Transcriptome analysis reveals markers of aberrantly activated innate immunity in vitiligo lesional and non-lesional skin

Richard Yu, Raewyn Broady, Yuanshen Huang, Yang Wang, Jie Yu, Min Gao, Megan Levings, Shencai Wei, Shengquan Zhang, Aie Xu, Mingwan Su, Jan Dutz, Xuejun Zhang, Youwen Zhou, Richard Yu, Raewyn Broady, Yuanshen Huang, Yang Wang, Jie Yu, Min Gao, Megan Levings, Shencai Wei, Shengquan Zhang, Aie Xu, Mingwan Su, Jan Dutz, Xuejun Zhang, Youwen Zhou

Abstract

Background: Vitiligo is characterized by the death of melanocytes in the skin. This is associated with the presence of T cell infiltrates in the lesional borders. However, at present, there is no detailed and systematic characterization on whether additional cellular or molecular changes are present inside vitiligo lesions. Further, it is unknown if the normal appearing non-lesional skin of vitiligo patients is in fact normal. The purpose of this study is to systematically characterize the molecular and cellular characteristics of the lesional and non-lesional skin of vitiligo patients.

Methods and materials: Paired lesional and non-lesional skin biopsies from twenty-three vitiligo patients and normal skin biopsies from sixteen healthy volunteers were obtained with informed consent. The following aspects were analyzed: (1) transcriptome changes present in vitiligo skin using DNA microarrays and qRT-PCR; (2) abnormal cellular infiltrates in vitiligo skin explant cultures using flow cytometry; and (3) distribution of the abnormal cellular infiltrates in vitiligo skin using immunofluorescence microscopy.

Results: Compared with normal skin, vitiligo lesional skin contained 17 genes (mostly melanocyte-specific genes) whose expression was decreased or absent. In contrast, the relative expression of 13 genes was up-regulated. The up-regulated genes point to aberrant activity of the innate immune system, especially natural killer cells in vitiligo. Strikingly, the markers of heightened innate immune responses were also found to be up-regulated in the non-lesional skin of vitiligo patients.

Conclusions and clinical implications: As the first systematic transcriptome characterization of the skin in vitiligo patients, this study revealed previously unknown molecular markers that strongly suggest aberrant innate immune activation in the microenvironment of vitiligo skin. Since these changes involve both lesional and non-lesional skin, our results suggest that therapies targeting the entire skin surface may improve treatment outcomes. Finally, this study revealed novel mediators that may facilitate future development of vitiligo therapies.

Conflict of interest statement

Competing Interests: This work was in part supported by funding from Astellas Pharma Research Competition, Canada. However, this does not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Transcriptome Analysis of Vitiligo and…
Figure 1. Transcriptome Analysis of Vitiligo and Normal Skin Biopsies.
A heat map is constructed by Gene Spring software (see methods) comparing the relative expression levels of the 30 significantly altered genes in vitiligo skin. Depicted are the expression levels of these genes in individual samples relative to their corresponding expression reference levels, which are the averages of expression in the 16 normal skin biopsies. Red squares: Genes with up-regulation in that sample compared with normal skin of healthy volunteers. Green squares: Genes with down-regulation in that specific sample compared with normal skin of healthy volunteers. Yellow squares: no significant change between the sample and the normal skin of healthy volunteers.
Figure 2. Explant culture analysis of natural…
Figure 2. Explant culture analysis of natural killer cell infiltrates in biopsies of vitiligo lesional and non-lesional skin.
Natural killer (NK) cells from 6 pairs of vitiligo skin explants (lesional and non-lesional) and 5 normal skin explants were cultured on Cellfoam matrices (see methods section) and analyzed using flow cytometry, with the gate set on total live cells A: Skin-resident CD56bright CD3-ve NK cells in normal control skin, vitiligo non-lesional skin and lesional skin by scatter plot. B: Further gating on the CD56bright CD3-ve cells revealed that majority of the NK cells in vitiligo skin were granzyme B-positive. C: Dot plot of all samples analyzed for CD56bright CD3- natural killer cells. The difference in the proportion of resident natural killer cells between normal skin and the respective vitiligo non-lesional and lesional skin is statistically significant (p = 0.0043; mean ± SEM). Comparisons between the respective groups are indicated in the figure by lines with an asterisk (*) denoting statistical significance (p<0.05). Abbreviations: NS: normal skin; NLS: non-lesional skin; LS: lesional-skin.
Figure 3. Distribution of natural killer cells…
Figure 3. Distribution of natural killer cells and cytotoxic T cells in vitiligo lesional and non-lesional skin.
Skin biopsies taken from 12 vitiligo patients and 6 normal individuals were subjected to immunofluorescence analysis of natural killer (NK) cells. A: Micrographs showing natural killer cells (CD3−/NKG2D+) (red) present in vitiligo lesional and non-lesional skin but absent from the normal skin of healthy volunteers. Some NK cells are in close proximity to the basal epidermal layer where melanocytes reside (arrows). In addition, increased numbers of cytotoxic T cells (CD3+/NKG2D+) (yellow: co-localization of red and green) as well as non-cytotoxic T cells (CD3+/NKG2D−) (green) were also found in both vitiligo peri-lesional and lesional skin. B: Quantification of cells demonstrates a statistically significant increase in NK cells, cytotoxic T cells and non-cytotoxic T cells in vitiligo non-lesional skin (p = 0.0021, 0.0015, 0.001; mean ± SEM) and lesional skin (p = 0.021, 0.0017, 0.0023; mean ± SEM) as compared with normal skin. Color keys: Green: CD3 (a pan-T cell marker); Red: NKG2D (NK cell activation receptor); and blue: DAPI (nuclear stain). Comparisons between the respective groups are indicated in the figure by lines with an asterisk (*) denoting statistical significance (p<0.05). Abbreviations: NS: normal skin; NLS: non-lesional skin; LS: lesional-skin. Magnification: 400×; scale bar: 20 µm.

References

    1. Whitton ME, Ashcroft DM, Gonzalez U (2008) Therapeutic interventions for vitiligo. J Am Acad Dermatol 59: 713–717.
    1. Grimes PE (2005) New insights and new therapies in vitiligo. JAMA 293: 730–735.
    1. Abu Tahir M, Pramod K, Ansari SH, Ali J (2010) Current remedies for vitiligo. Autoimmun Rev 9: 516–520.
    1. Porter JR, Beuf AH (1991) Racial variation in reaction to physical stigma: a study of degree of disturbance by vitiligo among black and white patients. J Health Soc Behav 32: 192–204.
    1. Thompson AR, Kent G, Smith JA (2002) Living with vitiligo: dealing with difference. Br J Health Psychol 7: 213–225.
    1. Jin Y, Mailloux CM, Gowan K, Riccardi SL, LaBerge G, et al. (2007) NALP1 in vitiligo-associated multiple autoimmune disease. N Engl J Med 356: 1216–1225.
    1. Spritz RA (2011) The genetics of vitiligo. J Invest Dermatol 131: E18–20.
    1. Jin Y, Birlea SA, Fain PR, Gowan K, Riccardi SL, et al. (2010) Variant of TYR and autoimmunity susceptibility loci in generalized vitiligo. N Engl J Med 362: 1686–1697.
    1. Jin Y, Birlea SA, Fain PR, Spritz RA (2007) Genetic variations in NALP1 are associated with generalized vitiligo in a Romanian population. J Invest Dermatol 127: 2558–2562.
    1. Zhu KJ, Lv YM, Yin XY, Wang ZX, Sun LD, et al. (2011) Psoriasis regression analysis of MHC loci identifies shared genetic variants with vitiligo. PLoS One 6: e23089.
    1. Quan C, Ren YQ, Xiang LH, Sun LD, Xu AE, et al. (2010) Genome-wide association study for vitiligo identifies susceptibility loci at 6q27 and the MHC. Nat Genet 42: 614–618.
    1. Ren Y, Yang S, Xu S, Gao M, Huang W, et al. (2009) Genetic variation of promoter sequence modulates XBP1 expression and genetic risk for vitiligo. PLoS Genet 5: e1000523.
    1. Liang Y, Yang S, Zhou Y, Gui J, Ren Y, et al. (2007) Evidence for two susceptibility loci on chromosomes 22q12 and 6p21-p22 in Chinese generalized vitiligo families. J Invest Dermatol 127: 2552–2557.
    1. Wang X, Erf GF (2003) Melanocyte-specific cell mediated immune response in vitiliginous Smyth line chickens. J Autoimmun 21: 149–160.
    1. Slominski A, Paus R, Bomirski A (1989) Hypothesis: possible role for the melatonin receptor in vitiligo: discussion paper. J R Soc Med 82: 539–541.
    1. Xu S, Zhou Y, Yang S, Ren Y, Zhang C, et al. (2010) Platelet-derived growth factor receptor alpha gene mutations in vitiligo vulgaris. Acta Derm Venereol 90: 131–135.
    1. Spritz RA, Gowan K, Bennett DC, Fain PR (2004) Novel vitiligo susceptibility loci on chromosomes 7 (AIS2) and 8 (AIS3), confirmation of SLEV1 on chromosome 17, and their roles in an autoimmune diathesis. Am J Hum Genet 74: 188–191.
    1. Zhang XJ, Chen JJ, Liu JB (2005) The genetic concept of vitiligo. J Dermatol Sci 39: 137–146.
    1. Boelaert K, Newby PR, Simmonds MJ, Holder RL, Carr-Smith JD, et al. (2010) Prevalence and relative risk of other autoimmune diseases in subjects with autoimmune thyroid disease. Am J Med 123: e181–189, 183, e181-189.
    1. Deretzi G, Kountouras J, Koutlas E, Zavos C, Polyzos S, et al. (2010) Familial prevalence of autoimmune disorders in multiple sclerosis in Northern Greece. Mult Scler 16: 1091–1101.
    1. Kocer B, Nazliel B, Oztas M, Batur HZ (2009) Vitiligo and multiple sclerosis in a patient treated with interferon beta-1a: a case report. Eur J Neurol 16: e78–79.
    1. Ramagopalan SV, Dyment DA, Valdar W, Herrera BM, Criscuoli M, et al. (2007) Autoimmune disease in families with multiple sclerosis: a population-based study. Lancet Neurol 6: 604–610.
    1. Rashtak S, Pittelkow MR (2008) Skin involvement in systemic autoimmune diseases. Curr Dir Autoimmun 10: 344–358.
    1. Harris JE, Harris TH, Weninger W, Wherry EJ, Hunter CA, et al. (2012) A Mouse Model of Vitiligo with Focused Epidermal Depigmentation Requires IFN-gamma for Autoreactive CD8(+) T-Cell Accumulation in the Skin. J Invest Dermatol
    1. Glassman SJ (2011) Vitiligo, reactive oxygen species and T-cells. Clin Sci (Lond) 120: 99–120.
    1. Kemp EH, Emhemad S, Akhtar S, Watson PF, Gawkrodger DJ, et al. (2011) Autoantibodies against tyrosine hydroxylase in patients with non-segmental (generalised) vitiligo. Exp Dermatol 20: 35–40.
    1. Le Poole IC, Luiten RM (2008) Autoimmune etiology of generalized vitiligo. Curr Dir Autoimmun 10: 227–243.
    1. Wang CQ, Cruz-Inigo AE, Fuentes-Duculan J, Moussai D, Gulati N, et al. (2011) Th17 cells and activated dendritic cells are increased in vitiligo lesions. PLoS One 6: e18907.
    1. Kitamura R, Tsukamoto K, Harada K, Shimizu A, Shimada S, et al. (2004) Mechanisms underlying the dysfunction of melanocytes in vitiligo epidermis: role of SCF/KIT protein interactions and the downstream effector, MITF-M. J Pathol 202: 463–475.
    1. Kingo K, Aunin E, Karelson M, Philips MA, Ratsep R, et al. (2007) Gene expression analysis of melanocortin system in vitiligo. J Dermatol Sci 48: 113–122.
    1. Kingo K, Aunin E, Karelson M, Ratsep R, Silm H, et al. (2008) Expressional changes in the intracellular melanogenesis pathways and their possible role in the pathogenesis of vitiligo. J Dermatol Sci 52: 39–46.
    1. Stromberg S, Bjorklund MG, Asplund A, Rimini R, Lundeberg J, et al. (2008) Transcriptional profiling of melanocytes from patients with vitiligo vulgaris. Pigment Cell Melanoma Res 21: 162–171.
    1. Spritz RA (2007) The genetics of generalized vitiligo and associated autoimmune diseases. Pigment Cell Res 20: 271–278.
    1. Wang Y, Su M, Zhou LL, Tu P, Zhang X, et al. (2011) Deficiency of SATB1 expression in Sezary cells causes apoptosis resistance by regulating FasL/CD95L transcription. Blood 117: 3826–3835.
    1. Kennah E, Ringrose A, Zhou LL, Esmailzadeh S, Qian H, et al. (2009) Identification of tyrosine kinase, HCK, and tumor suppressor, BIN1, as potential mediators of AHI-1 oncogene in primary and transformed CTCL cells. Blood 113: 4646–4655.
    1. Ringrose A, Zhou Y, Pang E, Zhou L, Lin AE, et al. (2006) Evidence for an oncogenic role of AHI-1 in Sezary syndrome, a leukemic variant of human cutaneous T-cell lymphomas. Leukemia 20: 1593–1601.
    1. Huang da W, Sherman BT, Lempicki RA (2009) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4: 44–57.
    1. Huang da W, Sherman BT, Lempicki RA (2009) Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res 37: 1–13.
    1. Zhou Y, Lee HS, Kooshesh F, Fujisawa H, Sauder DN, et al. (1996) Effects of UVB irradiation on keratinocyte growth factor (KGF) and receptor (KGFR) expression in cultured human keratinocytes. Exp Dermatol 5: 138–144.
    1. Broady R, Yu J, Chow V, Tantiworawit A, Kang C, et al. (2010) Cutaneous GVHD is associated with the expansion of tissue-localized Th1 and not Th17 cells. Blood 116: 5748–5751.
    1. Clark RA, Chong BF, Mirchandani N, Yamanaka K, Murphy GF, et al. (2006) A novel method for the isolation of skin resident T cells from normal and diseased human skin. J Invest Dermatol 126: 1059–1070.
    1. Yuan L, Zhou LJ, Li WM (2010) Oxidative stress-induced apoptosis is mediated by calpains in alcoholic cardiomyopathy. Int J Cardiol
    1. Dorn GW 2nd (2007) Periostin and myocardial repair, regeneration, and recovery. N Engl J Med 357: 1552–1554.
    1. Afonina IS, Cullen SP, Martin SJ (2010) Cytotoxic and non-cytotoxic roles of the CTL/NK protease granzyme B. Immunol Rev 235: 105–116.
    1. van den Boorn JG, Konijnenberg D, Dellemijn TA, van der Veen JP, Bos JD, et al. (2009) Autoimmune destruction of skin melanocytes by perilesional T cells from vitiligo patients. J Invest Dermatol 129: 2220–2232.
    1. Hoek KS, Schlegel NC, Eichhoff OM, Widmer DS, Praetorius C, et al. (2008) Novel MITF targets identified using a two-step DNA microarray strategy. Pigment Cell Melanoma Res 21: 665–676.
    1. Nonaka D, Chiriboga L, Rubin BP (2008) Sox10: a pan-schwannian and melanocytic marker. Am J Surg Pathol 32: 1291–1298.
    1. Kamholz J, Sessa M, Scherer S, Vogelbacker H, Mokuno K, et al. (1992) Structure and expression of proteolipid protein in the peripheral nervous system. J Neurosci Res 31: 231–244.
    1. Al'Abadie MS, Warren MA, Bleehen SS, Gawkrodger DJ (1995) Morphologic observations on the dermal nerves in vitiligo: an ultrastructural study. Int J Dermatol 34: 837–840.
    1. Breathnach AS, Bor S, Wyllie LM (1966) Electron microscopy of peripheral nerve terminals and marginal melanocytes in vitiligo. J Invest Dermatol 47: 125–140.
    1. Champsaur M, Lanier LL (2010) Effect of NKG2D ligand expression on host immune responses. Immunol Rev 235: 267–285.
    1. Huang H, Wang X, Zhang Y, Zheng X, Wei H, et al. (2010) Up-regulation of NKG2F receptor, a functionally unknown killer receptor, of human natural killer cells by interleukin-2 and interleukin-15. Oncol Rep 24: 1043–1048.
    1. Kim DK, Kabat J, Borrego F, Sanni TB, You CH, et al. (2004) Human NKG2F is expressed and can associate with DAP12. Mol Immunol 41: 53–62.
    1. French AR, Yokoyama WM (2004) Natural killer cells and autoimmunity. Arthritis Res Ther 6: 8–14.
    1. Perricone R, Perricone C, De Carolis C, Shoenfeld Y (2008) NK cells in autoimmunity: a two-edg'd weapon of the immune system. Autoimmun Rev 7: 384–390.
    1. Dalbeth N, Callan MF (2002) A subset of natural killer cells is greatly expanded within inflamed joints. Arthritis Rheum 46: 1763–1772.
    1. Pridgeon C, Lennon GP, Pazmany L, Thompson RN, Christmas SE, et al. (2003) Natural killer cells in the synovial fluid of rheumatoid arthritis patients exhibit a CD56bright,CD94bright,CD158negative phenotype. Rheumatology (Oxford) 42: 870–878.
    1. Schepis D, Gunnarsson I, Eloranta ML, Lampa J, Jacobson SH, et al. (2009) Increased proportion of CD56bright natural killer cells in active and inactive systemic lupus erythematosus. Immunology 126: 140–146.
    1. Yamagiwa S, Kamimura H, Ichida T (2009) Natural killer cell receptors and their ligands in liver diseases. Med Mol Morphol 42: 1–8.
    1. Karlsen TH, Boberg KM, Olsson M, Sun JY, Senitzer D, et al. (2007) Particular genetic variants of ligands for natural killer cell receptors may contribute to the HLA associated risk of primary sclerosing cholangitis. J Hepatol 46: 899–906.
    1. Trachtenberg EA (2009) Understanding the role of natural killer cell receptors and their human leukocyte antigen ligands in multiple sclerosis. Ann Neurol 65: 626–628.
    1. Jones CD, Guckian M, el-Ghorr AA, Gibbs NK, Norval M (1996) Effects of phototherapy on the production of cytokines by peripheral blood mononuclear cells and on systemic antibody responses in patients with psoriasis. Photodermatol Photoimmunol Photomed 12: 204–210.
    1. Durham-Pierre DG, Walters CS, Halder RM, Pham HN, Vanderpool EA (1995) Natural killer cell and lymphokine-activated killer cell activity against melanocytes in vitiligo. J Am Acad Dermatol 33: 26–30.
    1. Ghoneum M, Grimes PE, Gill G, Kelly AP (1987) Natural cell-mediated cytotoxicity in vitiligo. J Am Acad Dermatol 17: 600–605.
    1. Mozzanica N, Villa ML, Foppa S, Vignati G, Cattaneo A, et al. (1992) Plasma alpha-melanocyte-stimulating hormone, beta-endorphin, met-enkephalin, and natural killer cell activity in vitiligo. J Am Acad Dermatol 26: 693–700.
    1. Mozzanica N, Frigerio U, Negri M, Tadini G, Villa ML, et al. (1989) Circadian rhythm of natural killer cell activity in vitiligo. J Am Acad Dermatol 20: 591–596.
    1. Basak PY, Adiloglu AK, Koc IG, Tas T, Akkaya VB (2008) Evaluation of activatory and inhibitory natural killer cell receptors in non-segmental vitiligo: a flow cytometric study. J Eur Acad Dermatol Venereol 22: 970–976.
    1. Tak PP, Kummer JA, Hack CE, Daha MR, Smeets TJ, et al. (1994) Granzyme-positive cytotoxic cells are specifically increased in early rheumatoid synovial tissue. Arthritis Rheum 37: 1735–1743.
    1. Ronday HK, van der Laan WH, Tak PP, de Roos JA, Bank RA, et al. (2001) Human granzyme B mediates cartilage proteoglycan degradation and is expressed at the invasive front of the synovium in rheumatoid arthritis. Rheumatology (Oxford) 40: 55–61.
    1. Goldbach-Mansky R, Suson S, Wesley R, Hack CE, El-Gabalawy HS, et al. (2005) Raised granzyme B levels are associated with erosions in patients with early rheumatoid factor positive rheumatoid arthritis. Ann Rheum Dis 64: 715–721.
    1. Darrah E, Rosen A (2010) Granzyme B cleavage of autoantigens in autoimmunity. Cell Death Differ 17: 624–632.
    1. Long EO, Rajagopalan S (2002) Stress signals activate natural killer cells. J Exp Med 196: 1399–1402.
    1. Strid J, Roberts SJ, Filler RB, Lewis JM, Kwong BY, et al. (2008) Acute upregulation of an NKG2D ligand promotes rapid reorganization of a local immune compartment with pleiotropic effects on carcinogenesis. Nat Immunol 9: 146–154.
    1. Boissy RE, Spritz RA (2009) Frontiers and controversies in the pathobiology of vitiligo: separating the wheat from the chaff. Exp Dermatol 18: 583–585.
    1. Renedo M, Arce I, Montgomery K, Roda-Navarro P, Lee E, et al. (2000) A sequence-ready physical map of the region containing the human natural killer gene complex on chromosome 12p12.3-p13.2. Genomics 65: 129–136.
    1. Welte S, Kuttruff S, Waldhauer I, Steinle A (2006) Mutual activation of natural killer cells and monocytes mediated by NKp80-AICL interaction. Nat Immunol 7: 1334–1342.
    1. Kuttruff S, Koch S, Kelp A, Pawelec G, Rammensee HG, et al. (2009) NKp80 defines and stimulates a reactive subset of CD8 T cells. Blood 113: 358–369.
    1. Jacob SE, Blyumin M (2008) Vitiligo-like hypopigmentation with poliosis following treatment of superficial basal cell carcinoma with imiquimod. Dermatol Surg 34: 844–845.
    1. Serrao VV, Paris FR, Feio AB (2008) Genital vitiligo-like depigmentation following use of imiquimod 5% cream. Eur J Dermatol 18: 342–343.
    1. Senel E, Seckin D (2007) Imiquimod-induced vitiligo-like depigmentation. Indian J Dermatol Venereol Leprol 73: 423.

Source: PubMed

3
Prenumerera