Skip to main content
Top
Published in: Dermatology and Therapy 1/2024

Open Access 06-01-2024 | Atopic Dermatitis | Review

Cutaneous Components Leading to Pruritus, Pain, and Neurosensitivity in Atopic Dermatitis: A Narrative Review

Authors: Sonja Ständer, Thomas Luger, Brian Kim, Ethan Lerner, Martin Metz, Roni Adiri, Juliana M. Canosa, Amy Cha, Gil Yosipovitch

Published in: Dermatology and Therapy | Issue 1/2024

Login to get access

Abstract

Atopic dermatitis (AD) is a chronic, relapsing immunoinflammatory skin condition characterized by sensations such as pruritis, pain, and neuronal hypersensitivity. The mechanisms underlying these sensations are multifactorial and involve complex crosstalk among several cutaneous components. This review explores the role these components play in the pathophysiology of atopic dermatitis. In the skin intercellular spaces, sensory nerves interact with keratinocytes and immune cells via myriad mediators and receptors. These interactions generate action potentials that transmit pruritis and pain signals from the peripheral nervous system to the brain. Keratinocytes, the most abundant cell type in the epidermis, are key effector cells, triggering crosstalk with immune cells and sensory neurons to elicit pruritis, pain, and inflammation. Filaggrin expression by keratinocytes is reduced in atopic dermatitis, causing a weakened skin barrier and elevated skin pH. Fibroblasts are the main cell type in the dermis and, in atopic dermatitis, appear to reduce keratinocyte differentiation, further weakening the skin barrier. Fibroblasts and mast cells promote inflammation while dermal dendritic cells appear to attenuate inflammation. Inflammatory cytokines and chemokines play a major role in AD pathogenesis. Type 2 immune responses typically generate pruritis, and the type 1 and type 3 responses generate pain. Type 2 responses and increased skin pH contribute to barrier dysfunction and promote dysbiosis of the skin microbiome, causing the proliferation of Staphyloccocus aureus. In conclusion, understanding the dynamic interactions between cutaneous components in AD could drive the development of therapies to improve the quality of life for patients with AD.
Literature
1.
go back to reference Boguniewicz M, Fonacier L, Guttman-Yassky E, Ong PY, Silverberg J, Farrar JR. Atopic dermatitis yardstick: practical recommendations for an evolving therapeutic landscape. Ann Allergy Asthma Immunol. 2018;120:10-22.e2.PubMedCrossRef Boguniewicz M, Fonacier L, Guttman-Yassky E, Ong PY, Silverberg J, Farrar JR. Atopic dermatitis yardstick: practical recommendations for an evolving therapeutic landscape. Ann Allergy Asthma Immunol. 2018;120:10-22.e2.PubMedCrossRef
4.
go back to reference Bylund S, Kobyletzki LB, Svalstedt M, Svensson Å. Prevalence and incidence of atopic dermatitis: a systematic review. Acta Derm Venereol. 2020;100:adv00160.PubMedCrossRef Bylund S, Kobyletzki LB, Svalstedt M, Svensson Å. Prevalence and incidence of atopic dermatitis: a systematic review. Acta Derm Venereol. 2020;100:adv00160.PubMedCrossRef
5.
go back to reference Murota H, Koike Y, Morisaki H, Matsumoto M, Takenaka M. Exacerbating factors and disease burden in patients with atopic dermatitis. Allergol Int. 2022;71:25–30.PubMedCrossRef Murota H, Koike Y, Morisaki H, Matsumoto M, Takenaka M. Exacerbating factors and disease burden in patients with atopic dermatitis. Allergol Int. 2022;71:25–30.PubMedCrossRef
7.
go back to reference von Kobyletzki LB, Thomas KS, Schmitt J, Chalmers JR, Deckert S, Aoki V, et al. What factors are important to patients when assessing treatment response: an international cross-sectional survey. Acta Derm Venereol. 2017;97:86–90.CrossRef von Kobyletzki LB, Thomas KS, Schmitt J, Chalmers JR, Deckert S, Aoki V, et al. What factors are important to patients when assessing treatment response: an international cross-sectional survey. Acta Derm Venereol. 2017;97:86–90.CrossRef
8.
go back to reference Li JX, Dong RJ, Zeng YP. Characteristics, mechanism, and management of pain in atopic dermatitis: a literature review. Clin Transl Allergy. 2021;11: e12079.PubMedPubMedCentralCrossRef Li JX, Dong RJ, Zeng YP. Characteristics, mechanism, and management of pain in atopic dermatitis: a literature review. Clin Transl Allergy. 2021;11: e12079.PubMedPubMedCentralCrossRef
9.
go back to reference Weidinger S, Beck LA, Bieber T, Kabashima K, Irvine AD. Atopic dermatitis. Nat Rev Dis Primers. 2018;4:1.PubMedCrossRef Weidinger S, Beck LA, Bieber T, Kabashima K, Irvine AD. Atopic dermatitis. Nat Rev Dis Primers. 2018;4:1.PubMedCrossRef
10.
go back to reference Misery L, Belloni Fortina A, El Hachem M, Chernyshov P, von Kobyletzki L, Heratizadeh A, et al. A position paper on the management of itch and pain in atopic dermatitis from the international society of atopic dermatitis (isad)/oriented patient-education network in dermatology (opened) task force. J Eur Acad Dermatol Venereol. 2021;35:787–96.PubMedCrossRef Misery L, Belloni Fortina A, El Hachem M, Chernyshov P, von Kobyletzki L, Heratizadeh A, et al. A position paper on the management of itch and pain in atopic dermatitis from the international society of atopic dermatitis (isad)/oriented patient-education network in dermatology (opened) task force. J Eur Acad Dermatol Venereol. 2021;35:787–96.PubMedCrossRef
12.
go back to reference Vakharia PP, Chopra R, Sacotte R, Patel KR, Singam V, Patel N, et al. Burden of skin pain in atopic dermatitis. Ann Allergy Asthma Immunol. 2017;119(548–52): e3. Vakharia PP, Chopra R, Sacotte R, Patel KR, Singam V, Patel N, et al. Burden of skin pain in atopic dermatitis. Ann Allergy Asthma Immunol. 2017;119(548–52): e3.
13.
go back to reference Huet F, Shourick J, Séité S, Taïeb C, Misery L. Pain in atopic dermatitis: an online population-based survey. Acta Derm Venereol. 2020;100:adv00198.PubMedCrossRef Huet F, Shourick J, Séité S, Taïeb C, Misery L. Pain in atopic dermatitis: an online population-based survey. Acta Derm Venereol. 2020;100:adv00198.PubMedCrossRef
14.
go back to reference Dawn A, Papoiu AD, Chan YH, Rapp SR, Rassette N, Yosipovitch G. Itch characteristics in atopic dermatitis: results of a web-based questionnaire. Br J Dermatol. 2009;160:642–4.PubMedCrossRef Dawn A, Papoiu AD, Chan YH, Rapp SR, Rassette N, Yosipovitch G. Itch characteristics in atopic dermatitis: results of a web-based questionnaire. Br J Dermatol. 2009;160:642–4.PubMedCrossRef
15.
go back to reference Pojawa-Gołąb M, Reich A. Skin pain in patients with atopic dermatitis or psoriasis: a web-based survey. Acta Derm Venereol. 2020;100:adv00258.PubMedCrossRef Pojawa-Gołąb M, Reich A. Skin pain in patients with atopic dermatitis or psoriasis: a web-based survey. Acta Derm Venereol. 2020;100:adv00258.PubMedCrossRef
16.
go back to reference Maarouf M, Kromenacker B, Capozza KL, Kempton D, Hendricks A, Tran K, et al. Pain and itch are dual burdens in atopic dermatitis. Dermatitis. 2018;29:278–81.PubMedCrossRef Maarouf M, Kromenacker B, Capozza KL, Kempton D, Hendricks A, Tran K, et al. Pain and itch are dual burdens in atopic dermatitis. Dermatitis. 2018;29:278–81.PubMedCrossRef
17.
go back to reference Andersen HH, Elberling J, Sølvsten H, Yosipovitch G, Arendt-Nielsen L. Nonhistaminergic and mechanical itch sensitization in atopic dermatitis. Pain. 2017;158:1780–91.PubMedCrossRef Andersen HH, Elberling J, Sølvsten H, Yosipovitch G, Arendt-Nielsen L. Nonhistaminergic and mechanical itch sensitization in atopic dermatitis. Pain. 2017;158:1780–91.PubMedCrossRef
18.
go back to reference Rosen JD, Fostini AC, Chan YH, Nattkemper LA, Yosipovitch G. Cross-sectional study of clinical distinctions between neuropathic and inflammatory pruritus. J Am Acad Dermatol. 2018;79:1143–4.PubMedCrossRef Rosen JD, Fostini AC, Chan YH, Nattkemper LA, Yosipovitch G. Cross-sectional study of clinical distinctions between neuropathic and inflammatory pruritus. J Am Acad Dermatol. 2018;79:1143–4.PubMedCrossRef
20.
go back to reference Liu T, Ji RR. New insights into the mechanisms of itch: are pain and itch controlled by distinct mechanisms? Pflugers Arch. 2013;465:1671–85.PubMedCrossRef Liu T, Ji RR. New insights into the mechanisms of itch: are pain and itch controlled by distinct mechanisms? Pflugers Arch. 2013;465:1671–85.PubMedCrossRef
21.
go back to reference Silverberg JI, Gelfand JM, Margolis DJ, Boguniewicz M, Fonacier L, Grayson MH, et al. Pain is a common and burdensome symptom of atopic dermatitis in united states adults. J Allergy Clin Immunol Pract. 2019;7(2699–706): e7. Silverberg JI, Gelfand JM, Margolis DJ, Boguniewicz M, Fonacier L, Grayson MH, et al. Pain is a common and burdensome symptom of atopic dermatitis in united states adults. J Allergy Clin Immunol Pract. 2019;7(2699–706): e7.
22.
go back to reference Tominaga M, Takamori K. Peripheral itch sensitization in atopic dermatitis. Allergol Int. 2022;71:265–77.PubMedCrossRef Tominaga M, Takamori K. Peripheral itch sensitization in atopic dermatitis. Allergol Int. 2022;71:265–77.PubMedCrossRef
23.
go back to reference Yosipovitch G, Berger T, Fassett MS. Neuroimmune interactions in chronic itch of atopic dermatitis. J Eur Acad Dermatol Venereol. 2020;34:239–50.PubMedCrossRef Yosipovitch G, Berger T, Fassett MS. Neuroimmune interactions in chronic itch of atopic dermatitis. J Eur Acad Dermatol Venereol. 2020;34:239–50.PubMedCrossRef
24.
go back to reference Ikoma A, Steinhoff M, Ständer S, Yosipovitch G, Schmelz M. The neurobiology of itch. Nat Rev Neurosci. 2006;7:535–47.PubMedCrossRef Ikoma A, Steinhoff M, Ständer S, Yosipovitch G, Schmelz M. The neurobiology of itch. Nat Rev Neurosci. 2006;7:535–47.PubMedCrossRef
25.
26.
go back to reference Salimian J, Salehi Z, Ahmadi A, Emamvirdizadeh A, Davoudi SM, Karimi M, et al. Atopic dermatitis: Molecular, cellular, and clinical aspects. Mol Biol Rep. 2022;49:3333–48.PubMedCrossRef Salimian J, Salehi Z, Ahmadi A, Emamvirdizadeh A, Davoudi SM, Karimi M, et al. Atopic dermatitis: Molecular, cellular, and clinical aspects. Mol Biol Rep. 2022;49:3333–48.PubMedCrossRef
27.
go back to reference Lawton S. Skin 1: The structure and functions of the skin. Nurs Times. 2019;115:30–3. Lawton S. Skin 1: The structure and functions of the skin. Nurs Times. 2019;115:30–3.
28.
go back to reference Mollanazar NK, Smith PK, Yosipovitch G. Mediators of chronic pruritus in atopic dermatitis: getting the itch out? Clin Rev Allergy Immunol. 2016;51:263–92.PubMedCrossRef Mollanazar NK, Smith PK, Yosipovitch G. Mediators of chronic pruritus in atopic dermatitis: getting the itch out? Clin Rev Allergy Immunol. 2016;51:263–92.PubMedCrossRef
31.
go back to reference Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D. The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature. 1997;389:816–24.PubMedCrossRef Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D. The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature. 1997;389:816–24.PubMedCrossRef
32.
go back to reference Yosipovitch G, Rosen JD, Hashimoto T. Itch: from mechanism to (novel) therapeutic approaches. J Allergy Clin Immunol. 2018;142:1375–90.PubMedCrossRef Yosipovitch G, Rosen JD, Hashimoto T. Itch: from mechanism to (novel) therapeutic approaches. J Allergy Clin Immunol. 2018;142:1375–90.PubMedCrossRef
33.
go back to reference Han L, Ma C, Liu Q, Weng HJ, Cui Y, Tang Z, et al. A subpopulation of nociceptors specifically linked to itch. Nat Neurosci. 2013;16:174–82.PubMedCrossRef Han L, Ma C, Liu Q, Weng HJ, Cui Y, Tang Z, et al. A subpopulation of nociceptors specifically linked to itch. Nat Neurosci. 2013;16:174–82.PubMedCrossRef
34.
go back to reference Nattkemper LA, Tey HL, Valdes-Rodriguez R, Lee H, Mollanazar NK, Albornoz C, et al. The genetics of chronic itch: gene expression in the skin of patients with atopic dermatitis and psoriasis with severe itch. J Invest Dermatol. 2018;138:1311–7.PubMedCrossRef Nattkemper LA, Tey HL, Valdes-Rodriguez R, Lee H, Mollanazar NK, Albornoz C, et al. The genetics of chronic itch: gene expression in the skin of patients with atopic dermatitis and psoriasis with severe itch. J Invest Dermatol. 2018;138:1311–7.PubMedCrossRef
35.
go back to reference Trier AM, Mack MR, Kim BS. The neuroimmune axis in skin sensation, inflammation, and immunity. J Immunol. 2019;202:2829–35.PubMedCrossRef Trier AM, Mack MR, Kim BS. The neuroimmune axis in skin sensation, inflammation, and immunity. J Immunol. 2019;202:2829–35.PubMedCrossRef
37.
go back to reference Shin SM, Moehring F, Itson-Zoske B, Fan F, Stucky CL, Hogan QH, et al. Piezo2 mechanosensitive ion channel is located to sensory neurons and nonneuronal cells in rat peripheral sensory pathway: implications in pain. Pain. 2021;162:2750–68.PubMedPubMedCentralCrossRef Shin SM, Moehring F, Itson-Zoske B, Fan F, Stucky CL, Hogan QH, et al. Piezo2 mechanosensitive ion channel is located to sensory neurons and nonneuronal cells in rat peripheral sensory pathway: implications in pain. Pain. 2021;162:2750–68.PubMedPubMedCentralCrossRef
38.
39.
go back to reference Tominaga M, Ogawa H, Takamori K. Decreased production of semaphorin 3a in the lesional skin of atopic dermatitis. Br J Dermatol. 2008;158:842–4.PubMedCrossRef Tominaga M, Ogawa H, Takamori K. Decreased production of semaphorin 3a in the lesional skin of atopic dermatitis. Br J Dermatol. 2008;158:842–4.PubMedCrossRef
40.
go back to reference Nakahara T, Kido-Nakahara M, Tsuji G, Furue M. Basics and recent advances in the pathophysiology of atopic dermatitis. J Dermatol. 2021;48:130–9.PubMedCrossRef Nakahara T, Kido-Nakahara M, Tsuji G, Furue M. Basics and recent advances in the pathophysiology of atopic dermatitis. J Dermatol. 2021;48:130–9.PubMedCrossRef
41.
go back to reference Guseva D, Rüdrich U, Kotnik N, Gehring M, Patsinakidis N, Agelopoulos K, et al. Neuronal branching of sensory neurons is associated with bdnf-positive eosinophils in atopic dermatitis. Clin Exp Allergy. 2020;50:577–84.PubMedCrossRef Guseva D, Rüdrich U, Kotnik N, Gehring M, Patsinakidis N, Agelopoulos K, et al. Neuronal branching of sensory neurons is associated with bdnf-positive eosinophils in atopic dermatitis. Clin Exp Allergy. 2020;50:577–84.PubMedCrossRef
42.
go back to reference Sugiura H, Omoto M, Hirota Y, Danno K, Uehara M. Density and fine structure of peripheral nerves in various skin lesions of atopic dermatitis. Arch Dermatol Res. 1997;289:125–31.PubMedCrossRef Sugiura H, Omoto M, Hirota Y, Danno K, Uehara M. Density and fine structure of peripheral nerves in various skin lesions of atopic dermatitis. Arch Dermatol Res. 1997;289:125–31.PubMedCrossRef
43.
go back to reference Urashima R, Mihara M. Cutaneous nerves in atopic dermatitis. A histological, immunohistochemical and electron microscopic study. Virchows Arch. 1998;432:363–70.PubMedCrossRef Urashima R, Mihara M. Cutaneous nerves in atopic dermatitis. A histological, immunohistochemical and electron microscopic study. Virchows Arch. 1998;432:363–70.PubMedCrossRef
44.
go back to reference Tsutsumi M, Kitahata H, Fukuda M, Kumamoto J, Goto M, Denda S, et al. Numerical and comparative three-dimensional structural analysis of peripheral nerve fibres in epidermis of patients with atopic dermatitis. Br J Dermatol. 2016;174:191–4.PubMedCrossRef Tsutsumi M, Kitahata H, Fukuda M, Kumamoto J, Goto M, Denda S, et al. Numerical and comparative three-dimensional structural analysis of peripheral nerve fibres in epidermis of patients with atopic dermatitis. Br J Dermatol. 2016;174:191–4.PubMedCrossRef
45.
go back to reference Tan Y, Ng WJ, Lee SZX, Lee BTK, Nattkemper LA, Yosipovitch G, et al. 3-dimensional optical clearing and imaging of pruritic atopic dermatitis and psoriasis skin reveals downregulation of epidermal innervation. J Invest Dermatol. 2019;139:1201–4.PubMedCrossRef Tan Y, Ng WJ, Lee SZX, Lee BTK, Nattkemper LA, Yosipovitch G, et al. 3-dimensional optical clearing and imaging of pruritic atopic dermatitis and psoriasis skin reveals downregulation of epidermal innervation. J Invest Dermatol. 2019;139:1201–4.PubMedCrossRef
46.
go back to reference Das P, Mounika P, Yellurkar ML, Prasanna VS, Sarkar S, Velayutham R, et al. Keratinocytes: An enigmatic factor in atopic dermatitis. Cells. 2022;11:1683.PubMedPubMedCentralCrossRef Das P, Mounika P, Yellurkar ML, Prasanna VS, Sarkar S, Velayutham R, et al. Keratinocytes: An enigmatic factor in atopic dermatitis. Cells. 2022;11:1683.PubMedPubMedCentralCrossRef
47.
go back to reference Haftek M. Epidermal barrier disorders and corneodesmosome defects. Cell Tissue Res. 2015;360:483–90.PubMedCrossRef Haftek M. Epidermal barrier disorders and corneodesmosome defects. Cell Tissue Res. 2015;360:483–90.PubMedCrossRef
48.
go back to reference Furue M. Regulation of filaggrin, loricrin, and involucrin by il-4, il-13, il-17a, il-22, ahr, and nrf2: pathogenic implications in atopic dermatitis. Int J Mol Sci. 2020;21:5382.PubMedPubMedCentralCrossRef Furue M. Regulation of filaggrin, loricrin, and involucrin by il-4, il-13, il-17a, il-22, ahr, and nrf2: pathogenic implications in atopic dermatitis. Int J Mol Sci. 2020;21:5382.PubMedPubMedCentralCrossRef
49.
go back to reference Moosbrugger-Martinz V, Leprince C, Méchin MC, Simon M, Blunder S, Gruber R, et al. Revisiting the roles of filaggrin in atopic dermatitis. Int J Mol Sci. 2022;23:5318.PubMedPubMedCentralCrossRef Moosbrugger-Martinz V, Leprince C, Méchin MC, Simon M, Blunder S, Gruber R, et al. Revisiting the roles of filaggrin in atopic dermatitis. Int J Mol Sci. 2022;23:5318.PubMedPubMedCentralCrossRef
50.
go back to reference Agelopoulos K, Pereira MP, Wiegmann H, Ständer S. Cutaneous neuroimmune crosstalk in pruritus. Trends Mol Med. 2022;28:452–62.PubMedCrossRef Agelopoulos K, Pereira MP, Wiegmann H, Ständer S. Cutaneous neuroimmune crosstalk in pruritus. Trends Mol Med. 2022;28:452–62.PubMedCrossRef
51.
go back to reference Larkin C, Chen W, Szabó IL, Shan C, Dajnoki Z, Szegedi A, et al. Novel insights into the trpv3-mediated itch in atopic dermatitis. J Allergy Clin Immunol. 2021;147:1110-4.e5.PubMedCrossRef Larkin C, Chen W, Szabó IL, Shan C, Dajnoki Z, Szegedi A, et al. Novel insights into the trpv3-mediated itch in atopic dermatitis. J Allergy Clin Immunol. 2021;147:1110-4.e5.PubMedCrossRef
52.
go back to reference van den Bogaard EH, Esser C, Perdew GH. The aryl hydrocarbon receptor at the forefront of host-microbe interactions in the skin: a perspective on current knowledge gaps and directions for future research and therapeutic applications. Exp Dermatol. 2021;30:1477–83.PubMedPubMedCentralCrossRef van den Bogaard EH, Esser C, Perdew GH. The aryl hydrocarbon receptor at the forefront of host-microbe interactions in the skin: a perspective on current knowledge gaps and directions for future research and therapeutic applications. Exp Dermatol. 2021;30:1477–83.PubMedPubMedCentralCrossRef
53.
go back to reference Wilson SR, Thé L, Batia LM, Beattie K, Katibah GE, McClain SP, et al. The epithelial cell-derived atopic dermatitis cytokine tslp activates neurons to induce itch. Cell. 2013;155:285–95.PubMedPubMedCentralCrossRef Wilson SR, Thé L, Batia LM, Beattie K, Katibah GE, McClain SP, et al. The epithelial cell-derived atopic dermatitis cytokine tslp activates neurons to induce itch. Cell. 2013;155:285–95.PubMedPubMedCentralCrossRef
54.
go back to reference Guo CJ, Mack MR, Oetjen LK, Trier AM, Council ML, Pavel AB, et al. Kallikrein 7 promotes atopic dermatitis-associated itch independently of skin inflammation. J Invest Dermatol. 2020;140:1244-52.e4.PubMedCrossRef Guo CJ, Mack MR, Oetjen LK, Trier AM, Council ML, Pavel AB, et al. Kallikrein 7 promotes atopic dermatitis-associated itch independently of skin inflammation. J Invest Dermatol. 2020;140:1244-52.e4.PubMedCrossRef
55.
go back to reference Hashimoto T, Mishra SK, Olivry T, Yosipovitch G. Periostin, an emerging player in itch sensation. J Invest Dermatol. 2021;141:2338–43.PubMedCrossRef Hashimoto T, Mishra SK, Olivry T, Yosipovitch G. Periostin, an emerging player in itch sensation. J Invest Dermatol. 2021;141:2338–43.PubMedCrossRef
56.
go back to reference Mishra SK, Wheeler JJ, Pitake S, Ding H, Jiang C, Fukuyama T, et al. Periostin activation of integrin receptors on sensory neurons induces allergic itch. Cell Rep. 2020;31: 107472.PubMedPubMedCentralCrossRef Mishra SK, Wheeler JJ, Pitake S, Ding H, Jiang C, Fukuyama T, et al. Periostin activation of integrin receptors on sensory neurons induces allergic itch. Cell Rep. 2020;31: 107472.PubMedPubMedCentralCrossRef
57.
go back to reference Steinhoff M, Ahmad F, Pandey A, Datsi A, AlHammadi A, Al-Khawaga S, et al. Neuroimmune communication regulating pruritus in atopic dermatitis. J Allergy Clin Immunol. 2022;149:1875–98.PubMedCrossRef Steinhoff M, Ahmad F, Pandey A, Datsi A, AlHammadi A, Al-Khawaga S, et al. Neuroimmune communication regulating pruritus in atopic dermatitis. J Allergy Clin Immunol. 2022;149:1875–98.PubMedCrossRef
58.
go back to reference Yu S, Zhou Y, Follansbee T, Hwang ST. Immune mediators and therapies for pruritus in atopic dermatitis and psoriasis. J Cutane Immunol All. 2019;2:4–14.CrossRef Yu S, Zhou Y, Follansbee T, Hwang ST. Immune mediators and therapies for pruritus in atopic dermatitis and psoriasis. J Cutane Immunol All. 2019;2:4–14.CrossRef
59.
go back to reference Hefti FF, Rosenthal A, Walicke PA, Wyatt S, Vergara G, Shelton DL, et al. Novel class of pain drugs based on antagonism of ngf. Trends Pharmacol Sci. 2006;27:85–91.PubMedCrossRef Hefti FF, Rosenthal A, Walicke PA, Wyatt S, Vergara G, Shelton DL, et al. Novel class of pain drugs based on antagonism of ngf. Trends Pharmacol Sci. 2006;27:85–91.PubMedCrossRef
60.
go back to reference Muhl L, Genové G, Leptidis S, Liu J, He L, Mocci G, et al. Single-cell analysis uncovers fibroblast heterogeneity and criteria for fibroblast and mural cell identification and discrimination. Nat Commun. 2020;11:3953.PubMedPubMedCentralCrossRef Muhl L, Genové G, Leptidis S, Liu J, He L, Mocci G, et al. Single-cell analysis uncovers fibroblast heterogeneity and criteria for fibroblast and mural cell identification and discrimination. Nat Commun. 2020;11:3953.PubMedPubMedCentralCrossRef
61.
go back to reference He H, Suryawanshi H, Morozov P, Gay-Mimbrera J, Del Duca E, Kim HJ, et al. Single-cell transcriptome analysis of human skin identifies novel fibroblast subpopulation and enrichment of immune subsets in atopic dermatitis. J Allergy Clin Immunol. 2020;145:1615–28.PubMedCrossRef He H, Suryawanshi H, Morozov P, Gay-Mimbrera J, Del Duca E, Kim HJ, et al. Single-cell transcriptome analysis of human skin identifies novel fibroblast subpopulation and enrichment of immune subsets in atopic dermatitis. J Allergy Clin Immunol. 2020;145:1615–28.PubMedCrossRef
62.
go back to reference Berroth A, Kühnl J, Kurschat N, Schwarz A, Stäb F, Schwarz T, et al. Role of fibroblasts in the pathogenesis of atopic dermatitis. J Allergy Clin Immunol. 2013;131:1547–54.PubMedCrossRef Berroth A, Kühnl J, Kurschat N, Schwarz A, Stäb F, Schwarz T, et al. Role of fibroblasts in the pathogenesis of atopic dermatitis. J Allergy Clin Immunol. 2013;131:1547–54.PubMedCrossRef
63.
go back to reference Levi-Schaffer F, Gibbs BF, Hallgren J, Pucillo C, Redegeld F, Siebenhaar F, et al. Selected recent advances in understanding the role of human mast cells in health and disease. J Allergy Clin Immunol. 2022;149:1833–44.PubMedCrossRef Levi-Schaffer F, Gibbs BF, Hallgren J, Pucillo C, Redegeld F, Siebenhaar F, et al. Selected recent advances in understanding the role of human mast cells in health and disease. J Allergy Clin Immunol. 2022;149:1833–44.PubMedCrossRef
64.
go back to reference Schuler CF, Billi AC, Maverakis E, Tsoi LC, Gudjonsson JE. Novel insights into atopic dermatitis. J Allergy Clin Immunol. 2022;151:1145–54.PubMedCrossRef Schuler CF, Billi AC, Maverakis E, Tsoi LC, Gudjonsson JE. Novel insights into atopic dermatitis. J Allergy Clin Immunol. 2022;151:1145–54.PubMedCrossRef
65.
go back to reference Wernersson S, Pejler G. Mast cell secretory granules: armed for battle. Nat Rev Immunol. 2014;14:478–94.PubMedCrossRef Wernersson S, Pejler G. Mast cell secretory granules: armed for battle. Nat Rev Immunol. 2014;14:478–94.PubMedCrossRef
66.
go back to reference Honda T, Kabashima K. Prostanoids and leukotrienes in the pathophysiology of atopic dermatitis and psoriasis. Int Immunol. 2019;31:589–95.PubMedCrossRef Honda T, Kabashima K. Prostanoids and leukotrienes in the pathophysiology of atopic dermatitis and psoriasis. Int Immunol. 2019;31:589–95.PubMedCrossRef
68.
go back to reference Meixiong J, Anderson M, Limjunyawong N, Sabbagh MF, Hu E, Mack MR, et al. Activation of mast-cell-expressed mas-related g-protein-coupled receptors drives non-histaminergic itch. Immunity. 2019;50:1163-71.e5.PubMedPubMedCentralCrossRef Meixiong J, Anderson M, Limjunyawong N, Sabbagh MF, Hu E, Mack MR, et al. Activation of mast-cell-expressed mas-related g-protein-coupled receptors drives non-histaminergic itch. Immunity. 2019;50:1163-71.e5.PubMedPubMedCentralCrossRef
70.
go back to reference Navratilova E, Porreca F. Substance p and inflammatory pain: getting it wrong and right simultaneously. Neuron. 2019;101:353–5.PubMedCrossRef Navratilova E, Porreca F. Substance p and inflammatory pain: getting it wrong and right simultaneously. Neuron. 2019;101:353–5.PubMedCrossRef
71.
72.
go back to reference West HC, Bennett CL. Redefining the role of langerhans cells as immune regulators within the skin. Front Immunol. 2017;8:1941.PubMedCrossRef West HC, Bennett CL. Redefining the role of langerhans cells as immune regulators within the skin. Front Immunol. 2017;8:1941.PubMedCrossRef
74.
go back to reference Rajesh A, Wise L, Hibma M. The role of langerhans cells in pathologies of the skin. Immunol Cell Biol. 2019;97:700–13.PubMedCrossRef Rajesh A, Wise L, Hibma M. The role of langerhans cells in pathologies of the skin. Immunol Cell Biol. 2019;97:700–13.PubMedCrossRef
75.
go back to reference Wollenberg A, Kraft S, Hanau D, Bieber T. Immunomorphological and ultrastructural characterization of langerhans cells and a novel, inflammatory dendritic epidermal cell (idec) population in lesional skin of atopic eczema. J Invest Dermatol. 1996;106:446–53.PubMedCrossRef Wollenberg A, Kraft S, Hanau D, Bieber T. Immunomorphological and ultrastructural characterization of langerhans cells and a novel, inflammatory dendritic epidermal cell (idec) population in lesional skin of atopic eczema. J Invest Dermatol. 1996;106:446–53.PubMedCrossRef
76.
go back to reference Iwamoto K, Nümm TJ, Koch S, Herrmann N, Leib N, Bieber T. Langerhans and inflammatory dendritic epidermal cells in atopic dermatitis are tolerized toward tlr2 activation. Allergy. 2018;73:2205–13.PubMedCrossRef Iwamoto K, Nümm TJ, Koch S, Herrmann N, Leib N, Bieber T. Langerhans and inflammatory dendritic epidermal cells in atopic dermatitis are tolerized toward tlr2 activation. Allergy. 2018;73:2205–13.PubMedCrossRef
77.
go back to reference Li D, Lei H, Li Z, Li H, Wang Y, Lai Y. A novel lipopeptide from skin commensal activates tlr2/cd36-p38 mapk signaling to increase antibacterial defense against bacterial infection. PLoS ONE. 2013;8: e58288.PubMedPubMedCentralCrossRef Li D, Lei H, Li Z, Li H, Wang Y, Lai Y. A novel lipopeptide from skin commensal activates tlr2/cd36-p38 mapk signaling to increase antibacterial defense against bacterial infection. PLoS ONE. 2013;8: e58288.PubMedPubMedCentralCrossRef
78.
go back to reference Li W, Yosipovitch G. The role of the microbiome and microbiome-derived metabolites in atopic dermatitis and non-histaminergic itch. Am J Clin Dermatol. 2020;21:44–50.PubMedPubMedCentralCrossRef Li W, Yosipovitch G. The role of the microbiome and microbiome-derived metabolites in atopic dermatitis and non-histaminergic itch. Am J Clin Dermatol. 2020;21:44–50.PubMedPubMedCentralCrossRef
79.
80.
go back to reference Kiguchi N, Kobayashi Y, Kishioka S. Chemokines and cytokines in neuroinflammation leading to neuropathic pain. Curr Opin Pharmacol. 2012;12:55–61.PubMedCrossRef Kiguchi N, Kobayashi Y, Kishioka S. Chemokines and cytokines in neuroinflammation leading to neuropathic pain. Curr Opin Pharmacol. 2012;12:55–61.PubMedCrossRef
81.
go back to reference Kwatra SG, Misery L, Clibborn C, Steinhoff M. Molecular and cellular mechanisms of itch and pain in atopic dermatitis and implications for novel therapeutics. Clin Transl Immunology. 2022;11: e1390.PubMedPubMedCentralCrossRef Kwatra SG, Misery L, Clibborn C, Steinhoff M. Molecular and cellular mechanisms of itch and pain in atopic dermatitis and implications for novel therapeutics. Clin Transl Immunology. 2022;11: e1390.PubMedPubMedCentralCrossRef
82.
go back to reference Hashimoto T, Yokozeki H, Karasuyama H, Satoh T. Il-31-generating network in atopic dermatitis comprising macrophages, basophils, thymic stromal lymphopoietin, and periostin. J Allergy Clin Immunol. 2023;151:737-46.e6.PubMedCrossRef Hashimoto T, Yokozeki H, Karasuyama H, Satoh T. Il-31-generating network in atopic dermatitis comprising macrophages, basophils, thymic stromal lymphopoietin, and periostin. J Allergy Clin Immunol. 2023;151:737-46.e6.PubMedCrossRef
83.
go back to reference Furue M, Yamamura K, Kido-Nakahara M, Nakahara T, Fukui Y. Emerging role of interleukin-31 and interleukin-31 receptor in pruritus in atopic dermatitis. Allergy. 2018;73:29–36.PubMedCrossRef Furue M, Yamamura K, Kido-Nakahara M, Nakahara T, Fukui Y. Emerging role of interleukin-31 and interleukin-31 receptor in pruritus in atopic dermatitis. Allergy. 2018;73:29–36.PubMedCrossRef
84.
go back to reference Feld M, Garcia R, Buddenkotte J, Katayama S, Lewis K, Muirhead G, et al. The pruritus- and th2-associated cytokine il-31 promotes growth of sensory nerves. J Allergy Clin Immunol. 2016;138(500–8): e24. Feld M, Garcia R, Buddenkotte J, Katayama S, Lewis K, Muirhead G, et al. The pruritus- and th2-associated cytokine il-31 promotes growth of sensory nerves. J Allergy Clin Immunol. 2016;138(500–8): e24.
85.
go back to reference Oetjen LK, Mack MR, Feng J, Whelan TM, Niu H, Guo CJ, et al. Sensory neurons co-opt classical immune signaling pathways to mediate chronic itch. Cell. 2017;171(217–28):e13. Oetjen LK, Mack MR, Feng J, Whelan TM, Niu H, Guo CJ, et al. Sensory neurons co-opt classical immune signaling pathways to mediate chronic itch. Cell. 2017;171(217–28):e13.
86.
go back to reference Beck LA, Cork MJ, Amagai M, De Benedetto A, Kabashima K, Hamilton JD, et al. Type 2 inflammation contributes to skin barrier dysfunction in atopic dermatitis. JID Innov. 2022;2:100131.PubMedPubMedCentralCrossRef Beck LA, Cork MJ, Amagai M, De Benedetto A, Kabashima K, Hamilton JD, et al. Type 2 inflammation contributes to skin barrier dysfunction in atopic dermatitis. JID Innov. 2022;2:100131.PubMedPubMedCentralCrossRef
87.
90.
go back to reference Paller AS, Kong HH, Seed P, Naik S, Scharschmidt TC, Gallo RL, et al. The microbiome in patients with atopic dermatitis. J Allergy Clin Immunol. 2019;143:26–35.PubMedCrossRef Paller AS, Kong HH, Seed P, Naik S, Scharschmidt TC, Gallo RL, et al. The microbiome in patients with atopic dermatitis. J Allergy Clin Immunol. 2019;143:26–35.PubMedCrossRef
91.
go back to reference Kobayashi T, Glatz M, Horiuchi K, Kawasaki H, Akiyama H, Kaplan DH, et al. Dysbiosis and staphylococcus aureus colonization drives inflammation in atopic dermatitis. Immunity. 2015;42:756–66.PubMedPubMedCentralCrossRef Kobayashi T, Glatz M, Horiuchi K, Kawasaki H, Akiyama H, Kaplan DH, et al. Dysbiosis and staphylococcus aureus colonization drives inflammation in atopic dermatitis. Immunity. 2015;42:756–66.PubMedPubMedCentralCrossRef
92.
go back to reference Ali SM, Yosipovitch G. Skin ph: from basic science to basic skin care. Acta Derm Venereol. 2013;93:261–7.PubMedCrossRef Ali SM, Yosipovitch G. Skin ph: from basic science to basic skin care. Acta Derm Venereol. 2013;93:261–7.PubMedCrossRef
93.
go back to reference Azimi E, Reddy VB, Lerner EA. Brief communication: Mrgprx2, atopic dermatitis and red man syndrome. Itch (Phila). 2017;2:e5.PubMed Azimi E, Reddy VB, Lerner EA. Brief communication: Mrgprx2, atopic dermatitis and red man syndrome. Itch (Phila). 2017;2:e5.PubMed
94.
go back to reference Chiu IM, Heesters BA, Ghasemlou N, Von Hehn CA, Zhao F, Tran J, et al. Bacteria activate sensory neurons that modulate pain and inflammation. Nature. 2013;501:52–7.PubMedPubMedCentralCrossRef Chiu IM, Heesters BA, Ghasemlou N, Von Hehn CA, Zhao F, Tran J, et al. Bacteria activate sensory neurons that modulate pain and inflammation. Nature. 2013;501:52–7.PubMedPubMedCentralCrossRef
95.
96.
go back to reference Ständer S, Kwatra SG, Silverberg JI, Simpson EL, Thyssen JP, Yosipovitch G, et al. Early itch response with abrocitinib is associated with later efficacy outcomes in patients with moderate-to-severe atopic dermatitis: subgroup analysis of the randomized phase iii jade compare trial. Am J Clin Dermatol. 2022;24(1):97–107.PubMedPubMedCentralCrossRef Ständer S, Kwatra SG, Silverberg JI, Simpson EL, Thyssen JP, Yosipovitch G, et al. Early itch response with abrocitinib is associated with later efficacy outcomes in patients with moderate-to-severe atopic dermatitis: subgroup analysis of the randomized phase iii jade compare trial. Am J Clin Dermatol. 2022;24(1):97–107.PubMedPubMedCentralCrossRef
97.
go back to reference Han Y, Woo YR, Cho SH, Lee JD, Kim HS. Itch and janus kinase inhibitors. Acta Derm Venereol. 2023;103:adv00869.PubMedCrossRef Han Y, Woo YR, Cho SH, Lee JD, Kim HS. Itch and janus kinase inhibitors. Acta Derm Venereol. 2023;103:adv00869.PubMedCrossRef
99.
go back to reference Simpson EL, Bieber T, Guttman-Yassky E, Beck LA, Blauvelt A, Cork MJ, et al. Two phase 3 trials of dupilumab versus placebo in atopic dermatitis. N Engl J Med. 2016;375:2335–48.PubMedCrossRef Simpson EL, Bieber T, Guttman-Yassky E, Beck LA, Blauvelt A, Cork MJ, et al. Two phase 3 trials of dupilumab versus placebo in atopic dermatitis. N Engl J Med. 2016;375:2335–48.PubMedCrossRef
Metadata
Title
Cutaneous Components Leading to Pruritus, Pain, and Neurosensitivity in Atopic Dermatitis: A Narrative Review
Authors
Sonja Ständer
Thomas Luger
Brian Kim
Ethan Lerner
Martin Metz
Roni Adiri
Juliana M. Canosa
Amy Cha
Gil Yosipovitch
Publication date
06-01-2024
Publisher
Springer Healthcare
Published in
Dermatology and Therapy / Issue 1/2024
Print ISSN: 2193-8210
Electronic ISSN: 2190-9172
DOI
https://doi.org/10.1007/s13555-023-01081-0

Other articles of this Issue 1/2024

Dermatology and Therapy 1/2024 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.