Content » Vol 96, Issue 6

Investigative Report

MicroRNA-155 is Dysregulated in the Skin of Patients with Vitiligo and Inhibits Melanogenesis-associated Genes in Melanocytes and Keratinocytes

Liisi Šahmatova1,2, Stoyan Tankov1,3, Ele Prans3, Alar Aab3, Helen Hermann3, Paula Reemann3, Maire Pihlap3, Maire Karelson1,2, Kristi Abram1,2, Kai Kisand3, Külli Kingo1,2 and Ana Rebane3

1Department of Dermatology and 3Institute of Biomedicine and Translational Medicine, University of Tartu, and 2Dermatology Clinic, Tartu University Hospital, Tartu, Estonia

Little is known about the functions of microRNAs (miRNAs) in skin pigmentation disorders. The aim of this study was to investigate the expression and potential role of miRNAs in vitiligo. Of 12 studied miRNAs with proven functions in cell proliferation, differentiation, immune responses and melanogenesis, miR-99b, miR-125b, miR-155 and miR-199a-3p were found to be increased and miR-145 was found to be decreased in the skin of patients with vitiligo. Combined pathway and target analysis revealed melanogenesis-associated targets for miR-99b, miR-125b, miR-155 and miR-199a-3p. In situ hybridization analysis demonstrated increased expression of miR-155 in the epidermis of patients with vitiligo. Correspondingly, miR-155 was induced by vitiligo-associated cytokines in human primary melanocytes and keratinocytes. When overexpressed, miR-155 inhibited the expression of melanogenesis-associated genes and altered interferon-regulated genes in melanocytes and keratinocytes. In conclusion, this study demonstrates that the expression of miRNAs is dysregulated in the skin of patients with vitiligo and suggests that miR-155 contributes to the pathogenesis of vitiligo. Key words: vitiligo; miRNA; skin; melanocytes; keratinocytes.

Accepted Mar 3, 2016; Epub ahead of print Mar 4, 2016

Acta Derm Venereol 2016; 96: XX–XX.

Ana Rebane, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14B, EE-50411 Tartu, Estonia. E-mail: ana.rebane@ut.ee

Vitiligo is a common acquired, idiopathic disease, characterized by the destruction of melanocytes (1). Although there are rarely any physical symptoms other than depigmented macules on the skin, patients with vitiligo, especially dark-skinned patients, have an impaired quality of life (2, 3). Many theories, e.g. autoimmunity, cytotoxic metabolites, neural and genetic theories, have been proposed to explain the mechanisms of pigmentation loss. In addition to melanocyte dysfunction, keratinocyte alteration plays a role; keratinocytes in depigmented epidermis are more vulnerable to apoptosis and produce lower amounts of melanogenic mediators than normal skin (4). However, the exact mechanism underlying this pigmentary disorder remains unknown (1) and this has held back progress in its treatment (5).

MicroRNAs (miRNAs) are approximately 22 nucleo­tide-long non-coding gene-regulatory RNA molecules that inhibit gene expression through translational repression or mRNA turnover. In mammalian cells, in most cases, miRNAs bind with partial complementarity to sequences in the 3’ untranslated region (3’UTR) of target messenger RNAs (mRNAs) for the suppression of gene expression. Each miRNA has multiple targets and each mRNA is simultaneously regulated by multiple miRNAs (6).

miRNAs have been intensively investigated in humans for more than 10 years and have been found to regulate most of the cellular processes, including cell proliferation, differentiation, development, signal transduction, metabolism, apoptosis and immune responses (7). For instance, miR-155 is known to target the suppressor of cytokine signalling (SOCS1) and there­by activates interferon signalling in CD8+ cytotoxic T cells (8). miRNA dysregulation is associated with the pathogenesis of various inflammatory diseases, including different inflammatory skin disorders, such as psoriasis (9), atopic dermatitis (7, 10) and allergic contact dermatitis (11). Aberrant expression of several miRNAs in the skin and serum of patients with vitiligo has been demonstrated by microarray analysis (12, 13). Using quantitative reverse transcription-PCR (qRT-PCR), dysregulation of miR-224-3p, miR-4712-3p and miR-3940-5p has been shown in peripheral blood mononuclear cells of patients with vitiligo (14). A single nucleotide polymorphism, rs11614913 in miR-196-a-2, has been found to be associated with vitiligo (15). How­ever, little is known about the role of miRNAs in the pathogenesis of vitiligo.

The aim of this study was to investigate the potential role of previously known melanocyte-, keratinocyte-, immunity-, cell proliferation-, differentiation- and apoptosis-associated miRNAs in vitiligo. We measured their expression in the lesional and non-lesional skin of patients with vitiligo and in cultured unstimulated and cytokine-treated melanocytes and keratinocytes, and assessed the possible targets of the dysregulated miRNAs.

MATERIALS AND METHODS

Patients

A total of 17 patients with non-segmental vitiligo (5 with active and 12 with stable vitiligo) and 17 control subjects were included in the study. All participants were unrelated Caucasian individuals living in Estonia. The study was approved by the research ethics committee of the University of Tartu. A detailed description of the patients is provided in Appendix SI1.

Cell culture experiments and miRNA and mRNA expression analysis

Human melanocytes from paediatric foreskin were cultivated in melanocyte growth medium M2 with supplement mix (PromoCell, Heidelberg, Germany). Pooled, normal human epidermal keratinocytes (Promocell, Heidelberg, Germany) were cultured in keratinocyte-serum-free medium (SFM) with supplements (Life Technologies, Grand Island, New York, USA). In situ hybridization (ISH) was performed on 10-µm sections of frozen skin biopsy specimens using miR-155, miRCURY LNA™ Detection Probe for hsa-miR-155 (88072-15) (Exiqon, Vedbaek, Denmark) according to the manufacturer’s protocol. The transfection protocol, RNA purification, qRT-PCR and ISH are detailed in Appendix SI1.

miRNA target selection, pathway analysis and statistics

Putative targets were selected using Targetscan 6.2 (http://www.targetscan.org/) (16). The pathway analysis was performed with g:Profiler (http://biit.cs.ut.ee/gprofiler) (17). For statistical analysis, Fisher’s exact test, Student’s t-test and Mann-Whitney U test were applied. Details are given in Appendix SI1.

RESULTS

To investigate the potential role of miRNAs in vitiligo, we selected 12 miRNAs with known functions in the regulation of cell proliferation and differentiation (miR-10a, miR-99b, miR-125a, miR-125b and miR-199a-3p), immune responses (miR-146a, miR-146b, miR-155, miR-223 and miR-511), skin homeostasis (miR-203) and melanogenesis (miR-145) (7, 9, 18) (see also Appendix SI1 and Table SI1). The expression of the selected miRNAs was detectable by qRT-PCR in the skin from control subjects. The most abundantly expressed miRNA was miR-125b, whose expression level was approximately 3,800 times higher than that of miR-511, the miRNA with the lowest expression level (mean threshold cycle 29.5) (Fig. 1A). From this set of miRNAs, we found miR-99b, miR-155, miR-199a-3p, miR-125b and miR-145 to be dysregulated in skin from patients with vitiligo compared with skin from control subjects, and miR-146b to be differentially expressed in the lesional compared with non-lesional skin from patients with vitiligo (Fig. 1B). miR-99b expression levels were increased in vitiligo lesional (p < 0.001) and non-lesional skin (p < 0.01) compared with the control subjects. Highly significant upregulation of miR-155 (p < 0.01), miR-199a-3p (p < 0.01), significant upregulation of miR-125b (p < 0.05) and downregulation of miR-145 (p < 0.05) expression was detected exclusively in vitiligo lesional skin compared with the control subjects. In the cases of miR-155 (p < 0.05), miR-199a-3p (p < 0.05) and miR-146b (p < 0.05), the expression levels were higher in vitiligo lesional skin than in vitiligo non-lesional skin. The expression levels of dysregulated miRNAs did not differ statistically significantly between patients with active and stable vitiligo (data not shown). There were no statistically significant differences in the expression levels of miR-10a, miR-125a, miR-146a, miR-203, miR-223 and miR-511 between the groups (data not shown).

To analyse whether the differentially expressed miRNAs, miR-99b, miR-125b, miR-145, miR-155 and miR-199a-3p, can influence cellular processes associated with vitiligo, we performed pathway analysis for conserved and the best-scored miRNA targets expressed in the skin. Four miRNAs, miR-99b, miR-125b, miR-155 and miR-199a-3p, were found to have predicted direct targets belonging to either the gene ontology (GO) or human phenotype (HP) ontology groups or to Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways related to melanocyte differentiation, melanogenesis, melanosome structure and localization, and skin pigmentation (Table SII1). Putative miR-145 targets included genes involved in the regulation of stress-activated mitogen-activated protein kinase (MAPK), c-Jun N-terminal kinase (JNK) and transforming growth factor (TGF)-β signalling pathways. All 5 of the differentially expressed miRNAs also had putative targets belonging to functional groups that might indirectly influence cellular processes in vitiligo, including abnormalities in skin physiology, inflammatory abnormalities of the skin, programmed cell death, tight junction interactions, cell growth and cell cycle progression (data not shown).

We next focused our experiments on miR-155, as it was predicted to target multiple important melanogenesis-associated genes (Table SII1) and has previously been shown to contribute to the activation of interferon signalling (8). We first performed in situ hybridization on skin samples from patients with vitiligo and control individuals. The expression of miR-155 was detected in stratum basale, where melanocytes and proliferating keratinocytes are located, as well as in stratum spinosum of the epidermis of patients with vitiligo. In line with RT-qPCR results (Fig. 1B), no signal in one donor and faint positive signal of miR-155 was detected in the epidermis of control skin (Fig. 2A).

4681fig1.tif

Fig. 1. Relative expression of miRNAs in the skin of control subjects and patients with vitiligo. (A) The results in control skin are represented as boxes with whiskers showing the minimum and maximum and are shown relative to the level of miR-511 (=1). (B) Selected individual miRNA levels in control subjects (CS =1) and lesional skin (VLS) and non-lesional skin (VNLS) of patients with vitiligo. Mean ± standard error of the mean (SEM) is indicated. *p < 0.05; **p < 0.01; and ***p < 0.001.

4681Fig2.tif

Fig. 2. Expression of miR-155 in the skin, melanocytes and keratinocytes. (A) Lesional skin from 2 patients with vitiligo and the skin from 2 control subjects was used for in situ hybridization with miR-155-specific probe. Red colour indicates nuclear fast red staining, blue colour miR-155 expression, bar=50 μm. (B, C) The expression of miR-155 in tumour necrosis factor alpha (TNF-α)-, interferon (IFN)-γ-, IFN-α- and interleukin (IL)-1β-treated (B) melanocytes and (C) keratinocytes is shown relative to the expression levels in unstimulated cells at each indicated time-point. Results are displayed as the mean ± standard error of the mean (SEM). Data are from 3 different stimulations. *p < 0.05; **p < 0.01; and ***p < 0.001.

We next studied whether the cytokines, which are known to be associated with the pathogenesis of vitiligo, (1, 19, 20) influence the expression of miR-155 in melanocytes and keratinocytes. We treated human melanocytes and keratinocytes with tumour necrosis factor alpha (TNF-α), interferon (IFN)-γ, IFN-α and interleukin (IL)-1β and measured the relative expression of miR-155 using qRT-PCR. In melanocytes, the expression of miR-155 was significantly upregulated by TNF-α, IFN-γ, IFN-α and IL-1β after 24 h (p < 0.001, p < 0.01, p < 0.05 and p < 0.01, respectively) and 48 h (p < 0.001, p < 0.01, p < 0.01 and p < 0.001, respectively) of stimulation (Fig. 2B). In keratinocytes, the expression of miR-155 was significantly upregulated by TNF-α and IFN-α (p < 0.001 and p < 0.001, respectively) after 24 h of stimulation and then downregulated (p < 0.01) at the 48 h time-point. In response to IFN-γ, miR-155 was upregulated at both 24 h (p < 0.01) and 48 h (p < 0.05) time-points. Significant upregulation by IL-1β was only observed after 48 h of stimulation (p < 0.001) (Fig. 2C).

To test whether miR-155 may influence the development of vitiligo by targeting of melanogenesis-associated genes and modulating the interferon-regulated genes in melanocytes, we performed miR-155 overexpression experiments in melanocytes and keratinocytes. We first transfected human primary melanocytes with miR-155 and the control mimic (Fig. 3A), stimulated the cells with IFN-γ or left unstimulated, and measured relative expression of selected putative miR-155 targets and interferon-regulated genes with RT-qPCR. The melanogenesis-associated genes, such as tyrosinase-related protein 1 (TYRP1), syndecan binding protein (SDCBP), tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein epsilon (YWHAE), and a gene associated with melanocyte differentiation, SRY (sex determining region Y)-Box 10 (SOX10), were included in the analysis. Of IFN-γ-inducible genes, we analysed the effect of miR-155 on interferon-induced transmembrane protein 1 (IFITM1), interferon regulatory factor 1 (IRF1) (21) and previously described miR-155 direct target SOCS1. Fig. 3B and C demonstrate that the overexpression of miR-155 suppressed the expression of SOX10(p < 0.05), TYRP1 (p < 0.01) and YWHAE (p < 0.01) in unstimulated melanocytes. The mRNA level of SDCBP was inhibited both in unstimulated and IFN-γ-stimulated melanocytes with p < 0.01 and p < 0.05, respectively. Of interferon-inducible genes, SOCS1 was suppressed by miR-155, both in unstimulated (p < 0.01) and stimulated (p < 0.05) melanocytes. IRF1 was decreased (p < 0.001) and IFITM1 was increased (p < 0.05) in unstimulated melanocytes transfected with miR-155 compared with the control transfection. IFITM1 and IRF1 were strongly induced in response to IFN-γ, and IFITM1 was further increased (p < 0.01) upon overexpression of miR-155. For IRF1, no difference between miR-155 and control-transfected cells was observed when the cells were stimulated by IFN-γ (Fig. 3D).

In the similar overexpression experiment in keratinocytes, miR-155 suppressed the expression of YWHAE (p < 0.05) in unstimulated conditions (Fig. 3E). Among interferon-inducible genes, SOCS1 and IRF1 were suppressed by overexpression of miR-155, both in unstimulated (p < 0.05 and p < 0.01, respectively) and stimulated (both with p < 0.05) keratinocytes. Similarly to the effect in melanocytes, IFITM1 was increased in unstimulated (p < 0.001) and IFN-γ-stimulated (p < 0.001) keratinocytes transfected with miR-155 compared with the control transfections (Fig. 3F). In conclusion, these results demonstrate that miR-155 has the capacity to impact melanogenesis and inflammatory responses in vitiligo directly through its effect in melanocytes and keratinocytes.

4681fig3.tif

Fig. 3. miR-155 inhibits melanogenesis-associated targets and modulates interferon-inducible genes in melanocytes and keratinocytes. (A–D) Human primary melanocytes and (E, F) keratinocytes were transfected either with control (cont) or hsa-miR-155-5p mimic for 24 h and then stimulated with interferon (IFN)-γ for 48 h or left unstimulated (us). Relative expression compared with unstimulated control-transfected cells (=1) is shown. Data represent mean ± standard error of the mean (SEM). *p < 0.05; **p < 0.01; and ***p < 0.001.

DISCUSSION

Little is known about the functions of miRNA in the pathogenesis of vitiligo. This study revealed that miR-99b, miR-155, miR-199a-3p, miR-125b and miR-145 are dysregulated in the skin of patients with vitiligo, of which miR-99b, miR-125b, miR-155 and miR-199a-3p were found to have putative targets associated with melanocyte differentiation and melanogenesis. Among the dysregulated miRNAs, we demonstrated increased expression of miR-155 in the epidermis of patients with vitiligo. Thus, miR-155 was induced by vitiligo-associated cytokines TNF-α, IFN-α, IFN-γ and IL-1β in human primary melanocytes and keratinocytes. When overexpressed, miR-155 inhibited the expression of genes known to affect melanocyte differentiation and melanogenesis, such as TYRP1, YWHAE, SDCBP and SOX10 in melanocytes, and YWHAE in keratinocytes. In addition, miR-155 overexpression altered the levels of interferon-regulated genes SOCS1, IRF1 and IFITM1 in melanocytes and keratinocytes. Our results suggest that miR-155 and other miRNAs contribute to the pathogenesis of vitiligo.

Previously, miRNA expression profiles of peripheral blood mononuclear cells (14), serum (13) and skin (12) of patients with vitiligo have been studied. We report here for the first time the aberrant expression of miR-125b, miR-155 and miR-199a-3p and confirm the altered expression of miR-99b, in the skin of patients with vitiligo. In addition, miR-146b was significantly upregulated in vitiligo lesional skin compared with non-lesional skin. An increased expression level of miR-145 and miR-10a in the non-lesional skin from patients with vitiligo has been reported previously (12). The results of our study did not confirm these differences, as miR-145 was downregulated in vitiligo lesional skin and miR-10a showed no differences between the studied groups. This might be due to differences in participant characteristics or technical approach. For the other studied miRNAs, miR-125a, miR-203, miR-223 and miR-511, we did not detect any differences in the expression levels in the skin of control subjects and patients with vitiligo. Previously, the expression of miR-10a and miR-125b was observed to be downregulated and miR-223 upregulated in serum from patients with vitiligo (13).

Consistent with its overexpression in the epidermis, miR-155 was induced in response to TNF-α, IFN-α, IFN-γ and IL-1β, the proinflammatory cytokines that have been previously reported to be associated with vitiligo pathogenesis (see also Appendix SI1, Table SIII1) in cultured melanocytes and keratinocytes. miR-155 was overexpressed in the skin from patients with atopic dermatitis, mainly due to the presence of immune cells (22). In our study, in line with stimulation experiments in melanocytes and keratinocytes, miR-155 was observed to be increased in the epidermis of lesional vitiligo skin, when analysed by in situ hybridization. This suggests that the presence of inflammatory cytokines in the skin of patients with vitiligo modulates inflammatory responses and activates the expression of miR-155 in melanocytes and keratinocytes.

miR-155 is known as a proinflammatory miRNA, which, among other targets, inhibits suppressor of cytokine signalling 1 (SOCS1). This results in the activation of the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway and, consequently, of types I and II interferon signalling (23). Here, we found that miR-155 modulates several interferon-inducible genes, such as SOCS1, IFITM1 and IRF1, as well as melanogenesis-associated genes, TYRP1, YWHAE, SDCBP and SOX10, in melanocytes and keratinocytes, which suggests that miR-155 contributes to the pathogenesis of vitiligo, both through targeting of melanogenesis-associated targets and via modulation of interferon signalling (see also Appendix SI1, Table SIV1).

In addition to miR-155, other dysregulated miRNAs might contribute to the development of vitiligo. For example, miR-125b is downregulated after the induction of pigmentation in melanocytes (18) and miR-125b mimics have been shown to inhibit expression of pigmentation-related genes (24). Similar to miR-125b, the expression of miR-145 has been shown to be reduced in cultured pigment cells after induction of pigmentation (18). The pathway analysis in our study detected possible direct targets for miR-145 among the MAPK, JNK and TGF-β pathways, which can interfere with the viability and functionality of melanocytes (25). Although no significant overlap was revealed between miR-145 targets and genes associated with pigmentation, we detected the presence of binding sites for miR-145 in the mRNAs of melanogenesis-associated genes ras-related protein rab-27A (RAB27A), SRY (sex determining region Y)-Box9 (SOX9) and fascin actin-bundling protein 1 (FCSN1) using Targetscan (data not shown). This is in line with previous results demonstrating the influence of miR-145 on genes involved in the pigmentation process in miR-145-transfected cells (18).

In conclusion, this study demonstrates that miRNAs miR-99b, miR-125b, miR-145, miR-155 and miR-199a-3p are dysregulated in the skin of patients with vitiligo, and shows that miR-155 has the capacity to modulate melanogenesis-associated and interferon-inducible genes in melanocytes and keratinocytes. Further studies are needed to clarify whether miR-155 and other dysregulated miRNAs described in the present study are suitable diagnostic markers and/or targets for the treatment of vitiligo.

ACKNOWLEDGEMENTS

This work was supported by personal research funds PUT177 and PUT214, an institutional research grant IUT2-2 from the Estonian Research Council, European Regional Fund with the Archimedes Foundation and EU structural assistance grants SARMP12219T and SARBS12096T.

1http://www.medicaljournals.se/acta/content/?doi=10.2340/00015555-2394

 

The authors declare no conflicts of interest.

REFERENCES

 

1. Ezzedine K, Eleftheriadou V, Whitton M, van Geel N. Vitiligo. Lancet 2015; 386: 74–84.

2. Silverberg JI, Silverberg NB. Association between vitiligo extent and distribution and quality-of-life impairment. JAMA Dermatol 2013; 149: 159–164.

3. Karelson M, Silm H, Kingo K. Quality of life and emotional state in vitiligo in an Estonian sample: comparison with psoriasis and healthy controls. Acta Derm Venereol 2013; 93: 446–450.

4. Moretti S, Fabbri P, Baroni G, Berti S, Bani D, Berti E, et al. Keratinocyte dysfunction in vitiligo epidermis: cytokine microenvironment and correlation to keratinocyte apoptosis. Histol Histopathol 2009; 24: 849–857.

5. Taieb A, Alomar A, Bohm M, Dell’anna ML, De Pase A, Eleftheriadou V, et al. Guidelines for the management of vitiligo: the European Dermatology Forum consensus. Br J Dermatol 2013; 168: 5–19.

6. Jonas S, Izaurralde E. Towards a molecular understanding of microRNA-mediated gene silencing. Nature Rev 2015; 16: 421–433.

7. Rebane A, Akdis CA. MicroRNAs: Essential players in the regulation of inflammation. J Allergy Clin Immunol 2013; 132: 15–26.

8. Dudda JC, Salaun B, Ji Y, Palmer DC, Monnot GC, Merck E, et al. MicroRNA-155 is required for effector CD8+ T cell responses to virus infection and cancer. Immunity 2013; 38: 742–753.

9. Sonkoly E, Wei T, Janson PC, Sääf A, Lundeberg L, Tengvall-Linder M, et al. MicroRNAs: novel regulators involved in the pathogenesis of psoriasis? PLoS One 2007; 2: e610.

10. Rebane A, Runnel T, Aab A, Maslovskaja J, Ruckert B, Zimmermann M, et al. MicroRNA-146a alleviates chronic skin inflammation in atopic dermatitis through suppression of innate immune responses in keratinocytes. J Allergy Clin Immunol 2014; 134: 836–847 e811.

11. Vennegaard MT, Bonefeld CM, Hagedorn PH, Bangsgaard N, Lovendorf MB, Odum N, et al. Allergic contact dermatitis induces upregulation of identical microRNAs in humans and mice. Contact Dermatitis 2012; 67: 298–305.

12. Mansuri MS, Singh M, Dwivedi M, Laddha NC, Marfatia YS, Begum R. MicroRNA profiling reveals differentially expressed microRNA signatures from the skin of patients with nonsegmental vitiligo. Br J Dermatol 2014; 171: 1263–1267.

13. Shi YL, Weiland M, Li J, Hamzavi I, Henderson M, Huggins RH, et al. MicroRNA expression profiling identifies potential serum biomarkers for non-segmental vitiligo. Pigment Cell Melanoma Res 2013; 26: 418–421.

14. Wang Y, Wang K, Liang J, Yang H, Dang N, Yang X, et al. Differential expression analysis of miRNA in peripheral blood mononuclear cells of patients with non-segmental vitiligo. J Dermatol 2015; 42: 193–197.

15. Huang Y, Yi X, Jian Z, Wei C, Li S, Cai C, et al. A single-nucleotide polymorphism of miR-196a-2 and vitiligo: an association study and functional analysis in a Han Chinese population. Pigment Cell Melanoma Res 2013; 26: 338–347.

16. Friedman RC, Farh KK, Burge CB, Bartel DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 2009; 19: 92–105.

17. Reimand J, Kull M, Peterson H, Hansen J, Vilo J. Profiler – a web-based toolset for functional profiling of gene lists from large-scale experiments. Nucleic Acids Res 2007; 35: W193–200.

18. Dynoodt P, Mestdagh P, Van Peer G, Vandesompele J, Goossens K, Peelman LJ, et al. Identification of miR-145 as a key regulator of the pigmentary process. J Invest Dermatol 2013; 133: 201–209.

19. Reimann E, Kingo K, Karelson M, Reemann P, Loite U, Sulakatko H, et al. The mRNA expression profile of cytokines connected to the regulation of melanocyte functioning in vitiligo skin biopsy samples and peripheral blood mononuclear cells. Hum Immunol 2012; 73: 393–398.

20. Bertolotti A, Boniface K, Vergier B, Mossalayi D, Taieb A, Ezzedine K, et al. Type I interferon signature in the initiation of the immune response in vitiligo. Pigment Cell Melanoma Res 2014; 27: 398–407.

21. Rebane A, Zimmermann M, Aab A, Baurecht H, Koreck A, Karelson M, et al. Mechanisms of IFN-gamma-induced apoptosis of human skin keratinocytes in patients with atopic dermatitis. J Allergy Clin Immunol 2012; 129: 1297–1306.

22. Sonkoly E, Janson P, Majuri ML, Savinko T, Fyhrquist N, Eidsmo L, et al. MiR-155 is overexpressed in patients with atopic dermatitis and modulates T-cell proliferative responses by targeting cytotoxic T lymphocyte-associated antigen 4. J Allergy Clin Immunol 2010; 126: 581–589.e581–520.

23. Wang P, Hou J, Lin L, Wang C, Liu X, Li D, et al. Inducible microRNA-155 feedback promotes type I IFN signaling in antiviral innate immunity by targeting suppressor of cytokine signaling 1. J Immunol 2010; 185: 6226–6233.

24. Kim KH, Bin BH, Kim J, Dong SE, Park PJ, Choi H, et al. Novel inhibitory function of miR-125b in melanogenesis. Pigment Cell Melan Res 2014; 27: 140–144.

25. Bellei B, Pitisci A, Ottaviani M, Ludovici M, Cota C, Luzi F, et al. Vitiligo: a possible model of degenerative diseases. PLoS One 2013; 8: e59782.

26. Reemann P, Reimann E, Ilmjarv S, Porosaar O, Silm H, Jaks V, et al. Melanocytes in the skin – comparative whole transcriptome analysis of main skin cell types. PLoS One 2014; 9: e115717.

27. Park H, Huang X, Lu C, Cairo MS, Zhou X. MicroRNA-146a and microRNA-146b regulate human dendritic cell apoptosis and cytokine production by targeting TRAF6 and IRAK1 proteins. J Biol Chem 2015; 290: 2831–2841.

28. Taganov KD, Boldin MP, Chang KJ, Baltimore D. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci U S A 2006; 103: 12481–12486.

29. Etzrodt M, Cortez-Retamozo V, Newton A, Zhao J, Ng A, Wildgruber M, et al. Regulation of monocyte functional heterogeneity by miR-146a and Relb. Cell Reports 2012; 1: 317–324.

30. Crone SG, Jacobsen A, Federspiel B, Bardram L, Krogh A, Lund AH, et al. microRNA-146a inhibits G protein-coupled receptor-mediated activation of NF-kappaB by targeting CARD10 and COPS8 in gastric cancer. Mol Cancer 2012; 11: 71.

31. Evel-Kabler K, Song XT, Aldrich M, Huang XF, Chen SY. SOCS1 restricts dendritic cells’ ability to break self tolerance and induce antitumor immunity by regulating IL-12 production and signaling. J Clin Investig 2006; 116: 90–100.

32. Xu N, Brodin P, Wei T, Meisgen F, Eidsmo L, Nagy N, et al. MiR-125b, a microRNA downregulated in psoriasis, modulates keratinocyte proliferation by targeting FGFR2. J Invest Dermatol 2011; 131: 1521–1529.

33. Jin Y, Tymen SD, Chen D, Fang ZJ, Zhao Y, Dragas D, et al. MicroRNA-99 family targets AKT/mTOR signaling pathway in dermal wound healing. PLoS One 2013; 8: e64434.

34. Yi R, O’Carroll D, Pasolli HA, Zhang Z, Dietrich FS, Tarakhovsky A, et al. Morphogenesis in skin is governed by discrete sets of differentially expressed microRNAs. Nat Genet 2006; 38: 356–362.

35. Shatseva T, Lee DY, Deng Z, Yang BB. MicroRNA miR-199a-3p regulates cell proliferation and survival by targeting caveolin-2. J Cell Sci 2011; 124: 2826–2836.

36. Jia L, Li YF, Wu GF, Song ZY, Lu HZ, Song CC, et al. MiRNA-199a-3p Regulates C2C12 Myoblast Differentiation through IGF-1/AKT/mTOR Signal Pathway. Int J Mol Sci 2013; 15: 296–308.

37. Noguchi S, Kumazaki M, Yasui Y, Mori T, Yamada N, Akao Y. MicroRNA-203 regulates melanosome transport and tyrosinase expression in melanoma cells by targeting kinesin superfamily protein 5b. J Invest Dermatol 2014; 134: 461–469.

38. Nissan X, Denis JA, Saidani M, Lemaitre G, Peschanski M, Baldeschi C. miR-203 modulates epithelial differentiation of human embryonic stem cells towards epidermal stratification. Dev Biol 2011; 356: 506–515.

39. Sonkoly E, Wei T, Pavez Lorie E, Suzuki H, Kato M, Torma H, et al. Protein kinase C-dependent upregulation of miR-203 induces the differentiation of human keratinocytes. J Invest Dermatol 2010; 130: 124–134.

40. Tserel L, Runnel T, Kisand K, Pihlap M, Bakhoff L, Kolde R, et al. MicroRNA expression profiles of human blood monocyte-derived dendritic cells and macrophages reveal miR-511 as putative positive regulator of toll-like receptor 4. J Biol Chem 2011; 286: 26487–26495.

41. Lovendorf MB, Mitsui H, Zibert JR, Ropke MA, Hafner M, Dyring-Andersen B, et al. Laser capture microdissection followed by next-generation sequencing identifies disease-related microRNAs in psoriatic skin that reflect systemic microRNA changes in psoriasis. Exp Dermatol 2015; 24: 187–193.

42. Huang H, Xie C, Sun X, Ritchie RP, Zhang J, Chen YE. miR-10a contributes to retinoid acid-induced smooth muscle cell differentiation. J Biol Chem 2010; 285: 9383–9389.

43. Xue X, Feng T, Yao S, Wolf KJ, Liu CG, Liu X, et al. Microbiota downregulates dendritic cell expression of miR-10a, which targets IL-12/IL-23p40. J Immunol 2011; 187: 5879–5886.

44. Takahashi H, Kanno T, Nakayamada S, Hirahara K, Sciume G, Muljo SA, et al. TGF-beta and retinoic acid induce the microRNA miR-10a, which targets Bcl-6 and constrains the plasticity of helper T cells. Nature Immunol 2012; 13: 587–595.

45. Camara-Lemarroy CR, Salas-Alanis JC. The role of tumor necrosis factor-alpha in the pathogenesis of vitiligo. Am J Clin Dermatol 2013; 14: 343–350.

46. Arya V, Bansal M, Girard L, Arya S, Valluri A. Vitiligo at injection site of PEG-IFN-alpha 2a in two patients with chronic hepatitis c: case report and literature review. Case Rep Dermatol 2010; 2: 156–164.

47. Li W, Xin H, Ge L, Song H, Cao W. Induction of vitiligo after imiquimod treatment of condylomata acuminata. BMC Infect Dis 2014; 14: 329.

48. Harris JE, Harris TH, Weninger W, Wherry EJ, Hunter CA, Turka LA. A mouse model of vitiligo with focused epidermal depigmentation requires IFN-gamma for autoreactive CD8(+) T-cell accumulation in the skin. J Invest Dermatol 2012; 132: 1869–1876.

49. Rashighi M, Agarwal P, Richmond JM, Harris TH, Dresser K, Su MW, et al. CXCL10 is critical for the progression and maintenance of depigmentation in a mouse model of vitiligo. Sci Transl Med 2014; 6: 223ra223.

50. Agarwal P, Rashighi M, Essien KI, Richmond JM, Randall L, Pazoki-Toroudi H, et al. Simvastatin prevents and reverses depigmentation in a mouse model of vitiligo. J Invest Dermatol 2015; 135: 1080–1088.

51. Paulos CM, Wrzesinski C, Kaiser A, Hinrichs CS, Chieppa M, Cassard L, et al. Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling. J Clin Invest 2007; 117: 2197–2204.

52. Tu CX, Gu JS, Lin XR. Increased interleukin-6 and granulocyte-macrophage colony stimulating factor levels in the sera of patients with non-segmental vitiligo. J Dermatol Sci 2003; 31: 73–78.

53. Laddha NC, Dwivedi M, Mansuri MS, Singh M, Patel HH, Agarwal N, et al. Association of neuropeptide Y (NPY), interleukin-1B (IL1B) genetic variants and correlation of IL1B transcript levels with vitiligo susceptibility. PLoS One 2014; 9: e107020.

54. Hearing VJ. Biogenesis of pigment granules: a sensitive way to regulate melanocyte function. J Dermatol Sci 2005; 37: 3–14.

55. Yatsu A, Ohbayashi N, Tamura K, Fukuda M. Syntaxin-3 is required for melanosomal localization of Tyrp1 in melanocytes. J Invest Dermatol 2013; 133: 2237–2246.

56. Nishioka E, Funasaka Y, Kondoh H, Chakraborty AK, Mishima Y, Ichihashi M. Expression of tyrosinase, TRP-1 and TRP-2 in ultraviolet-irradiated human melanomas and melanocytes: TRP-2 protects melanoma cells from ultraviolet B induced apoptosis. Melanoma Res 1999; 9: 433–443.

57. 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.

58.El Hajj P, Gilot D, Migault M, Theunis A, van Kempen LC, Sales F, et al. SNPs at miR-155 binding sites of TYRP1 explain discrepancy between mRNA and protein and refine TYRP1 prognostic value in melanoma. Br J Cancer 2015; 113: 91–98.

59. Jacobsen KK, Kleppe R, Johansson S, Zayats T, Haavik J. Epistatic and gene wide effects in YWHA and aromatic amino hydroxylase genes across ADHD and other common neuropsychiatric disorders: association with YWHAE. Am J Med Genet B Neuropsychiatr Genet 2015 Jul 14. [Epub ahead of print]

60. Delevoye C. Melanin transfer: the keratinocytes are more than gluttons. J Invest Dermatol 2014; 134: 877–879.

61. Philley JV, Kannan A, Dasgupta S. MDA-9/Syntenin Control. J Cell Physiol 2016; 231: 545–550.

62. Fufa TD, Harris ML, Watkins-Chow DE, Levy D, Gorkin DU, Gildea DE, et al. Genomic analysis reveals distinct mechanisms and functional classes of SOX10-regulated genes in melanocytes. Hum Mol Genet 2015; 24: 5433–5450.