediment for nutrients. They can be found on the ocean floor under hundreds of feet of water or in shallow sandy waters along the coast, but they most commonly are found living among coral reefs. Sea cucumbers look just as they sound—shaped like cucumbers or sausages, ranging from under 1 inch to upwards of 6 feet in length depending on the specific species (Figure 1). They have a group of tentacles around the mouth used for filtering sediment, and they move about the ocean floor on tubular feet protruding through the body wall, similar to a sea star.
Beneficial Properties and Cultural Relevance
Although more than 1200 species of sea cucumbers have been identified thus far, only about 20 of these are edible.2 The most common of the edible species is Stichopus japonicus, which can be found off the coasts of Korea, China, Japan, and Russia. This particular species most commonly is used in traditional dishes and is divided into 3 groups based on the color: red, green, or black. The price and taste of sea cucumbers varies based on the color, with red being the most expensive.2 The body wall of the sea cucumber is cleaned, repeatedly boiled, and dried until edible. It is considered a delicacy, not only in food but also in pharmaceutical forms, as it is comprised of a variety of vitamins, minerals, and other nutrients that are thought to provide anticancer, anticoagulant, antioxidant, antifungal, and anti-inflammatory properties. Components of the body wall include collagen, mucopolysaccharides, peptides, gelatin, glycosaminoglycans, glycosides (including various holotoxins), hydroxylates, saponins, and fatty acids.2 The regenerative properties of the sea cucumber also are important in future biomedical developments.
Toxic Properties
Although sea cucumbers have proven to have many beneficial properties, at least 30 species also produce potent toxins that pose a danger to both humans and other wildlife.3 The toxins are collectively referred to as holothurin; however, specific species actually produce a variety of holothurin toxins with unique chemical structures. Each toxin is a variation of a specific triterpene glycoside called saponins, which are common glycosides in the plant world. Holothurin was the first saponin to be found in animals. The only animals known to contain holothurin are the echinoderms, including sea cucumbers and sea stars.1 Holothurins A and B are the 2 groups of holothurin toxins produced specifically by sea cucumbers. The toxins are composed of roughly 60% glycosides and pigment; 30% free amino acids (alanine, arginine, cysteine, glycine, glutamic acid, histidine, serine, and valine); 5% to 10% insoluble proteins; and 1% cholesterol, salts, and polypeptides.3
Holothurins are concentrated in granules within specialized structures of the sea cucumber called Cuvierian tubules, which freely float in the posterior coelomic cavity of the sea cucumber and are attached at the base of the respiratory tree. It is with these tubules that sea cucumbers utilize a unique defensive mechanism. Upon disturbance, the sea cucumber will turn its posterior end to the threat and squeeze its body in a series of violent contractions, inducing a tear in the cloacal wall.4 The tubules pass through this tear, are autotomized from the attachment point at the respiratory tree, and are finally expelled through the anus onto the predator and into the surrounding waters. The tubules are both sticky on contact and poisonous due to the holothurin, allowing the sea cucumber to crawl away from the threat unscathed. Over time, the tubules will regenerate, allowing the sea cucumber to protect itself again in the face of future danger.
Aside from direct disturbance by a threat, sea cucumbers also are known to undergo evisceration due to high temperatures and oxygen deficiency.3 Species that lack Cuvierian tubules can still produce holothurin toxins, though the toxins are secreted onto the outer surface of the body wall and mainly pose a risk with direct contact undiluted by seawater.5 The toxin induces a neural blockade in other sea creatures through its interaction with ion channels. On Asian islands, sea cucumbers have been exploited for this ability and commonly are thrown into tidal pools by fishermen to paralyze fish for easier capture.1
Effects on Human Skin
In humans, the holothurin toxins of sea cucumbers cause an acute irritant dermatitis upon contact with the skin.6 Fishermen or divers handling sea cucumbers without gloves may present with an irritant contact dermatitis characterized by marked erythema and swelling (Figure 2).6-8 Additionally, holothurin toxins can cause irritation of the mucous membranes of the eyes and mouth. Contact with the mucous membranes of the eyes can induce a painful conjunctivitis that may result in blindness.6,8 Ingestion of large quantities of sea cucumber can produce an anticoagulant effect, and toxins in some species act similar to cardiac glycosides.3,9
In addition to their own toxins, sea cucumbers also can secrete undigested nematocysts of previously consumed cnidarians through the integument.7,10 In this case, the result of direct contact with the body wall is similar to a jellyfish sting in addition to the irritant contact dermatitis caused by the holothurin toxin.
Treatment and Prevention
Irritant dermatitis resulting from contact with a holothurin toxin is first treated with cleansing of the affected area at the time of exposure with generous amounts of seawater or preferably hot seawater and soap. Most marine toxins are inactivated by heat, but holothurin is partially heat stable. Vinegar or isopropyl alcohol also have been used.9 The result is removal of the slime containing the holothurin toxin rather than deactivation of the toxin. Although this alone may relieve symptoms, dermatitis also may be addressed with topical anesthetics, corticosteroids, or, if a severe reaction has occurred, systemic steroids.9
Conjunctivitis should be addressed with copious irrigation with tap water and topical anesthesia. Following proper irrigation, providers may choose to follow up with fluorescein staining to rule out corneal injury.10
The dermatologic effects of holothurin toxins can be prevented with the use of gloves and diving masks or goggles. Proper protective wear should be utilized not only when directly handling sea cucumbers but also when swimming in water where sea cucumbers may be present. Systemic toxicity can be prevented by proper cooking, as holothurin toxins are only partially heat resistant and also are hydrolyzed into nontoxic products by gastric acid. Additionally, the species of the sea cucumber should be confirmed prior to consumption, as edible species are known to contain less toxin.1
Conclusion
Although sea cucumbers have ecologic, culinary, and pharmaceutical value, they also can pose a threat to both humans and wildlife. The holothurin toxins produced by sea cucumbers cause a painful contact dermatitis and can lead to conjunctivitis and even blindness following eye exposure. Although the toxin is broken down into nontoxic metabolites by gastric acid, large amounts of potent variants can induce systemic effects. Individuals who come in contact with sea cucumbers, such as fishermen and divers, should utilize proper protection including gloves and protective eyewear.
Sea cucumbers—commonly known as trepang in Indonesia, namako in Japan, and hai shen in China, where they are treasured as a food delicacy—are sea creatures belonging to the phylum Echinodermata, class Holothuridea, and family Cucumariidae . 1,2 They are an integral part of a variety of marine habitats, serving as cleaners as they filter through sediment for nutrients. They can be found on the ocean floor under hundreds of feet of water or in shallow sandy waters along the coast, but they most commonly are found living among coral reefs. Sea cucumbers look just as they sound—shaped like cucumbers or sausages, ranging from under 1 inch to upwards of 6 feet in length depending on the specific species (Figure 1). They have a group of tentacles around the mouth used for filtering sediment, and they move about the ocean floor on tubular feet protruding through the body wall, similar to a sea star.
Beneficial Properties and Cultural Relevance
Although more than 1200 species of sea cucumbers have been identified thus far, only about 20 of these are edible.2 The most common of the edible species is Stichopus japonicus, which can be found off the coasts of Korea, China, Japan, and Russia. This particular species most commonly is used in traditional dishes and is divided into 3 groups based on the color: red, green, or black. The price and taste of sea cucumbers varies based on the color, with red being the most expensive.2 The body wall of the sea cucumber is cleaned, repeatedly boiled, and dried until edible. It is considered a delicacy, not only in food but also in pharmaceutical forms, as it is comprised of a variety of vitamins, minerals, and other nutrients that are thought to provide anticancer, anticoagulant, antioxidant, antifungal, and anti-inflammatory properties. Components of the body wall include collagen, mucopolysaccharides, peptides, gelatin, glycosaminoglycans, glycosides (including various holotoxins), hydroxylates, saponins, and fatty acids.2 The regenerative properties of the sea cucumber also are important in future biomedical developments.
Toxic Properties
Although sea cucumbers have proven to have many beneficial properties, at least 30 species also produce potent toxins that pose a danger to both humans and other wildlife.3 The toxins are collectively referred to as holothurin; however, specific species actually produce a variety of holothurin toxins with unique chemical structures. Each toxin is a variation of a specific triterpene glycoside called saponins, which are common glycosides in the plant world. Holothurin was the first saponin to be found in animals. The only animals known to contain holothurin are the echinoderms, including sea cucumbers and sea stars.1 Holothurins A and B are the 2 groups of holothurin toxins produced specifically by sea cucumbers. The toxins are composed of roughly 60% glycosides and pigment; 30% free amino acids (alanine, arginine, cysteine, glycine, glutamic acid, histidine, serine, and valine); 5% to 10% insoluble proteins; and 1% cholesterol, salts, and polypeptides.3
Holothurins are concentrated in granules within specialized structures of the sea cucumber called Cuvierian tubules, which freely float in the posterior coelomic cavity of the sea cucumber and are attached at the base of the respiratory tree. It is with these tubules that sea cucumbers utilize a unique defensive mechanism. Upon disturbance, the sea cucumber will turn its posterior end to the threat and squeeze its body in a series of violent contractions, inducing a tear in the cloacal wall.4 The tubules pass through this tear, are autotomized from the attachment point at the respiratory tree, and are finally expelled through the anus onto the predator and into the surrounding waters. The tubules are both sticky on contact and poisonous due to the holothurin, allowing the sea cucumber to crawl away from the threat unscathed. Over time, the tubules will regenerate, allowing the sea cucumber to protect itself again in the face of future danger.
Aside from direct disturbance by a threat, sea cucumbers also are known to undergo evisceration due to high temperatures and oxygen deficiency.3 Species that lack Cuvierian tubules can still produce holothurin toxins, though the toxins are secreted onto the outer surface of the body wall and mainly pose a risk with direct contact undiluted by seawater.5 The toxin induces a neural blockade in other sea creatures through its interaction with ion channels. On Asian islands, sea cucumbers have been exploited for this ability and commonly are thrown into tidal pools by fishermen to paralyze fish for easier capture.1
Effects on Human Skin
In humans, the holothurin toxins of sea cucumbers cause an acute irritant dermatitis upon contact with the skin.6 Fishermen or divers handling sea cucumbers without gloves may present with an irritant contact dermatitis characterized by marked erythema and swelling (Figure 2).6-8 Additionally, holothurin toxins can cause irritation of the mucous membranes of the eyes and mouth. Contact with the mucous membranes of the eyes can induce a painful conjunctivitis that may result in blindness.6,8 Ingestion of large quantities of sea cucumber can produce an anticoagulant effect, and toxins in some species act similar to cardiac glycosides.3,9
In addition to their own toxins, sea cucumbers also can secrete undigested nematocysts of previously consumed cnidarians through the integument.7,10 In this case, the result of direct contact with the body wall is similar to a jellyfish sting in addition to the irritant contact dermatitis caused by the holothurin toxin.
Treatment and Prevention
Irritant dermatitis resulting from contact with a holothurin toxin is first treated with cleansing of the affected area at the time of exposure with generous amounts of seawater or preferably hot seawater and soap. Most marine toxins are inactivated by heat, but holothurin is partially heat stable. Vinegar or isopropyl alcohol also have been used.9 The result is removal of the slime containing the holothurin toxin rather than deactivation of the toxin. Although this alone may relieve symptoms, dermatitis also may be addressed with topical anesthetics, corticosteroids, or, if a severe reaction has occurred, systemic steroids.9
Conjunctivitis should be addressed with copious irrigation with tap water and topical anesthesia. Following proper irrigation, providers may choose to follow up with fluorescein staining to rule out corneal injury.10
The dermatologic effects of holothurin toxins can be prevented with the use of gloves and diving masks or goggles. Proper protective wear should be utilized not only when directly handling sea cucumbers but also when swimming in water where sea cucumbers may be present. Systemic toxicity can be prevented by proper cooking, as holothurin toxins are only partially heat resistant and also are hydrolyzed into nontoxic products by gastric acid. Additionally, the species of the sea cucumber should be confirmed prior to consumption, as edible species are known to contain less toxin.1
Conclusion
Although sea cucumbers have ecologic, culinary, and pharmaceutical value, they also can pose a threat to both humans and wildlife. The holothurin toxins produced by sea cucumbers cause a painful contact dermatitis and can lead to conjunctivitis and even blindness following eye exposure. Although the toxin is broken down into nontoxic metabolites by gastric acid, large amounts of potent variants can induce systemic effects. Individuals who come in contact with sea cucumbers, such as fishermen and divers, should utilize proper protection including gloves and protective eyewear.
- Burnett K, Fenner P, Williamson J. Venomous and Poisonous Marine Animals: A Medical and Biological Handbook. University of New South Wales Press; 1996.
- Oh GW, Ko SC, Lee DH, et al. Biological activities and biomedical potential of sea cucumber (Stichopus japonicus): a review. Fisheries Aquatic Sci. 2017;20:28.
- Nigrelli RF, Jakowska S. Effects of holothurian, a steroid saponin from the Bahamian sea cucumber (Actinopyga agassizi), on various biological systems. Ann NY Acad Sci. 1960;90:884-892.
- Demeuldre M, Hennebert E, Bonneel M, et al. Mechanical adaptability of sea cucumber Cuvierian tubules involves a mutable collagenous tissue. J Exp Biol. 2017;220:2108-2119.
- Matranga V, ed. Echinodermata: Progress in Molecular and Subcellular Biology. Springer; 2005.
- Tlougan, BE, Podjasek, JO, Adams BB. Aquatic sports dermatoses. part 2—in the water: saltwater dermatoses. Int J Dermatol. 2010;49:994-1002.
- Bonamonte D, Verni P, Filoni A, et al. Dermatitis caused by echinoderms. In: Bonamonte D, Angelini G, eds. Springer; 2016:59-72.
- Haddad V Jr. Medical Emergencies Caused by Aquatic Animals: A Zoological and Clinical Guide. Springer International Publishing; 2016.
- French LK, Horowitz BZ. Marine vertebrates, cnidarians, and mollusks. In: Brent J, Burkhart K, Dargan P, et al, eds. Critical Care Toxicology. Springer; 2017:1-30.
- Smith ML. Skin problems from marine echinoderms. Dermatol Ther. 2002;15:30-33.
- Burnett K, Fenner P, Williamson J. Venomous and Poisonous Marine Animals: A Medical and Biological Handbook. University of New South Wales Press; 1996.
- Oh GW, Ko SC, Lee DH, et al. Biological activities and biomedical potential of sea cucumber (Stichopus japonicus): a review. Fisheries Aquatic Sci. 2017;20:28.
- Nigrelli RF, Jakowska S. Effects of holothurian, a steroid saponin from the Bahamian sea cucumber (Actinopyga agassizi), on various biological systems. Ann NY Acad Sci. 1960;90:884-892.
- Demeuldre M, Hennebert E, Bonneel M, et al. Mechanical adaptability of sea cucumber Cuvierian tubules involves a mutable collagenous tissue. J Exp Biol. 2017;220:2108-2119.
- Matranga V, ed. Echinodermata: Progress in Molecular and Subcellular Biology. Springer; 2005.
- Tlougan, BE, Podjasek, JO, Adams BB. Aquatic sports dermatoses. part 2—in the water: saltwater dermatoses. Int J Dermatol. 2010;49:994-1002.
- Bonamonte D, Verni P, Filoni A, et al. Dermatitis caused by echinoderms. In: Bonamonte D, Angelini G, eds. Springer; 2016:59-72.
- Haddad V Jr. Medical Emergencies Caused by Aquatic Animals: A Zoological and Clinical Guide. Springer International Publishing; 2016.
- French LK, Horowitz BZ. Marine vertebrates, cnidarians, and mollusks. In: Brent J, Burkhart K, Dargan P, et al, eds. Critical Care Toxicology. Springer; 2017:1-30.
- Smith ML. Skin problems from marine echinoderms. Dermatol Ther. 2002;15:30-33.
Practice Points
- Sea cucumbers produce a toxin known as holothurin, which is contained in specialized structures called Cuvierian tubules and secreted onto the outer surface of the body wall. Some species also eject portions of their toxic inner organs through the anus as a defensive mechanism.
- In humans, the holothurin toxins cause an acute irritant dermatitis upon contact with the skin and a painful chemical conjunctivitis upon contact with the eyes.
- In addition to their own toxin, sea cucumbers also can secrete undigested nematocysts of previously consumed cnidarians through their integument, causing additional effects on human skin.
- The dermatologic effects of sea cucumbers can be prevented with the use of gloves and swim masks or goggles.
What’s Eating You? Culex Mosquitoes and West Nile Virus
What is West Nile virus? How is it contracted, and who can become infected?
West Nile virus (WNV) is a single-stranded RNA virus of the Flaviviridae family and Flavivirus genus, a lineage that also includes the yellow fever, dengue, Zika, Japanese encephalitis, and Saint Louis encephalitis viruses.1 Birds serve as the reservoir hosts of WNV, and mosquitoes acquire the virus during feeding.2 West Nile virus then is transmitted to humans primarily by bites from Culex mosquitoes, which are especially prevalent in wooded areas during peak mosquito season (summer through early fall in North America).1 Mosquitoes also can infect horses; however, humans and horses are dead-end hosts, meaning they do not pass the virus on to other biting mosquitoes.3 There also have been rare reports of transmission of WNV through blood and donation as well as mother-to-baby transmission.2
What is the epidemiology of WNV in the United States?
Since the introduction of WNV to the United States in 1999, it has become an important public health concern, with 48,183 cases and 2163 deaths reported since 1999.2,3 In 2018, Nebraska had the highest number of cases of WNV (n=251), followed by California (n=217), North Dakota (n=204), Illinois (n=176), and South Dakota (n=169).3 West Nile virus is endemic to all 48 contiguous states and Canada, though the Great Plains region is especially affected by WNV due to several factors, such as a greater percentage of rural land, forests, and irrigated areas.4 The Great Plains region also has been thought to be an ecological niche for a more virulent species (Culex tarsalis) compared to other regions in the United States.5
The annual incidence of WNV in the United States peaked in 2003 at 9862 cases (up from 62 cases in 1999), then declined gradually until 2008 to 2011, during which the incidence was stable at 700 to 1100 new cases per year. However, there was a resurgence of cases (n=5674) in 2012 that steadied at around 2200 cases annually in subsequent years.6 Although there likely are several factors affecting WNV incidence trends in the United States, interannual changes in temperature and precipitation have been described. An increased mean annual temperature (from September through October, the end of peak mosquito season) and an increased temperature in winter months (from January through March, prior to peak mosquito season) have both been associated with an increased incidence of WNV.7 An increased temperature is thought to increase population numbers of mosquitoes both by increasing reproductive rates and creating ideal breeding environments via pooled water areas.8 Depending on the region, both above average and below average precipitation levels in the United States can increase WNV incidence the following year.7,9
What are the signs and symptoms of WNV infection?
Up to 80% of those infected with WNV are asymptomatic.3 After an incubation period of roughly 2 to 14 days, the remaining 20% may develop symptoms of West Nile fever (WNF), typically a self-limited illness that consists of 3 to 10 days of nonspecific symptoms such as fever, headache, fatigue, muscle pain and/or weakness, eye pain, gastrointestinal tract upset, and a macular rash that usually presents on the trunk or extremities.1,3 Less than 1% of patients affected by WNV develop neuroinvasive disease, including meningitis, encephalitis, and/or acute flaccid paralysis.10 West Nile virus neuroinvasive disease can cause permanent neurologic sequelae such as muscle weakness, confusion, memory loss, and fatigue; it carries a mortality rate of 10% to 30%, which is mainly dependent on older age and immunosuppression status.1,10
What is the reported spectrum of cutaneous findings in WNV?
Of the roughly 20% of patients infected with WNV that develop WNF, approximately 25% to 50% will develop an associated rash.1 It most commonly is described as a morbilliform or maculopapular rash located on the chest, back, and arms, usually sparing the palms and soles, though 1 case report noted involvement with these areas (Figure).11,12 It typically appears 5 days after symptom onset, can be associated with defervescence, and lasts less than a week.1,13 Pruritus and dysesthesia are sometimes present.13 Other rare presentations that have been reported include an ill-defined pseudovesicular rash with erythematous papules on the palms and pink, scaly, psoriasiform papules on the feet and thighs, as well as neuroinvasive WNV leading to purpura fulminans.14,15 A diffuse, erythematous, petechial rash on the face, neck, trunk, and extremities was reported in a pediatric patient, but there have been no reports of a petechial rash associated with WNV in adult patients.16 These findings suggest some potential variability in the presentation of the WNV rash.
What role does the presence of rash play diagnostically and prognostically?
The rash of WNV has been implicated as a potential prognostic factor in predicting more favorable outcomes.17 Using 2002 data from the Illinois Department of Public Health and 2003 data from the Colorado Department of Public Health, Huhn and Dworkin17 found the age-adjusted risk of encephalitis and death to be decreased in WNV patients with a rash (relative risk, 0.44; 95% CI, 0.21-0.92). The reasons for this are not definitively known, but we hypothesize that the rash may prompt patients to seek earlier medical attention or indicate a more robust immune response. Additionally, a rash in WNV more commonly is seen in younger patients, whereas WNV neuroinvasive disease is more common in older patients, who also tend to have worse outcomes.10 One study found rash to be the only symptom that demonstrated a significant association with seropositivity (overall risk=6.35; P<.05; 95% CI, 3.75-10.80) by multivariate analysis.18
How is WNV diagnosed? What are the downsides to WNV testing?
Given that the presenting symptoms of WNV and WNF are nonspecific, it becomes challenging to arrive at the diagnosis based solely on physical examination. As such, the patient’s clinical and epidemiologic history, such as timing, pattern, and appearance of the rash or recent history of mosquito bites, is key to arriving at the correct diagnosis. With clinical suspicion, possible diagnostic tests include an IgM enzyme-linked immunosorbent assay (ELISA) for WNV, a plaque reduction neutralization test (PNRT), and blood polymerase chain reaction (PCR).
An ELISA is a confirmatory test to detect IgM antibodies to WNV in the serum. Because IgM seroconversion typically occurs between days 4 and 10 of symptom onset, there is a high probability of initial false-negative testing within the first 8 days after symptom onset.19,20 Clinical understanding of this fact is imperative, as an initial negative ELISA does not rule out WNV, and a retest is warranted if clinical suspicion is high. In addition to a high initial false-negative rate with ELISA, there are several other limitations to note. IgM antibodies remain elevated for 1 to 3 months or possibly up to a year in immunocompromised patients.1 Due to this, false positives may be present if there was a recent prior infection. Enzyme-linked immunosorbent assay may not distinguish from different flaviviruses, including the yellow fever, dengue, Zika, Japanese encephalitis, and Saint Louis encephalitis viruses. Seropositivity has been estimated in some states, including 1999 data from New York (2.6%), 2003 data from Nebraska (9.5%), and 2012-2014 data from Connecticut (8.5%).21-23 Regional variance may be expected, as there also were significant differences in WNV seropositivity between different regions in Nebraska (P<.001).23
Because ELISA testing for WNV has readily apparent flaws, other tests have been utilized in its diagnosis. The PNRT is the most specific test, and it works by measuring neutralizing antibody titers for different flaviviruses. It has the ability to determine cross-reactivity with other flaviviruses; however, it does not discriminate between a current infection and a prior infection or prior flavivirus vaccine (ie, yellow fever vaccine). Despite this, a positive PNRT can lend credibility to a positive ELISA test and determine specificity for WNV for those with no prior flavivirus exposure.24 According to the Centers for Disease Control and Prevention (CDC), this test can be performed by the CDC or in reference laboratories designated by the CDC.3 Additionally, some state health laboratories may perform PRNTs.
Viral detection with PCR currently is used to screen blood donations and may be beneficial for immunocompromised patients that lack the ability to form a robust antibody response or if a patient presents early, as PCR works best within the first week of symptom onset.1 Tilley et al25 showed that a combination of PCR and ELISA were able to accurately predict 94.2% of patients (180/191) with documented WNV on a first blood sample compared to 45% and 58.1% for only viral detection or ELISA, respectively. Based on costs from a Midwest academic center, antibody detection tests are around $100 while PCR may range from $500 to $1000 and is only performed in reference laboratories. Although these tests remain in the repertoire for WNV diagnosis, financial stewardship is important.
If there are symptoms of photophobia, phonophobia, nuchal rigidity, loss of consciousness, or marked personality changes, a lumbar puncture for WNV IgM in the cerebrospinal fluid can be performed. As with most viral infections, cerebrospinal fluid findings normally include an elevated protein and lymphocyte count, but neutrophils may be predominantly elevated if the infection is early in its course.26
What are the management options?
To date, there is no curative treatment for WNV, and management is largely supportive. For WNF, over-the-counter pain medications may be helpful to reduce fever and pain. If more severe disease develops, hospitalization for further supportive care may be needed.27 If meningitis or encephalitis is suspected, broad-spectrum antibiotics may need to be started until other common etiologies are ruled out.28
How can you prevent WNV infection?
Disease prevention largely consists of educating the public to avoid heavily wooded areas, especially in areas of high prevalence and during peak months, and to use protective clothing and insect repellant that has been approved by the Environmental Protection Agency.3 Insect repellants approved by the Environmental Protection Agency contain ingredients such as DEET (N, N-diethyl-meta-toluamide), picaridin, IR3535 (ethyl butylacetylaminopropionate), and oil of lemon eucalyptus, which have been proven safe and effective.29 Patients also can protect their homes by using window screens and promptly repairing screens with holes.3
What is the differential diagnosis for WNV?
The differential diagnosis for fever with generalized maculopapular rash broadly ranges from viral etiologies (eg, WNV, Zika, measles), to tick bites (eg, Rocky Mountain spotted fever, ehrlichiosis), to drug-induced rashes. A detailed patient history inquiring on recent sick contacts, travel (WNV in the Midwest, ehrlichiosis in the Southeast), environmental exposures (ticks, mosquitoes), and new medications (typically 7–10 days after starting) is imperative to narrow the differential.30 In addition, the distribution, timing, and clinical characteristics of the rash may aid in diagnosis, along with an appropriately correlated clinical picture. West Nile virus likely will present in the summer in mid central geographic locations and often develops on the trunk and extremities as a blanching, generalized, maculopapular rash around 5 days after symptom onset or with defervescence.1
- Petersen LR. Clinical manifestations and diagnosis of West Nile virus infection. UpToDate website. Updated August 7, 2020. Accessed April 16, 2021. https://www.uptodate.com/contents/clinical-manifestations-and-diagnosis-of-west-nile-virus-infection?search=clinical-manifestations-and-diagnosis-of-west-nile-virusinfection.&source=search_result&selectedTitle=1~78&usage_type=default&display_rank=1
- Sampathkumar P. West Nile virus: epidemiology, clinical presentation, diagnosis, and prevention. Mayo Clin Proc. 2003;78:1137-1144.
- Centers for Disease Control and Prevention. West Nile virus. Updated June 3, 2020. Accessed April 16, 2021. https://www.cdc.gov/westnile/index.html
- Chuang TW, Hockett CW, Kightlinger L, et al. Landscape-level spatial patterns of West Nile virus risk in the northern Great Plains. Am J Trop Med Hyg. 2012;86:724-731.
- Wimberly MC, Hildreth MB, Boyte SP, et al. Ecological niche of the 2003 West Nile virus epidemic in the northern great plains of the United States. PLoS One. 2008;3:E3744. doi:10.1371/journal.pone.0003744
- Centers for Disease Control and Prevention. West Nile virus disease cases reported to CDC by state of residence, 1999-2019. Accessed April 26, 2021. https://www.cdc.gov/westnile/resources/pdfs/data/West-Nile-virus-disease-cases-by-state_1999-2019-P.pdf
- Hahn MB, Monaghan AJ, Hayden MH, et al. Meteorological conditions associated with increased incidence of West Nile virus disease in the United States, 2004–2012. Am J Trop Med Hyg. 2015;92:1013-1022.
- Brown CM, DeMaria A Jr. The resurgence of West Nile virus. Ann Intern Med. 2012;157:823-824.
- Landesman WJ, Allan BF, Langerhans RB, et al. Inter-annual associations between precipitation and human incidence of West Nile virus in the United States. Vector Borne Zoonotic Dis. 2007;7:337-343.
- Hart J Jr, Tillman G, Kraut MA, et al. West Nile virus neuroinvasive disease: neurological manifestations and prospective longitudinal outcomes. BMC Infect Dis. 2014;14:248.
- Wu JJ, Huang DB, Tyring SK. West Nile virus rash on the palms and soles of the feet. J Eur Acad Dermatol Venereol. 2006;20:1393-1394.
- Sejvar J. Clinical manifestations and outcomes of West Nile virus infection. Viruses. 2014;6:606-623.
- Ferguson DD, Gershman K, LeBailly A, et al. Characteristics of the rash associated with West Nile virus fever. Clin Infect Dis. 2005;41:1204-1207.
- Marszalek R, Chen A, Gjede J. Psoriasiform eruption in the setting of West Nile virus. J Am Acad Dermatol. 2014;70:AB4. doi:10.1016/j.jaad.2014.01.017
- Shah S, Fite LP, Lane N, et al. Purpura fulminans associated with acute West Nile virus encephalitis. J Clin Virol. 2016;75:1-4.
- Civen R, Villacorte F, Robles DT, et al. West Nile virus infection in the pediatric population. Pediatr Infect Dis J. 2006;25:75-78.
- Huhn GD, Dworkin MS. Rash as a prognostic factor in West Nile virus disease. Clin Infect Dis. 2006;43:388-389.
- Murphy TD, Grandpre J, Novick SL, et al. West Nile virus infection among health-fair participants, Wyoming 2003: assessment of symptoms and risk factors. Vector Borne Zoonotic Dis. 2005;5:246-251.
- Prince HE, Tobler LH, Lapé-Nixon M, et al. Development and persistence of West Nile virus–specific immunoglobulin M (IgM), IgA, and IgG in viremic blood donors. J Clin Microbiol. 2005;43:4316-4320.
- Busch MP, Kleinman SH, Tobler LH, et al. Virus and antibody dynamics in acute West Nile Virus infection. J Infect Dis. 2008;198:984-993.
- Mostashari F, Bunning ML, Kitsutani PT, et al. Epidemic West Nile encephalitis, New York, 1999: results of a household-based seroepidemiological survey. Lancet. 2001;358:261-264.
- Cahill ME, Yao Y, Nock D, et al. West Nile virus seroprevalence, Connecticut, USA, 2000–2014. Emerg Infect Dis. 2017;23:708-710.
- Schweitzer BK, Kramer WL, Sambol AR, et al. Geographic factors contributing to a high seroprevalence of West Nile virus-specific antibodies in humans following an epidemic. Clin Vaccine Immunol. 2006;13:314-318.
- Maeda A, Maeda J. Review of diagnostic plaque reduction neutralization tests for flavivirus infection. Vet J. 2013;195:33-40.
- Tilley PA, Fox JD, Jayaraman GC, et al. Nucleic acid testing for west nile virus RNA in plasma enhances rapid diagnosis of acute infection in symptomatic patients. J Infect Dis. 2006;193:1361-1364.
- Petersen LR, Brault AC, Nasci RS. West Nile virus: review of the literature. JAMA. 2013;310:308-315.
- Yu A, Ferenczi E, Moussa K, et al. Clinical spectrum of West Nile virus neuroinvasive disease. Neurohospitalist. 2020;10:43-47.
- Michaelis M, Kleinschmidt MC, Doerr HW, et al. Minocycline inhibits West Nile virus replication and apoptosis in human neuronal cells. J Antimicrob Chemother. 2007;60:981-986.
- United State Environmental Protection Agency. Skin-applied repellent ingredients. https://www.epa.gov/insect-repellents/skin-applied-repellent-ingredients. Accessed April 16, 2021.
- Muzumdar S, Rothe MJ, Grant-Kels JM. The rash with maculopapules and fever in adults. Clin Dermatol. 2019;37:109-118.
What is West Nile virus? How is it contracted, and who can become infected?
West Nile virus (WNV) is a single-stranded RNA virus of the Flaviviridae family and Flavivirus genus, a lineage that also includes the yellow fever, dengue, Zika, Japanese encephalitis, and Saint Louis encephalitis viruses.1 Birds serve as the reservoir hosts of WNV, and mosquitoes acquire the virus during feeding.2 West Nile virus then is transmitted to humans primarily by bites from Culex mosquitoes, which are especially prevalent in wooded areas during peak mosquito season (summer through early fall in North America).1 Mosquitoes also can infect horses; however, humans and horses are dead-end hosts, meaning they do not pass the virus on to other biting mosquitoes.3 There also have been rare reports of transmission of WNV through blood and donation as well as mother-to-baby transmission.2
What is the epidemiology of WNV in the United States?
Since the introduction of WNV to the United States in 1999, it has become an important public health concern, with 48,183 cases and 2163 deaths reported since 1999.2,3 In 2018, Nebraska had the highest number of cases of WNV (n=251), followed by California (n=217), North Dakota (n=204), Illinois (n=176), and South Dakota (n=169).3 West Nile virus is endemic to all 48 contiguous states and Canada, though the Great Plains region is especially affected by WNV due to several factors, such as a greater percentage of rural land, forests, and irrigated areas.4 The Great Plains region also has been thought to be an ecological niche for a more virulent species (Culex tarsalis) compared to other regions in the United States.5
The annual incidence of WNV in the United States peaked in 2003 at 9862 cases (up from 62 cases in 1999), then declined gradually until 2008 to 2011, during which the incidence was stable at 700 to 1100 new cases per year. However, there was a resurgence of cases (n=5674) in 2012 that steadied at around 2200 cases annually in subsequent years.6 Although there likely are several factors affecting WNV incidence trends in the United States, interannual changes in temperature and precipitation have been described. An increased mean annual temperature (from September through October, the end of peak mosquito season) and an increased temperature in winter months (from January through March, prior to peak mosquito season) have both been associated with an increased incidence of WNV.7 An increased temperature is thought to increase population numbers of mosquitoes both by increasing reproductive rates and creating ideal breeding environments via pooled water areas.8 Depending on the region, both above average and below average precipitation levels in the United States can increase WNV incidence the following year.7,9
What are the signs and symptoms of WNV infection?
Up to 80% of those infected with WNV are asymptomatic.3 After an incubation period of roughly 2 to 14 days, the remaining 20% may develop symptoms of West Nile fever (WNF), typically a self-limited illness that consists of 3 to 10 days of nonspecific symptoms such as fever, headache, fatigue, muscle pain and/or weakness, eye pain, gastrointestinal tract upset, and a macular rash that usually presents on the trunk or extremities.1,3 Less than 1% of patients affected by WNV develop neuroinvasive disease, including meningitis, encephalitis, and/or acute flaccid paralysis.10 West Nile virus neuroinvasive disease can cause permanent neurologic sequelae such as muscle weakness, confusion, memory loss, and fatigue; it carries a mortality rate of 10% to 30%, which is mainly dependent on older age and immunosuppression status.1,10
What is the reported spectrum of cutaneous findings in WNV?
Of the roughly 20% of patients infected with WNV that develop WNF, approximately 25% to 50% will develop an associated rash.1 It most commonly is described as a morbilliform or maculopapular rash located on the chest, back, and arms, usually sparing the palms and soles, though 1 case report noted involvement with these areas (Figure).11,12 It typically appears 5 days after symptom onset, can be associated with defervescence, and lasts less than a week.1,13 Pruritus and dysesthesia are sometimes present.13 Other rare presentations that have been reported include an ill-defined pseudovesicular rash with erythematous papules on the palms and pink, scaly, psoriasiform papules on the feet and thighs, as well as neuroinvasive WNV leading to purpura fulminans.14,15 A diffuse, erythematous, petechial rash on the face, neck, trunk, and extremities was reported in a pediatric patient, but there have been no reports of a petechial rash associated with WNV in adult patients.16 These findings suggest some potential variability in the presentation of the WNV rash.
What role does the presence of rash play diagnostically and prognostically?
The rash of WNV has been implicated as a potential prognostic factor in predicting more favorable outcomes.17 Using 2002 data from the Illinois Department of Public Health and 2003 data from the Colorado Department of Public Health, Huhn and Dworkin17 found the age-adjusted risk of encephalitis and death to be decreased in WNV patients with a rash (relative risk, 0.44; 95% CI, 0.21-0.92). The reasons for this are not definitively known, but we hypothesize that the rash may prompt patients to seek earlier medical attention or indicate a more robust immune response. Additionally, a rash in WNV more commonly is seen in younger patients, whereas WNV neuroinvasive disease is more common in older patients, who also tend to have worse outcomes.10 One study found rash to be the only symptom that demonstrated a significant association with seropositivity (overall risk=6.35; P<.05; 95% CI, 3.75-10.80) by multivariate analysis.18
How is WNV diagnosed? What are the downsides to WNV testing?
Given that the presenting symptoms of WNV and WNF are nonspecific, it becomes challenging to arrive at the diagnosis based solely on physical examination. As such, the patient’s clinical and epidemiologic history, such as timing, pattern, and appearance of the rash or recent history of mosquito bites, is key to arriving at the correct diagnosis. With clinical suspicion, possible diagnostic tests include an IgM enzyme-linked immunosorbent assay (ELISA) for WNV, a plaque reduction neutralization test (PNRT), and blood polymerase chain reaction (PCR).
An ELISA is a confirmatory test to detect IgM antibodies to WNV in the serum. Because IgM seroconversion typically occurs between days 4 and 10 of symptom onset, there is a high probability of initial false-negative testing within the first 8 days after symptom onset.19,20 Clinical understanding of this fact is imperative, as an initial negative ELISA does not rule out WNV, and a retest is warranted if clinical suspicion is high. In addition to a high initial false-negative rate with ELISA, there are several other limitations to note. IgM antibodies remain elevated for 1 to 3 months or possibly up to a year in immunocompromised patients.1 Due to this, false positives may be present if there was a recent prior infection. Enzyme-linked immunosorbent assay may not distinguish from different flaviviruses, including the yellow fever, dengue, Zika, Japanese encephalitis, and Saint Louis encephalitis viruses. Seropositivity has been estimated in some states, including 1999 data from New York (2.6%), 2003 data from Nebraska (9.5%), and 2012-2014 data from Connecticut (8.5%).21-23 Regional variance may be expected, as there also were significant differences in WNV seropositivity between different regions in Nebraska (P<.001).23
Because ELISA testing for WNV has readily apparent flaws, other tests have been utilized in its diagnosis. The PNRT is the most specific test, and it works by measuring neutralizing antibody titers for different flaviviruses. It has the ability to determine cross-reactivity with other flaviviruses; however, it does not discriminate between a current infection and a prior infection or prior flavivirus vaccine (ie, yellow fever vaccine). Despite this, a positive PNRT can lend credibility to a positive ELISA test and determine specificity for WNV for those with no prior flavivirus exposure.24 According to the Centers for Disease Control and Prevention (CDC), this test can be performed by the CDC or in reference laboratories designated by the CDC.3 Additionally, some state health laboratories may perform PRNTs.
Viral detection with PCR currently is used to screen blood donations and may be beneficial for immunocompromised patients that lack the ability to form a robust antibody response or if a patient presents early, as PCR works best within the first week of symptom onset.1 Tilley et al25 showed that a combination of PCR and ELISA were able to accurately predict 94.2% of patients (180/191) with documented WNV on a first blood sample compared to 45% and 58.1% for only viral detection or ELISA, respectively. Based on costs from a Midwest academic center, antibody detection tests are around $100 while PCR may range from $500 to $1000 and is only performed in reference laboratories. Although these tests remain in the repertoire for WNV diagnosis, financial stewardship is important.
If there are symptoms of photophobia, phonophobia, nuchal rigidity, loss of consciousness, or marked personality changes, a lumbar puncture for WNV IgM in the cerebrospinal fluid can be performed. As with most viral infections, cerebrospinal fluid findings normally include an elevated protein and lymphocyte count, but neutrophils may be predominantly elevated if the infection is early in its course.26
What are the management options?
To date, there is no curative treatment for WNV, and management is largely supportive. For WNF, over-the-counter pain medications may be helpful to reduce fever and pain. If more severe disease develops, hospitalization for further supportive care may be needed.27 If meningitis or encephalitis is suspected, broad-spectrum antibiotics may need to be started until other common etiologies are ruled out.28
How can you prevent WNV infection?
Disease prevention largely consists of educating the public to avoid heavily wooded areas, especially in areas of high prevalence and during peak months, and to use protective clothing and insect repellant that has been approved by the Environmental Protection Agency.3 Insect repellants approved by the Environmental Protection Agency contain ingredients such as DEET (N, N-diethyl-meta-toluamide), picaridin, IR3535 (ethyl butylacetylaminopropionate), and oil of lemon eucalyptus, which have been proven safe and effective.29 Patients also can protect their homes by using window screens and promptly repairing screens with holes.3
What is the differential diagnosis for WNV?
The differential diagnosis for fever with generalized maculopapular rash broadly ranges from viral etiologies (eg, WNV, Zika, measles), to tick bites (eg, Rocky Mountain spotted fever, ehrlichiosis), to drug-induced rashes. A detailed patient history inquiring on recent sick contacts, travel (WNV in the Midwest, ehrlichiosis in the Southeast), environmental exposures (ticks, mosquitoes), and new medications (typically 7–10 days after starting) is imperative to narrow the differential.30 In addition, the distribution, timing, and clinical characteristics of the rash may aid in diagnosis, along with an appropriately correlated clinical picture. West Nile virus likely will present in the summer in mid central geographic locations and often develops on the trunk and extremities as a blanching, generalized, maculopapular rash around 5 days after symptom onset or with defervescence.1
What is West Nile virus? How is it contracted, and who can become infected?
West Nile virus (WNV) is a single-stranded RNA virus of the Flaviviridae family and Flavivirus genus, a lineage that also includes the yellow fever, dengue, Zika, Japanese encephalitis, and Saint Louis encephalitis viruses.1 Birds serve as the reservoir hosts of WNV, and mosquitoes acquire the virus during feeding.2 West Nile virus then is transmitted to humans primarily by bites from Culex mosquitoes, which are especially prevalent in wooded areas during peak mosquito season (summer through early fall in North America).1 Mosquitoes also can infect horses; however, humans and horses are dead-end hosts, meaning they do not pass the virus on to other biting mosquitoes.3 There also have been rare reports of transmission of WNV through blood and donation as well as mother-to-baby transmission.2
What is the epidemiology of WNV in the United States?
Since the introduction of WNV to the United States in 1999, it has become an important public health concern, with 48,183 cases and 2163 deaths reported since 1999.2,3 In 2018, Nebraska had the highest number of cases of WNV (n=251), followed by California (n=217), North Dakota (n=204), Illinois (n=176), and South Dakota (n=169).3 West Nile virus is endemic to all 48 contiguous states and Canada, though the Great Plains region is especially affected by WNV due to several factors, such as a greater percentage of rural land, forests, and irrigated areas.4 The Great Plains region also has been thought to be an ecological niche for a more virulent species (Culex tarsalis) compared to other regions in the United States.5
The annual incidence of WNV in the United States peaked in 2003 at 9862 cases (up from 62 cases in 1999), then declined gradually until 2008 to 2011, during which the incidence was stable at 700 to 1100 new cases per year. However, there was a resurgence of cases (n=5674) in 2012 that steadied at around 2200 cases annually in subsequent years.6 Although there likely are several factors affecting WNV incidence trends in the United States, interannual changes in temperature and precipitation have been described. An increased mean annual temperature (from September through October, the end of peak mosquito season) and an increased temperature in winter months (from January through March, prior to peak mosquito season) have both been associated with an increased incidence of WNV.7 An increased temperature is thought to increase population numbers of mosquitoes both by increasing reproductive rates and creating ideal breeding environments via pooled water areas.8 Depending on the region, both above average and below average precipitation levels in the United States can increase WNV incidence the following year.7,9
What are the signs and symptoms of WNV infection?
Up to 80% of those infected with WNV are asymptomatic.3 After an incubation period of roughly 2 to 14 days, the remaining 20% may develop symptoms of West Nile fever (WNF), typically a self-limited illness that consists of 3 to 10 days of nonspecific symptoms such as fever, headache, fatigue, muscle pain and/or weakness, eye pain, gastrointestinal tract upset, and a macular rash that usually presents on the trunk or extremities.1,3 Less than 1% of patients affected by WNV develop neuroinvasive disease, including meningitis, encephalitis, and/or acute flaccid paralysis.10 West Nile virus neuroinvasive disease can cause permanent neurologic sequelae such as muscle weakness, confusion, memory loss, and fatigue; it carries a mortality rate of 10% to 30%, which is mainly dependent on older age and immunosuppression status.1,10
What is the reported spectrum of cutaneous findings in WNV?
Of the roughly 20% of patients infected with WNV that develop WNF, approximately 25% to 50% will develop an associated rash.1 It most commonly is described as a morbilliform or maculopapular rash located on the chest, back, and arms, usually sparing the palms and soles, though 1 case report noted involvement with these areas (Figure).11,12 It typically appears 5 days after symptom onset, can be associated with defervescence, and lasts less than a week.1,13 Pruritus and dysesthesia are sometimes present.13 Other rare presentations that have been reported include an ill-defined pseudovesicular rash with erythematous papules on the palms and pink, scaly, psoriasiform papules on the feet and thighs, as well as neuroinvasive WNV leading to purpura fulminans.14,15 A diffuse, erythematous, petechial rash on the face, neck, trunk, and extremities was reported in a pediatric patient, but there have been no reports of a petechial rash associated with WNV in adult patients.16 These findings suggest some potential variability in the presentation of the WNV rash.
What role does the presence of rash play diagnostically and prognostically?
The rash of WNV has been implicated as a potential prognostic factor in predicting more favorable outcomes.17 Using 2002 data from the Illinois Department of Public Health and 2003 data from the Colorado Department of Public Health, Huhn and Dworkin17 found the age-adjusted risk of encephalitis and death to be decreased in WNV patients with a rash (relative risk, 0.44; 95% CI, 0.21-0.92). The reasons for this are not definitively known, but we hypothesize that the rash may prompt patients to seek earlier medical attention or indicate a more robust immune response. Additionally, a rash in WNV more commonly is seen in younger patients, whereas WNV neuroinvasive disease is more common in older patients, who also tend to have worse outcomes.10 One study found rash to be the only symptom that demonstrated a significant association with seropositivity (overall risk=6.35; P<.05; 95% CI, 3.75-10.80) by multivariate analysis.18
How is WNV diagnosed? What are the downsides to WNV testing?
Given that the presenting symptoms of WNV and WNF are nonspecific, it becomes challenging to arrive at the diagnosis based solely on physical examination. As such, the patient’s clinical and epidemiologic history, such as timing, pattern, and appearance of the rash or recent history of mosquito bites, is key to arriving at the correct diagnosis. With clinical suspicion, possible diagnostic tests include an IgM enzyme-linked immunosorbent assay (ELISA) for WNV, a plaque reduction neutralization test (PNRT), and blood polymerase chain reaction (PCR).
An ELISA is a confirmatory test to detect IgM antibodies to WNV in the serum. Because IgM seroconversion typically occurs between days 4 and 10 of symptom onset, there is a high probability of initial false-negative testing within the first 8 days after symptom onset.19,20 Clinical understanding of this fact is imperative, as an initial negative ELISA does not rule out WNV, and a retest is warranted if clinical suspicion is high. In addition to a high initial false-negative rate with ELISA, there are several other limitations to note. IgM antibodies remain elevated for 1 to 3 months or possibly up to a year in immunocompromised patients.1 Due to this, false positives may be present if there was a recent prior infection. Enzyme-linked immunosorbent assay may not distinguish from different flaviviruses, including the yellow fever, dengue, Zika, Japanese encephalitis, and Saint Louis encephalitis viruses. Seropositivity has been estimated in some states, including 1999 data from New York (2.6%), 2003 data from Nebraska (9.5%), and 2012-2014 data from Connecticut (8.5%).21-23 Regional variance may be expected, as there also were significant differences in WNV seropositivity between different regions in Nebraska (P<.001).23
Because ELISA testing for WNV has readily apparent flaws, other tests have been utilized in its diagnosis. The PNRT is the most specific test, and it works by measuring neutralizing antibody titers for different flaviviruses. It has the ability to determine cross-reactivity with other flaviviruses; however, it does not discriminate between a current infection and a prior infection or prior flavivirus vaccine (ie, yellow fever vaccine). Despite this, a positive PNRT can lend credibility to a positive ELISA test and determine specificity for WNV for those with no prior flavivirus exposure.24 According to the Centers for Disease Control and Prevention (CDC), this test can be performed by the CDC or in reference laboratories designated by the CDC.3 Additionally, some state health laboratories may perform PRNTs.
Viral detection with PCR currently is used to screen blood donations and may be beneficial for immunocompromised patients that lack the ability to form a robust antibody response or if a patient presents early, as PCR works best within the first week of symptom onset.1 Tilley et al25 showed that a combination of PCR and ELISA were able to accurately predict 94.2% of patients (180/191) with documented WNV on a first blood sample compared to 45% and 58.1% for only viral detection or ELISA, respectively. Based on costs from a Midwest academic center, antibody detection tests are around $100 while PCR may range from $500 to $1000 and is only performed in reference laboratories. Although these tests remain in the repertoire for WNV diagnosis, financial stewardship is important.
If there are symptoms of photophobia, phonophobia, nuchal rigidity, loss of consciousness, or marked personality changes, a lumbar puncture for WNV IgM in the cerebrospinal fluid can be performed. As with most viral infections, cerebrospinal fluid findings normally include an elevated protein and lymphocyte count, but neutrophils may be predominantly elevated if the infection is early in its course.26
What are the management options?
To date, there is no curative treatment for WNV, and management is largely supportive. For WNF, over-the-counter pain medications may be helpful to reduce fever and pain. If more severe disease develops, hospitalization for further supportive care may be needed.27 If meningitis or encephalitis is suspected, broad-spectrum antibiotics may need to be started until other common etiologies are ruled out.28
How can you prevent WNV infection?
Disease prevention largely consists of educating the public to avoid heavily wooded areas, especially in areas of high prevalence and during peak months, and to use protective clothing and insect repellant that has been approved by the Environmental Protection Agency.3 Insect repellants approved by the Environmental Protection Agency contain ingredients such as DEET (N, N-diethyl-meta-toluamide), picaridin, IR3535 (ethyl butylacetylaminopropionate), and oil of lemon eucalyptus, which have been proven safe and effective.29 Patients also can protect their homes by using window screens and promptly repairing screens with holes.3
What is the differential diagnosis for WNV?
The differential diagnosis for fever with generalized maculopapular rash broadly ranges from viral etiologies (eg, WNV, Zika, measles), to tick bites (eg, Rocky Mountain spotted fever, ehrlichiosis), to drug-induced rashes. A detailed patient history inquiring on recent sick contacts, travel (WNV in the Midwest, ehrlichiosis in the Southeast), environmental exposures (ticks, mosquitoes), and new medications (typically 7–10 days after starting) is imperative to narrow the differential.30 In addition, the distribution, timing, and clinical characteristics of the rash may aid in diagnosis, along with an appropriately correlated clinical picture. West Nile virus likely will present in the summer in mid central geographic locations and often develops on the trunk and extremities as a blanching, generalized, maculopapular rash around 5 days after symptom onset or with defervescence.1
- Petersen LR. Clinical manifestations and diagnosis of West Nile virus infection. UpToDate website. Updated August 7, 2020. Accessed April 16, 2021. https://www.uptodate.com/contents/clinical-manifestations-and-diagnosis-of-west-nile-virus-infection?search=clinical-manifestations-and-diagnosis-of-west-nile-virusinfection.&source=search_result&selectedTitle=1~78&usage_type=default&display_rank=1
- Sampathkumar P. West Nile virus: epidemiology, clinical presentation, diagnosis, and prevention. Mayo Clin Proc. 2003;78:1137-1144.
- Centers for Disease Control and Prevention. West Nile virus. Updated June 3, 2020. Accessed April 16, 2021. https://www.cdc.gov/westnile/index.html
- Chuang TW, Hockett CW, Kightlinger L, et al. Landscape-level spatial patterns of West Nile virus risk in the northern Great Plains. Am J Trop Med Hyg. 2012;86:724-731.
- Wimberly MC, Hildreth MB, Boyte SP, et al. Ecological niche of the 2003 West Nile virus epidemic in the northern great plains of the United States. PLoS One. 2008;3:E3744. doi:10.1371/journal.pone.0003744
- Centers for Disease Control and Prevention. West Nile virus disease cases reported to CDC by state of residence, 1999-2019. Accessed April 26, 2021. https://www.cdc.gov/westnile/resources/pdfs/data/West-Nile-virus-disease-cases-by-state_1999-2019-P.pdf
- Hahn MB, Monaghan AJ, Hayden MH, et al. Meteorological conditions associated with increased incidence of West Nile virus disease in the United States, 2004–2012. Am J Trop Med Hyg. 2015;92:1013-1022.
- Brown CM, DeMaria A Jr. The resurgence of West Nile virus. Ann Intern Med. 2012;157:823-824.
- Landesman WJ, Allan BF, Langerhans RB, et al. Inter-annual associations between precipitation and human incidence of West Nile virus in the United States. Vector Borne Zoonotic Dis. 2007;7:337-343.
- Hart J Jr, Tillman G, Kraut MA, et al. West Nile virus neuroinvasive disease: neurological manifestations and prospective longitudinal outcomes. BMC Infect Dis. 2014;14:248.
- Wu JJ, Huang DB, Tyring SK. West Nile virus rash on the palms and soles of the feet. J Eur Acad Dermatol Venereol. 2006;20:1393-1394.
- Sejvar J. Clinical manifestations and outcomes of West Nile virus infection. Viruses. 2014;6:606-623.
- Ferguson DD, Gershman K, LeBailly A, et al. Characteristics of the rash associated with West Nile virus fever. Clin Infect Dis. 2005;41:1204-1207.
- Marszalek R, Chen A, Gjede J. Psoriasiform eruption in the setting of West Nile virus. J Am Acad Dermatol. 2014;70:AB4. doi:10.1016/j.jaad.2014.01.017
- Shah S, Fite LP, Lane N, et al. Purpura fulminans associated with acute West Nile virus encephalitis. J Clin Virol. 2016;75:1-4.
- Civen R, Villacorte F, Robles DT, et al. West Nile virus infection in the pediatric population. Pediatr Infect Dis J. 2006;25:75-78.
- Huhn GD, Dworkin MS. Rash as a prognostic factor in West Nile virus disease. Clin Infect Dis. 2006;43:388-389.
- Murphy TD, Grandpre J, Novick SL, et al. West Nile virus infection among health-fair participants, Wyoming 2003: assessment of symptoms and risk factors. Vector Borne Zoonotic Dis. 2005;5:246-251.
- Prince HE, Tobler LH, Lapé-Nixon M, et al. Development and persistence of West Nile virus–specific immunoglobulin M (IgM), IgA, and IgG in viremic blood donors. J Clin Microbiol. 2005;43:4316-4320.
- Busch MP, Kleinman SH, Tobler LH, et al. Virus and antibody dynamics in acute West Nile Virus infection. J Infect Dis. 2008;198:984-993.
- Mostashari F, Bunning ML, Kitsutani PT, et al. Epidemic West Nile encephalitis, New York, 1999: results of a household-based seroepidemiological survey. Lancet. 2001;358:261-264.
- Cahill ME, Yao Y, Nock D, et al. West Nile virus seroprevalence, Connecticut, USA, 2000–2014. Emerg Infect Dis. 2017;23:708-710.
- Schweitzer BK, Kramer WL, Sambol AR, et al. Geographic factors contributing to a high seroprevalence of West Nile virus-specific antibodies in humans following an epidemic. Clin Vaccine Immunol. 2006;13:314-318.
- Maeda A, Maeda J. Review of diagnostic plaque reduction neutralization tests for flavivirus infection. Vet J. 2013;195:33-40.
- Tilley PA, Fox JD, Jayaraman GC, et al. Nucleic acid testing for west nile virus RNA in plasma enhances rapid diagnosis of acute infection in symptomatic patients. J Infect Dis. 2006;193:1361-1364.
- Petersen LR, Brault AC, Nasci RS. West Nile virus: review of the literature. JAMA. 2013;310:308-315.
- Yu A, Ferenczi E, Moussa K, et al. Clinical spectrum of West Nile virus neuroinvasive disease. Neurohospitalist. 2020;10:43-47.
- Michaelis M, Kleinschmidt MC, Doerr HW, et al. Minocycline inhibits West Nile virus replication and apoptosis in human neuronal cells. J Antimicrob Chemother. 2007;60:981-986.
- United State Environmental Protection Agency. Skin-applied repellent ingredients. https://www.epa.gov/insect-repellents/skin-applied-repellent-ingredients. Accessed April 16, 2021.
- Muzumdar S, Rothe MJ, Grant-Kels JM. The rash with maculopapules and fever in adults. Clin Dermatol. 2019;37:109-118.
- Petersen LR. Clinical manifestations and diagnosis of West Nile virus infection. UpToDate website. Updated August 7, 2020. Accessed April 16, 2021. https://www.uptodate.com/contents/clinical-manifestations-and-diagnosis-of-west-nile-virus-infection?search=clinical-manifestations-and-diagnosis-of-west-nile-virusinfection.&source=search_result&selectedTitle=1~78&usage_type=default&display_rank=1
- Sampathkumar P. West Nile virus: epidemiology, clinical presentation, diagnosis, and prevention. Mayo Clin Proc. 2003;78:1137-1144.
- Centers for Disease Control and Prevention. West Nile virus. Updated June 3, 2020. Accessed April 16, 2021. https://www.cdc.gov/westnile/index.html
- Chuang TW, Hockett CW, Kightlinger L, et al. Landscape-level spatial patterns of West Nile virus risk in the northern Great Plains. Am J Trop Med Hyg. 2012;86:724-731.
- Wimberly MC, Hildreth MB, Boyte SP, et al. Ecological niche of the 2003 West Nile virus epidemic in the northern great plains of the United States. PLoS One. 2008;3:E3744. doi:10.1371/journal.pone.0003744
- Centers for Disease Control and Prevention. West Nile virus disease cases reported to CDC by state of residence, 1999-2019. Accessed April 26, 2021. https://www.cdc.gov/westnile/resources/pdfs/data/West-Nile-virus-disease-cases-by-state_1999-2019-P.pdf
- Hahn MB, Monaghan AJ, Hayden MH, et al. Meteorological conditions associated with increased incidence of West Nile virus disease in the United States, 2004–2012. Am J Trop Med Hyg. 2015;92:1013-1022.
- Brown CM, DeMaria A Jr. The resurgence of West Nile virus. Ann Intern Med. 2012;157:823-824.
- Landesman WJ, Allan BF, Langerhans RB, et al. Inter-annual associations between precipitation and human incidence of West Nile virus in the United States. Vector Borne Zoonotic Dis. 2007;7:337-343.
- Hart J Jr, Tillman G, Kraut MA, et al. West Nile virus neuroinvasive disease: neurological manifestations and prospective longitudinal outcomes. BMC Infect Dis. 2014;14:248.
- Wu JJ, Huang DB, Tyring SK. West Nile virus rash on the palms and soles of the feet. J Eur Acad Dermatol Venereol. 2006;20:1393-1394.
- Sejvar J. Clinical manifestations and outcomes of West Nile virus infection. Viruses. 2014;6:606-623.
- Ferguson DD, Gershman K, LeBailly A, et al. Characteristics of the rash associated with West Nile virus fever. Clin Infect Dis. 2005;41:1204-1207.
- Marszalek R, Chen A, Gjede J. Psoriasiform eruption in the setting of West Nile virus. J Am Acad Dermatol. 2014;70:AB4. doi:10.1016/j.jaad.2014.01.017
- Shah S, Fite LP, Lane N, et al. Purpura fulminans associated with acute West Nile virus encephalitis. J Clin Virol. 2016;75:1-4.
- Civen R, Villacorte F, Robles DT, et al. West Nile virus infection in the pediatric population. Pediatr Infect Dis J. 2006;25:75-78.
- Huhn GD, Dworkin MS. Rash as a prognostic factor in West Nile virus disease. Clin Infect Dis. 2006;43:388-389.
- Murphy TD, Grandpre J, Novick SL, et al. West Nile virus infection among health-fair participants, Wyoming 2003: assessment of symptoms and risk factors. Vector Borne Zoonotic Dis. 2005;5:246-251.
- Prince HE, Tobler LH, Lapé-Nixon M, et al. Development and persistence of West Nile virus–specific immunoglobulin M (IgM), IgA, and IgG in viremic blood donors. J Clin Microbiol. 2005;43:4316-4320.
- Busch MP, Kleinman SH, Tobler LH, et al. Virus and antibody dynamics in acute West Nile Virus infection. J Infect Dis. 2008;198:984-993.
- Mostashari F, Bunning ML, Kitsutani PT, et al. Epidemic West Nile encephalitis, New York, 1999: results of a household-based seroepidemiological survey. Lancet. 2001;358:261-264.
- Cahill ME, Yao Y, Nock D, et al. West Nile virus seroprevalence, Connecticut, USA, 2000–2014. Emerg Infect Dis. 2017;23:708-710.
- Schweitzer BK, Kramer WL, Sambol AR, et al. Geographic factors contributing to a high seroprevalence of West Nile virus-specific antibodies in humans following an epidemic. Clin Vaccine Immunol. 2006;13:314-318.
- Maeda A, Maeda J. Review of diagnostic plaque reduction neutralization tests for flavivirus infection. Vet J. 2013;195:33-40.
- Tilley PA, Fox JD, Jayaraman GC, et al. Nucleic acid testing for west nile virus RNA in plasma enhances rapid diagnosis of acute infection in symptomatic patients. J Infect Dis. 2006;193:1361-1364.
- Petersen LR, Brault AC, Nasci RS. West Nile virus: review of the literature. JAMA. 2013;310:308-315.
- Yu A, Ferenczi E, Moussa K, et al. Clinical spectrum of West Nile virus neuroinvasive disease. Neurohospitalist. 2020;10:43-47.
- Michaelis M, Kleinschmidt MC, Doerr HW, et al. Minocycline inhibits West Nile virus replication and apoptosis in human neuronal cells. J Antimicrob Chemother. 2007;60:981-986.
- United State Environmental Protection Agency. Skin-applied repellent ingredients. https://www.epa.gov/insect-repellents/skin-applied-repellent-ingredients. Accessed April 16, 2021.
- Muzumdar S, Rothe MJ, Grant-Kels JM. The rash with maculopapules and fever in adults. Clin Dermatol. 2019;37:109-118.
Practice Points
- Dermatologists should be aware of the most common rash associated with West Nile virus (WNV), which is a nonspecific maculopapular rash appearing on the trunk and extremities around 5 days after the onset of fever, fatigue, and other nonspecific symptoms.
- Rash may serve as a prognostic indicator for improved outcomes in WNV due to its association with decreased risk of encephalitis and death.
- An IgM enzyme-linked immunosorbent assay for WNV initially may yield false-negative results, as the development of detectable antibodies against the virus may take up to 8 days after symptom onset.
Botanical Briefs: Phytophotodermatitis Is an Occupational and Recreational Dermatosis in the Limelight
Phytophotodermatitis (PPD) is a nonallergic contact dermatitis and thus is independent of the immune system, so prior sensitization is not required.1-3 It sometimes is known by colorful names such as margarita photodermatitis, in which a slice of lime in a refreshing summer drink may be etiologic,4,5 or berloque dermatitis, caused by exposure to perfumes containing bergapten (5-methoxypsoralen).6,7 Phytophotodermatitis may develop when phototoxic agents such as furocoumarins, which protect plants from fungal pathogens, and psoralens are applied to the skin followed by exposure to UV light, more specifically in the UVA range of 320 to 400 nm. Thus, these chemicals produce a phototoxic rather than photoallergic reaction, leading to cellular damage. Furocoumarins and psoralens often are found in plants such as celery and figs as well as in citrus fruits such as limes, lemons, and grapefruits. Exposure may be cryptic, as the patient may not consider or mention the eruption as possibly caused by activities such as soaking one’s feet in a folk remedy containing fig leaves.7,8 Once these phototoxic agents come in contact with the skin, the symptoms of PPD may arise within 24 hours of exposure, beginning as an acute dermatitis with erythema, edema, vesicles, or bullae accompanied by pain and itching.
Etiology
Phytophotodermatitis is caused by exposure to several different types of plants, including Ficus carica (common fig), the genus Citrus (eg, lime, lemon), or Pastina sativa (wild parsnip). Each of these contain furocoumarins and psoralens—phototoxic agents that cause cellular damage with epidermal necrosis and resultant pain when the skin is exposed to UVA light.1-4 There are 2 types of photochemical reactions in PPD: type I reactions occur in the absence of oxygen, whereas oxygen is present in type II reactions. Both damage cell membranes and DNA, which then results in DNA interstrand cross-linking between the psoralen furan ring and the thymine or cytosine of DNA, activating arachidonic acid metabolic pathways to produce cell death.1
Epidemiology
The incidence of PPD is unknown due to the high variability of reactions in individuals spanning from children to the elderly. It can be caused by many different wild and domestic plants in many areas of the world and can affect any individual regardless of age, race, gender, or ethnicity. Some individuals may be affected by hyperpigmentation without prominent inflammation.8 Diagnosis of PPD can be challenging, and an occupation and recreational history of exposure or recent travel with possible contact with plants may be required.
Occupational Dermatitis
Recreational Dermatitis
Phytophotodermatitis may be caused by exposure to phototoxic agents during leisure activities. Recreational exposure can occur almost anywhere, including in the kitchen, backyard, park, or woods, as well as at the beach. One notable culprit in recreational PPD is cooking with limes, parsley, or parsnips—plants that often are employed as garnishes in dishes, allowing early exposure of juices on the hands. Individuals who garden recreationally should be aware of ornamental plants such as hogweed and figs, which are notorious for causing PPD.13 Children’s camp counselors should have knowledge of PPD, as children have considerable curiosity and may touch or play with attractive plants such as hogweed. Children enjoying sports in parks can accidentally fall onto or be exposed to wild parsnip or hogweed growing nearby and wake up the next day with erythema and burning.14 Photoprotection is important, but sunscreens containing carrot extract can produce PPD.15 Widespread PPD over 80% of the body surface area due to sunbathing after applying fig leaf tea as a tanning agent has been described.16 Eating figs does not cause photosensitization unless the juice is smeared onto the skin. Margarita dermatitis and “Mexican beer dermatitis” can occur due to limes and other citrus fruits being used as ingredients in summer drinks.5 Similarly, preparing sangria may produce PPD from lime and lemon juices.17 In one report, hiking in Corsica resulted in PPD following incidental contact with the endemic plant Peucedanum paniculatum.18
Perfume (Berloque) Dermatitis
Perfume dermatitis, or berloque dermatitis, is a type of PPD for which the name is derived from the German word berlock or the French word berloque meaning trinket or charm; it was first described in 1925 by Rosenthal7 with regard to pendantlike streaks of pigmentation on the neck, face, arms, or trunk. The dermatitis develops due to bergapten, a component of bergamot oil, which is derived from the rind of Citrus bergamia. Many perfumes contain bergamot oil, but the incidence of this condition has been diminished due to use of artificial bergamot oil.6
Clinical Manifestation
Phytophotodermatitis is first evident as erythematous patches that appear within 24 hours of initial exposure to a phototoxic agent and UVA light, sometimes with a burning sensation. Solar exposure within 48 hours of sufficient plant exposure is required. Perfuse sweating may enhance the reaction.19 Rarely, it first may be seen with the sudden appearance of
Differential Diagnosis
Phytophotodermatitis may resemble other types of dermatitis, particularly other forms of contact dermatitis such poison ivy, and occasionally other environmental simulants such as jellyfish stings.1-6,20,21 Photosensitizing disorders including porphyria cutanea tarda, pseudoporphyria, and lupus erythematosus must be distinguished from PPD.22-24 Photosensitizing medications such tetracyclines, thiazide diuretics, sulfonamides, griseofulvin, and sulfonylureas should be considered. Airborne contact dermatitis may resemble PPD, as when poison ivy is burned and is exposed to the skin in sites of airborne contact.20 Excessive solar exposure is popular, particularly among adolescents, so sunburn and sunburnlike reactions can be noteworthy.25,26
Treatment
Phytophotodermatitis can be treated with topical steroids, sometimes adding an oral antihistamine, and occasionally oral steroids.2-4 Localized pain or a burning sensation should respond to therapy. Alternatively, a cold compress applied to the skin can relieve the pain and pruritus, and the burn can be debrided and dressed daily with silver sulfadiazine plus an oral nonsteroidal anti-inflammatory drug. This eruption should be self-limited as long as it is recognized early and the cause avoided. Management of acute exposure includes prompt application of soap and water and avoidance of UV light exposure for 48 to 72 hours to prevent psoralen photoactivation.
Because PPD is essentially a chemical burn, a burn protocol and possible referral to a burn center may be needed, whether the reaction is acute or widespread.11,12,14,27,28 Surgical debridement and skin grafting rarely may be mandated.14 Postinflammatory hyperpigmentation may ensue as the dermatitis resolves but is not common.
The best approach for PPD is prevention (Figure 2). Individuals who are at risk should be aware of their surroundings and potential plants of concern and employ personal protective equipment to shield the skin from plant sap, which should be promptly removed if it comes in contact with the skin.
- Zhang R, Zhu W. Phytophotodermatitis due to Chinese herbal medicine decoction. Indian J Dermatol. 2011;56:329-331.
- Harshman J, Quan Y, Hsiang D. Phytophotodermatitis: rash with many faces. Can Fam Physician. 2017;63:938-940.
- Imen MS, Ahmadabadi A, Tavousi SH, et al. The curious cases of burn by fig tree leaves. Indian J Dermatol. 2019;64:71-73.
- Hankinson A, Lloyd B, Alweis R. Lime-induced phytophotodermatitis [published online September 29, 2014]. J Community Hosp Intern Med Perspect. doi:10.3402/jchimp.v4.25090
- Abramowitz AI, Resnik KS, Cohen KR. Margarita photodermatitis. N Engl J Med. 2013;328:891.
- Quaak MS, Martens H, Hassing RJ, et al. The sunny side of lime. J Travel Med. 2012;19:327-328.
- Rosenthal O. Berloque dermatitis: Berliner Dermatologische Gesellschaft. Dermatol Zeitschrift. 1925;42:295.
- Choi JY, Hwang S, Lee SH, et al. Asymptomatic hyperpigmentation without preceding inflammation as a clinical feature of citrus fruits–induced phytophotodermatitis. Ann Dermatol. 2018;30:75-78.
- Wynn P, Bell S. Phytophotodermatitis in grounds operatives. Occup Med (Lond). 2005;55:393-395.
- Klimaszyk P, Klimaszyk D, Piotrowiak M, et al. Unusual complications after occupational exposure to giant hogweed (Heracleum mantegazzianum): a case report. Int J Occup Med Environ Health. 2014;27:141-144.
- Downs JW, Cumpston KL, Feldman MJ. Giant hogweed phytophotodermatitis. Clin Toxicol (Phila). 2019;57:822-823.
- Maso MJ, Ruszkowski AM, Bauerle J, et al. Celery phytophotodermatitis in a chef. Arch Dermatol. 1991;127:912-913.
- Derraik JG, Rademaker M. Phytophotodermatitis caused by contact with a fig tree (Ficus carica). New Zealand Med J. 2007;120:U2720.
- Chan JC, Sullivan PJ, O’Sullivan MJ, et al. Full thickness burn caused by exposure to giant hogweed: delayed presentation, histological features and surgical management. J Plast Reconstr Aesthet Surg. 2011;64:128-130.
- Bosanac SS, Clark AK, Sivamani RK. Phytophotodermatitis related to carrot extract–containing sunscreen. Dermatol Online J. 2018;24:1-3.
- Sforza M, Andjelkov K, Zaccheddu R. Severe burn on 81% of body surface after sun tanning. Ulus Travma Acil Cerrahi Derg. 2013;19:383-384.
- Mioduszewski M, Beecker J. Phytophotodermatitis from making sangria: a phototoxic reaction to lime and lemon juice. CMAJ. 2015;187:756.
- Torrents R, Schmitt C, Domangé B, et al. Phytophotodermatitis with Peucedanum paniculatum: an endemic species to Corsica. Clin Toxicol (Phila). 2019;57:68-69.
- Sarhane KA, Ibrahim A, Fagan SP, et al. Phytophotodermatitis. Eplasty. 2013;13:ic57.
- DeLeo VA, Suarez SM, Maso MJ. Photoallergic contact dermatitis. results of photopatch testing in New York, 1985 to 1990. Arch Dermatol. 1992;128:1513-1518.
- Kimyon RS, Warshaw EM. Airborne allergic contact dermatitis: management and responsible allergens on the American Contact Dermatitis Society Core Series. Dermatitis. 2019;30:106-115.
- Miteva L, Broshtilova V, Schwartz RA. Unusual clinical manifestations of chronic discoid lupus erythematosus. Serbian J Dermatol Venereol. 2014;6:69-72.
- Handler NS, Handler MZ, Stephany MP, et al. Porphyria cutanea tarda: an intriguing genetic disease and marker. Int J Dermatol. 2017;56:E106-E117.
- Papadopoulos AJ, Schwartz RA, Fekete Z, et al. Pseudoporphyria: an atypical variant resembling toxic epidermal necrolysis. J Cutan Med Surg. 2001;5:479-485.
- Jasterzbski TJ, Janniger EJ, Schwartz RA. Adolescent tanning practices: understanding the popularity of excessive ultraviolet light exposure. In: Or
anje A, Al-Mutairi N, Shwayder T, eds. Practical Pediatric Dermatology. Controversies in Diagnosis and Treatment. Springer Verlag; 2016:177-185. - Lai YC, Janniger EJ, Schwartz RA. Solar protection policy in school children: proposals for progress. In: Oranje A, Al-Mutairi N, Shwayder T, eds. Practical Pediatric Dermatology. Controversies in Diagnosis and Treatment. Springer Verlag; 2016:165-176.
- Lagey K, Duinslaeger L, Vanderkelen A. Burns induced by plants. Burns. 1995;21:542-543.
- Redgrave N, Solomon J. Severe phytophotodermatitis from fig sap: a little known phenomenon. BMJ Case Rep. 2021;14:e238745.
Phytophotodermatitis (PPD) is a nonallergic contact dermatitis and thus is independent of the immune system, so prior sensitization is not required.1-3 It sometimes is known by colorful names such as margarita photodermatitis, in which a slice of lime in a refreshing summer drink may be etiologic,4,5 or berloque dermatitis, caused by exposure to perfumes containing bergapten (5-methoxypsoralen).6,7 Phytophotodermatitis may develop when phototoxic agents such as furocoumarins, which protect plants from fungal pathogens, and psoralens are applied to the skin followed by exposure to UV light, more specifically in the UVA range of 320 to 400 nm. Thus, these chemicals produce a phototoxic rather than photoallergic reaction, leading to cellular damage. Furocoumarins and psoralens often are found in plants such as celery and figs as well as in citrus fruits such as limes, lemons, and grapefruits. Exposure may be cryptic, as the patient may not consider or mention the eruption as possibly caused by activities such as soaking one’s feet in a folk remedy containing fig leaves.7,8 Once these phototoxic agents come in contact with the skin, the symptoms of PPD may arise within 24 hours of exposure, beginning as an acute dermatitis with erythema, edema, vesicles, or bullae accompanied by pain and itching.
Etiology
Phytophotodermatitis is caused by exposure to several different types of plants, including Ficus carica (common fig), the genus Citrus (eg, lime, lemon), or Pastina sativa (wild parsnip). Each of these contain furocoumarins and psoralens—phototoxic agents that cause cellular damage with epidermal necrosis and resultant pain when the skin is exposed to UVA light.1-4 There are 2 types of photochemical reactions in PPD: type I reactions occur in the absence of oxygen, whereas oxygen is present in type II reactions. Both damage cell membranes and DNA, which then results in DNA interstrand cross-linking between the psoralen furan ring and the thymine or cytosine of DNA, activating arachidonic acid metabolic pathways to produce cell death.1
Epidemiology
The incidence of PPD is unknown due to the high variability of reactions in individuals spanning from children to the elderly. It can be caused by many different wild and domestic plants in many areas of the world and can affect any individual regardless of age, race, gender, or ethnicity. Some individuals may be affected by hyperpigmentation without prominent inflammation.8 Diagnosis of PPD can be challenging, and an occupation and recreational history of exposure or recent travel with possible contact with plants may be required.
Occupational Dermatitis
Recreational Dermatitis
Phytophotodermatitis may be caused by exposure to phototoxic agents during leisure activities. Recreational exposure can occur almost anywhere, including in the kitchen, backyard, park, or woods, as well as at the beach. One notable culprit in recreational PPD is cooking with limes, parsley, or parsnips—plants that often are employed as garnishes in dishes, allowing early exposure of juices on the hands. Individuals who garden recreationally should be aware of ornamental plants such as hogweed and figs, which are notorious for causing PPD.13 Children’s camp counselors should have knowledge of PPD, as children have considerable curiosity and may touch or play with attractive plants such as hogweed. Children enjoying sports in parks can accidentally fall onto or be exposed to wild parsnip or hogweed growing nearby and wake up the next day with erythema and burning.14 Photoprotection is important, but sunscreens containing carrot extract can produce PPD.15 Widespread PPD over 80% of the body surface area due to sunbathing after applying fig leaf tea as a tanning agent has been described.16 Eating figs does not cause photosensitization unless the juice is smeared onto the skin. Margarita dermatitis and “Mexican beer dermatitis” can occur due to limes and other citrus fruits being used as ingredients in summer drinks.5 Similarly, preparing sangria may produce PPD from lime and lemon juices.17 In one report, hiking in Corsica resulted in PPD following incidental contact with the endemic plant Peucedanum paniculatum.18
Perfume (Berloque) Dermatitis
Perfume dermatitis, or berloque dermatitis, is a type of PPD for which the name is derived from the German word berlock or the French word berloque meaning trinket or charm; it was first described in 1925 by Rosenthal7 with regard to pendantlike streaks of pigmentation on the neck, face, arms, or trunk. The dermatitis develops due to bergapten, a component of bergamot oil, which is derived from the rind of Citrus bergamia. Many perfumes contain bergamot oil, but the incidence of this condition has been diminished due to use of artificial bergamot oil.6
Clinical Manifestation
Phytophotodermatitis is first evident as erythematous patches that appear within 24 hours of initial exposure to a phototoxic agent and UVA light, sometimes with a burning sensation. Solar exposure within 48 hours of sufficient plant exposure is required. Perfuse sweating may enhance the reaction.19 Rarely, it first may be seen with the sudden appearance of
Differential Diagnosis
Phytophotodermatitis may resemble other types of dermatitis, particularly other forms of contact dermatitis such poison ivy, and occasionally other environmental simulants such as jellyfish stings.1-6,20,21 Photosensitizing disorders including porphyria cutanea tarda, pseudoporphyria, and lupus erythematosus must be distinguished from PPD.22-24 Photosensitizing medications such tetracyclines, thiazide diuretics, sulfonamides, griseofulvin, and sulfonylureas should be considered. Airborne contact dermatitis may resemble PPD, as when poison ivy is burned and is exposed to the skin in sites of airborne contact.20 Excessive solar exposure is popular, particularly among adolescents, so sunburn and sunburnlike reactions can be noteworthy.25,26
Treatment
Phytophotodermatitis can be treated with topical steroids, sometimes adding an oral antihistamine, and occasionally oral steroids.2-4 Localized pain or a burning sensation should respond to therapy. Alternatively, a cold compress applied to the skin can relieve the pain and pruritus, and the burn can be debrided and dressed daily with silver sulfadiazine plus an oral nonsteroidal anti-inflammatory drug. This eruption should be self-limited as long as it is recognized early and the cause avoided. Management of acute exposure includes prompt application of soap and water and avoidance of UV light exposure for 48 to 72 hours to prevent psoralen photoactivation.
Because PPD is essentially a chemical burn, a burn protocol and possible referral to a burn center may be needed, whether the reaction is acute or widespread.11,12,14,27,28 Surgical debridement and skin grafting rarely may be mandated.14 Postinflammatory hyperpigmentation may ensue as the dermatitis resolves but is not common.
The best approach for PPD is prevention (Figure 2). Individuals who are at risk should be aware of their surroundings and potential plants of concern and employ personal protective equipment to shield the skin from plant sap, which should be promptly removed if it comes in contact with the skin.
Phytophotodermatitis (PPD) is a nonallergic contact dermatitis and thus is independent of the immune system, so prior sensitization is not required.1-3 It sometimes is known by colorful names such as margarita photodermatitis, in which a slice of lime in a refreshing summer drink may be etiologic,4,5 or berloque dermatitis, caused by exposure to perfumes containing bergapten (5-methoxypsoralen).6,7 Phytophotodermatitis may develop when phototoxic agents such as furocoumarins, which protect plants from fungal pathogens, and psoralens are applied to the skin followed by exposure to UV light, more specifically in the UVA range of 320 to 400 nm. Thus, these chemicals produce a phototoxic rather than photoallergic reaction, leading to cellular damage. Furocoumarins and psoralens often are found in plants such as celery and figs as well as in citrus fruits such as limes, lemons, and grapefruits. Exposure may be cryptic, as the patient may not consider or mention the eruption as possibly caused by activities such as soaking one’s feet in a folk remedy containing fig leaves.7,8 Once these phototoxic agents come in contact with the skin, the symptoms of PPD may arise within 24 hours of exposure, beginning as an acute dermatitis with erythema, edema, vesicles, or bullae accompanied by pain and itching.
Etiology
Phytophotodermatitis is caused by exposure to several different types of plants, including Ficus carica (common fig), the genus Citrus (eg, lime, lemon), or Pastina sativa (wild parsnip). Each of these contain furocoumarins and psoralens—phototoxic agents that cause cellular damage with epidermal necrosis and resultant pain when the skin is exposed to UVA light.1-4 There are 2 types of photochemical reactions in PPD: type I reactions occur in the absence of oxygen, whereas oxygen is present in type II reactions. Both damage cell membranes and DNA, which then results in DNA interstrand cross-linking between the psoralen furan ring and the thymine or cytosine of DNA, activating arachidonic acid metabolic pathways to produce cell death.1
Epidemiology
The incidence of PPD is unknown due to the high variability of reactions in individuals spanning from children to the elderly. It can be caused by many different wild and domestic plants in many areas of the world and can affect any individual regardless of age, race, gender, or ethnicity. Some individuals may be affected by hyperpigmentation without prominent inflammation.8 Diagnosis of PPD can be challenging, and an occupation and recreational history of exposure or recent travel with possible contact with plants may be required.
Occupational Dermatitis
Recreational Dermatitis
Phytophotodermatitis may be caused by exposure to phototoxic agents during leisure activities. Recreational exposure can occur almost anywhere, including in the kitchen, backyard, park, or woods, as well as at the beach. One notable culprit in recreational PPD is cooking with limes, parsley, or parsnips—plants that often are employed as garnishes in dishes, allowing early exposure of juices on the hands. Individuals who garden recreationally should be aware of ornamental plants such as hogweed and figs, which are notorious for causing PPD.13 Children’s camp counselors should have knowledge of PPD, as children have considerable curiosity and may touch or play with attractive plants such as hogweed. Children enjoying sports in parks can accidentally fall onto or be exposed to wild parsnip or hogweed growing nearby and wake up the next day with erythema and burning.14 Photoprotection is important, but sunscreens containing carrot extract can produce PPD.15 Widespread PPD over 80% of the body surface area due to sunbathing after applying fig leaf tea as a tanning agent has been described.16 Eating figs does not cause photosensitization unless the juice is smeared onto the skin. Margarita dermatitis and “Mexican beer dermatitis” can occur due to limes and other citrus fruits being used as ingredients in summer drinks.5 Similarly, preparing sangria may produce PPD from lime and lemon juices.17 In one report, hiking in Corsica resulted in PPD following incidental contact with the endemic plant Peucedanum paniculatum.18
Perfume (Berloque) Dermatitis
Perfume dermatitis, or berloque dermatitis, is a type of PPD for which the name is derived from the German word berlock or the French word berloque meaning trinket or charm; it was first described in 1925 by Rosenthal7 with regard to pendantlike streaks of pigmentation on the neck, face, arms, or trunk. The dermatitis develops due to bergapten, a component of bergamot oil, which is derived from the rind of Citrus bergamia. Many perfumes contain bergamot oil, but the incidence of this condition has been diminished due to use of artificial bergamot oil.6
Clinical Manifestation
Phytophotodermatitis is first evident as erythematous patches that appear within 24 hours of initial exposure to a phototoxic agent and UVA light, sometimes with a burning sensation. Solar exposure within 48 hours of sufficient plant exposure is required. Perfuse sweating may enhance the reaction.19 Rarely, it first may be seen with the sudden appearance of
Differential Diagnosis
Phytophotodermatitis may resemble other types of dermatitis, particularly other forms of contact dermatitis such poison ivy, and occasionally other environmental simulants such as jellyfish stings.1-6,20,21 Photosensitizing disorders including porphyria cutanea tarda, pseudoporphyria, and lupus erythematosus must be distinguished from PPD.22-24 Photosensitizing medications such tetracyclines, thiazide diuretics, sulfonamides, griseofulvin, and sulfonylureas should be considered. Airborne contact dermatitis may resemble PPD, as when poison ivy is burned and is exposed to the skin in sites of airborne contact.20 Excessive solar exposure is popular, particularly among adolescents, so sunburn and sunburnlike reactions can be noteworthy.25,26
Treatment
Phytophotodermatitis can be treated with topical steroids, sometimes adding an oral antihistamine, and occasionally oral steroids.2-4 Localized pain or a burning sensation should respond to therapy. Alternatively, a cold compress applied to the skin can relieve the pain and pruritus, and the burn can be debrided and dressed daily with silver sulfadiazine plus an oral nonsteroidal anti-inflammatory drug. This eruption should be self-limited as long as it is recognized early and the cause avoided. Management of acute exposure includes prompt application of soap and water and avoidance of UV light exposure for 48 to 72 hours to prevent psoralen photoactivation.
Because PPD is essentially a chemical burn, a burn protocol and possible referral to a burn center may be needed, whether the reaction is acute or widespread.11,12,14,27,28 Surgical debridement and skin grafting rarely may be mandated.14 Postinflammatory hyperpigmentation may ensue as the dermatitis resolves but is not common.
The best approach for PPD is prevention (Figure 2). Individuals who are at risk should be aware of their surroundings and potential plants of concern and employ personal protective equipment to shield the skin from plant sap, which should be promptly removed if it comes in contact with the skin.
- Zhang R, Zhu W. Phytophotodermatitis due to Chinese herbal medicine decoction. Indian J Dermatol. 2011;56:329-331.
- Harshman J, Quan Y, Hsiang D. Phytophotodermatitis: rash with many faces. Can Fam Physician. 2017;63:938-940.
- Imen MS, Ahmadabadi A, Tavousi SH, et al. The curious cases of burn by fig tree leaves. Indian J Dermatol. 2019;64:71-73.
- Hankinson A, Lloyd B, Alweis R. Lime-induced phytophotodermatitis [published online September 29, 2014]. J Community Hosp Intern Med Perspect. doi:10.3402/jchimp.v4.25090
- Abramowitz AI, Resnik KS, Cohen KR. Margarita photodermatitis. N Engl J Med. 2013;328:891.
- Quaak MS, Martens H, Hassing RJ, et al. The sunny side of lime. J Travel Med. 2012;19:327-328.
- Rosenthal O. Berloque dermatitis: Berliner Dermatologische Gesellschaft. Dermatol Zeitschrift. 1925;42:295.
- Choi JY, Hwang S, Lee SH, et al. Asymptomatic hyperpigmentation without preceding inflammation as a clinical feature of citrus fruits–induced phytophotodermatitis. Ann Dermatol. 2018;30:75-78.
- Wynn P, Bell S. Phytophotodermatitis in grounds operatives. Occup Med (Lond). 2005;55:393-395.
- Klimaszyk P, Klimaszyk D, Piotrowiak M, et al. Unusual complications after occupational exposure to giant hogweed (Heracleum mantegazzianum): a case report. Int J Occup Med Environ Health. 2014;27:141-144.
- Downs JW, Cumpston KL, Feldman MJ. Giant hogweed phytophotodermatitis. Clin Toxicol (Phila). 2019;57:822-823.
- Maso MJ, Ruszkowski AM, Bauerle J, et al. Celery phytophotodermatitis in a chef. Arch Dermatol. 1991;127:912-913.
- Derraik JG, Rademaker M. Phytophotodermatitis caused by contact with a fig tree (Ficus carica). New Zealand Med J. 2007;120:U2720.
- Chan JC, Sullivan PJ, O’Sullivan MJ, et al. Full thickness burn caused by exposure to giant hogweed: delayed presentation, histological features and surgical management. J Plast Reconstr Aesthet Surg. 2011;64:128-130.
- Bosanac SS, Clark AK, Sivamani RK. Phytophotodermatitis related to carrot extract–containing sunscreen. Dermatol Online J. 2018;24:1-3.
- Sforza M, Andjelkov K, Zaccheddu R. Severe burn on 81% of body surface after sun tanning. Ulus Travma Acil Cerrahi Derg. 2013;19:383-384.
- Mioduszewski M, Beecker J. Phytophotodermatitis from making sangria: a phototoxic reaction to lime and lemon juice. CMAJ. 2015;187:756.
- Torrents R, Schmitt C, Domangé B, et al. Phytophotodermatitis with Peucedanum paniculatum: an endemic species to Corsica. Clin Toxicol (Phila). 2019;57:68-69.
- Sarhane KA, Ibrahim A, Fagan SP, et al. Phytophotodermatitis. Eplasty. 2013;13:ic57.
- DeLeo VA, Suarez SM, Maso MJ. Photoallergic contact dermatitis. results of photopatch testing in New York, 1985 to 1990. Arch Dermatol. 1992;128:1513-1518.
- Kimyon RS, Warshaw EM. Airborne allergic contact dermatitis: management and responsible allergens on the American Contact Dermatitis Society Core Series. Dermatitis. 2019;30:106-115.
- Miteva L, Broshtilova V, Schwartz RA. Unusual clinical manifestations of chronic discoid lupus erythematosus. Serbian J Dermatol Venereol. 2014;6:69-72.
- Handler NS, Handler MZ, Stephany MP, et al. Porphyria cutanea tarda: an intriguing genetic disease and marker. Int J Dermatol. 2017;56:E106-E117.
- Papadopoulos AJ, Schwartz RA, Fekete Z, et al. Pseudoporphyria: an atypical variant resembling toxic epidermal necrolysis. J Cutan Med Surg. 2001;5:479-485.
- Jasterzbski TJ, Janniger EJ, Schwartz RA. Adolescent tanning practices: understanding the popularity of excessive ultraviolet light exposure. In: Or
anje A, Al-Mutairi N, Shwayder T, eds. Practical Pediatric Dermatology. Controversies in Diagnosis and Treatment. Springer Verlag; 2016:177-185. - Lai YC, Janniger EJ, Schwartz RA. Solar protection policy in school children: proposals for progress. In: Oranje A, Al-Mutairi N, Shwayder T, eds. Practical Pediatric Dermatology. Controversies in Diagnosis and Treatment. Springer Verlag; 2016:165-176.
- Lagey K, Duinslaeger L, Vanderkelen A. Burns induced by plants. Burns. 1995;21:542-543.
- Redgrave N, Solomon J. Severe phytophotodermatitis from fig sap: a little known phenomenon. BMJ Case Rep. 2021;14:e238745.
- Zhang R, Zhu W. Phytophotodermatitis due to Chinese herbal medicine decoction. Indian J Dermatol. 2011;56:329-331.
- Harshman J, Quan Y, Hsiang D. Phytophotodermatitis: rash with many faces. Can Fam Physician. 2017;63:938-940.
- Imen MS, Ahmadabadi A, Tavousi SH, et al. The curious cases of burn by fig tree leaves. Indian J Dermatol. 2019;64:71-73.
- Hankinson A, Lloyd B, Alweis R. Lime-induced phytophotodermatitis [published online September 29, 2014]. J Community Hosp Intern Med Perspect. doi:10.3402/jchimp.v4.25090
- Abramowitz AI, Resnik KS, Cohen KR. Margarita photodermatitis. N Engl J Med. 2013;328:891.
- Quaak MS, Martens H, Hassing RJ, et al. The sunny side of lime. J Travel Med. 2012;19:327-328.
- Rosenthal O. Berloque dermatitis: Berliner Dermatologische Gesellschaft. Dermatol Zeitschrift. 1925;42:295.
- Choi JY, Hwang S, Lee SH, et al. Asymptomatic hyperpigmentation without preceding inflammation as a clinical feature of citrus fruits–induced phytophotodermatitis. Ann Dermatol. 2018;30:75-78.
- Wynn P, Bell S. Phytophotodermatitis in grounds operatives. Occup Med (Lond). 2005;55:393-395.
- Klimaszyk P, Klimaszyk D, Piotrowiak M, et al. Unusual complications after occupational exposure to giant hogweed (Heracleum mantegazzianum): a case report. Int J Occup Med Environ Health. 2014;27:141-144.
- Downs JW, Cumpston KL, Feldman MJ. Giant hogweed phytophotodermatitis. Clin Toxicol (Phila). 2019;57:822-823.
- Maso MJ, Ruszkowski AM, Bauerle J, et al. Celery phytophotodermatitis in a chef. Arch Dermatol. 1991;127:912-913.
- Derraik JG, Rademaker M. Phytophotodermatitis caused by contact with a fig tree (Ficus carica). New Zealand Med J. 2007;120:U2720.
- Chan JC, Sullivan PJ, O’Sullivan MJ, et al. Full thickness burn caused by exposure to giant hogweed: delayed presentation, histological features and surgical management. J Plast Reconstr Aesthet Surg. 2011;64:128-130.
- Bosanac SS, Clark AK, Sivamani RK. Phytophotodermatitis related to carrot extract–containing sunscreen. Dermatol Online J. 2018;24:1-3.
- Sforza M, Andjelkov K, Zaccheddu R. Severe burn on 81% of body surface after sun tanning. Ulus Travma Acil Cerrahi Derg. 2013;19:383-384.
- Mioduszewski M, Beecker J. Phytophotodermatitis from making sangria: a phototoxic reaction to lime and lemon juice. CMAJ. 2015;187:756.
- Torrents R, Schmitt C, Domangé B, et al. Phytophotodermatitis with Peucedanum paniculatum: an endemic species to Corsica. Clin Toxicol (Phila). 2019;57:68-69.
- Sarhane KA, Ibrahim A, Fagan SP, et al. Phytophotodermatitis. Eplasty. 2013;13:ic57.
- DeLeo VA, Suarez SM, Maso MJ. Photoallergic contact dermatitis. results of photopatch testing in New York, 1985 to 1990. Arch Dermatol. 1992;128:1513-1518.
- Kimyon RS, Warshaw EM. Airborne allergic contact dermatitis: management and responsible allergens on the American Contact Dermatitis Society Core Series. Dermatitis. 2019;30:106-115.
- Miteva L, Broshtilova V, Schwartz RA. Unusual clinical manifestations of chronic discoid lupus erythematosus. Serbian J Dermatol Venereol. 2014;6:69-72.
- Handler NS, Handler MZ, Stephany MP, et al. Porphyria cutanea tarda: an intriguing genetic disease and marker. Int J Dermatol. 2017;56:E106-E117.
- Papadopoulos AJ, Schwartz RA, Fekete Z, et al. Pseudoporphyria: an atypical variant resembling toxic epidermal necrolysis. J Cutan Med Surg. 2001;5:479-485.
- Jasterzbski TJ, Janniger EJ, Schwartz RA. Adolescent tanning practices: understanding the popularity of excessive ultraviolet light exposure. In: Or
anje A, Al-Mutairi N, Shwayder T, eds. Practical Pediatric Dermatology. Controversies in Diagnosis and Treatment. Springer Verlag; 2016:177-185. - Lai YC, Janniger EJ, Schwartz RA. Solar protection policy in school children: proposals for progress. In: Oranje A, Al-Mutairi N, Shwayder T, eds. Practical Pediatric Dermatology. Controversies in Diagnosis and Treatment. Springer Verlag; 2016:165-176.
- Lagey K, Duinslaeger L, Vanderkelen A. Burns induced by plants. Burns. 1995;21:542-543.
- Redgrave N, Solomon J. Severe phytophotodermatitis from fig sap: a little known phenomenon. BMJ Case Rep. 2021;14:e238745.
Practice Points
- Phytophotodermatitis (PPD) can be both an occupational and recreational dermatosis.
- Phytophotodermatitis is a nonallergic contact dermatitis and thus is independent of the immune system, so prior sensitization is not required.
- Individuals who work with plants should be aware of PPD and methods of prevention.
- Phytophotodermatitis may be evident only as asymptomatic hyperpigmentation.
Erethism Mercurialis and Reactions to Elemental Mercury
Evidence of human exposure to mercury dates as far back as the Egyptians in 1500
Mercury release in the environment primarily is a function of human activity, including coal-fired power plants, residential heating, and mining.9,10 Mercury from these sources is commonly found in the sediment of lakes and bays, where it is enzymatically converted to methylmercury by aquatic microorganisms; subsequent food chain biomagnification results in elevated mercury levels in apex predators. Substantial release of mercury into the environment also can be attributed to health care facilities from their use of thermometers containing 0.5 to 3 g of elemental mercury,11 blood pressure monitors, and medical waste incinerators.5
Mercury has been reported as the second most common cause of heavy metal poisoning after lead.12 Standards from the US Food and Drug Administration dictate that methylmercury levels in fish and wheat products must not exceed 1 ppm.13 Most plant and animal food sources contain methylmercury at levels between 0.0001 and 0.01 ppm; mercury concentrations are especially high in tuna, averaging 0.4 ppm, while larger predatory fish contain levels in excess of 1 ppm.14 The use of mercury-containing cosmetic products also presents a substantial exposure risk to consumers.5,10 In one study, 3.3% of skin-lightening creams and soaps purchased within the United States contained concentrations of mercury exceeding 1000 ppm.15
We describe a case of mercury toxicity resulting from intentional injection of liquid mercury into the right antecubital fossa in a suicide attempt.
Case Report
A 31-year-old woman presented to the family practice center for evaluation of a firm stained area on the skin of the right arm. She reported increasing anxiety, depression, tremors, irritability, and difficulty concentrating over the last 6 months. She denied headache and joint or muscle pain. Four years earlier, she had broken apart a thermometer and injected approximately 0.7 mL of its contents into the right arm in a suicide attempt. She intended to inject the thermometer’s contents directly into a vein, but the material instead entered the surrounding tissue. She denied notable pain or itching overlying the injection site. Her medications included aripiprazole and buspirone. She noted that she smoked half a pack of cigarettes per day and had a history of methamphetamine abuse. She was homeless and unemployed. Physical examination revealed an anxious tremulous woman with an erythematous to bluish gray, firm plaque on the right antecubital fossa (Figure 1). There were no notable tremors and no gait disturbance.
Her blood mercury level was greater than 100 µg/L and urine mercury was 477 µg/g (reference ranges, 1–8 μg/L and 4–5 μg/L, respectively). A radiograph of the right elbow area revealed scattered punctate foci of increased density within or overlying the anterolateral elbow soft tissues. She was diagnosed with mercury granuloma causing chronic mercury elevation. She underwent excision of the granuloma (Figure 2) with endovascular surgery via an elliptical incision. The patient was subsequently lost to follow-up.
Comment
Elemental mercury is a silver liquid at room temperature that spontaneously evaporates to form mercury vapor, an invisible, odorless, toxic gas. Accidental cutaneous exposure typically is safely managed by washing exposed skin with soap and water,16 though there is a potential risk for systemic absorption, especially when the skin is inflamed. When metallic mercury is subcutaneously injected, it is advised to promptly excise all subcutaneous areas containing mercury, regardless of any symptoms of systemic toxicity. Patients should subsequently be monitored for signs of both central nervous system (CNS) and renal deficits, undergo chelation therapy when systemic effects are apparent, and finally receive psychiatric consultation and treatment when necessary.17
Inorganic mercury compounds are formed when elemental mercury combines with sulfur or oxygen and often take the form of mercury salts, which appear as white crystals.16 These salts occur naturally in the environment and are used in pesticides, antiseptics, and skin-lightening creams and soaps.18
Methylmercury is a highly toxic, organic compound that is capable of crossing the placental and blood-brain barriers. It is the most common organic mercury compound found in the environment.16 Most humans have trace amounts of methylmercury in their bodies, typically as a result of consuming seafood.5
Exposure to mercury most commonly occurs through chronic consumption of methylmercury in seafood or acute inhalation of elemental mercury vapors.9 Iatrogenic cases of mercury exposure via injection also have been reported in the literature, including a case resulting in acute poisoning due to peritoneal lavage with mercury bichloride.19 Acute mercury-induced pulmonary damage typically resolves completely. However, there have been reported cases of exposure progressing to interstitial emphysema, pneumatocele, pneumothorax, pneumomediastinum, interstitial fibrosis, and chronic respiratory insufficiency, with examples of fatal acute respiratory distress syndrome being reported.5,16,20 Although individuals who inhale mercury vapors initially may be unaware of exposure due to little upper airway irritation, symptoms following an initial acute exposure may include ptyalism, a metallic taste, dysphagia, enteritis, diarrhea, nausea, renal damage, and CNS effects.16 Additionally, exposure may lead to confusion with signs and symptoms of metal fume fever, including shortness of breath, pleuritic chest pain, stomatitis, lethargy, and vomiting.20
Chronic exposure to mercury vapor can result in accumulation of mercury in the body, leading to neuropsychiatric, dermatologic, oropharyngeal, and renal manifestations. Sore throat, fever, headache, fatigue, dyspnea, chest pain, and pneumonitis are common.16 Typically, low-level exposure to elemental mercury does not lead to long-lasting health effects. However, individuals exposed to high-level elemental mercury vapors may require hospitalization. Treatment of acute mercury poisoning consists of removing the source of exposure, followed by cardiopulmonary support.16
Specific assays for mercury levels in blood and urine are useful to assess the level of exposure and risk to the patient. Blood mercury concentrations of 20 µg/L or below are considered within reference range; however, once blood and urine concentrations of mercury exceed 100 µg/L, clinical signs of acute mercury poisoning typically manifest.21 Chest radiographs can reveal pulmonary damage, while complete blood cell count, metabolic panel, and urinalysis can assess damage to other organs. Neuropsychiatric testing and nerve conduction studies may provide objective evidence of CNS toxicity. Assays for N-acetyl-β-D-glucosaminidase can provide an indication of early renal tubular dysfunction.16
Elemental mercury is not absorbed from the gastrointestinal tract, posing minimal risk for acute toxicity from ingestion. Generally, less than 10% of ingested inorganic mercury is absorbed from the gut, while elemental mercury is nonabsorbable.10 If an individual ingests a large amount of mercury, it may persist in the gastrointestinal tract for an extended period. Mercury is radiopaque, and abdominal radiographs should be obtained in all cases of ingestion.16
Mercury is toxic to the CNS and peripheral nervous system, resulting in erethism mercurialis, a constellation of neuropsychologic signs and symptoms including restlessness, irritability, insomnia, emotional lability, difficulty concentrating, and impaired memory. In severe cases, delirium and psychosis may develop. Other CNS effects include tremors, paresthesia, dysarthria, neuromuscular changes, headaches, polyneuropathy, and cerebellar ataxia, as well as ophthalmologic and audiologic impairment.5,16
Upon inhalation exposure, patients with respiratory concerns should be given oxygen. Bronchospasms are treated with bronchodilators; however, if multiple chemical exposures are suspected, bronchial-sensitizing agents may pose additional risks. Corticosteroids and antibiotics have been recommended for treatment of chemical pneumonitis, but their efficacy has not been substantiated.16
Skin reactions associated with skin contact to elemental mercury are rare. However, hives and dermatitis have been observed following accidental contact with inorganic mercury compounds.5 Manifestation in children chronically exposed to mercury includes a nonallergic hypersensitivity (acrodynia),5,17 which is characterized by pain and dusky pink discoloration in the hands and feet, most often seen in children chronically exposed to mercury absorbed from vapor inhalation or cutaneous exposure.16
Renal conditions associated with acute inhalation of elemental mercury vapor include proteinuria, nephrotic syndrome, temporary tubular dysfunction, acute tubular necrosis, and oliguric renal failure.16 Chronic exposure to inorganic mercury compounds also has been reported to cause renal damage.5 Chelation therapy should be performed for any symptomatic patient with a clear history of acute elemental mercury exposure.16 The most frequently used chelation agent in cases of acute inorganic mercury exposures is dimercaprol. In rare cases of mercury intoxication, hemodialysis is required in the treatment of renal failure and to expedite removal of dimercaprol-mercury complexes.16
Cardiovascular symptoms associated with acute inhalation of high levels of elemental mercury include tachycardia and hypertension.16 Increases in blood pressure, palpitations, and heart rate also have been observed in instances of acute elemental mercury exposure. Studies show that exposure to mercury increases both the risk for acute myocardial infarction as well as death from coronary heart and cardiovascular diseases.5
Conclusion
Mercury poisoning presents with varied neuropsychologic signs and symptoms. Our case provides insight into a unique route of exposure for mercury toxicity. In addition to the unusual presentation of a mercury granuloma, our case illustrates how surgical techniques can aid in removal of cutaneous reservoirs in the setting of percutaneous exposure.
- History of mercury. Government of Canada website. Modified April 26, 2010. Accessed March 11, 2021. https://www.canada.ca/en/environment-climate-change/services/pollutants/mercury-environment/about/history.html
- Dartmouth Toxic Metals Superfund Research Program website. Accessed March 11, 2021. https://sites.dartmouth.edu/toxmetal/
- Norn S, Permin H, Kruse E, et al. Mercury—a major agent in the history of medicine and alchemy [in Danish]. Dan Medicinhist Arbog. 2008;36:21-40.
- Waldron HA. Did the Mad Hatter have mercury poisoning? Br Med J (Clin Res Ed). 1983;287:1961.
- Poulin J, Gibb H. Mercury: assessing the environmental burden of disease at national and local levels. WHO Environmental Burden of Disease Series No. 16. World Health Organization; 2008.
- Charcot JM. Clinical lectures of the diseases of the nervous system. In: Kinnier Wilson SA. The Landmark Library of Neurology and Neurosurgery. Gryphon Editions; 1994:186.
- Kinnier Wilson SA. Neurology. In: Kinnier Wilson SA. The Landmark Library of Neurology and Neurosurgery. Gryphon Editions; 1994:739-740.
- Harada M. Minamata disease: methylmercury poisoning in Japan caused by environmental pollution. Crit Rev Toxicol. 1995;25:1-24.
- Mercury and health. World Health Organization website. Updated March 31, 2017. Accessed March 12, 2021. http://www.whoint/mediacentre/factsheets/fs361/en/
- Olson DA. Mercury toxicity. Updated November 5, 2018. Accessed March 12, 2021.http://emedicine.medscape.com/article/1175560-overview
- Mercury thermometers. Environmental Protection Agency website. Updated June 26, 2018. https://www.epa.gov/mercury/mercury-thermometers
- Jao-Tan C, Pope E. Cutaneous poisoning syndromes in children: a review. Curr Opin Pediatr. 2006;18:410-416.
- US Department of Health and Human Services: Public Health Service Agency for Toxic Substances and Disease Registry. Toxicological profile for mercury: regulations and advisories. Published March 1999. Accessed March 23, 2021. https://www.atsdr.cdc.gov/toxprofiles/tp46.pdf
- US Food and Drug Administration. Mercury levels in commercial fish and shellfish (1990-2012). Updated October 25, 2017. Accessed March 16, 2021. https://www.fda.gov/food/metals-and-your-food/mercury-levels-commercial-fish-and-shellfish-1990-2012
- Hamann CR, Boonchai W, Wen L, et al. Spectrometric analysis of mercury content in 549 skin-lightening products: is mercury toxicity a hidden global health hazard? J Am Acad Dermatol. 2014;70:281-287.e3.
- Mercury. Managing Hazardous Materials Incidents. Agency for Toxic Substances and Disease Registry website. Accessed March 16, 2021. https://www.atsdr.cdc.gov/MHMI/mmg46.pdf
- Krohn IT, Solof A, Mobini J, et al. Subcutaneous injection of metallic mercury. JAMA. 1980;243:548-549.
- Lai O, Parsi KK, Wu D, et al. Mercury toxicity presenting acrodynia and a papulovesicular eruption in a 5-year-old girl. Dermatol Online J. 2016;16;22:13030/qt6444r7nc.
- Dolianiti M, Tasiopoulou K, Kalostou A, et al. Mercury bichloride iatrogenic poisoning: a case report. J Clin Toxicol. 2016;6:2. doi:10.4172/2161-0495.1000290
- Broussard LA, Hammett-Stabler CA, Winecker RE, et al. The toxicology of mercury. Lab Med. 2002;33:614-625. doi:10.1309/5HY1-V3NE-2LFL-P9MT
- Byeong-Jin Y, Byoung-Gwon K, Man-Joong J, et al. Evaluation of mercury exposure levels, clinical diagnosis and treatment for mercury intoxication. Ann Occup Environ Med. 2016;28:5.
Evidence of human exposure to mercury dates as far back as the Egyptians in 1500
Mercury release in the environment primarily is a function of human activity, including coal-fired power plants, residential heating, and mining.9,10 Mercury from these sources is commonly found in the sediment of lakes and bays, where it is enzymatically converted to methylmercury by aquatic microorganisms; subsequent food chain biomagnification results in elevated mercury levels in apex predators. Substantial release of mercury into the environment also can be attributed to health care facilities from their use of thermometers containing 0.5 to 3 g of elemental mercury,11 blood pressure monitors, and medical waste incinerators.5
Mercury has been reported as the second most common cause of heavy metal poisoning after lead.12 Standards from the US Food and Drug Administration dictate that methylmercury levels in fish and wheat products must not exceed 1 ppm.13 Most plant and animal food sources contain methylmercury at levels between 0.0001 and 0.01 ppm; mercury concentrations are especially high in tuna, averaging 0.4 ppm, while larger predatory fish contain levels in excess of 1 ppm.14 The use of mercury-containing cosmetic products also presents a substantial exposure risk to consumers.5,10 In one study, 3.3% of skin-lightening creams and soaps purchased within the United States contained concentrations of mercury exceeding 1000 ppm.15
We describe a case of mercury toxicity resulting from intentional injection of liquid mercury into the right antecubital fossa in a suicide attempt.
Case Report
A 31-year-old woman presented to the family practice center for evaluation of a firm stained area on the skin of the right arm. She reported increasing anxiety, depression, tremors, irritability, and difficulty concentrating over the last 6 months. She denied headache and joint or muscle pain. Four years earlier, she had broken apart a thermometer and injected approximately 0.7 mL of its contents into the right arm in a suicide attempt. She intended to inject the thermometer’s contents directly into a vein, but the material instead entered the surrounding tissue. She denied notable pain or itching overlying the injection site. Her medications included aripiprazole and buspirone. She noted that she smoked half a pack of cigarettes per day and had a history of methamphetamine abuse. She was homeless and unemployed. Physical examination revealed an anxious tremulous woman with an erythematous to bluish gray, firm plaque on the right antecubital fossa (Figure 1). There were no notable tremors and no gait disturbance.
Her blood mercury level was greater than 100 µg/L and urine mercury was 477 µg/g (reference ranges, 1–8 μg/L and 4–5 μg/L, respectively). A radiograph of the right elbow area revealed scattered punctate foci of increased density within or overlying the anterolateral elbow soft tissues. She was diagnosed with mercury granuloma causing chronic mercury elevation. She underwent excision of the granuloma (Figure 2) with endovascular surgery via an elliptical incision. The patient was subsequently lost to follow-up.
Comment
Elemental mercury is a silver liquid at room temperature that spontaneously evaporates to form mercury vapor, an invisible, odorless, toxic gas. Accidental cutaneous exposure typically is safely managed by washing exposed skin with soap and water,16 though there is a potential risk for systemic absorption, especially when the skin is inflamed. When metallic mercury is subcutaneously injected, it is advised to promptly excise all subcutaneous areas containing mercury, regardless of any symptoms of systemic toxicity. Patients should subsequently be monitored for signs of both central nervous system (CNS) and renal deficits, undergo chelation therapy when systemic effects are apparent, and finally receive psychiatric consultation and treatment when necessary.17
Inorganic mercury compounds are formed when elemental mercury combines with sulfur or oxygen and often take the form of mercury salts, which appear as white crystals.16 These salts occur naturally in the environment and are used in pesticides, antiseptics, and skin-lightening creams and soaps.18
Methylmercury is a highly toxic, organic compound that is capable of crossing the placental and blood-brain barriers. It is the most common organic mercury compound found in the environment.16 Most humans have trace amounts of methylmercury in their bodies, typically as a result of consuming seafood.5
Exposure to mercury most commonly occurs through chronic consumption of methylmercury in seafood or acute inhalation of elemental mercury vapors.9 Iatrogenic cases of mercury exposure via injection also have been reported in the literature, including a case resulting in acute poisoning due to peritoneal lavage with mercury bichloride.19 Acute mercury-induced pulmonary damage typically resolves completely. However, there have been reported cases of exposure progressing to interstitial emphysema, pneumatocele, pneumothorax, pneumomediastinum, interstitial fibrosis, and chronic respiratory insufficiency, with examples of fatal acute respiratory distress syndrome being reported.5,16,20 Although individuals who inhale mercury vapors initially may be unaware of exposure due to little upper airway irritation, symptoms following an initial acute exposure may include ptyalism, a metallic taste, dysphagia, enteritis, diarrhea, nausea, renal damage, and CNS effects.16 Additionally, exposure may lead to confusion with signs and symptoms of metal fume fever, including shortness of breath, pleuritic chest pain, stomatitis, lethargy, and vomiting.20
Chronic exposure to mercury vapor can result in accumulation of mercury in the body, leading to neuropsychiatric, dermatologic, oropharyngeal, and renal manifestations. Sore throat, fever, headache, fatigue, dyspnea, chest pain, and pneumonitis are common.16 Typically, low-level exposure to elemental mercury does not lead to long-lasting health effects. However, individuals exposed to high-level elemental mercury vapors may require hospitalization. Treatment of acute mercury poisoning consists of removing the source of exposure, followed by cardiopulmonary support.16
Specific assays for mercury levels in blood and urine are useful to assess the level of exposure and risk to the patient. Blood mercury concentrations of 20 µg/L or below are considered within reference range; however, once blood and urine concentrations of mercury exceed 100 µg/L, clinical signs of acute mercury poisoning typically manifest.21 Chest radiographs can reveal pulmonary damage, while complete blood cell count, metabolic panel, and urinalysis can assess damage to other organs. Neuropsychiatric testing and nerve conduction studies may provide objective evidence of CNS toxicity. Assays for N-acetyl-β-D-glucosaminidase can provide an indication of early renal tubular dysfunction.16
Elemental mercury is not absorbed from the gastrointestinal tract, posing minimal risk for acute toxicity from ingestion. Generally, less than 10% of ingested inorganic mercury is absorbed from the gut, while elemental mercury is nonabsorbable.10 If an individual ingests a large amount of mercury, it may persist in the gastrointestinal tract for an extended period. Mercury is radiopaque, and abdominal radiographs should be obtained in all cases of ingestion.16
Mercury is toxic to the CNS and peripheral nervous system, resulting in erethism mercurialis, a constellation of neuropsychologic signs and symptoms including restlessness, irritability, insomnia, emotional lability, difficulty concentrating, and impaired memory. In severe cases, delirium and psychosis may develop. Other CNS effects include tremors, paresthesia, dysarthria, neuromuscular changes, headaches, polyneuropathy, and cerebellar ataxia, as well as ophthalmologic and audiologic impairment.5,16
Upon inhalation exposure, patients with respiratory concerns should be given oxygen. Bronchospasms are treated with bronchodilators; however, if multiple chemical exposures are suspected, bronchial-sensitizing agents may pose additional risks. Corticosteroids and antibiotics have been recommended for treatment of chemical pneumonitis, but their efficacy has not been substantiated.16
Skin reactions associated with skin contact to elemental mercury are rare. However, hives and dermatitis have been observed following accidental contact with inorganic mercury compounds.5 Manifestation in children chronically exposed to mercury includes a nonallergic hypersensitivity (acrodynia),5,17 which is characterized by pain and dusky pink discoloration in the hands and feet, most often seen in children chronically exposed to mercury absorbed from vapor inhalation or cutaneous exposure.16
Renal conditions associated with acute inhalation of elemental mercury vapor include proteinuria, nephrotic syndrome, temporary tubular dysfunction, acute tubular necrosis, and oliguric renal failure.16 Chronic exposure to inorganic mercury compounds also has been reported to cause renal damage.5 Chelation therapy should be performed for any symptomatic patient with a clear history of acute elemental mercury exposure.16 The most frequently used chelation agent in cases of acute inorganic mercury exposures is dimercaprol. In rare cases of mercury intoxication, hemodialysis is required in the treatment of renal failure and to expedite removal of dimercaprol-mercury complexes.16
Cardiovascular symptoms associated with acute inhalation of high levels of elemental mercury include tachycardia and hypertension.16 Increases in blood pressure, palpitations, and heart rate also have been observed in instances of acute elemental mercury exposure. Studies show that exposure to mercury increases both the risk for acute myocardial infarction as well as death from coronary heart and cardiovascular diseases.5
Conclusion
Mercury poisoning presents with varied neuropsychologic signs and symptoms. Our case provides insight into a unique route of exposure for mercury toxicity. In addition to the unusual presentation of a mercury granuloma, our case illustrates how surgical techniques can aid in removal of cutaneous reservoirs in the setting of percutaneous exposure.
Evidence of human exposure to mercury dates as far back as the Egyptians in 1500
Mercury release in the environment primarily is a function of human activity, including coal-fired power plants, residential heating, and mining.9,10 Mercury from these sources is commonly found in the sediment of lakes and bays, where it is enzymatically converted to methylmercury by aquatic microorganisms; subsequent food chain biomagnification results in elevated mercury levels in apex predators. Substantial release of mercury into the environment also can be attributed to health care facilities from their use of thermometers containing 0.5 to 3 g of elemental mercury,11 blood pressure monitors, and medical waste incinerators.5
Mercury has been reported as the second most common cause of heavy metal poisoning after lead.12 Standards from the US Food and Drug Administration dictate that methylmercury levels in fish and wheat products must not exceed 1 ppm.13 Most plant and animal food sources contain methylmercury at levels between 0.0001 and 0.01 ppm; mercury concentrations are especially high in tuna, averaging 0.4 ppm, while larger predatory fish contain levels in excess of 1 ppm.14 The use of mercury-containing cosmetic products also presents a substantial exposure risk to consumers.5,10 In one study, 3.3% of skin-lightening creams and soaps purchased within the United States contained concentrations of mercury exceeding 1000 ppm.15
We describe a case of mercury toxicity resulting from intentional injection of liquid mercury into the right antecubital fossa in a suicide attempt.
Case Report
A 31-year-old woman presented to the family practice center for evaluation of a firm stained area on the skin of the right arm. She reported increasing anxiety, depression, tremors, irritability, and difficulty concentrating over the last 6 months. She denied headache and joint or muscle pain. Four years earlier, she had broken apart a thermometer and injected approximately 0.7 mL of its contents into the right arm in a suicide attempt. She intended to inject the thermometer’s contents directly into a vein, but the material instead entered the surrounding tissue. She denied notable pain or itching overlying the injection site. Her medications included aripiprazole and buspirone. She noted that she smoked half a pack of cigarettes per day and had a history of methamphetamine abuse. She was homeless and unemployed. Physical examination revealed an anxious tremulous woman with an erythematous to bluish gray, firm plaque on the right antecubital fossa (Figure 1). There were no notable tremors and no gait disturbance.
Her blood mercury level was greater than 100 µg/L and urine mercury was 477 µg/g (reference ranges, 1–8 μg/L and 4–5 μg/L, respectively). A radiograph of the right elbow area revealed scattered punctate foci of increased density within or overlying the anterolateral elbow soft tissues. She was diagnosed with mercury granuloma causing chronic mercury elevation. She underwent excision of the granuloma (Figure 2) with endovascular surgery via an elliptical incision. The patient was subsequently lost to follow-up.
Comment
Elemental mercury is a silver liquid at room temperature that spontaneously evaporates to form mercury vapor, an invisible, odorless, toxic gas. Accidental cutaneous exposure typically is safely managed by washing exposed skin with soap and water,16 though there is a potential risk for systemic absorption, especially when the skin is inflamed. When metallic mercury is subcutaneously injected, it is advised to promptly excise all subcutaneous areas containing mercury, regardless of any symptoms of systemic toxicity. Patients should subsequently be monitored for signs of both central nervous system (CNS) and renal deficits, undergo chelation therapy when systemic effects are apparent, and finally receive psychiatric consultation and treatment when necessary.17
Inorganic mercury compounds are formed when elemental mercury combines with sulfur or oxygen and often take the form of mercury salts, which appear as white crystals.16 These salts occur naturally in the environment and are used in pesticides, antiseptics, and skin-lightening creams and soaps.18
Methylmercury is a highly toxic, organic compound that is capable of crossing the placental and blood-brain barriers. It is the most common organic mercury compound found in the environment.16 Most humans have trace amounts of methylmercury in their bodies, typically as a result of consuming seafood.5
Exposure to mercury most commonly occurs through chronic consumption of methylmercury in seafood or acute inhalation of elemental mercury vapors.9 Iatrogenic cases of mercury exposure via injection also have been reported in the literature, including a case resulting in acute poisoning due to peritoneal lavage with mercury bichloride.19 Acute mercury-induced pulmonary damage typically resolves completely. However, there have been reported cases of exposure progressing to interstitial emphysema, pneumatocele, pneumothorax, pneumomediastinum, interstitial fibrosis, and chronic respiratory insufficiency, with examples of fatal acute respiratory distress syndrome being reported.5,16,20 Although individuals who inhale mercury vapors initially may be unaware of exposure due to little upper airway irritation, symptoms following an initial acute exposure may include ptyalism, a metallic taste, dysphagia, enteritis, diarrhea, nausea, renal damage, and CNS effects.16 Additionally, exposure may lead to confusion with signs and symptoms of metal fume fever, including shortness of breath, pleuritic chest pain, stomatitis, lethargy, and vomiting.20
Chronic exposure to mercury vapor can result in accumulation of mercury in the body, leading to neuropsychiatric, dermatologic, oropharyngeal, and renal manifestations. Sore throat, fever, headache, fatigue, dyspnea, chest pain, and pneumonitis are common.16 Typically, low-level exposure to elemental mercury does not lead to long-lasting health effects. However, individuals exposed to high-level elemental mercury vapors may require hospitalization. Treatment of acute mercury poisoning consists of removing the source of exposure, followed by cardiopulmonary support.16
Specific assays for mercury levels in blood and urine are useful to assess the level of exposure and risk to the patient. Blood mercury concentrations of 20 µg/L or below are considered within reference range; however, once blood and urine concentrations of mercury exceed 100 µg/L, clinical signs of acute mercury poisoning typically manifest.21 Chest radiographs can reveal pulmonary damage, while complete blood cell count, metabolic panel, and urinalysis can assess damage to other organs. Neuropsychiatric testing and nerve conduction studies may provide objective evidence of CNS toxicity. Assays for N-acetyl-β-D-glucosaminidase can provide an indication of early renal tubular dysfunction.16
Elemental mercury is not absorbed from the gastrointestinal tract, posing minimal risk for acute toxicity from ingestion. Generally, less than 10% of ingested inorganic mercury is absorbed from the gut, while elemental mercury is nonabsorbable.10 If an individual ingests a large amount of mercury, it may persist in the gastrointestinal tract for an extended period. Mercury is radiopaque, and abdominal radiographs should be obtained in all cases of ingestion.16
Mercury is toxic to the CNS and peripheral nervous system, resulting in erethism mercurialis, a constellation of neuropsychologic signs and symptoms including restlessness, irritability, insomnia, emotional lability, difficulty concentrating, and impaired memory. In severe cases, delirium and psychosis may develop. Other CNS effects include tremors, paresthesia, dysarthria, neuromuscular changes, headaches, polyneuropathy, and cerebellar ataxia, as well as ophthalmologic and audiologic impairment.5,16
Upon inhalation exposure, patients with respiratory concerns should be given oxygen. Bronchospasms are treated with bronchodilators; however, if multiple chemical exposures are suspected, bronchial-sensitizing agents may pose additional risks. Corticosteroids and antibiotics have been recommended for treatment of chemical pneumonitis, but their efficacy has not been substantiated.16
Skin reactions associated with skin contact to elemental mercury are rare. However, hives and dermatitis have been observed following accidental contact with inorganic mercury compounds.5 Manifestation in children chronically exposed to mercury includes a nonallergic hypersensitivity (acrodynia),5,17 which is characterized by pain and dusky pink discoloration in the hands and feet, most often seen in children chronically exposed to mercury absorbed from vapor inhalation or cutaneous exposure.16
Renal conditions associated with acute inhalation of elemental mercury vapor include proteinuria, nephrotic syndrome, temporary tubular dysfunction, acute tubular necrosis, and oliguric renal failure.16 Chronic exposure to inorganic mercury compounds also has been reported to cause renal damage.5 Chelation therapy should be performed for any symptomatic patient with a clear history of acute elemental mercury exposure.16 The most frequently used chelation agent in cases of acute inorganic mercury exposures is dimercaprol. In rare cases of mercury intoxication, hemodialysis is required in the treatment of renal failure and to expedite removal of dimercaprol-mercury complexes.16
Cardiovascular symptoms associated with acute inhalation of high levels of elemental mercury include tachycardia and hypertension.16 Increases in blood pressure, palpitations, and heart rate also have been observed in instances of acute elemental mercury exposure. Studies show that exposure to mercury increases both the risk for acute myocardial infarction as well as death from coronary heart and cardiovascular diseases.5
Conclusion
Mercury poisoning presents with varied neuropsychologic signs and symptoms. Our case provides insight into a unique route of exposure for mercury toxicity. In addition to the unusual presentation of a mercury granuloma, our case illustrates how surgical techniques can aid in removal of cutaneous reservoirs in the setting of percutaneous exposure.
- History of mercury. Government of Canada website. Modified April 26, 2010. Accessed March 11, 2021. https://www.canada.ca/en/environment-climate-change/services/pollutants/mercury-environment/about/history.html
- Dartmouth Toxic Metals Superfund Research Program website. Accessed March 11, 2021. https://sites.dartmouth.edu/toxmetal/
- Norn S, Permin H, Kruse E, et al. Mercury—a major agent in the history of medicine and alchemy [in Danish]. Dan Medicinhist Arbog. 2008;36:21-40.
- Waldron HA. Did the Mad Hatter have mercury poisoning? Br Med J (Clin Res Ed). 1983;287:1961.
- Poulin J, Gibb H. Mercury: assessing the environmental burden of disease at national and local levels. WHO Environmental Burden of Disease Series No. 16. World Health Organization; 2008.
- Charcot JM. Clinical lectures of the diseases of the nervous system. In: Kinnier Wilson SA. The Landmark Library of Neurology and Neurosurgery. Gryphon Editions; 1994:186.
- Kinnier Wilson SA. Neurology. In: Kinnier Wilson SA. The Landmark Library of Neurology and Neurosurgery. Gryphon Editions; 1994:739-740.
- Harada M. Minamata disease: methylmercury poisoning in Japan caused by environmental pollution. Crit Rev Toxicol. 1995;25:1-24.
- Mercury and health. World Health Organization website. Updated March 31, 2017. Accessed March 12, 2021. http://www.whoint/mediacentre/factsheets/fs361/en/
- Olson DA. Mercury toxicity. Updated November 5, 2018. Accessed March 12, 2021.http://emedicine.medscape.com/article/1175560-overview
- Mercury thermometers. Environmental Protection Agency website. Updated June 26, 2018. https://www.epa.gov/mercury/mercury-thermometers
- Jao-Tan C, Pope E. Cutaneous poisoning syndromes in children: a review. Curr Opin Pediatr. 2006;18:410-416.
- US Department of Health and Human Services: Public Health Service Agency for Toxic Substances and Disease Registry. Toxicological profile for mercury: regulations and advisories. Published March 1999. Accessed March 23, 2021. https://www.atsdr.cdc.gov/toxprofiles/tp46.pdf
- US Food and Drug Administration. Mercury levels in commercial fish and shellfish (1990-2012). Updated October 25, 2017. Accessed March 16, 2021. https://www.fda.gov/food/metals-and-your-food/mercury-levels-commercial-fish-and-shellfish-1990-2012
- Hamann CR, Boonchai W, Wen L, et al. Spectrometric analysis of mercury content in 549 skin-lightening products: is mercury toxicity a hidden global health hazard? J Am Acad Dermatol. 2014;70:281-287.e3.
- Mercury. Managing Hazardous Materials Incidents. Agency for Toxic Substances and Disease Registry website. Accessed March 16, 2021. https://www.atsdr.cdc.gov/MHMI/mmg46.pdf
- Krohn IT, Solof A, Mobini J, et al. Subcutaneous injection of metallic mercury. JAMA. 1980;243:548-549.
- Lai O, Parsi KK, Wu D, et al. Mercury toxicity presenting acrodynia and a papulovesicular eruption in a 5-year-old girl. Dermatol Online J. 2016;16;22:13030/qt6444r7nc.
- Dolianiti M, Tasiopoulou K, Kalostou A, et al. Mercury bichloride iatrogenic poisoning: a case report. J Clin Toxicol. 2016;6:2. doi:10.4172/2161-0495.1000290
- Broussard LA, Hammett-Stabler CA, Winecker RE, et al. The toxicology of mercury. Lab Med. 2002;33:614-625. doi:10.1309/5HY1-V3NE-2LFL-P9MT
- Byeong-Jin Y, Byoung-Gwon K, Man-Joong J, et al. Evaluation of mercury exposure levels, clinical diagnosis and treatment for mercury intoxication. Ann Occup Environ Med. 2016;28:5.
- History of mercury. Government of Canada website. Modified April 26, 2010. Accessed March 11, 2021. https://www.canada.ca/en/environment-climate-change/services/pollutants/mercury-environment/about/history.html
- Dartmouth Toxic Metals Superfund Research Program website. Accessed March 11, 2021. https://sites.dartmouth.edu/toxmetal/
- Norn S, Permin H, Kruse E, et al. Mercury—a major agent in the history of medicine and alchemy [in Danish]. Dan Medicinhist Arbog. 2008;36:21-40.
- Waldron HA. Did the Mad Hatter have mercury poisoning? Br Med J (Clin Res Ed). 1983;287:1961.
- Poulin J, Gibb H. Mercury: assessing the environmental burden of disease at national and local levels. WHO Environmental Burden of Disease Series No. 16. World Health Organization; 2008.
- Charcot JM. Clinical lectures of the diseases of the nervous system. In: Kinnier Wilson SA. The Landmark Library of Neurology and Neurosurgery. Gryphon Editions; 1994:186.
- Kinnier Wilson SA. Neurology. In: Kinnier Wilson SA. The Landmark Library of Neurology and Neurosurgery. Gryphon Editions; 1994:739-740.
- Harada M. Minamata disease: methylmercury poisoning in Japan caused by environmental pollution. Crit Rev Toxicol. 1995;25:1-24.
- Mercury and health. World Health Organization website. Updated March 31, 2017. Accessed March 12, 2021. http://www.whoint/mediacentre/factsheets/fs361/en/
- Olson DA. Mercury toxicity. Updated November 5, 2018. Accessed March 12, 2021.http://emedicine.medscape.com/article/1175560-overview
- Mercury thermometers. Environmental Protection Agency website. Updated June 26, 2018. https://www.epa.gov/mercury/mercury-thermometers
- Jao-Tan C, Pope E. Cutaneous poisoning syndromes in children: a review. Curr Opin Pediatr. 2006;18:410-416.
- US Department of Health and Human Services: Public Health Service Agency for Toxic Substances and Disease Registry. Toxicological profile for mercury: regulations and advisories. Published March 1999. Accessed March 23, 2021. https://www.atsdr.cdc.gov/toxprofiles/tp46.pdf
- US Food and Drug Administration. Mercury levels in commercial fish and shellfish (1990-2012). Updated October 25, 2017. Accessed March 16, 2021. https://www.fda.gov/food/metals-and-your-food/mercury-levels-commercial-fish-and-shellfish-1990-2012
- Hamann CR, Boonchai W, Wen L, et al. Spectrometric analysis of mercury content in 549 skin-lightening products: is mercury toxicity a hidden global health hazard? J Am Acad Dermatol. 2014;70:281-287.e3.
- Mercury. Managing Hazardous Materials Incidents. Agency for Toxic Substances and Disease Registry website. Accessed March 16, 2021. https://www.atsdr.cdc.gov/MHMI/mmg46.pdf
- Krohn IT, Solof A, Mobini J, et al. Subcutaneous injection of metallic mercury. JAMA. 1980;243:548-549.
- Lai O, Parsi KK, Wu D, et al. Mercury toxicity presenting acrodynia and a papulovesicular eruption in a 5-year-old girl. Dermatol Online J. 2016;16;22:13030/qt6444r7nc.
- Dolianiti M, Tasiopoulou K, Kalostou A, et al. Mercury bichloride iatrogenic poisoning: a case report. J Clin Toxicol. 2016;6:2. doi:10.4172/2161-0495.1000290
- Broussard LA, Hammett-Stabler CA, Winecker RE, et al. The toxicology of mercury. Lab Med. 2002;33:614-625. doi:10.1309/5HY1-V3NE-2LFL-P9MT
- Byeong-Jin Y, Byoung-Gwon K, Man-Joong J, et al. Evaluation of mercury exposure levels, clinical diagnosis and treatment for mercury intoxication. Ann Occup Environ Med. 2016;28:5.
Practice Points
- Chronic mercury granulomas can present as firm, erythematous to bluish gray plaques.
- Accidental skin contact to elemental mercury may cause urticaria and dermatitis.
- Blood mercury concentrations below 20 11µg/L are considered within reference range; once blood and urine concentrations exceed 100 11µg/L, clinical signs of acute mercury poisoning typically manifest.
- Mercury is toxic to the central and peripheral nervous systems, resulting in erethism mercurialis, a constellation of neuropsychologic signs and symptoms including restlessness, irritability, insomnia, emotional lability, difficulty concentrating, and impaired memory.
What’s Eating You? Black Butterfly (Hylesia nigricans)
The order Lepidoptera (phylum Arthropoda, class Hexapoda) is comprised of moths and butterflies.1 Lepidopterism refers to a range of adverse medical conditions resulting from contact with these insects, typically during the caterpillar (larval) stage. It involves multiple pathologic mechanisms, including direct toxicity of venom and mechanical irritant effects.2 Erucism has been defined as any reaction caused by contact with caterpillars or any reaction limited to the skin caused by contact with caterpillars, butterflies, or moths. Lepidopterism can mean any reaction to caterpillars or moths, referring only to reactions from contact with scales or hairs from adult moths or butterflies, or referring only to cases with systemic signs and symptoms (eg, rhinoconjunctival or asthmatic symptoms, angioedema and anaphylaxis, hemorrhagic diathesis) with or without cutaneous findings, resulting from contact with any lepidopteran source.1 Strictly speaking, erucism should refer to any reaction from caterpillars and lepidopterism to reactions from moths or butterflies. Because reactions to both larval and adult lepidoptera can cause a variety of either cutaneous and/or systemic symptoms, classifying reactions into erucism or lepidopterism is only of academic interest.1
We report a documented case of lepidopterism in a patient with acute cutaneous lesions following exposure to an adult-stage black butterfly (Hylesia nigricans).
Case Report
A 21-year-old oil well worker presented with pruritic skin lesions on the right arm and flank of 3 hours’ duration. He reported that a black butterfly had perched on his arm while he was working and left a considerable number of small yellowish hairs on the skin, after which an intense pruritus and skin lesions began to develop. He denied other associated symptoms. Physical examination revealed numerous 1- to 2-mm, nonconfluent, erythematous and edematous papules on the right forearm, arm (Figure 1A), and flank. Some abrasions of the skin due to scratching and crusting were noted (Figure 1B). A skin biopsy from the right arm showed a superficial perivascular dermatitis with a mixed infiltrate of polymorphonuclear predominance with eosinophils (Figure 2A). Importantly, a structure resembling an urticating spicule was identified in the stratum corneum (Figure 2B); spicules are located on the abdomen of arthropods and are associated with an inflammatory response in human skin.
Based on the patient’s history of butterfly exposure, clinical presentation of the lesions, and histopathologic findings demonstrating the presence of the spicules, the diagnosis of lepidopterism was confirmed. The patient was treated with oral antihistamines and topical steroids for 1 week with complete resolution of the lesions.
Comment
Epidemiology of Envenomation
Although many tropical insects carry infectious diseases, cutaneous injury can occur by other mechanisms, such as dermatitis caused by contact with the skin (erucism or lepidopterism). Caterpillar envenomation is common, but this phenomenon rarely has been studied due to few reported cases, which hinders a complete understanding of the problem.3
The order Lepidoptera comprises 2 suborders: Rhopalocera, with adult specimens that fly during the daytime (butterflies), and Heterocera, which are largely nocturnal (moths). The stages of development include egg, larva (caterpillar), pupa (chrysalis), and adult (imago), constituting a holometabolic life cycle.4 Adult butterflies and moths represent the reproductive stage of lepidoptera.
The pathology of lepidopterism is attributed to contact with fluids such as hemolymph and secretions from the spicules, with histamine being identified as the main causative component.3 During the reproductive stage, female insects approach light sources and release clouds of bristles from their abdomens that can penetrate human skin and cause an irritating dermatitis.5 Lepidopterism can occur following contact with bristles from insects of the Hylesia genus (Saturniidae family), such as in our patient.3,6 Epidemic outbreaks have been reported in several countries, mainly Argentina, Brazil, and Venezuela.5 The patient was located in Colombia, a country without any reported cases of lepidopterism from the black butterfly (H nigricans). Cutaneous reactions to lepidoptera insects come in many forms, most commonly presenting as a mild stinging reaction with a papular eruption, pruritic urticarial papules and plaques, or scaly erythematous papules and plaques in exposed areas.7
Histopathologic Findings
The histology of lepidoptera exposure is nonspecific, typically demonstrating epidermal edema, superficial perivascular lymphocytic infiltrate, and eosinophils. Epidermal necrosis and vasculitis rarely are seen. Embedded spines from Hylesia insects have been described.7 The histopathologic examination generally reveals a foreign body reaction in addition to granulomas.3
Therapy
The use of oral antihistamines is indicated for the control of pruritus, and topical treatment with cold compresses, baths, and corticosteroid creams is recommended.3,8,9
Conclusion
We report the case of a patient with lepidopterism, a rare entity confirmed histologically with documentation of a spicule in the stratum corneum in the patient’s biopsy. Changes due to urbanization and industrialization have a closer relationship with various animal species that are pathogenic to humans; therefore, we encourage dermatologists to be aware of these diseases.
- Hossler EW. Caterpillars and moths: part I. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:666.
- Redd JT, Voorhees RE, Török TJ. Outbreak of lepidopterism at a Boy Scout camp. J Am Acad Dermatol. 2007;56:952-955.
- 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.
- Cardoso AEC, Haddad V Jr. Accidents caused by lepidopterans (moth larvae and adult): study on the epidemiological, clinical and therapeutic aspects. An Bras Dermatol. 2005;80:571-578.
- Salomón AD, Simón D, Rimoldi JC, et al. Lepidopterism due to the butterfly Hylesia nigricans. preventive research-intervention in Buenos Aires. Medicina (B Aires). 2005;65:241-246.
- Moreira SC, Lima JC, Silva L, et al. Description of an outbreak of lepidopterism (dermatitis associated with contact with moths) among sailors in Salvador, State of Bahia. Rev Soc Bras Med Trop. 2007;40:591-593.
- Hossler EW. Caterpillars and moths: part II. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:666.
- Maier H, Spiegel W, Kinaciyan T, et al. The oak processionary caterpillar as the cause of an epidemic airborne disease: survey and analysis. Br J Dermatol. 2003;149:990-997.
- Herrera-Chaumont C, Sojo-Milano M, Pérez-Ybarra LM. Knowledge and practices on lepidopterism by Hylesia metabus (Cramer, 1775)(Lepidoptera: Saturniidae) in Yaguaraparo parish, Sucre state, northeastern Venezuela. Revista Biomédica. 2016;27:11-23.
The order Lepidoptera (phylum Arthropoda, class Hexapoda) is comprised of moths and butterflies.1 Lepidopterism refers to a range of adverse medical conditions resulting from contact with these insects, typically during the caterpillar (larval) stage. It involves multiple pathologic mechanisms, including direct toxicity of venom and mechanical irritant effects.2 Erucism has been defined as any reaction caused by contact with caterpillars or any reaction limited to the skin caused by contact with caterpillars, butterflies, or moths. Lepidopterism can mean any reaction to caterpillars or moths, referring only to reactions from contact with scales or hairs from adult moths or butterflies, or referring only to cases with systemic signs and symptoms (eg, rhinoconjunctival or asthmatic symptoms, angioedema and anaphylaxis, hemorrhagic diathesis) with or without cutaneous findings, resulting from contact with any lepidopteran source.1 Strictly speaking, erucism should refer to any reaction from caterpillars and lepidopterism to reactions from moths or butterflies. Because reactions to both larval and adult lepidoptera can cause a variety of either cutaneous and/or systemic symptoms, classifying reactions into erucism or lepidopterism is only of academic interest.1
We report a documented case of lepidopterism in a patient with acute cutaneous lesions following exposure to an adult-stage black butterfly (Hylesia nigricans).
Case Report
A 21-year-old oil well worker presented with pruritic skin lesions on the right arm and flank of 3 hours’ duration. He reported that a black butterfly had perched on his arm while he was working and left a considerable number of small yellowish hairs on the skin, after which an intense pruritus and skin lesions began to develop. He denied other associated symptoms. Physical examination revealed numerous 1- to 2-mm, nonconfluent, erythematous and edematous papules on the right forearm, arm (Figure 1A), and flank. Some abrasions of the skin due to scratching and crusting were noted (Figure 1B). A skin biopsy from the right arm showed a superficial perivascular dermatitis with a mixed infiltrate of polymorphonuclear predominance with eosinophils (Figure 2A). Importantly, a structure resembling an urticating spicule was identified in the stratum corneum (Figure 2B); spicules are located on the abdomen of arthropods and are associated with an inflammatory response in human skin.
Based on the patient’s history of butterfly exposure, clinical presentation of the lesions, and histopathologic findings demonstrating the presence of the spicules, the diagnosis of lepidopterism was confirmed. The patient was treated with oral antihistamines and topical steroids for 1 week with complete resolution of the lesions.
Comment
Epidemiology of Envenomation
Although many tropical insects carry infectious diseases, cutaneous injury can occur by other mechanisms, such as dermatitis caused by contact with the skin (erucism or lepidopterism). Caterpillar envenomation is common, but this phenomenon rarely has been studied due to few reported cases, which hinders a complete understanding of the problem.3
The order Lepidoptera comprises 2 suborders: Rhopalocera, with adult specimens that fly during the daytime (butterflies), and Heterocera, which are largely nocturnal (moths). The stages of development include egg, larva (caterpillar), pupa (chrysalis), and adult (imago), constituting a holometabolic life cycle.4 Adult butterflies and moths represent the reproductive stage of lepidoptera.
The pathology of lepidopterism is attributed to contact with fluids such as hemolymph and secretions from the spicules, with histamine being identified as the main causative component.3 During the reproductive stage, female insects approach light sources and release clouds of bristles from their abdomens that can penetrate human skin and cause an irritating dermatitis.5 Lepidopterism can occur following contact with bristles from insects of the Hylesia genus (Saturniidae family), such as in our patient.3,6 Epidemic outbreaks have been reported in several countries, mainly Argentina, Brazil, and Venezuela.5 The patient was located in Colombia, a country without any reported cases of lepidopterism from the black butterfly (H nigricans). Cutaneous reactions to lepidoptera insects come in many forms, most commonly presenting as a mild stinging reaction with a papular eruption, pruritic urticarial papules and plaques, or scaly erythematous papules and plaques in exposed areas.7
Histopathologic Findings
The histology of lepidoptera exposure is nonspecific, typically demonstrating epidermal edema, superficial perivascular lymphocytic infiltrate, and eosinophils. Epidermal necrosis and vasculitis rarely are seen. Embedded spines from Hylesia insects have been described.7 The histopathologic examination generally reveals a foreign body reaction in addition to granulomas.3
Therapy
The use of oral antihistamines is indicated for the control of pruritus, and topical treatment with cold compresses, baths, and corticosteroid creams is recommended.3,8,9
Conclusion
We report the case of a patient with lepidopterism, a rare entity confirmed histologically with documentation of a spicule in the stratum corneum in the patient’s biopsy. Changes due to urbanization and industrialization have a closer relationship with various animal species that are pathogenic to humans; therefore, we encourage dermatologists to be aware of these diseases.
The order Lepidoptera (phylum Arthropoda, class Hexapoda) is comprised of moths and butterflies.1 Lepidopterism refers to a range of adverse medical conditions resulting from contact with these insects, typically during the caterpillar (larval) stage. It involves multiple pathologic mechanisms, including direct toxicity of venom and mechanical irritant effects.2 Erucism has been defined as any reaction caused by contact with caterpillars or any reaction limited to the skin caused by contact with caterpillars, butterflies, or moths. Lepidopterism can mean any reaction to caterpillars or moths, referring only to reactions from contact with scales or hairs from adult moths or butterflies, or referring only to cases with systemic signs and symptoms (eg, rhinoconjunctival or asthmatic symptoms, angioedema and anaphylaxis, hemorrhagic diathesis) with or without cutaneous findings, resulting from contact with any lepidopteran source.1 Strictly speaking, erucism should refer to any reaction from caterpillars and lepidopterism to reactions from moths or butterflies. Because reactions to both larval and adult lepidoptera can cause a variety of either cutaneous and/or systemic symptoms, classifying reactions into erucism or lepidopterism is only of academic interest.1
We report a documented case of lepidopterism in a patient with acute cutaneous lesions following exposure to an adult-stage black butterfly (Hylesia nigricans).
Case Report
A 21-year-old oil well worker presented with pruritic skin lesions on the right arm and flank of 3 hours’ duration. He reported that a black butterfly had perched on his arm while he was working and left a considerable number of small yellowish hairs on the skin, after which an intense pruritus and skin lesions began to develop. He denied other associated symptoms. Physical examination revealed numerous 1- to 2-mm, nonconfluent, erythematous and edematous papules on the right forearm, arm (Figure 1A), and flank. Some abrasions of the skin due to scratching and crusting were noted (Figure 1B). A skin biopsy from the right arm showed a superficial perivascular dermatitis with a mixed infiltrate of polymorphonuclear predominance with eosinophils (Figure 2A). Importantly, a structure resembling an urticating spicule was identified in the stratum corneum (Figure 2B); spicules are located on the abdomen of arthropods and are associated with an inflammatory response in human skin.
Based on the patient’s history of butterfly exposure, clinical presentation of the lesions, and histopathologic findings demonstrating the presence of the spicules, the diagnosis of lepidopterism was confirmed. The patient was treated with oral antihistamines and topical steroids for 1 week with complete resolution of the lesions.
Comment
Epidemiology of Envenomation
Although many tropical insects carry infectious diseases, cutaneous injury can occur by other mechanisms, such as dermatitis caused by contact with the skin (erucism or lepidopterism). Caterpillar envenomation is common, but this phenomenon rarely has been studied due to few reported cases, which hinders a complete understanding of the problem.3
The order Lepidoptera comprises 2 suborders: Rhopalocera, with adult specimens that fly during the daytime (butterflies), and Heterocera, which are largely nocturnal (moths). The stages of development include egg, larva (caterpillar), pupa (chrysalis), and adult (imago), constituting a holometabolic life cycle.4 Adult butterflies and moths represent the reproductive stage of lepidoptera.
The pathology of lepidopterism is attributed to contact with fluids such as hemolymph and secretions from the spicules, with histamine being identified as the main causative component.3 During the reproductive stage, female insects approach light sources and release clouds of bristles from their abdomens that can penetrate human skin and cause an irritating dermatitis.5 Lepidopterism can occur following contact with bristles from insects of the Hylesia genus (Saturniidae family), such as in our patient.3,6 Epidemic outbreaks have been reported in several countries, mainly Argentina, Brazil, and Venezuela.5 The patient was located in Colombia, a country without any reported cases of lepidopterism from the black butterfly (H nigricans). Cutaneous reactions to lepidoptera insects come in many forms, most commonly presenting as a mild stinging reaction with a papular eruption, pruritic urticarial papules and plaques, or scaly erythematous papules and plaques in exposed areas.7
Histopathologic Findings
The histology of lepidoptera exposure is nonspecific, typically demonstrating epidermal edema, superficial perivascular lymphocytic infiltrate, and eosinophils. Epidermal necrosis and vasculitis rarely are seen. Embedded spines from Hylesia insects have been described.7 The histopathologic examination generally reveals a foreign body reaction in addition to granulomas.3
Therapy
The use of oral antihistamines is indicated for the control of pruritus, and topical treatment with cold compresses, baths, and corticosteroid creams is recommended.3,8,9
Conclusion
We report the case of a patient with lepidopterism, a rare entity confirmed histologically with documentation of a spicule in the stratum corneum in the patient’s biopsy. Changes due to urbanization and industrialization have a closer relationship with various animal species that are pathogenic to humans; therefore, we encourage dermatologists to be aware of these diseases.
- Hossler EW. Caterpillars and moths: part I. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:666.
- Redd JT, Voorhees RE, Török TJ. Outbreak of lepidopterism at a Boy Scout camp. J Am Acad Dermatol. 2007;56:952-955.
- 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.
- Cardoso AEC, Haddad V Jr. Accidents caused by lepidopterans (moth larvae and adult): study on the epidemiological, clinical and therapeutic aspects. An Bras Dermatol. 2005;80:571-578.
- Salomón AD, Simón D, Rimoldi JC, et al. Lepidopterism due to the butterfly Hylesia nigricans. preventive research-intervention in Buenos Aires. Medicina (B Aires). 2005;65:241-246.
- Moreira SC, Lima JC, Silva L, et al. Description of an outbreak of lepidopterism (dermatitis associated with contact with moths) among sailors in Salvador, State of Bahia. Rev Soc Bras Med Trop. 2007;40:591-593.
- Hossler EW. Caterpillars and moths: part II. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:666.
- Maier H, Spiegel W, Kinaciyan T, et al. The oak processionary caterpillar as the cause of an epidemic airborne disease: survey and analysis. Br J Dermatol. 2003;149:990-997.
- Herrera-Chaumont C, Sojo-Milano M, Pérez-Ybarra LM. Knowledge and practices on lepidopterism by Hylesia metabus (Cramer, 1775)(Lepidoptera: Saturniidae) in Yaguaraparo parish, Sucre state, northeastern Venezuela. Revista Biomédica. 2016;27:11-23.
- Hossler EW. Caterpillars and moths: part I. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:666.
- Redd JT, Voorhees RE, Török TJ. Outbreak of lepidopterism at a Boy Scout camp. J Am Acad Dermatol. 2007;56:952-955.
- 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.
- Cardoso AEC, Haddad V Jr. Accidents caused by lepidopterans (moth larvae and adult): study on the epidemiological, clinical and therapeutic aspects. An Bras Dermatol. 2005;80:571-578.
- Salomón AD, Simón D, Rimoldi JC, et al. Lepidopterism due to the butterfly Hylesia nigricans. preventive research-intervention in Buenos Aires. Medicina (B Aires). 2005;65:241-246.
- Moreira SC, Lima JC, Silva L, et al. Description of an outbreak of lepidopterism (dermatitis associated with contact with moths) among sailors in Salvador, State of Bahia. Rev Soc Bras Med Trop. 2007;40:591-593.
- Hossler EW. Caterpillars and moths: part II. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:666.
- Maier H, Spiegel W, Kinaciyan T, et al. The oak processionary caterpillar as the cause of an epidemic airborne disease: survey and analysis. Br J Dermatol. 2003;149:990-997.
- Herrera-Chaumont C, Sojo-Milano M, Pérez-Ybarra LM. Knowledge and practices on lepidopterism by Hylesia metabus (Cramer, 1775)(Lepidoptera: Saturniidae) in Yaguaraparo parish, Sucre state, northeastern Venezuela. Revista Biomédica. 2016;27:11-23.
Practice Points
- When contact with caterpillars, butterflies, or moths occurs, patients should be advised not to rub or scratch the area or attempt to remove or crush the insect with a bare hand, as this may further spread irritating setae or spines.
- Careful removal of the larva with forceps or a similar instrument, combined with tape stripping of the area and immediate washing with soap and water, can be effective in minimizing exposure.
- Contaminated clothing should be removed and laundered thoroughly.
Aquatic Antagonists: Sponge Dermatitis
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.
Sponges have been used in medicine for centuries. The first use in Western culture was recorded in 405
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.
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
- 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.
- 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.
- 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.
- 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.
- Reese E, Depenbrock P. Water envenomations and stings. Curr Sports Med Rep. 2014;13:126-131.
- Dormandy TL. Trace element analysis of hair. Br Med J (Clin Res Ed). 1986;293:975-976.
- Voultsiadou E. Sponges: an historical survey of their knowledge in Greek antiquity. J Mar Biol Assoc UK. 2007;87:1757-1763.
- 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
- Sagar S, Kaur M, Minneman KP. Antiviral lead compounds from marine sponges. Mar Drugs. 2010;8:2619-2638.
- 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.
- Elston DM. Aquatic antagonists: sponge dermatitis. Cutis. 2007;80:279-280.
- 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.
- 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.
- Burnett JW, Calton GJ, Morgan RJ. Dermatitis due to stinging sponges. Cutis. 1987;39:476.
- Kizer KW. Marine envenomations. J Toxicol Clin Toxicol. 1983;21:527-555.
- 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.
- Fromont J, Abdo DA. New species of Haliclona (Demospongiae: Haplosclerida: Chalinidae) from Western Australia. Zootaxa. 2014;3835:97-109.
- 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.
- 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.
- 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.
- Yaffee HS. Irritation from red sponge. N Engl J Med. 1970;282:51.
- Haddad V Jr. Environmental dermatology: skin manifestations of injuries caused by invertebrate aquatic animals. An Bras Dermatol. 2013;88:496-506.
- 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.
- 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.
- 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.
- Haddad V Jr, Lupi O, Lonza JP, et al. Tropical dermatology: marine and aquatic dermatology. J Am Acad Dermatol. 2009;61:733-750.
- Gaastra MT. Aquatic skin disorders. In: Faber WR, Hay RJ, Naafs B, eds. Imported Skin Diseases. 2nd ed. Wiley; 2012:283-292.
- Auerbach P. Envenomation by aquatic invertebrates. In: Auerbach P, ed. Wilderness Medicine. 6th ed. Elsevier Mosby; 2011;1596-1627.
- Sims JK, Irei MY. Human Hawaiian marine sponge poisoning. Hawaii Med J. 1979;38:263-270.
- Haddad V Jr. Aquatic animals of medical importance in Brazil. Rev Soc Bras Med Trop. 2003;36:591-597.
- Tlougan BE, Podjasek JO, Adams BB. Aquatic sports dermatoses. part 2—in the water: saltwater dermatoses. Int J Dermatol. 2010;49:994-1002.
- 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.
- Southcott R. Human injuries from invertebrate animals in the Australian seas. Clin Toxicol. 1970;3:617-636.
- Russell FE. Sponge injury—traumatic, toxic or allergic? N Engl J Med. 1970;282:753-754.
- Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337.
- Muraro A, Roberts G, Worm M, et al. Anaphylaxis: guidelines from the European Academy of Allergy and Clinical Immunology. Allergy. 2014;69:1026-1045.
- Kizer K, Auerbach P, Dwyer B. Marine envenomations: not just a problem of the tropics. Emerg Med Rep. 1985;6:129-135.
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.
Sponges have been used in medicine for centuries. The first use in Western culture was recorded in 405
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.
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.
Sponges have been used in medicine for centuries. The first use in Western culture was recorded in 405
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.
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
- 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.
- 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.
- 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.
- 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.
- Reese E, Depenbrock P. Water envenomations and stings. Curr Sports Med Rep. 2014;13:126-131.
- Dormandy TL. Trace element analysis of hair. Br Med J (Clin Res Ed). 1986;293:975-976.
- Voultsiadou E. Sponges: an historical survey of their knowledge in Greek antiquity. J Mar Biol Assoc UK. 2007;87:1757-1763.
- 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
- Sagar S, Kaur M, Minneman KP. Antiviral lead compounds from marine sponges. Mar Drugs. 2010;8:2619-2638.
- 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.
- Elston DM. Aquatic antagonists: sponge dermatitis. Cutis. 2007;80:279-280.
- 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.
- 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.
- Burnett JW, Calton GJ, Morgan RJ. Dermatitis due to stinging sponges. Cutis. 1987;39:476.
- Kizer KW. Marine envenomations. J Toxicol Clin Toxicol. 1983;21:527-555.
- 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.
- Fromont J, Abdo DA. New species of Haliclona (Demospongiae: Haplosclerida: Chalinidae) from Western Australia. Zootaxa. 2014;3835:97-109.
- 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.
- 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.
- 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.
- Yaffee HS. Irritation from red sponge. N Engl J Med. 1970;282:51.
- Haddad V Jr. Environmental dermatology: skin manifestations of injuries caused by invertebrate aquatic animals. An Bras Dermatol. 2013;88:496-506.
- 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.
- 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.
- 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.
- Haddad V Jr, Lupi O, Lonza JP, et al. Tropical dermatology: marine and aquatic dermatology. J Am Acad Dermatol. 2009;61:733-750.
- Gaastra MT. Aquatic skin disorders. In: Faber WR, Hay RJ, Naafs B, eds. Imported Skin Diseases. 2nd ed. Wiley; 2012:283-292.
- Auerbach P. Envenomation by aquatic invertebrates. In: Auerbach P, ed. Wilderness Medicine. 6th ed. Elsevier Mosby; 2011;1596-1627.
- Sims JK, Irei MY. Human Hawaiian marine sponge poisoning. Hawaii Med J. 1979;38:263-270.
- Haddad V Jr. Aquatic animals of medical importance in Brazil. Rev Soc Bras Med Trop. 2003;36:591-597.
- Tlougan BE, Podjasek JO, Adams BB. Aquatic sports dermatoses. part 2—in the water: saltwater dermatoses. Int J Dermatol. 2010;49:994-1002.
- 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.
- Southcott R. Human injuries from invertebrate animals in the Australian seas. Clin Toxicol. 1970;3:617-636.
- Russell FE. Sponge injury—traumatic, toxic or allergic? N Engl J Med. 1970;282:753-754.
- Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337.
- Muraro A, Roberts G, Worm M, et al. Anaphylaxis: guidelines from the European Academy of Allergy and Clinical Immunology. Allergy. 2014;69:1026-1045.
- Kizer K, Auerbach P, Dwyer B. Marine envenomations: not just a problem of the tropics. Emerg Med Rep. 1985;6:129-135.
- 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.
- 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.
- 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.
- 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.
- Reese E, Depenbrock P. Water envenomations and stings. Curr Sports Med Rep. 2014;13:126-131.
- Dormandy TL. Trace element analysis of hair. Br Med J (Clin Res Ed). 1986;293:975-976.
- Voultsiadou E. Sponges: an historical survey of their knowledge in Greek antiquity. J Mar Biol Assoc UK. 2007;87:1757-1763.
- 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
- Sagar S, Kaur M, Minneman KP. Antiviral lead compounds from marine sponges. Mar Drugs. 2010;8:2619-2638.
- 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.
- Elston DM. Aquatic antagonists: sponge dermatitis. Cutis. 2007;80:279-280.
- 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.
- 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.
- Burnett JW, Calton GJ, Morgan RJ. Dermatitis due to stinging sponges. Cutis. 1987;39:476.
- Kizer KW. Marine envenomations. J Toxicol Clin Toxicol. 1983;21:527-555.
- 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.
- Fromont J, Abdo DA. New species of Haliclona (Demospongiae: Haplosclerida: Chalinidae) from Western Australia. Zootaxa. 2014;3835:97-109.
- 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.
- 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.
- 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.
- Yaffee HS. Irritation from red sponge. N Engl J Med. 1970;282:51.
- Haddad V Jr. Environmental dermatology: skin manifestations of injuries caused by invertebrate aquatic animals. An Bras Dermatol. 2013;88:496-506.
- 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.
- 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.
- 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.
- Haddad V Jr, Lupi O, Lonza JP, et al. Tropical dermatology: marine and aquatic dermatology. J Am Acad Dermatol. 2009;61:733-750.
- Gaastra MT. Aquatic skin disorders. In: Faber WR, Hay RJ, Naafs B, eds. Imported Skin Diseases. 2nd ed. Wiley; 2012:283-292.
- Auerbach P. Envenomation by aquatic invertebrates. In: Auerbach P, ed. Wilderness Medicine. 6th ed. Elsevier Mosby; 2011;1596-1627.
- Sims JK, Irei MY. Human Hawaiian marine sponge poisoning. Hawaii Med J. 1979;38:263-270.
- Haddad V Jr. Aquatic animals of medical importance in Brazil. Rev Soc Bras Med Trop. 2003;36:591-597.
- Tlougan BE, Podjasek JO, Adams BB. Aquatic sports dermatoses. part 2—in the water: saltwater dermatoses. Int J Dermatol. 2010;49:994-1002.
- 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.
- Southcott R. Human injuries from invertebrate animals in the Australian seas. Clin Toxicol. 1970;3:617-636.
- Russell FE. Sponge injury—traumatic, toxic or allergic? N Engl J Med. 1970;282:753-754.
- Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337.
- Muraro A, Roberts G, Worm M, et al. Anaphylaxis: guidelines from the European Academy of Allergy and Clinical Immunology. Allergy. 2014;69:1026-1045.
- Kizer K, Auerbach P, Dwyer B. Marine envenomations: not just a problem of the tropics. Emerg Med Rep. 1985;6:129-135.
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.
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.
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.
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.
- Burrow M. How fleas jump. J Exp Biol. 2009;18:2881-2883.
- Buckland PC, Sandler JP. A biogeography of the human flea, Pulex irritans L (Siphonaptera: Pulicidae). J Biogeogr. 1989;16:115-120.
- Krasnov BR. Life cycles. In: Krasnov BR, ed. Functional and Evolutional Ecology of Fleas. Cambridge, MA: Cambridge Univ Press; 2008:45-67.
- 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.
- 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.
- Archibald WS, Kunitz SJ. Detection of plague by testing serums of dogs on the Navajo Reservation. HSMHA Health Rep. 1971;86:377-380.
- 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.
- Laudisoit A, Leirs H, Makundi RH, et al. Plague and the human flea, Tanzania. Emerg Infect Dis. 2007;13:687-693.
- Foucault C, Brouqui P, Raoult D. Bartonella quintana characteristics and clinical management. Emerg Infect Dis. 2006;12:217-223.
- 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.
- 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.
- 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.
- 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.
- 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.
- Rolain JM, Bourry, O, Davoust B, et al. Bartonella quintana and Rickettsia felis in Gabon. Emerg Infect Dis. 2005;11:1742-1744.
- 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.
- Brown L, Macaluso KR. Rickettsia felis, an emerging flea-borne rickettsiosis. Curr Trop Med Rep. 2016;3:27-39.
- 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.
- 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.
- Traversa D. Fleas infesting pets in the era of emerging extra-intestinal nematodes. Parasit Vectors. 2013;6:59.
- Rust MK. Insecticide resistance in fleas. Insects. 2016;7:10.
- 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.
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.
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.
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.
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.
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.
- Burrow M. How fleas jump. J Exp Biol. 2009;18:2881-2883.
- Buckland PC, Sandler JP. A biogeography of the human flea, Pulex irritans L (Siphonaptera: Pulicidae). J Biogeogr. 1989;16:115-120.
- Krasnov BR. Life cycles. In: Krasnov BR, ed. Functional and Evolutional Ecology of Fleas. Cambridge, MA: Cambridge Univ Press; 2008:45-67.
- 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.
- 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.
- Archibald WS, Kunitz SJ. Detection of plague by testing serums of dogs on the Navajo Reservation. HSMHA Health Rep. 1971;86:377-380.
- 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.
- Laudisoit A, Leirs H, Makundi RH, et al. Plague and the human flea, Tanzania. Emerg Infect Dis. 2007;13:687-693.
- Foucault C, Brouqui P, Raoult D. Bartonella quintana characteristics and clinical management. Emerg Infect Dis. 2006;12:217-223.
- 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.
- 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.
- 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.
- 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.
- 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.
- Rolain JM, Bourry, O, Davoust B, et al. Bartonella quintana and Rickettsia felis in Gabon. Emerg Infect Dis. 2005;11:1742-1744.
- 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.
- Brown L, Macaluso KR. Rickettsia felis, an emerging flea-borne rickettsiosis. Curr Trop Med Rep. 2016;3:27-39.
- 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.
- 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.
- Traversa D. Fleas infesting pets in the era of emerging extra-intestinal nematodes. Parasit Vectors. 2013;6:59.
- Rust MK. Insecticide resistance in fleas. Insects. 2016;7:10.
- 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.
- Burrow M. How fleas jump. J Exp Biol. 2009;18:2881-2883.
- Buckland PC, Sandler JP. A biogeography of the human flea, Pulex irritans L (Siphonaptera: Pulicidae). J Biogeogr. 1989;16:115-120.
- Krasnov BR. Life cycles. In: Krasnov BR, ed. Functional and Evolutional Ecology of Fleas. Cambridge, MA: Cambridge Univ Press; 2008:45-67.
- 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.
- 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.
- Archibald WS, Kunitz SJ. Detection of plague by testing serums of dogs on the Navajo Reservation. HSMHA Health Rep. 1971;86:377-380.
- 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.
- Laudisoit A, Leirs H, Makundi RH, et al. Plague and the human flea, Tanzania. Emerg Infect Dis. 2007;13:687-693.
- Foucault C, Brouqui P, Raoult D. Bartonella quintana characteristics and clinical management. Emerg Infect Dis. 2006;12:217-223.
- 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.
- 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.
- 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.
- 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.
- 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.
- Rolain JM, Bourry, O, Davoust B, et al. Bartonella quintana and Rickettsia felis in Gabon. Emerg Infect Dis. 2005;11:1742-1744.
- 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.
- Brown L, Macaluso KR. Rickettsia felis, an emerging flea-borne rickettsiosis. Curr Trop Med Rep. 2016;3:27-39.
- 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.
- 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.
- Traversa D. Fleas infesting pets in the era of emerging extra-intestinal nematodes. Parasit Vectors. 2013;6:59.
- Rust MK. Insecticide resistance in fleas. Insects. 2016;7:10.
- 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.
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.
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
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
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
- Bitam I, Dittmar K, Parola P, et al. Flea and flea-borne diseases. Int J Infect Dis. 2010;14:E667-E676.
- Mathison BA, Pritt BS. Laboratory identification of arthropod ectoparasites. Clin Microbiol Rev. 2014;27:48-67.
- Ligon BL. Plague: a review of its history and potential as a biological weapon. Semin Pediatr Infect Dis. 2006;17:161-170.
- Josko D. Yersinia pestis: still a plague in the 21st century. Clin Lab Sci. 2004;17:25-29.
- Plague around the world, 2010–2015. Wkly Epidemiol Rec. 2016;91:89-93.
- 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.
- 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.
- 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.
- 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.
- 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.
- Carniel E. Subtle genetic modifications transformed an enteropathogen into a flea-borne pathogen. Proc Natl Acad Sci U S A. 2014;111:18409-18410.
- 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.
- Aoyagi KL, Brooks BD, Bearden SW, et al. LPS modification promotes maintenance of Yersinia pestis in fleas. Microbiology. 2015;161:628-638.
- Civen R, Ngo V. Murine typhus: an unrecognized suburban vectorborne disease. Clin Infect Dis. 2008;46:913-918.
- 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.
- 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.
- 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.
- 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.
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
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
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
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
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
- Bitam I, Dittmar K, Parola P, et al. Flea and flea-borne diseases. Int J Infect Dis. 2010;14:E667-E676.
- Mathison BA, Pritt BS. Laboratory identification of arthropod ectoparasites. Clin Microbiol Rev. 2014;27:48-67.
- Ligon BL. Plague: a review of its history and potential as a biological weapon. Semin Pediatr Infect Dis. 2006;17:161-170.
- Josko D. Yersinia pestis: still a plague in the 21st century. Clin Lab Sci. 2004;17:25-29.
- Plague around the world, 2010–2015. Wkly Epidemiol Rec. 2016;91:89-93.
- 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.
- 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.
- 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.
- 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.
- 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.
- Carniel E. Subtle genetic modifications transformed an enteropathogen into a flea-borne pathogen. Proc Natl Acad Sci U S A. 2014;111:18409-18410.
- 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.
- Aoyagi KL, Brooks BD, Bearden SW, et al. LPS modification promotes maintenance of Yersinia pestis in fleas. Microbiology. 2015;161:628-638.
- Civen R, Ngo V. Murine typhus: an unrecognized suburban vectorborne disease. Clin Infect Dis. 2008;46:913-918.
- 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.
- 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.
- 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.
- 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.
- Bitam I, Dittmar K, Parola P, et al. Flea and flea-borne diseases. Int J Infect Dis. 2010;14:E667-E676.
- Mathison BA, Pritt BS. Laboratory identification of arthropod ectoparasites. Clin Microbiol Rev. 2014;27:48-67.
- Ligon BL. Plague: a review of its history and potential as a biological weapon. Semin Pediatr Infect Dis. 2006;17:161-170.
- Josko D. Yersinia pestis: still a plague in the 21st century. Clin Lab Sci. 2004;17:25-29.
- Plague around the world, 2010–2015. Wkly Epidemiol Rec. 2016;91:89-93.
- 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.
- 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.
- 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.
- 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.
- 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.
- Carniel E. Subtle genetic modifications transformed an enteropathogen into a flea-borne pathogen. Proc Natl Acad Sci U S A. 2014;111:18409-18410.
- 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.
- Aoyagi KL, Brooks BD, Bearden SW, et al. LPS modification promotes maintenance of Yersinia pestis in fleas. Microbiology. 2015;161:628-638.
- Civen R, Ngo V. Murine typhus: an unrecognized suburban vectorborne disease. Clin Infect Dis. 2008;46:913-918.
- 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.
- 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.
- 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.
- 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.
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.
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.5 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
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
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
- Villas-Boas IM, Bonfa G, Tambourgi DV. Venomous caterpillars: from inoculation apparatus to venom composition and envenomation. Toxicon. 2018;153:39-52.
- Hossler EW. Caterpillars and moths: part I. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:1-10; quiz 11-12.
- 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.
- 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.
- Pappano DA, Trout Fryxell R, Warren M. Oral mucosal envenomation of an infant by a puss caterpillar. Pediatr Emerg Care. 2017;33:424-426.
- Forrester MB. Megalopyge opercularis caterpillar stings reported to Texas poison centers. Wilderness Environ Med. 2018;29:215-220.
- Hossler EW. Caterpillars and moths: part II. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:13-28; quiz 29-30.
- Eagleman DM. Envenomation by the asp caterpillar (Megalopyge opercularis). Clin Toxicol (Phila). 2008;46:201-205.
- 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.
- Langley RL. Animal bites and stings reported by United States Poison Control Centers, 2001-2005. Wilderness Environ Med. 2008;19:7-14.
- 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.
- 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.
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.5 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
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
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.5 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
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
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
- Villas-Boas IM, Bonfa G, Tambourgi DV. Venomous caterpillars: from inoculation apparatus to venom composition and envenomation. Toxicon. 2018;153:39-52.
- Hossler EW. Caterpillars and moths: part I. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:1-10; quiz 11-12.
- 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.
- 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.
- Pappano DA, Trout Fryxell R, Warren M. Oral mucosal envenomation of an infant by a puss caterpillar. Pediatr Emerg Care. 2017;33:424-426.
- Forrester MB. Megalopyge opercularis caterpillar stings reported to Texas poison centers. Wilderness Environ Med. 2018;29:215-220.
- Hossler EW. Caterpillars and moths: part II. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:13-28; quiz 29-30.
- Eagleman DM. Envenomation by the asp caterpillar (Megalopyge opercularis). Clin Toxicol (Phila). 2008;46:201-205.
- 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.
- Langley RL. Animal bites and stings reported by United States Poison Control Centers, 2001-2005. Wilderness Environ Med. 2008;19:7-14.
- 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.
- 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.
- Villas-Boas IM, Bonfa G, Tambourgi DV. Venomous caterpillars: from inoculation apparatus to venom composition and envenomation. Toxicon. 2018;153:39-52.
- Hossler EW. Caterpillars and moths: part I. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:1-10; quiz 11-12.
- 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.
- 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.
- Pappano DA, Trout Fryxell R, Warren M. Oral mucosal envenomation of an infant by a puss caterpillar. Pediatr Emerg Care. 2017;33:424-426.
- Forrester MB. Megalopyge opercularis caterpillar stings reported to Texas poison centers. Wilderness Environ Med. 2018;29:215-220.
- Hossler EW. Caterpillars and moths: part II. dermatologic manifestations of encounters with Lepidoptera. J Am Acad Dermatol. 2010;62:13-28; quiz 29-30.
- Eagleman DM. Envenomation by the asp caterpillar (Megalopyge opercularis). Clin Toxicol (Phila). 2008;46:201-205.
- 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.
- Langley RL. Animal bites and stings reported by United States Poison Control Centers, 2001-2005. Wilderness Environ Med. 2008;19:7-14.
- 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.
- 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.
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.