Patho-immunological mechanisms of vitiligo: the role of the innate and adaptive immunities and environmental stress factors

Safa Faraj, Elizabeth Helen Kemp, David John Gawkrodger, Safa Faraj, Elizabeth Helen Kemp, David John Gawkrodger

Abstract

Epidermal melanocyte loss in vitiligo, triggered by stresses ranging from trauma to emotional stress, chemical exposure or metabolite imbalance, to the unknown, can stimulate oxidative stress in pigment cells, which secrete damage-associated molecular patterns that then initiate innate immune responses. Antigen presentation to melanocytes leads to stimulation of autoreactive T-cell responses, with further targeting of pigment cells. Studies show a pathogenic basis for cellular stress, innate immune responses and adaptive immunity in vitiligo. Improved understanding of the aetiological mechanisms in vitiligo has already resulted in successful use of the Jak inhibitors in vitiligo. In this review, we outline the current understanding of the pathological mechanisms in vitiligo and locate loci to which therapeutic attack might be directed.

Keywords: Th1/Th2 cells; antibodies; autoimmunity; cytokines; cytotoxic T cells.

© The Author(s) 2021. Published by Oxford University Press on behalf of the British Society for Immunology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.

Figures

Fig. 1
Fig. 1
Melanocytes are regularly exposed to environmental insults such as phenolic compounds can stimulate a state of melanocyte stress through interacting with tyrosinase and TYRP1, leading to production of DAMPs. DAMPs can then stimulate nearby dendritic cells through PRRs. Activated dendritic cells locally synthesize cytokines, inducing CD8+ T cell activation and recruitment to the skin. HMGB1 can activate ILCs with subsequent release of IFN-γ. In the progressive phase of vitiligo, melanocyte-reactive CD8+T cells produce interferon-gamma on encountering melanocyte antigens. These induce keratinocytes to secrete CXCL 9 and CXCL 10, resulting in additional recruitment of lymphocytes to the site through the CXCR3 receptor. CD8+ T cells, resident in vitiligo skin, are involved through the induction of IL-15, which results in the stimulation of IFN-γ, granzyme B and perforin. DAMPs; pathogens or damage-associated molecular patterns, TYRP1; tyrosinase-related protein-1, PRRs; pattern recognition receptors, HMGB1; high-mobility group protein B1, ILCs; innate lymphoid cells, IFN-γ; interferon (IFN)-γ, CXCR3; C-X-C chemokine receptor type 3, CXCL 9/10; C-X-C motif chemokine ligand 9/10, IL-15; interleukin-15.

References

    1. Ezzedine K, Lim HW, Suzuki T, et al. .; Vitiligo Global Issue Consensus Conference Panelists . Revised classification/nomenclature of vitiligo and related issues: the Vitiligo Global Issues Consensus Conference. Pigment Cell Melanoma Res 2012, 25, E1–13.
    1. Manga P, Elbuluk N, Orlow SJ.. Recent advances in understanding vitiligo. F1000Research 2016, 5, 2234.
    1. Al Abadie MS, Gawkrodger DJ.. Integrating neuronal involvement into the immune and genetic paradigm of vitiligo. Clin Exp Dermatol 2021, 46, 646–50.
    1. Zhang Y, Cai Y, Shi M, et al. .. The prevalence of vitiligo: a meta-analysis. PLoS One 2016, 11, e0163806.
    1. Kent G, Al’Abadie M.. Psychologic effects of vitiligo: a critical incident analysis. J Am Acad Dermatol 1996, 35, 895–8.
    1. Nogueira LS, Zancanaro PC, Azambuja RD.. Vitiligo and emotions. An Bras Dermatol 2009, 84, 41–5.
    1. Kostopoulou P, Jouary T, Quintard B, et al. .. Objective vs. subjective factors in the psychological impact of vitiligo: the experience from a French referral centre. Br J Dermatol 2009, 161, 128–33.
    1. Parsad D, Dogra S, Kanwar AJ.. Quality of life in patients with vitiligo. Health Qual Life Outcomes 2003, 1, 58.
    1. Porter J, Beuf A, Nordlund JJ, Lerner AB.. Personal responses of patients to vitiligo: the importance of the patient-physician interaction. Arch Dermatol 1978, 114, 1384–5.
    1. Sukan M, Maner F.. The problems in sexual functions of vitiligo and chronic urticaria patients. J Sex Marital Ther 2007, 33, 55–64.
    1. Alikhan A, Felsten LM, Daly M, Petronic-Rosic V.. Vitiligo: a comprehensive overview Part I. Introduction, epidemiology, quality of life, diagnosis, differential diagnosis, associations, histopathology, etiology, and work-up. J Am Acad Dermatol 2011, 65, 473–91.
    1. Parsad D, Pandhi R, Dogra S, Kanwar AJ, Kumar B.. Dermatology Life Quality Index score in vitiligo and its impact on the treatment outcome. Br J Dermatol 2003, 148, 373–4.
    1. Ezzedine K, Eleftheriadou V, Whitton M, van Geel N.. Vitiligo. Lancet 2015, 386, 74–84.
    1. Boissy RE, Manga P.. On the etiology of contact/occupational vitiligo. Pigment Cell Res 2004, 17, 208–14.
    1. Relke N, Gooderham M.. The use of Janus kinase inhibitors in vitiligo: a review of the literature. J Cutan Med Surg 2019, 23, 298–306.
    1. Tobin DJ, Swanson NN, Pittelkow MR, Peters EM, Schallreuter KU.. Melanocytes are not absent in lesional skin of long duration vitiligo. J Pathol 2000, 191, 407–16.
    1. Ackerman AB, Kerl H, Sánchez J.. A Clinical Atlas of 101 Common Skin Diseases: with Histopathologic Correlation. New York: Ardor Scribendi, 2000.
    1. van den Wijngaard R, Wankowicz-Kalinska A, Le Poole C, Tigges B, Westerhof W, Das P.. Local immune response in skin of generalized vitiligo patients. Destruction of melanocytes is associated with the prominent presence of CLA+ T cells at the perilesional site. Lab Invest 2000, 80, 1299–309.
    1. Puri N, Mojamdar M, Ramaiah A.. In vitro growth characteristics of melanocytes obtained from adult normal and vitiligo subjects. J Invest Dermatol 1987, 88, 434–8.
    1. Puri N, Mojamdar M, Ramaiah A.. Growth defects of melanocytes in culture from vitiligo subjects are spontaneously corrected in vivo in repigmenting subjects and can be partially corrected by the addition of fibroblast-derived growth factors in vitro. Arch Dermatol Res 1989, 281, 178–84.
    1. Dell’Anna ML, Mastrofrancesco A, Sala R, et al. .. Antioxidants and narrow band-UVB in the treatment of vitiligo: a double-blind placebo controlled trial. Clin Exp Dermatol 2007, 32, 631–6.
    1. Schallreuter KU, Moore J, Wood JM, et al. .. In vivo and in vitro evidence for hydrogen peroxide (H2O2) accumulation in the epidermis of patients with vitiligo and its successful removal by a UVB-activated pseudocatalase. J Investig Dermatol Symp Proc 1999, 4, 91–6.
    1. Medrano EE, Nordlund JJ.. Successful culture of adult human melanocytes obtained from normal and vitiligo donors. J Invest Dermatol 1990, 95, 441–5.
    1. Dell’Anna ML, Ottaviani M, Albanesi V, et al. .. Membrane lipid alterations as a possible basis for melanocyte degeneration in vitiligo. J Invest Dermatol 2007, 127, 1226–33.
    1. Shalbaf M, Gibbons NC, Wood JM, et al. .. Presence of epidermal allantoin further supports oxidative stress in vitiligo. Exp Dermatol 2008, 17, 761–70.
    1. Maresca V, Roccella M, Roccella F, et al. .. Increased sensitivity to peroxidative agents as a possible pathogenic factor of melanocyte damage in vitiligo. J Invest Dermatol 1997, 109, 310–3.
    1. Jimbow K, Chen H, Park JS, Thomas PD.. Increased sensitivity of melanocytes to oxidative stress and abnormal expression of tyrosinase-related protein in vitiligo. Br J Dermatol 2001, 144, 55–65.
    1. Gawkrodger DJ. Pseudocatalase and narrowband ultraviolet B for vitiligo: clearing the picture. Br J Dermatol 2009, 161, 721–2.
    1. Spielvogel RL, Kantor GR.. Pigmentary disorders of the skin. In: Elder DE, Elenitsas R, Johansson BL, Murphy GF (eds), Lever’s Histopathology of the Skin. Philadelphia: Lippincott Williams & Wilkins, 2005, 705–13.
    1. Gilhar A, Pillar T, Eidelman S, Etzioni A.. Vitiligo and idiopathic guttate hypomelanosis. Repigmentation of skin following engraftment onto nude mice. Arch Dermatol 1989, 125, 1363–6.
    1. Li W, Wang S, Xu AE.. Role of in vivo reflectance confocal microscopy in determining stability in vitiligo: a preliminary study. Indian J Dermatol 2013, 58, 429–32.
    1. Rashighi M, Harris JE.. Vitiligo pathogenesis and emerging treatments. Dermatol Clin 2017, 35, 257–65.
    1. Sravani PV, Babu NK, Gopal KV, et al. .. Determination of oxidative stress in vitiligo by measuring superoxide dismutase and catalase levels in vitiliginous and non-vitiliginous skin. Indian J Dermatol Venereol Leprol 2009, 75, 268–71.
    1. Schallreuter KU, Moore J, Wood JM, et al. .. Epidermal H(2)O(2) accumulation alters tetrahydrobiopterin (6BH4) recycling in vitiligo: identification of a general mechanism in regulation of all 6BH4-dependent processes? J Invest Dermatol 2001, 116, 167–74.
    1. Xie H, Zhou F, Liu L, et al. .. Vitiligo: How do oxidative stress-induced autoantigens trigger autoimmunity? J Dermatol Sci 2016, 81, 3–9.
    1. Wang Y, Li S, Li C.. Perspectives of new advances in the pathogenesis of vitiligo: from oxidative stress to autoimmunity. Med Sci Monit 2019, 25, 1017–23.
    1. Ghanem G, Fabrice J.. Tyrosinase related protein 1 (TYRP1/gp75) in human cutaneous melanoma. Mol Oncol 2011, 5, 150–5.
    1. Todd DJ, Lee AH, Glimcher LH.. The endoplasmic reticulum stress response in immunity and autoimmunity. Nat Rev Immunol 2008, 8, 663–74.
    1. Ren Y, Yang S, Xu S, et al. .. Genetic variation of promoter sequence modulates XBP1 expression and genetic risk for vitiligo. PLoS Genet 2009, 5, e1000523.
    1. Toosi S, Orlow SJ, Manga P.. Vitiligo-inducing phenols activate the unfolded protein response in melanocytes resulting in upregulation of IL6 and IL8. J Invest Dermatol 2012, 132, 2601–9.
    1. Singh M, Jadeja SD, Vaishnav J, et al. .. Investigation of the role of interleukin 6 in vitiligo pathogenesis. Immunol Invest 2020, 1–18.
    1. James EA, Pietropaolo M, Mamula MJ.. Immune recognition of β-Cells: Neoepitopes as key players in the loss of tolerance. Diabetes 2018, 67, 1035–42.
    1. Frisoli ML, Essien K, Harris JE.. Vitiligo: Mechanisms of pathogenesis and treatment. Annu Rev Immunol 2020, 38, 621–48.
    1. Doss RW, El-Rifaie AA, Abdel-Wahab AM, Gohary YM, Rashed LA.. Heat Shock Protein-70 expression in vitiligo and its relation to the disease activity. Indian J Dermatol 2016, 61, 408–12.
    1. Levandowski CB, Mailloux CM, Ferrara TM, et al. .. NLRP1 haplotypes associated with vitiligo and autoimmunity increase interleukin-1β processing via the NLRP1 inflammasome. Proc Natl Acad Sci USA 2013, 110, 2952–6.
    1. Richmond JM, Frisoli ML, Harris JE.. Innate immune mechanisms in vitiligo: danger from within. Curr Opin Immunol 2013, 25, 676–82.
    1. Yu R, Broady R, Huang Y, et al. .. Transcriptome analysis reveals markers of aberrantly activated innate immunity in vitiligo lesional and non-lesional skin. PLoS One 2012, 7, e51040.
    1. Sun L, Liu W, Zhang LJ.. The role of toll-like receptors in skin host defense, psoriasis, and atopic dermatitis. J Immunol Res 2019, 2019, 1824624.
    1. Mosenson JA, Zloza A, Nieland JD, et al. .. Mutant HSP70 reverses autoimmune depigmentation in vitiligo. Sci Transl Med 2013, 5, 174ra28.
    1. Denman CJ, McCracken J, Hariharan V, et al. .. HSP70i accelerates depigmentation in a mouse model of autoimmune vitiligo. J Invest Dermatol 2008, 128, 2041–8.
    1. Henning SW, Fernandez MF, Mahon JP, et al. .. HSP70iQ435A-encoding DNA repigments vitiligo lesions in sinclair swine. J Invest Dermatol 2018, 138, 2531–9.
    1. Zhang H, Wang W, Li Q, Huang W.. Fusion protein of ATPase domain of Hsc70 with TRP2 acting as a tumor vaccine against B16 melanoma. Immunol Lett 2006, 105, 167–73.
    1. Kroll TM, Bommiasamy H, Boissy RE, et al. .. 4-Tertiary butyl phenol exposure sensitizes human melanocytes to dendritic cell-mediated killing: relevance to vitiligo. J Invest Dermatol 2005, 124, 798–806.
    1. Mosenson JA, Flood K, Klarquist J, et al. .. Preferential secretion of inducible HSP70 by vitiligo melanocytes under stress. Pigment Cell Melanoma Res 2014, 27, 209–20.
    1. Zhang Y, Liu L, Jin L, et al. .. Oxidative stress-induced calreticulin expression and translocation: new insights into the destruction of melanocytes. J Invest Dermatol 2014, 134, 183–91.
    1. Kim JY, Lee EJ, Seo J, Oh SH.. Impact of high-mobility group box 1 on melanocytic survival and its involvement in the pathogenesis of vitiligo. Br J Dermatol 2017, 176, 1558–68.
    1. Cui T, Zhang W, Li S, et al. .. Oxidative Stress-Induced HMGB1 release from melanocytes: a paracrine mechanism underlying the cutaneous inflammation in vitiligo. J Invest Dermatol 2019, 139, 2174–84.e4.
    1. Andersson U, Yang H, Harris H.. High-mobility group box 1 protein (HMGB1) operates as an alarmin outside as well as inside cells. Semin Immunol 2018, 38, 40–8.
    1. Rodrigues M, Ezzedine K, Hamzavi I, Pandya AG, Harris JE; Vitiligo Working Group . New discoveries in the pathogenesis and classification of vitiligo. J Am Acad Dermatol 2017, 77, 1–13.
    1. van den Boorn JG, Picavet DI, van Swieten PF, et al. .. Skin-depigmenting agent monobenzone induces potent T-cell autoimmunity toward pigmented cells by tyrosinase haptenation and melanosome autophagy. J Invest Dermatol 2011, 131, 1240–51.
    1. Bertolotti A, Boniface K, Vergier B, et al. .. Type I interferon signature in the initiation of the immune response in vitiligo. Pigment Cell Melanoma Res 2014, 27, 398–407.
    1. Tulic MK, Cavazza E, Cheli Y, et al. .. Innate lymphocyte-induced CXCR3B-mediated melanocyte apoptosis is a potential initiator of T-cell autoreactivity in vitiligo. Nat Commun 2019, 10, 2178.
    1. Li S, Kang P, Zhang W, et al. .. Activated NLR family pyrin domain containing 3 (NLRP3) inflammasome in keratinocytes promotes cutaneous T-cell response in patients with vitiligo. J Allergy Clin Immunol 2020, 145, 632–45.
    1. Tang L, Zhou F.. Inflammasomes in common immune-related skin diseases. Front Immunol 2020, 11, 882.
    1. van den Boorn JG, Jakobs C, Hagen C, et al. .. Inflammasome-dependent induction of adaptive NK cell memory. Immunity 2016, 44, 1406–21.
    1. Thornberry NA, Bull HG, Calaycay JR, et al. .. A novel heterodimeric cysteine protease is required for interleukin-1 beta processing in monocytes. Nature 1992, 356, 768–74.
    1. Gu Y, Kuida K, Tsutsui H, et al. .. Activation of interferon-gamma inducing factor mediated by interleukin-1beta converting enzyme. Science 1997, 275, 206–9.
    1. Speeckaert R, van Geel N.. Vitiligo: An update on pathophysiology and treatment options. Am J Clin Dermatol 2017, 18, 733–44.
    1. Badri AM, Todd PM, Garioch JJ, Gudgeon JE, Stewart DG, Goudie RB.. An immunohistological study of cutaneous lymphocytes in vitiligo. J Pathol 1993, 170, 149–55.
    1. Wańkowicz-Kalińska A, van den Wijngaard RM, Tigges BJ, et al. .. Immunopolarization of CD4+ and CD8+ T cells to Type-1-like is associated with melanocyte loss in human vitiligo. Lab Invest 2003, 83, 683–95.
    1. Le Poole IC, van den Wijngaard RM, Westerhof W, Das PK.. Presence of T cells and macrophages in inflammatory vitiligo skin parallels melanocyte disappearance. Am J Pathol 1996, 148, 1219–28.
    1. Ogg GS, Rod Dunbar P, Romero P, Chen JL, Cerundolo V.. High frequency of skin-homing melanocyte-specific cytotoxic T lymphocytes in autoimmune vitiligo. J Exp Med 1998, 188, 1203–8.
    1. Strassner JP, Rashighi M, Ahmed Refat M, Richmond JM, Harris JE.. Suction blistering the lesional skin of vitiligo patients reveals useful biomarkers of disease activity. J Am Acad Dermatol 2017, 76, 847–855.e5.
    1. van Geel NA, Mollet IG, De Schepper S, et al. .. First histopathological and immunophenotypic analysis of early dynamic events in a patient with segmental vitiligo associated with halo nevi. Pigment Cell Melanoma Res 2010, 23, 375–84.
    1. Palermo B, Campanelli R, Garbelli S, et al. .. Specific cytotoxic T lymphocyte responses against Melan-A/MART1, tyrosinase and gp100 in vitiligo by the use of major histocompatibility complex/peptide tetramers: the role of cellular immunity in the etiopathogenesis of vitiligo. J Invest Dermatol 2001, 117, 326–32.
    1. van den Boorn JG, Konijnenberg D, Dellemijn TA, et al. .. Autoimmune destruction of skin melanocytes by perilesional T cells from vitiligo patients. J Invest Dermatol 2009, 129, 2220–32.
    1. Boniface K, Jacquemin C, Darrigade AS, et al. .. Vitiligo skin is imprinted with resident memory CD8 T cells expressing CXCR3. J Invest Dermatol 2018, 138, 355–64.
    1. Grimes PE, Morris R, Avaniss-Aghajani E, Soriano T, Meraz M, Metzger A.. Topical tacrolimus therapy for vitiligo: therapeutic responses and skin messenger RNA expression of proinflammatory cytokines. J Am Acad Dermatol 2004, 51, 52–61.
    1. Rashighi M, Agarwal P, Richmond JM, et al. .. CXCL10 is critical for the progression and maintenance of depigmentation in a mouse model of vitiligo. Sci Transl Med 2014, 6, 223ra23.
    1. Wang XX, Wang QQ, Wu JQ, et al. .. Increased expression of CXCR3 and its ligands in patients with vitiligo and CXCL10 as a potential clinical marker for vitiligo. Br J Dermatol 2016, 174, 1318–26.
    1. Harris JE, Harris TH, Weninger W, Wherry EJ, Hunter CA, Turka LA.. A mouse model of vitiligo with focused epidermal depigmentation requires IFN-γ for autoreactive CD8+ T-cell accumulation in the skin. J Invest Dermatol 2012, 132, 1869–76.
    1. Richmond JM, Bangari DS, Essien KI, et al. .. Keratinocyte-derived chemokines orchestrate T-cell positioning in the epidermis during vitiligo and may serve as biomarkers of disease. J Invest Dermatol 2017, 137, 350–8.
    1. Rosmarin D, Pandya AG, Lebwohl M, et al. .. Ruxolitinib cream for treatment of vitiligo: a randomised, controlled, phase 2 trial. Lancet 2020, 396, 110–20.
    1. Moraes-Vasconcelos D, Costa-Carvalho BT, Torgerson TR, Ochs HD.. Primary immune deficiency disorders presenting as autoimmune diseases: IPEX and APECED. J Clin Immunol 2008, 28 Suppl 1, S11–9.
    1. Birlea SA, Jin Y, Bennett DC, et al. .. Comprehensive association analysis of candidate genes for generalized vitiligo supports XBP1, FOXP3, and TSLP. J Invest Dermatol 2011, 131, 371–81.
    1. Lahl K, Loddenkemper C, Drouin C, et al. .. Selective depletion of Foxp3+ regulatory T cells induces a scurfy-like disease. J Exp Med 2007, 204, 57–63.
    1. Eby JM, Kang HK, Klarquist J, et al. .. Immune responses in a mouse model of vitiligo with spontaneous epidermal de- and repigmentation. Pigment Cell Melanoma Res 2014, 27, 1075–85.
    1. Gregg RK, Nichols L, Chen Y, Lu B, Engelhard VH.. Mechanisms of spatial and temporal development of autoimmune vitiligo in tyrosinase-specific TCR transgenic mice. J Immunol 2010, 184, 1909–17.
    1. Chatterjee S, Eby JM, Al-Khami AA, et al. .. A quantitative increase in regulatory T cells controls development of vitiligo. J Invest Dermatol 2014, 134, 1285–94.
    1. Eby JM, Kang HK, Tully ST, et al. .. CCL22 to activate treg migration and suppress depigmentation in vitiligo. J Invest Dermatol 2015, 135, 1574–80.
    1. Miao X, Xu R, Fan B, et al. .. PD-L1 reverses depigmentation in Pmel-1 vitiligo mice by increasing the abundance of Tregs in the skin. Sci Rep 2018, 8, 1605.
    1. Mukhatayev Z, Dellacecca ER, Cosgrove C, et al. .. Antigen specificity enhances disease control by tregs in vitiligo. Front Immunol 2020, 11, 581433.
    1. Lili Y, Yi W, Ji Y, Yue S, Weimin S, Ming L.. Global activation of CD8+ cytotoxic T lymphocytes correlates with an impairment in regulatory T cells in patients with generalized vitiligo. PLoS One 2012, 7, e37513.
    1. Klarquist J, Denman CJ, Hernandez C, et al. .. Reduced skin homing by functional Treg in vitiligo. Pigment Cell Melanoma Res 2010, 23, 276–86.
    1. Ben Ahmed M, Zaraa I, Rekik R, et al. .. Functional defects of peripheral regulatory T lymphocytes in patients with progressive vitiligo. Pigment Cell Melanoma Res 2012, 25, 99–109.
    1. Terras S, Gambichler T, Moritz RK, Altmeyer P, Lambert J.. Immunohistochemical analysis of FOXP3+ regulatory T cells in healthy human skin and autoimmune dermatoses. Int J Dermatol 2014, 53, 294–9.
    1. Abdallah M, Lotfi R, Othman W, Galal R.. Assessment of tissue FoxP3+, CD4+ and CD8+ T-cells in active and stable nonsegmental vitiligo. Int J Dermatol 2014, 53, 940–6.
    1. Maeda Y, Nishikawa H, Sugiyama D, et al. .. Detection of self-reactive CD8+ T cells with an anergic phenotype in healthy individuals. Science 2014, 346, 1536–40.
    1. Zhen Y, Yao L, Zhong S, Song Y, Cui Y, Li S.. Enhanced Th1 and Th17 responses in peripheral blood in active non-segmental vitiligo. Arch Dermatol Res 2016, 308, 703–10.
    1. Martins C, Darrigade AS, Jacquemin C, et al. .. Phenotype and function of circulating memory T cells in human vitiligo. Br J Dermatol 2020, 183, 899–908.
    1. Matos TR. Is targeting circulating T blood cells a therapeutic option for vitiligo? Br J Dermatol 2020, 183, 803.
    1. Speeckaert R, Mylle S, van Geel N.. IL-17A is not a treatment target in progressive vitiligo. Pigment Cell Melanoma Res 2019, 32, 842–7.
    1. Giordano D, Magri F, Persechino F, et al. .. Vitiligo with progressive repigmentation during secukinumab treatment in a patient with Psoriatic Arthritis: A case report. Case Rep Dermatol 2021, 13, 209–15.
    1. Cavalié M, Ezzedine K, Fontas E, et al. .. Maintenance therapy of adult vitiligo with 0.1% tacrolimus ointment: a randomized, double blind, placebo-controlled study. J Invest Dermatol 2015, 135, 970–4.
    1. Sitek JC, Loeb M, Ronnevig JR.. Narrowband UVB therapy for vitiligo: does the repigmentation last? J Eur Acad Dermatol Venereol 2007, 21, 891–6.
    1. Mueller SN, Gebhardt T, Carbone FR, Heath WR.. Memory T cell subsets, migration patterns, and tissue residence. Annu Rev Immunol 2013, 31, 137–61.
    1. Steinbach K, Vincenti I, Merkler D.. Resident-memory T cells in tissue-restricted immune responses: for better or worse? Front Immunol 2018, 9, 2827.
    1. Dijkgraaf FE, Matos TR, Hoogenboezem M, et al. .. Tissue patrol by resident memory CD8+ T cells in human skin. Nat Immunol 2019, 20, 756–64.
    1. Topham DJ, Reilly EC.. Tissue-resident memory CD8+ T cells: from phenotype to function. Front Immunol 2018, 9, 515.
    1. Frączek A, Owczarczyk-Saczonek A, Placek W.. The role of T(RM) cells in the pathogenesis of vitiligo-a review of the current state-of-the-art. Int J Mol Sci 2020, 21, 552.
    1. Mackay LK, Braun A, Macleod BL, et al. .. Cutting edge: CD69 interference with sphingosine-1-phosphate receptor function regulates peripheral T cell retention. J Immunol 2015, 194, 2059–63.
    1. Skon CN, Lee JY, Anderson KG, Masopust D, Hogquist KA, Jameson SC.. Transcriptional downregulation of S1pr1 is required for the establishment of resident memory CD8+ T cells. Nat Immunol 2013, 14, 1285–93.
    1. Mackay LK, Rahimpour A, Ma JZ, et al. .. The developmental pathway for CD103(+)CD8+ tissue-resident memory T cells of skin. Nat Immunol 2013, 14, 1294–301.
    1. Cheuk S, Schlums H, Gallais Sérézal I, et al. .. CD49a expression defines tissue-resident CD8+ T cells poised for cytotoxic function in human skin. Immunity 2017, 46, 287–300.
    1. Richmond JM, Strassner JP, Zapata L, et al. .. Antibody blockade of IL-15 signaling has the potential to durably reverse vitiligo. Sci Transl Med 2018, 10, eaam7710.
    1. Budagian V, Bulanova E, Paus R, Bulfone-Paus S.. IL-15/IL-15 receptor biology: a guided tour through an expanding universe. Cytokine Growth Factor Rev 2006, 17, 259–80.
    1. Jacquemin C, Martins C, Lucchese F, et al. .. NKG2D Defines a subset of skin effector memory CD8 T Cells with proinflammatory functions in vitiligo. J Invest Dermatol 2020, 140, 1143–1153.e5.
    1. Jiang X, Clark RA, Liu L, Wagers AJ, Fuhlbrigge RC, Kupper TS.. Skin infection generates non-migratory memory CD8+ T(RM) cells providing global skin immunity. Nature 2012, 483, 227–31.
    1. Mackay LK, Stock AT, Ma JZ, et al. .. Long-lived epithelial immunity by tissue-resident memory T (TRM) cells in the absence of persisting local antigen presentation. Proc Natl Acad Sci USA 2012, 109, 7037–42.
    1. Seidel JA, Vukmanovic-Stejic M, Muller-Durovic B, et al. .. Skin resident memory CD8+ T cells are phenotypically and functionally distinct from circulating populations and lack immediate cytotoxic function. Clin Exp Immunol 2018, 194, 79–92.
    1. McMaster SR, Wilson JJ, Wang H, Kohlmeier JE.. Airway-Resident Memory CD8 T Cells provide antigen-specific protection against respiratory virus challenge through rapid IFN-γ production. J Immunol 2015, 195, 203–9.
    1. Malik BT, Byrne KT, Vella JL, et al. .. Resident memory T cells in the skin mediate durable immunity to melanoma. Sci Immunol 2017, 2, eaam6346.
    1. Richmond JM, Strassner JP, Rashighi M, et al. .. Resident memory and recirculating memory T cells cooperate to maintain disease in a mouse model of vitiligo. J Invest Dermatol 2019, 139, 769–78.
    1. Matos TR, O’Malley JT, Lowry EL, et al. .. Clinically resolved psoriatic lesions contain psoriasis-specific IL-17-producing αβ T cell clones. J Clin Invest 2017, 127, 4031–41.
    1. Naughton GK, Eisinger M, Bystryn JC.. Antibodies to normal human melanocytes in vitiligo. J Exp Med 1983, 158, 246–51.
    1. Naughton GK, Reggiardo D, Bystryn JC.. Correlation between vitiligo antibodies and extent of depigmentation in vitiligo. J Am Acad Dermatol 1986, 15, 978–81.
    1. Rocha IM, Oliveira LJ, De Castro LC, et al. .. Recognition of melanoma cell antigens with antibodies present in sera from patients with vitiligo. Int J Dermatol 2000, 39, 840–3.
    1. Farrokhi S, Hojjat-Farsangi M, Noohpisheh MK, Tahmasbi R, Rezaei N.. Assessment of the immune system in 55 Iranian patients with vitiligo. J Eur Acad Dermatol Venereol 2005, 19, 706–11.
    1. Harning R, Cui J, Bystryn JC.. Relation between the incidence and level of pigment cell antibodies and disease activity in vitiligo. J Invest Dermatol 1991, 97, 1078–80.
    1. Yu HS, Kao CH, Yu CL.. Coexistence and relationship of antikeratinocyte and antimelanocyte antibodies in patients with non-segmental-type vitiligo. J Invest Dermatol 1993, 100, 823–8.
    1. Abu Tahir M, Pramod K, Ansari SH, Ali J.. Current remedies for vitiligo. Autoimmun Rev 2010, 9, 516–20.
    1. Xie P, Geoghegan W, Jordon R. Vitiligo autoantibodies. Studies of subclass distribution and complement activation. J Invest Dermatol 1991, 96, 627.
    1. Aronson PJ, Hashimoto K.. Association of IgA anti-melanoma antibodies in the sera of vitiligo patients with active disease. J Invest Dermatol 1987, 88, 475.
    1. Zhu MC, Liu CG, Wang DX, Zhan Z.. Detection of serum anti-melanocyte antibodies and identification of related antigens in patients with vitiligo. Genet Mol Res 2015, 14, 16060–73.
    1. Cui J, Arita Y, Bystryn JC.. Characterization of vitiligo antigens. Pigment Cell Res 1995, 8, 53–9.
    1. Park YK, Kim NS, Hann SK, Im S.. Identification of autoantibody to melanocytes and characterization of vitiligo antigen in vitiligo patients. J Dermatol Sci 1996, 11, 111–20.
    1. Song YH, Connor E, Li Y, Zorovich B, Balducci P, Maclaren N.. The role of tyrosinase in autoimmune vitiligo. Lancet 1994, 344, 1049–52.
    1. Baharav E, Merimski O, Shoenfeld Y, et al. .. Tyrosinase as an autoantigen in patients with vitiligo. Clin Exp Immunol 1996, 105, 84–8.
    1. Kemp EH, Gawkrodger DJ, MacNeil S, Watson PF, Weetman AP.. Detection of tyrosinase autoantibodies in patients with vitiligo using 35S-labeled recombinant human tyrosinase in a radioimmunoassay. J Invest Dermatol 1997, 109, 69–73.
    1. Kemp EH, Gawkrodger DJ, Watson PF, Weetman AP.. Immunoprecipitation of melanogenic enzyme autoantigens with vitiligo sera: evidence for cross-reactive autoantibodies to tyrosinase and tyrosinase-related protein-2 (TRP-2). Clin Exp Immunol 1997, 109, 495–500.
    1. Kemp EH, Gawkrodger DJ, Watson PF, Weetman AP.. Autoantibodies to human melanocyte-specific protein pmel17 in the sera of vitiligo patients: a sensitive and quantitative radioimmunoassay (RIA). Clin Exp Immunol 1998, 114, 333–8.
    1. Kemp EH, Waterman EA, Gawkrodger DJ, Watson PF, Weetman AP.. Autoantibodies to tyrosinase-related protein-1 detected in the sera of vitiligo patients using a quantitative radiobinding assay. Br J Dermatol 1998, 139, 798–805.
    1. Ruiz-Argüelles A, García-Carrasco M, Jimenez-Brito G, et al. .. Treatment of vitiligo with a chimeric monoclonal antibody to CD20: a pilot study. Clin Exp Immunol 2013, 174, 229–36.
    1. Mandry RC, Ortíz LJ, Lugo-Somolinos A, Sánchez JL.. Organ-specific autoantibodies in vitiligo patients and their relatives. Int J Dermatol 1996, 35, 18–21.
    1. Kemp EH, Waterman EA, Hawes BE, et al. .. The melanin-concentrating hormone receptor 1, a novel target of autoantibody responses in vitiligo. J Clin Invest 2002, 109, 923–30.
    1. Waterman EA, Gawkrodger DJ, Watson PF, Weetman AP, Kemp EH.. Autoantigens in vitiligo identified by the serological selection of a phage-displayed melanocyte cDNA expression library. J Invest Dermatol 2010, 130, 230–40.
    1. Faraj S, Gawkrodger D, Weetman A, Kemp EH.. An investigation of lamin A autoantibodies in vitiligo. J Invest Dermatol 2017, 137, S14.
    1. Norris DA, Kissinger RM, Naughton GM, Bystryn JC.. Evidence for immunologic mechanisms in human vitiligo: patients’ sera induce damage to human melanocytes in vitro by complement-mediated damage and antibody-dependent cellular cytotoxicity. J Invest Dermatol 1988, 90, 783–9.
    1. Gilhar A, Zelickson B, Ulman Y, Etzioni A.. In vivo destruction of melanocytes by the IgG fraction of serum from patients with vitiligo. J Invest Dermatol 1995, 105, 683–6.
    1. Gottumukkala RV, Gavalas NG, Akhtar S, et al. .. Function-blocking autoantibodies to the melanin-concentrating hormone receptor in vitiligo patients. Lab Invest 2006, 86, 781–9.
    1. Yohn JJ, Morelli JG, Walchak SJ, Rundell KB, Norris DA, Zamora MR.. Cultured human keratinocytes synthesize and secrete endothelin-1. J Invest Dermatol 1993, 100, 23–6.
    1. Kemp EH, Waterman EA, Weetman AP.. Autoimmune aspects of vitiligo. Autoimmunity 2001, 34, 65–77.
    1. Homey B, Assmann T, Vohr HW, et al. .. Topical FK506 suppresses cytokine and costimulatory molecule expression in epidermal and local draining lymph node cells during primary skin immune responses. J Immunol 1998, 160, 5331–40.
    1. Hartmann A, Bröcker EB, Hamm H.. Occlusive treatment enhances efficacy of tacrolimus 0.1% ointment in adult patients with vitiligo: results of a placebo-controlled 12-month prospective study. Acta Derm Venereol 2008, 88, 474–9.
    1. Gawkrodger DJ, Ormerod AD, Shaw L, et al. .. Vitiligo: concise evidence based guidelines on diagnosis and management. Postgrad Med J 2010, 86, 466–71.
    1. Lee CH, Wu SB, Hong CH, Yu HS, Wei YH.. Molecular mechanisms of UV-induced apoptosis and its effects on skin residential cells: the implication in UV-based phototherapy. Int J Mol Sci 2013, 14, 6414–35.
    1. Fishman P, Azizi E, Shoenfeld Y, et al. .. Vitiligo autoantibodies are effective against melanoma. Cancer 1993, 72, 2365–9.
    1. Cario-André M, Pain C, Gauthier Y, Taïeb A.. The melanocytorrhagic hypothesis of vitiligo tested on pigmented, stressed, reconstructed epidermis. Pigment Cell Res 2007, 20, 385–93.
    1. Kemp EH, Waterman EA, Weetman AP.. Immunological pathomechanisms in vitiligo. Expert Rev Mol Med 2001, 3, 1–22.
    1. Takechi Y, Hara I, Naftzger C, Xu Y, Houghton AN.. A melanosomal membrane protein is a cell surface target for melanoma therapy. Clin Cancer Res 1996, 2, 1837–42.
    1. Leonhardt RM, Vigneron N, Rahner C, Cresswell P.. Proprotein convertases process Pmel17 during secretion. J Biol Chem 2011, 286, 9321–37.
    1. Martínez-Lostao L, Anel A, Pardo J.. How do cytotoxic lymphocytes kill cancer cells? Clin Cancer Res 2015, 21, 5047–56.
    1. Hassin D, Garber OG, Meiraz A, Schiffenbauer YS, Berke G.. Cytotoxic T lymphocyte perforin and Fas ligand working in concert even when Fas ligand lytic action is still not detectable. Immunology 2011, 133, 190–6.
    1. Spritz RA, Andersen GH.. Genetics of Vitiligo. Dermatol Clin 2017, 35, 245–55.
    1. Bethune MT, Joglekar AV.. Personalized T cell-mediated cancer immunotherapy: progress and challenges. Curr Opin Biotechnol 2017, 48, 142–52.
    1. Leisegang M, Kammertoens T, Uckert W, Blankenstein T.. Targeting human melanoma neoantigens by T cell receptor gene therapy. J Clin Invest 2016, 126, 854–8.
    1. Singh M, Kotnis A, Jadeja SD, Mondal A, Mansuri MS, Begum R.. Cytokines: the yin and yang of vitiligo pathogenesis. Expert Rev Clin Immunol 2019, 15, 177–88.
    1. Singh S, Singh U, Pandey SS.. Serum concentration of IL-6, IL-2, TNF-α, and IFNγ in Vitiligo patients. Indian J Dermatol 2012, 57, 12–4.
    1. Miniati A, Weng Z, Zhang B, et al. .. Stimulated human melanocytes express and release interleukin-8, which is inhibited by luteolin: relevance to early vitiligo. Clin Exp Dermatol 2014, 39, 54–7.
    1. Gholijani N, Yazdani MR, Dastgheib L.. Predominant role of innate pro-inflammatory cytokines in vitiligo disease. Arch Dermatol Res 2020, 312, 123–31.
    1. Moretti S, Fabbri P, Baroni G, et al. .. Keratinocyte dysfunction in vitiligo epidermis: cytokine microenvironment and correlation to keratinocyte apoptosis. Histol Histopathol 2009, 24, 849–57.
    1. Boukhedouni N, Martins C, Darrigade A-S, et al. .. Type-1 cytokines regulate MMP-9 production and E-cadherin disruption to promote melanocyte loss in vitiligo. JCI Insight 2020, 5, e133772.
    1. Harris JE, Rashighi M, Nguyen N, Jabbari A, et al. .. Rapid skin repigmentation on oral ruxolitinib in a patient with coexistent vitiligo and alopecia areata (AA). J Am Acad Dermatol 2016, 74, 370–1.
    1. van Elsas A, Hurwitz AA, Allison JP.. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J Exp Med 1999, 190, 355–66.
    1. Fishman P, Merimski O, Baharav E, Shoenfeld Y.. Autoantibodies to tyrosinase: the bridge between melanoma and vitiligo. Cancer 1997, 79, 1461–4.
    1. Huang SK, Okamoto T, Morton DL, Hoon DS.. Antibody responses to melanoma/melanocyte autoantigens in melanoma patients. J Invest Dermatol 1998, 111, 662–7.
    1. Kirkin AF, Dzhandzhugazyan K, Zeuthen J.. Melanoma-associated antigens recognized by cytotoxic T lymphocytes. APMIS 1998, 106, 665–79.
    1. Edwards J, Wilmott JS, Madore J, et al. .. CD103 + tumor-resident CD8 + T cells are associated with improved survival in immunotherapy-naïve melanoma patients and expand significantly during anti-PD-1 treatment. Clinical Cancer Research 2018, 24, 3036.
    1. Teulings HE, Limpens J, Jansen SN, et al. .. Vitiligo-like depigmentation in patients with stage III-IV melanoma receiving immunotherapy and its association with survival: a systematic review and meta-analysis. J Clin Oncol 2015, 33, 773–81.
    1. Hua C, Boussemart L, Mateus C, et al. .. Association of vitiligo with tumor response in patients with metastatic melanoma treated with Pembrolizumab. JAMA Dermatol 2016, 152, 45–51.
    1. Yee C, Thompson JA, Roche P, et al. .. Melanocyte destruction after antigen-specific immunotherapy of melanoma: direct evidence of t cell-mediated vitiligo. J Exp Med 2000, 192, 1637–44.
    1. Lo JA, Kawakubo M, Juneja VR, et al. .. Epitope spreading toward wild-type melanocyte-lineage antigens rescues suboptimal immune checkpoint blockade responses. Sci Transl Med 2021, 13, eabd8636.
    1. Tatli AM, Besen AA, Kalender ME, et al. .. Association of vitiligo and response in patients with metastatic malignant melanoma on temozolomide. Tumori 2015, 101, e67–9.
    1. Bergqvist C, Ezzedine K.. Vitiligo: a review. Dermatology 2020, 236, 571–92.
    1. Jin Y, Birlea SA, Fain PR, et al. .. Variant of TYR and autoimmunity susceptibility loci in generalized vitiligo. N Engl J Med 2010, 362, 1686–97.
    1. Spritz RA. The genetics of generalized vitiligo: autoimmune pathways and an inverse relationship with malignant melanoma. Genome Med 2010, 2, 78.
    1. Gudbjartsson DF, Sulem P, Stacey SN, et al. .. ASIP and TYR pigmentation variants associate with cutaneous melanoma and basal cell carcinoma. Nat Genet 2008, 40, 886–91.
    1. Alkhateeb A, Fain PR, Thody A, Bennett DC, Spritz RA.. Epidemiology of vitiligo and associated autoimmune diseases in Caucasian probands and their families. Pigment Cell Res 2003, 16, 208–14.
    1. Picardo M, Dell’Anna ML, Ezzedine K, et al. .. Vitiligo. Nat Rev Dis Primers 2015, 1, 15011.
    1. Lerner AB. Vitiligo. J Invest Dermatol 1959, 32, 285–310.
    1. Jin Y, Birlea SA, Fain PR, et al. .. Genome-wide association analyses identify 13 new susceptibility loci for generalized vitiligo. Nat Genet 2012, 44, 676–80.
    1. Tang XF, Zhang Z, Hu DY, et al. .. Association analyses identify three susceptibility Loci for vitiligo in the Chinese Han population. J Invest Dermatol 2013, 133, 403–10.
    1. Liu JB, Li M, Chen H, et al. .. Association of vitiligo with HLA-A2: a meta-analysis. J Eur Acad Dermatol Venereol 2007, 21, 205–13.
    1. Jin Y, Hayashi M, Fain PR, et al. .. Major association of vitiligo with HLA-A∗02:01 in Japanese. Pigment Cell Melanoma Res 2015, 28, 360–2.
    1. Fain PR, Babu SR, Bennett DC, Spritz RA.. HLA class II haplotype DRB1∗04-DQB1∗0301 contributes to risk of familial generalized vitiligo and early disease onset. Pigment Cell Res 2006, 19, 51–7.
    1. Quan C, Ren YQ, Xiang LH, et al. .. Genome-wide association study for vitiligo identifies susceptibility loci at 6q27 and the MHC. Nat Genet 2010, 42, 614–8.
    1. Tazi-Ahnini R, McDonagh AJ, Wengraf DA, et al. .. The autoimmune regulator gene (AIRE) is strongly associated with vitiligo. Br J Dermatol 2008, 159, 591–6.
    1. Oftedal BE, Hellesen A, Erichsen MM, et al. .. Dominant mutations in the autoimmune regulator AIRE are associated with common organ-specific autoimmune diseases. Immunity 2015, 42, 1185–96.
    1. Kemp EH, Ajjan RA, Waterman EA, et al. .. Analysis of a microsatellite polymorphism of the cytotoxic T-lymphocyte antigen-4 gene in patients with vitiligo. Br J Dermatol 1999, 140, 73–8.
    1. Birlea SA, Laberge GS, Procopciuc LM, Fain PR, Spritz RA.. CTLA4 and generalized vitiligo: two genetic association studies and a meta-analysis of published data. Pigment Cell Melanoma Res 2009, 22, 230–4.
    1. Pehlivan S, Ozkinay F, Alper S, et al. .. Association between IL4 (-590), ACE (I)/(D), CCR5 (Delta32), CTLA4 (+49) and IL1-RN (VNTR in intron 2) gene polymorphisms and vitiligo. Eur J Dermatol 2009, 19, 126–8.
    1. Li M, Sun D, Li C, et al. .. Functional polymorphisms of the FAS gene associated with risk of vitiligo in Chinese populations: a case-control analysis. J Invest Dermatol 2008, 128, 2820–4.
    1. Jin Y, Birlea SA, Fain PR, et al. .. Common variants in FOXP1 are associated with generalized vitiligo. Nat Genet 2010, 42, 576–8.
    1. Ferrara TM, Jin Y, Gowan K, Fain PR, Spritz RA.. Risk of generalized vitiligo is associated with the common 55R-94A-247H variant haplotype of GZMB (encoding granzyme B). J Invest Dermatol 2013, 133, 1677–9.
    1. Jin Y, Birlea SA, Fain PR, Spritz RA.. Genetic variations in NALP1 are associated with generalized vitiligo in a Romanian population. J Invest Dermatol 2007, 127, 2558–62.
    1. Jin Y, Mailloux CM, Gowan K, et al. .. NALP1 in vitiligo-associated multiple autoimmune disease. N Engl J Med 2007, 356, 1216–25.
    1. Alkhateeb A, Qarqaz F.. Genetic association of NALP1 with generalized vitiligo in Jordanian Arabs. Arch Dermatol Res 2010, 302, 631–4.
    1. Cantón I, Akhtar S, Gavalas NG, et al. .. A single-nucleotide polymorphism in the gene encoding lymphoid protein tyrosine phosphatase (PTPN22) confers susceptibility to generalised vitiligo. Genes Immun 2005, 6, 584–7.
    1. LaBerge GS, Bennett DC, Fain PR, Spritz RA.. PTPN22 is genetically associated with risk of generalized vitiligo, but CTLA4 is not. J Invest Dermatol 2008, 128, 1757–62.
    1. Laberge GS, Birlea SA, Fain PR, Spritz RA.. The PTPN22-1858C>T (R620W) functional polymorphism is associated with generalized vitiligo in the Romanian population. Pigment Cell Melanoma Res 2008, 21, 206–8.
    1. Laddha NC, Dwivedi M, Shajil EM, Prajapati H, Marfatia YS, Begum R.. Association of PTPN22 1858C/T polymorphism with vitiligo susceptibility in Gujarat population. J Dermatol Sci 2008, 49, 260–2.
    1. Song GG, Kim JH, Lee YH.. The CTLA-4 + 49 A/G, CT60 A/G and PTPN22 1858 C/T polymorphisms and susceptibility to vitiligo: a meta-analysis. Mol Biol Rep 2013, 40, 2985–93.
    1. Garcia-Melendez ME, Salinas-Santander M, Sanchez-Dominguez C, et al. .. Protein tyrosine phosphatase PTPN22 + 1858C/T polymorphism is associated with active vitiligo. Exp Ther Med 2014, 8, 1433–7.
    1. Na GY, Lee KH, Kim MK, Lee SJ, Kim DW, Kim JC.. Polymorphisms in the melanocortin-1 receptor (MC1R) and agouti signaling protein (ASIP) genes in Korean vitiligo patients. Pigment Cell Res 2003, 16, 383–7.
    1. Alkhateeb A, Fain PR, Spritz RA.. Candidate functional promoter variant in the FOXD3 melanoblast developmental regulator gene in autosomal dominant vitiligo. J Invest Dermatol 2005, 125, 388–91.
    1. Jin Y, Ferrara T, Gowan K, et al. .. Next-generation DNA re-sequencing identifies common variants of TYR and HLA-A that modulate the risk of generalized vitiligo via antigen presentation. J Invest Dermatol 2012, 132, 1730–3.
    1. Sun Y, Zuo X, Zheng X, et al. .. A comprehensive association analysis confirms ZMIZ1 to be a susceptibility gene for vitiligo in Chinese population. J Med Genet 2014, 51, 345–53.
    1. Jin SY, Park HH, Li GZ, et al. .. Association of angiotensin converting enzyme gene I/D polymorphism of vitiligo in Korean population. Pigment Cell Res 2004, 17, 84–6.
    1. Wood JM, Gibbons NC, Chavan B, Schallreuter KU.. Computer simulation of heterogeneous single nucleotide polymorphisms in the catalase gene indicates structural changes in the enzyme active site, NADPH-binding and tetramerization domains: a genetic predisposition for an altered catalase in patients with vitiligo? Exp Dermatol 2008, 17, 366–71.
    1. Liu L, Li C, Gao J, et al. .. Promoter variant in the catalase gene is associated with vitiligo in Chinese people. J Invest Dermatol 2010, 130, 2647–53.
    1. Mansuri MS, Jadeja SD, Singh M, Laddha NC, Dwivedi M, Begum R.. The catalase gene promoter and 5’-untranslated region variants lead to altered gene expression and enzyme activity in vitiligo. Br J Dermatol 2017, 177, 1590–600.
    1. Kim HJ, Choi CP, Uhm YK, et al. .. The association between endothelin-1 gene polymorphisms and susceptibility to vitiligo in a Korean population. Exp Dermatol 2007, 16, 561–6.
    1. Gokhale BB, Mehta LN.. Histopathology of vitiliginous skin. Int J Dermatol 1983, 22, 477–80.
    1. Nordlands JJ. Vitiligo. In: Thiers BH, Dobson RL (eds), In: Pathogenesis of Skin Diseases. New York: Churchill Livingstone, 1986, 99–128.
    1. Al’Abadie MS, Warren MA, Bleehen SS, Gawkrodger DJ.. Morphologic observations on the dermal nerves in vitiligo: an ultrastructural study. Int J Dermatol 1995, 34, 837–40.
    1. Al’Abadie MS, Senior HJ, Bleehen SS, Gawkrodger DJ.. Neuropeptide and neuronal marker studies in vitiligo. Br J Dermatol 1994, 131, 160–5.
    1. Dimitrijević M, Stanojević S.. The intriguing mission of neuropeptide Y in the immune system. Amino Acids 2013, 45, 41–53.
    1. Ganea D, Hooper KM, Kong W.. The neuropeptide vasoactive intestinal peptide: direct effects on immune cells and involvement in inflammatory and autoimmune diseases. Acta Physiol (Oxf) 2015, 213, 442–52.
    1. Taams LS. Neuroimmune interactions: how the nervous and immune systems influence each other. Clin Exp Immunol 2019, 197, 276–7.
    1. Kundu RV, Mhlaba JM, Rangel SM, Le Poole IC.. The convergence theory for vitiligo: A reappraisal. Exp Dermatol 2019, 28, 647–55.
    1. Cucchi ML, Frattini P, Santagostino G, Orecchia G.. Higher plasma catecholamine and metabolite levels in the early phase of nonsegmental vitiligo. Pigment Cell Res 2000, 13, 28–32.
    1. Gauthier Y, Cario-Andre M, Lepreux S, Pain C, Taïeb A.. Melanocyte detachment after skin friction in non lesional skin of patients with generalized vitiligo. Br J Dermatol 2003, 148, 95–101.
    1. Pichler R, Sfetsos K, Badics B, Gutenbrunner S, Auböck J.. Vitiligo patients present lower plasma levels of alpha-melanotropin immunoreactivities. Neuropeptides 2006, 40, 177–83.
    1. Harris JE. Chemical-Induced Vitiligo. Dermatol Clin 2017, 35, 151–61.
    1. Westerhof W, d’Ischia M.. Vitiligo puzzle: the pieces fall in place. Pigment Cell Res 2007, 20, 345–59.
    1. Namazi MR. Neurogenic dysregulation, oxidative stress, autoimmunity, and melanocytorrhagy in vitiligo: can they be interconnected? Pigment Cell Res 2007, 20, 360–3.

Source: PubMed

3
Suscribir