Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Apr 1.
Published in final edited form as: Autoimmun Rev. 2016 Jan 21;15(4):397–404. doi: 10.1016/j.autrev.2016.01.004

The Role of IL-17 in Vitiligo: A Review

Rasnik K Singh a, Kristina M Lee b, Ivan Vujkovic-Cvijin c, Derya Ucmak b, Benjamin Farahnik d, Michael Abrouk e, Mio Nakamura f, Tian Hao Zhu g, Tina Bhutani b, Maria Wei b, Wilson Liao b
PMCID: PMC4769658  NIHMSID: NIHMS753598  PMID: 26804758

Abstract

IL-17 is involved in the pathogenesis of several autoimmune diseases, however its role in vitiligo has not been well defined. Emerging human and mouse studies have demonstrated that systemic, tissue, and cellular levels of IL-17 are elevated in vitiligo. Many studies have also shown significant positive correlations between these levels and disease activity, extent, and severity. Treatments that improve vitiligo, such as ultraviolet B phototherapy, also modulate IL-17 levels. This review synthesizes our current understanding of how IL-17 may influence the pathogenesis of autoimmune vitiligo at the molecular level. This has implications for defining new vitiligo biomarkers and treatments.

Keywords: Interleukin-17, IL-17, Vitiligo

1. Introduction

The cytokine interleukin-17 has been increasingly implicated in the pathogenesis of several immune-mediated diseases. Whether interleukin-17 plays a role in the etiology of vitiligo, an autoimmune pigmentary skin disorder, has not been well defined in the literature. In this review, we synthesize molecular data from vitiligo patients and animal models of vitiligo to demonstrate that interleukin-17 may play an important role in vitiligo pathogenesis. This has implications not only for our understanding of the biology of vitiligo, but also for defining new biomarkers and treatments.

2. Interleukin-17

Interleukin-17 is a family of six cytokines that includes IL-17A through IL-17F[1]. IL-17A (also known as IL-17) and IL-17F are the most homologous to one another, as well as the best studied for their roles in immune modulation[2]. The two cytokines are expressed by the Th17 subset of CD4+ helper T cells[3] and signal through IL-17RA and IL-17RC, heterodimeric receptors chiefly expressed on tissue fibroblasts and epithelial cells[4]. Despite strong sequence homology and similarly controlled expression patterns, IL-17A and IL-17F regulate distinct responses in vivo. For instance, studies in mice selectively deficient for either IL-17A or IL-17F have demonstrated that only IL-17A is required for the induction of experimental autoimmune encephalomyelitis (EAE). On the other hand, IL-17F and not IL-17A deficiency leads to an aberrant airway neutrophil response to allergen stimulus[5].

2.1. IL-17 in inflammatory disorders

Several inflammatory disorders have been attributed in part to pathogenic effects of the interleukin-17 family. Psoriasis is a chronic inflammatory skin condition marked by rapid epithelial cell turnover, dermal angiogenesis, and multi-immune cell infiltration. Elevated levels of IL-17A and IL-17F, along with other pro-inflammatory Th17 type cytokines, have been found in the sera and skin of psoriatic patients[6]. Phase III data evaluating the efficacy of secukinumab, an anti-IL-17A drug FDA-approved for psoriasis, have revealed that the drug has high efficacy with PASI75 (the proportion of patients in a trial achieving at least 75% reduction in psoriasis severity) ranging from 67% to 91% based on the dose given[710]. Phase I and II studies of anti-IL17A therapy in psoriasis have also documented dose-dependent reductions in keratinocyte proliferation, epidermal hyperplasia, and immune cell infiltration with simultaneous improvements in cellular and molecular disease biomarkers[1114]. Additional clinical trials of biologic therapies antagonizing IL-17 or IL-17 receptor have also shown high efficacy in psoriasis[1517].

Rheumatoid arthritis (RA) is characterized by chronic inflammation of joint synovial fluid that damages adjacent cartilage and bone over time. Elevated levels of IL-17 are observed in the sera and synovial fluid of patients, particularly in areas with larger T-cell populations[18]. Furthermore, these levels correlate with disease severity and duration[19]. Human clinical trials of IL-17 blockade in RA have demonstrated a favorable response profile to the therapy, with reduced inflammatory marker levels, improved quality of life scores, and significant response rates[20]. Additional support for the cytokine’s role in disease promotion comes from mouse models constructed with an IL-17 knockout. These mice develop considerably less arthritis[21,22]. Moreover, introduction of anti-IL-17 neutralizing antibodies directly reduces synovial inflammation and bone and cartilage erosion in mice with arthritis[23,24].

IL-17 has also recently been implicated in the pathogenesis of multiple sclerosis (MS), a non-rheumatic condition in which neurons of the central nervous system undergo autoimmune demyelination. MS subjects exhibit higher blood and CSF levels of IL-17 as compared to controls[25]. An intersection of the Th1 and Th17 pathways has been observed in mouse models of MS, wherein the transcription factors that govern both lineages are co-expressed in autoreactive CD4+ T cells that cross the blood brain barrier (BBB) and induce pathologic demyelination[26]. IL-17, in particular, disrupts junctions of the BBB via increased production of reactive oxygen species (ROS)[27].

Primary Sjogren’s syndrome (pSS) is an autoimmune disorder marked by widespread chronic inflammation of exocrine glands that impairs tissue integrity and glandular secretion. Studies have reported a direct correlation between periductal infiltrating IL-17+ mononuclear cells and extent of exocrine gland involvement. In addition, higher IL-17 mRNA expression has been associated with greater severity minor salivary gland lesions. Lastly, lacrimal IL-17 is increased in patients with pSS relative to those with dry eye syndrome of other etiologies. Although yet unemployed, it has been suggested that targeting the IL-17/Th17 axis represents a credible therapeutic approach to pSS[28].

The role of IL-17 in inflammatory bowel disease remains controversial. Although once only thought of as proinflammatory, IL-17 has recently also been recognized to play a protective role in the intestine. A few studies using experimental IL-17A knock-out mice have demonstrated that these animals may undergo worsening of dextran sulfate sodium-induced colitis[29,30]. More importantly, clinical studies of antibodies targeting IL-17A and IL-17 receptor in Crohn’s disease unexpectedly showed these agents to be either ineffective or disease worsening[31,32]. To explain this, it has been proposed that IL-17 inhibition in the intestines may paradoxically lead to an enhanced Th1 response marked by increased IFN-γ production[33]. On the other hand, functional studies of the small-molecule inhibitors Vidofludimus and Tofacitinib in IBD patients, which antagonize IL-17 among their other functions, have demonstrated efficacy. It is possible that the less selective IL-17 inhibition achieved by these small-molecule inhibitors is a more effective therapeutic strategy for IBD, although the reasons why are still unclear[34].

3. Vitiligo

Vitiligo is a disease of the pigment-producing cells, or melanocytes, that can result in varying patterns and degrees of skin depigmentation. The effect of genetics is thought to be complex and multifactorial, with several vitiligo susceptibility loci identified by genome-wide association studies [3538]. A study examining monozygotic twins reported a vitiligo concordance rate of 23%, suggesting a strong environmental contribution to the pathogenesis; however the etiology of vitiligo remains poorly understood[39]. Several groups have explored a neural hypothesis, uncovering so-called neurogenic inflammatory mediators such as NGF and NPY that may be directly toxic to melanocytes[40,41]. Antibody-mediated, cell-mediated, and cytokine-mediated mechanisms have all accumulated supportive evidence for an autoimmune hypothesis. Autoantibodies against tyrosine hydroxylase and various pigment cell-surface antigens that are specific to melanocytes have been identified in the sera of vitiligo patients [42,43]. Additionally, studies have supported a melanocyte-specific immune reaction driven by cytotoxic CD8+ T cells[44,45]. Lastly, Th1, Th2, and more recently Th17 type cytokines have been significantly quantified in the sera and skin of patients with vitiligo[46,47]. A final hypothesis suggests that hyperproduction of ROS may occur in affected melanocytes, directly damaging critical cellular components[48]. It appears that vitiligo pathogenesis involves a complex interplay between multiple systems, including genetic, neural, autoimmune, and redox pathways.

4. IL-17 Dynamics in Vitiligo

4.1. Human Studies

Several human studies have investigated the roles that Th17 cells and IL-17, the signature Th17 cytokine, play in vitiligo (Table 1):

Table 1.

Human studies examining IL-17 biomarkers in vitiligo

Study Patient Demographics Biomarkers examined Key Results Implications
Zhou et al, 201550 45 patients with NSV1; 45 healthy controls Th17 cell frequency (blood)
IL-17A and Th17 cytokines (serum)
Increased frequency of Th17 cells in NSV group (P= .001); positively correlated with BSA2 of lesions (P=.014)
Increased expression of IL-17 in NSV group (P= .0145)
Role for Th17 cells in Vit3 pathogenesis
Jandus et al, 200851 5 patients with Vit; 10 patients with PsA4; 10 patients with AS5; 10 patients with RA6; 25 healthy controls Th17 cell frequency (blood) No increased frequency of Th17 cells in Vit vs control group Sample size not sufficiently powered
Bassiouny et al, 201152 30 patients with Vit; 20 healthy controls IL-17 cytokine (serum)
IL-17 mRNA (skin)
Increased expression of IL-17 in serum and skin of Vit group (P< .001 for both); positively correlated with disease duration and BSA of lesions (P < .05) Role for IL-17 in Vit pathogenesis, extent, and severity
Khan et al, 201253 45 patients with Vit; 45 healthy controls IL-17 cytokine (serum) Increased expression of IL-17 in Vit group (P= .001)
Ratio of IL-4: IL-17 significantly lower in patients
Vit may involve immune system shift away from Th2 and towards Th17
Basak et al, 200954 40 patients with Vit; 40 healthy controls IL-17 cytokine (serum) SS7 negative correlation between IL-17 and age of onset in Vit group (P= .011); positive correlation with BSA of lesions (P= .038) Role for IL-17 in Vit pathogenesis and extent
Elela et al, 201355 84 patients with NSV; 80 healthy controls IL-17 cytokine (serum)
IL-17 mRNA (skin)
Increased expression of IL-17 in serum and skin (P< .001 for both); positively correlated with VASI8 and VIDA9 Role for IL-17 in Vit pathogenesis, extent, and severity
Tembhre et al, 201356 60 patients with active Vit; 20 patients with stable Vit; 25 patients with Vit tx with NB-UVB; 70 healthy controls IL-17 cytokine (serum) Increased baseline expression of IL-17 in active Vit vs stable Vit vs control group (P= .001)
Decreased expression of IL-17 in Vit group after NB-UVB (P< .001)
Part of NB-UVB10 therapy efficacy may be through reduction of IL-17 expression
Habeb et al, 201358 15 patients with Vit; 15 healthy controls IL-17 mRNA (leukocytes) Increased expression of IL-17 in Vit group (P< .01) Role for IL-17 in Vit pathogenesis
Esmaeili et al, 201157 15 patients with Vit; 15 healthy controls IL-17 mRNA (leukocytes) Increased expression of IL-17 in Vit vs control group, not SS (P= 0.05) Role for IL-17 in Vit pathogenesis
Van den boorn et al, 200960 15 patients with Vit; biopsies given melanocyte antigen-specific stimulation IL-17-secreting T cell frequency (skin) Increased frequency of IL-17 producing CD8+ T cells after stimulation Role for IL-17 in Vit pathogenesis
Wang et al, 201161 20 patients with NSV IL-17 mRNA (skin)
IL-17-secreting T cell frequency (skin)
Increased expression of IL-17 in lesions vs non-lesions (P< .0069)
Increased frequency of IL-17 producing T cells in lesions vs non-lesions (P< .05)
Direct tissue evidence implicating active Th17 cells and IL-17 in Vit skin lesions
Kotobuki et al, 201262 23 patients with Vit; melanocytes tx11 with IL-17A Th17 cell frequency (skin)
MITF12 mRNA (melanocytes)
IL-1, IL-6, TNF-a mRNA (melanocytes)
Increased frequency of Th17 cells (P< .05)
Decreased expression of MITF and the cytokines after tx with IL-17 (P< .05)
IL-17 contributes to local depigmentation through several mechanisms
Hegazy et al, 201463 20 patients with NSV tx with NB-UVB; 20 healthy controls IL-17 mRNA (skin) Increased baseline expression of IL-17 in NSV group lesional (P=.003) and perilesional (P= .001) skin
Decreased expression of IL-17 after NB-UVB in lesional and perilesional skin (P < .05 for both)
IL-17 positively correlated with VASI
Part of NB-UVB therapy efficacy may be through reduction of IL-17 expression
Zhan et al, 201466 51 patients with Vit tx with CO2 laser
51 patients with Vit tx with BCG PSN13+ CO2 laser
Th17 cell frequency (blood)
IL-17 cytokine (serum)
IL-17 mRNA (skin)
Decreased frequency of Th17 cells after NB-UVB (P< .05)
Decreased expression of IL-17 in serum and skin after NB-UVB (P< .05)
CO2 laser as potential therapy for Vit by reducing IL-17 and Th17
1

NSV, non-segmental vitiligo;

2

BSA, body surface area;

3

Vit, vitiligo;

4

PsA, psoriatic arthritis;

5

AS, ankylosing spondylitis;

6

RA, rheumatoid arthritis;

7

SS, statistically significant;

8

VASI, vitiligo area scoring index;

9

VIDA, vitiligo disease activity score;

10

NB-UVB, narrow-band ultraviolet B;

11

Tx, treated;

12

MITF, microphthalmia-associated transcription factor;

13

BCG + PSN, Balillus Calmette-Guerin fraction

4.1.1. Peripheral blood Th17 cells in vitiligo

Th17 cells are a subset of CD4+ T cells that secrete several immune modulatory substances, including IL-17, IL-21, IL-22, GM-CSF, and CCL20. These cells and their effector molecules have been implicated in the pathogenesis of various autoimmune diseases[49]. Two studies have investigated whether circulating Th17 cells are elevated in vitiligo patients. A cross-sectional study of 45 patients with active non-segmental vitiligo (NSV) compared to 45 age-, gender-, and race-matched healthy controls used flow cytometry to quantify peripheral blood Th17 cells. The abundance of these IL-17 producing cells was increased in vitiligo relative to control subjects (p=0.001) and positively correlated with affected body surface area (r=0.0615, p<0.001)[50]. A second cross-sectional study of 5 vitiligo patients did not report similar findings; however, their sample size was not sufficiently powered[51].

4.1.2. Serum levels of IL-17 in vitiligo

Seven studies quantified serum IL-17, with six showing strong evidence of higher levels in vitiligo and one not demonstrating any difference between vitiligo and control subjects. Zhou et al. detected significantly higher serum IL-17 in 45 subjects with active NSV versus 45 controls using ELISA (p = 0.0145)[50]. Two other studies reported similar findings, with 7-fold higher levels on average in vitiligo subjects[52,53]. One of these additionally uncovered significant positive correlations between serum IL-17 and both disease duration (r=0.42, p=0.02) and extent of body area involvement of vitiligo (r=0.65, p<0.001)[52].

Serum IL-17 levels have also been shown to be negatively correlated with age of disease onset (r = −0.397, p = 0.011) and positively correlated with extent of body area involvement (r = 0.329, p= 0.038) in 40 patients with vitiligo[54].

Systemic IL-17A was quantified by both ELISA and quantitative real-time PCR (qRT-PCR) in a case-control study of 84 NSV patients and 80 healthy controls. Vitiligo subjects were noted to have nearly 2-fold higher serum IL-17 compared to controls (p<0.001) that positively correlated with both Vitiligo Area Scoring Index (VASI) and Vitiligo Disease Activity (VDA)[55].

Another case-control study similarly quantified peripheral IL-17 levels of 80 vitiligo patients and 70 healthy controls, but also further subdivided the vitiligo group based on disease stage (60 active versus 20 stable). Serum IL-17 was noted to be 1.4-fold higher on average in the pooled vitiligo versus control groups and 1.2-fold higher in active versus stable stage vitiligo subjects (p=0.001 for both)[56]. In contrast, two other groups did not note any correlation between serum IL-17 and disease stage, although they had smaller sample sizes of only 15 vitiligo patients each[57,58]. Nevertheless, Habeb and colleagues did still report significantly higher serum IL-17 in vitiligo subjects compared to age- and sex-matched controls, as detected by RT-PCR (p<0.01). They also noted a significant difference in systemic IL-17 between subjects with positive versus negative family history of vitiligo and patients with early versus late onset of vitiligo[58].

4.1.3. IL-17+ mononuclear cells in vitiligo lesions

In addition to studies examining inflammatory markers within peripheral blood, a number of studies have focused on whether such markers are also elevated within affected skin tissue. Histologic examination of vitiliginous skin has revealed that the perilesional margins contain an infiltrate of activated cytotoxic T cells[59]. Provision of melanocyte antigen-specific stimulation to T cells isolated from perilesional skin biopsies of vitiligo has been noted to induce substantial numbers of IL-17-producing CD8+ T cells in response from one patient[60]. Additional immunohistochemical and immunofluorescent studies have examined leading edge biopsies of vitiligo lesions and demonstrated greater numbers of IL-17A+ and IL-17A receptor+ staining in lesional compared to non-lesional vitiliginous skin[61,62].

4.1.4. Tissue expression of IL-17 mRNA in vitiligo lesions

Six studies have investigated whether tissue IL-17 mRNA expression is higher in vitiligo lesions compared to non-lesions. Lesional expression of IL-17 mRNA in NSV has been reported to be up to 7-fold higher, with perilesional expression 3-fold higher compared to non-lesional control skin[52,55,63]. The extent of body surface affected by vitiligo (r=0.48, p<0.05), disease duration (r=0.45, p<0.015), and serum IL-17 (r=0.54, p=0.002) have also been noted to positively correlate with tissue IL-17 expression[52]. Additionally, two studies demonstrated consistently higher IL-17 mRNA expression in leading edge biopsies of vitiliginous lesions compared to non-lesions via qRT-PCR[60,61].

Lastly, a trend toward increased IL-17 gene expression in the neutrophils of 15 vitiligo patients has also been reported[57].

4.1.5. NB-UVB therapy and IL-17, Th17 in vitiligo

Narrow-band ultraviolet B (NB-UVB) light therapy is considered a cornerstone of vitiligo treatment[64,65]. To explore a functional role for IL-17 and Th17 cells in vitiligo, a few studies have measured the effect of NB-UVB treatment on the two. One study found that after 12 weeks of NB-UVB treatment, IL-17 expression decreased by 33% lesionally and 50% perilesionally (p<0.05 for both). This was accompanied by significant reductions in VASI (p<0.001). Moreover, VASI was significantly positively correlated with systemic IL-17 levels at baseline and post-treatment[63].

Another group noted that serum IL-17 remained higher in vitiligo patients post NB-UVB relative to healthy controls, but that treated patients had a 7.5% reduction compared to their untreated counterparts (p<0.001) Their results suggest that NB-UVB treatment abrogates IL-17A secretion, which may in turn be responsible for the clinical improvement of vitiligo seen with NB-UVB therapy[56].

CO2 laser therapy has also been noted to lead to a significant reduction in circulating Th17 lymphocytes, serum IL-17, and IL-17 mRNA expression within cutaneous lesions[66].

4.1.6. Direct effects of exogenous IL-17 on melanocytes

To expand upon the correlative studies reporting elevated IL-17 in the sera and skin of vitiligo patients, one group investigated the direct functional effects of exogenous IL-17 on melanocytes. Kotobuki et al. showed that expression of MITF, an essential transcriptional regulator of melanogenesis, and its downstream genes was reduced by more than 10% in melanocytes treated with IL-17 (p<0.01). This trend was also seen for expression of the anti-apoptotic BCL2 molecule, with more than 20% reduction post IL-17-treatment (p<0.05). The authors also noted an approximately 30% decrease in melanin production from IL-17-treated melanocytes (p<0.05). Furthermore, IL-17 was observed to cause morphological shrinking of melanocytes, further contributing to decreased pigment production. Finally, IL-17 robustly induced expression of the pro-inflammatory cytokines IL-1β, IL-6, and TNF-α in skin-resident keratinocytes and fibroblasts in a dose-dependent fashion (p<0.05). The authors suggested that IL-17 contributes to local depigmentation in autoimmune vitiligo via its antagonism of factors related to melanocyte function, reduction in melanogenesis, and dramatic induction of other Th17 type cytokines from dermal fibroblasts and keratinocytes[62].

4.2. Mouse Studies

Mouse models of vitiligo have also investigated the impact of IL-17 on disease development (Table 2).

Table 2.

Mouse studies examining IL-17 biomarkers in vitiligo

Study Mouse Line Biomarkers Examined Key Results Implications
Eby et al, 201467 Triple transgenic mice expressing epidermal melanocytes, melanocyte-reactive T cells, and HLA-A21, IL-17 expression (splenocytes) (spontaneous and after stimulation)
IL-17 expression (T cells) (spontaneous and after Ag2 presentation)
IL-17-producing T cell frequency (spontaneous and after Ag presentation)
Increased IL-17 expression from Vit3 splenocytes spontaneously (P= .0281) and after stimulation (P= .0091) compared to control
Increased expression of IL-17 from Vit T cells spontaneously (P= .0184) and after Ag presentation (P= .0004) compared to control
Increased frequency of IL-17-producing T cells in Vit spontaneously (P = .0281) and after Ag presentation (P= .039) compared to control
IL-17 levels are increased in mouse models of autoimmune Vit at baseline and after Ag-specific TCR4 stimulation
Chatterjee et al, 201468 h3TA2 transgenic mice with T cells with HLA-A2-restricted human tyrosinase peptide reactive TCR IL-17 expression (splenocytes)
IL-17+ cell frequency (skin)
Increased expression of IL-17 from TCR-cognate peptide stimulated splenocytes vs control peptide stimulated splenocytes (P< .01)
Increased frequency of IL-17+ cells in Treg5-depleted mice compared to Treg-preserved mice (P< .0001)
Reduced Tregs cause depigmentation mediated in part via upregulation of IL-17
Nelson et al, 201569 ICOS6-deficient and ICOSL7-deficient mice Autoimmune Vit development ICOS agonist promoted Vit development compared to IgG8 control (p=0.003)
ICOS antagonist impaired capacity to induce Vit (p<0.05)
ICOS directly modulates IL-17 expression, which in turn modulates Vit development
1

HLA-A2, human leukocyte antigen-A2;

2

Ag, antigen;

3

Vit; vitiligo;

4

TCR, T-cell receptor;

5

Treg, regulatory T cell;

6

ICOS, inducible costimulator;

7

ICOSL, inducible costimulator ligand;

8

IgG, immunoglobulin G

Eby and colleagues used triple transgenic “Vitesse” mice expressing epidermal melanocytes, melanocyte-reactive T cells, and HLA-A2 to rapidly induce depigmentation in a manner analogous to human vitiligo. These mice had 2.5-fold increased numbers of spontaneous and 6.3-fold increased numbers of inducible IL-17A-producing mononuclear cells compared to the control mice (p=0.0281 and p=0.0091, respectively). Vitesse splenocytes also exhibited significantly higher IL-17A secretion (p=0.0281). The authors noted 3-fold increased antigen-specific IL-17A secretion from Vitesse T cells upon stimulation with cognate peptide (p=0.0184). Finally, the number of circulating IL-17 secreting T cells as well as IL-17 tissue expression increased 1.3-fold (p=0.0039) and 27.2-fold (p=0.0004), respectively in response to tyrosinase presentation, while the number of IFN-gamma-secreting cells did not[67].

Similar trends have been reported in an IFN-gamma-deficient transgenic mouse model expressing a tyrosinase-reactive T cell receptor (TCR). Upon depletion of Tregs, which normally function to keep auto-reactive T cells in check, the transgenic TCRs had increased IL-17 secretion (p<0.01). This trend was also seen after antigen-specific TCR stimulation with cognate peptide (p<0.01). Additionally, there were increased numbers of IL-17+ T cells seen infiltrating the skin of these mice (p<0.01)[68].

Lastly the inducible costimulator (ICOS), which is important for the functional maintenance of IL-17-secreting T cells, has also been used to study development of autoimmune vitiligo in mice. Infusion of an ICOS agonist has been reported to enhance the capacity to induce autoimmune vitiligo compared to an IgG control (p=0.003), while infusion of an ICOS antagonist impairs the capacity to induce autoimmune vitiligo (p<0.05)[69].

5. Putative roles for IL-17 in vitiligo pathogenesis

Th17 cells are critical mediators of defense against extracellular microbes, and as such can coordinate a wide array of physiological responses from a variety of cell types. Numerous facets of their role in this defense, mediated largely by their prototypical effector molecule IL-17, may exacerbate autoimmune inflammation in vitiligo. IL-17 is a potent producer of the chemokine CCL20, a homing molecule that can attract cytotoxic CD8+ T cells from systemic circulation into peripheral tissues[7072]. CD8+ T cells kill self-cells upon antigen recognition and form a critical component of adaptive immunity. These cells have been implicated in the direct killing of melanocytes in vitiligo models[60,73], and thus their recruitment by IL-17 may play an important role in disease pathogenesis[44,7476].

IL-17 stimulates endothelial expression of E- and P-selectins as well as the adhesion molecules ICAM-1 and VCAM-1, to enhance neutrophil migration [77]. IL-17 also stimulates keratinocytes to release several chemokines that result in further T cell, neutrophil, macrophage, and dendritic cell influx[78]. The presence of infiltrating macrophages and T cells has been shown to coincide with loss of melanocytes[79]. In addition, neutrophil influx promotes production of several ROS[80]. Oxidative stress induced by these intermediates, in turn, has been associated with vitiligo. ROS accumulation may be directly toxic to critical cell components, resulting in melanocyte destruction and subsequent skin depigmentation[48]. Interestingly NB-UVB, which has demonstrated clinical efficacy in treating vitiligo[64,65], helps relieve oxidative stress and restore oxidant-antioxidant balance, possibly mediated in part through down-regulation of IL-17[81]. As demonstrated in psoriasis, NB-UVB down-regulates both local and systemic IL-17 resulting in decreased inflammation and clinical improvement of psoriatic lesions[82,83]. Studies reviewed in this article have corroborated that NB-UVB leads to significant reductions in IL-17 expression and simultaneous clinical improvement of vitiliginous lesions, as well. This trend is suggestive of the substantial role that IL-17 may play in vitiligo pathogenesis and may explain the efficacy of NB-UVB treatment.

Studies on melanoma, a cancer of melanocytes, have also shed light on putative roles for the Th17 program in an anti-melanocyte immune response. Melanocyte-specific T cells were able to clear melanoma and induce vitiligo best when cultured in Th17-polarizing conditions, as opposed to conditions that favored Th1 or Th2 differentiation[73]. As noted previously, IL-17 has been shown to antagonize melanogenesis by decreasing expression of MITF and also promote melanocyte death by down-regulating BCL2[62]. Indeed, melanocyte apoptosis is thought to be one mechanism of depigmentation in vitiligo[84].

IL-17 also induces the production of angiogenic factors such as vascular endothelial growth factor (VEGF)[85]. VEGF is critical in generating new blood vessels and increasing permeability of systemic circulation for the passage of immune cells into peripheral tissues[86,87]. These functions are thought to explain the critical role of VEGF in inflammation, and may also be operative in enhancing migration of melanocyte-reactive T cells to cutaneous melanocytes in vitiligo. Importantly, increased dermal angiogenesis has been documented in vitiligo, primarily in the center of lesions[88,89].

6. Conclusion

In summary, review of the literature suggests that IL-17 is significantly correlated with autoimmune vitiligo and may be an integral factor in its progression and severity. Human studies have demonstrated an increased frequency of circulating Th17 cells and higher serum IL-17 in vitiligo patients that positively correlates with disease duration, extent, and activity. Vitiliginous lesions contain significantly higher numbers of IL-17-secreting Th17 and CD8+ cells as compared to unaffected skin in both healthy controls and patients. Several studies have also demonstrated dramatically increased expression of IL-17 mRNA within vitiliginous lesions that also positively correlates with disease duration and extent. NB-UVB therapy, which has been shown to clinically improve vitiligo, reduces Th17 cell abundance as well as serum and tissue IL-17 levels. Finally, exogenous IL-17 directly antagonizes factors related to melanocyte function and survival, possibly contributing to clinical depigmentation. Mouse studies have further demonstrated that depigmentation correlates with greater IL-17A secretion, which modulates vitiligo development.

Further clinical studies are needed to determine the implications of directly targeting the IL-17 pathway in order to treat vitiligo. IL-17 inhibition has been effective for psoriasis, but resulted in a worsening of Crohn’s disease. It is important to note, however, that the primary sites of inflammation differ between the two diseases (skin versus intestines). Therefore, clinical studies examining the therapeutic effect and any adverse events of IL-17 blockade on vitiligo should be pursued based on current molecular data. Moreover, IL-17 or IL-17 pathway molecules could be investigated as a surrogate marker to measure the response to established treatments in vitiligo, such as NB-UVB therapy. Together, the reviewed studies indicate a strong correlation between IL-17 activity and autoimmune vitiligo; however, further studies are needed to investigate a potential causal role.

Highlights.

  • IL-17 and the Th17 program have been implicated in the pathogenesis of several autoimmune and inflammatory disorders.

  • Several human and mouse studies have shown that IL-17 is significantly correlated with vitiligo and may play a role in its pathogenesis.

  • Serum IL-17 may have utility as a surrogate marker to measure response to UVB and other therapies in vitiligo.

  • Clinical studies may be warranted to investigate IL-17 blockade as a potential therapeutic target in vitiligo.

Footnotes

Conflicts of Interest

Dr. Liao receives funding from the NIH (R01AR065174, U01AI119125) and serves as a research investigator for Abbvie, Janssen, and Novartis.

Dr. Bhutani serves as a research investigator for Abbvie, Janssen, and Merck.

The other authors (Ms. Rasnik K. Singh, Ms. Kristina M. Lee, Dr. Ivan Vujkovic-Cvijin, Dr. Derya Ucmak, Mr. Benjamin Farahnik, Mr. Michael Abrouk, Dr. Mio Nakamura, Mr. Tian Hao Zhu, and Dr. Maria Wei) report no conflicts of interest, financial or other.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Singh RP, Hasan S, Sharma S, Nagra S, Yamaguchi DT, Wong DT, et al. Th17 cells in inflammation and autoimmunity. Autoimmun Rev. 2014;13:1174–81. doi: 10.1016/j.autrev.2014.08.019. [DOI] [PubMed] [Google Scholar]
  • 2.Jin W, Dong C. IL-17 cytokines in immunity and inflammation. Emerg Microbes Infect. 2013;2:e60. doi: 10.1038/emi.2013.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Weaver CT, Harrington LE, Mangan PR, Gavrieli M, Murphy KM. Th17: an effector CD4 T cell lineage with regulatory T cell ties. Immunity. 2006;24:677–88. doi: 10.1016/j.immuni.2006.06.002. [DOI] [PubMed] [Google Scholar]
  • 4.Wright JF, Bennett F, Li B, Brooks J, Luxenberg DP, Whitters MJ, et al. The human IL-17F/IL-17A heterodimeric cytokine signals through the IL-17RA/IL-17RC receptor complex. J Immunol. 2008;181:2799–805. doi: 10.4049/jimmunol.181.4.2799. [DOI] [PubMed] [Google Scholar]
  • 5.Yang XO, Chang SH, Park H, Nurieva R, Shah B, Acero L, et al. Regulation of inflammatory responses by IL-17F. J Exp Med. 2008;205:1063–75. doi: 10.1084/jem.20071978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Martin DA, Towne JE, Kricorian G, Klekotka P, Gudjonsson JE, Krueger JG, et al. The emerging role of IL-17 in the pathogenesis of psoriasis: preclinical and clinical findings. J Invest Dermatol. 2013;133:17–26. doi: 10.1038/jid.2012.194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Langley RG, Elewski BE, Lebwohl M, Reich K, Griffiths CEM, Papp K, et al. Secukinumab in Plaque Psoriasis — Results of Two Phase 3 Trials. N Engl J Med. 2014;371:326–38. doi: 10.1056/NEJMoa1314258. [DOI] [PubMed] [Google Scholar]
  • 8.Blauvelt A, Prinz JC, Gottlieb AB, Kingo K, Sofen H, Ruer-Mulard M, et al. Secukinumab administration by pre-filled syringe: efficacy, safety and usability results from a randomized controlled trial in psoriasis (FEATURE) Br J Dermatol. 2015;172:484–93. doi: 10.1111/bjd.13348. [DOI] [PubMed] [Google Scholar]
  • 9.Thaçi D, Blauvelt A, Reich K, Tsai T-F, Vanaclocha F, Kingo K, et al. Secukinumab is superior to ustekinumab in clearing skin of subjects with moderate to severe plaque psoriasis: CLEAR, a randomized controlled trial. J Am Acad Dermatol. 2015;73:400–9. doi: 10.1016/j.jaad.2015.05.013. [DOI] [PubMed] [Google Scholar]
  • 10.Paul C, Lacour J-P, Tedremets L, Kreutzer K, Jazayeri S, Adams S, et al. Efficacy, safety and usability of secukinumab administration by autoinjector/pen in psoriasis: a randomized, controlled trial (JUNCTURE) J Eur Acad Dermatology Venereol. 2015;29:1082–90. doi: 10.1111/jdv.12751. [DOI] [PubMed] [Google Scholar]
  • 11.Krueger JG, Fretzin S, Suárez-Fariñas M, Haslett PA, Phipps KM, Cameron GS, et al. IL-17A is essential for cell activation and inflammatory gene circuits in subjects with psoriasis. J Allergy Clin Immunol. 2012;130:145–54.e9. doi: 10.1016/j.jaci.2012.04.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Chandrakumar SF, Yeung J. Interleukin-17 antagonists in the treatment of psoriasis. J Cutan Med Surg. n.d;19:109–14. doi: 10.2310/7750.2014.14038. [DOI] [PubMed] [Google Scholar]
  • 13.Lønnberg AS, Zachariae C, Skov L. Targeting of interleukin-17 in the treatment of psoriasis. Clin Cosmet Investig Dermatol. 2014;7:251–9. doi: 10.2147/CCID.S67534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Brown G, Malakouti M, Wang E, Koo JY, Levin E. Anti-IL-17 phase II data for psoriasis: A review. J Dermatolog Treat. 2015;26:32–6. doi: 10.3109/09546634.2013.878448. [DOI] [PubMed] [Google Scholar]
  • 15.Gordon K. Ixekizumab for Treatment of Moderate-to-Severe Plaque Psoriasis: 60-Week Results From a Double-Blind Phase 3 Induction and Randomized Withdrawal Study (UNCOVER-1). Presented at the 73rd Annual Meeting of the American Academy of Dermatology; 2015 March 20; 2015. [Google Scholar]
  • 16.Griffiths CEM, Reich K, Lebwohl M, van de Kerkhof P, Paul C, Menter A, et al. Comparison of ixekizumab with etanercept or placebo in moderate-to-severe psoriasis (UNCOVER-2 and UNCOVER-3): results from two phase 3 randomised trials. Lancet. 2015;386:541–51. doi: 10.1016/S0140-6736(15)60125-8. [DOI] [PubMed] [Google Scholar]
  • 17.Lebwohl M, Strober B, Menter A, Gordon K, Weglowska J, Puig L, et al. Phase 3 Studies Comparing Brodalumab with Ustekinumab in Psoriasis. N Engl J Med. 2015;373:1318–28. doi: 10.1056/NEJMoa1503824. [DOI] [PubMed] [Google Scholar]
  • 18.Hwang S-Y, Kim H-Y. Expression of IL-17 homologs and their receptors in the synovial cells of rheumatoid arthritis patients. Mol Cells. 2005;19:180–4. [PubMed] [Google Scholar]
  • 19.Metawi SA, Abbas D, Kamal MM, Ibrahim MK. Serum and synovial fluid levels of interleukin-17 in correlation with disease activity in patients with RA. Clin Rheumatol. 2011;30:1201–7. doi: 10.1007/s10067-011-1737-y. [DOI] [PubMed] [Google Scholar]
  • 20.Kellner H. Targeting interleukin-17 in patients with active rheumatoid arthritis: rationale and clinical potential. Ther Adv Musculoskelet Dis. 2013;5:141–52. doi: 10.1177/1759720X13485328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Nakae S, Nambu A, Sudo K, Iwakura Y. Suppression of Immune Induction of Collagen-Induced Arthritis in IL-17-Deficient Mice. J Immunol. 2003;171:6173–7. doi: 10.4049/jimmunol.171.11.6173. [DOI] [PubMed] [Google Scholar]
  • 22.Novelli L, Chimenti MS, Chiricozzi A, Perricone R. The new era for the treatment of psoriasis and psoriatic arthritis: Perspectives and validated strategies. Autoimmun Rev. 2014;13:64–9. doi: 10.1016/j.autrev.2013.08.006. [DOI] [PubMed] [Google Scholar]
  • 23.Lubberts E, Koenders MI, Oppers-Walgreen B, van den Bersselaar L, Coenen-de Roo CJJ, Joosten LAB, et al. Treatment with a neutralizing anti-murine interleukin-17 antibody after the onset of collagen-induced arthritis reduces joint inflammation, cartilage destruction, and bone erosion. Arthritis Rheum. 2004;50:650–9. doi: 10.1002/art.20001. [DOI] [PubMed] [Google Scholar]
  • 24.Bush KA, Farmer KM, Walker JS, Kirkham BW. Reduction of joint inflammation and bone erosion in rat adjuvant arthritis by treatment with interleukin-17 receptor IgG1 Fc fusion protein. Arthritis Rheum. 2002;46:802–5. doi: 10.1002/art.10173. [DOI] [PubMed] [Google Scholar]
  • 25.Matusevicius D, Kivisäkk P, He B, Kostulas N, Ozenci V, Fredrikson S, et al. Interleukin-17 mRNA expression in blood and CSF mononuclear cells is augmented in multiple sclerosis. Mult Scler. 1999;5:101–4. doi: 10.1177/135245859900500206. [DOI] [PubMed] [Google Scholar]
  • 26.Rostami A, Ciric B. Role of Th17 cells in the pathogenesis of CNS inflammatory demyelination. J Neurol Sci. 2013;333:76–87. doi: 10.1016/j.jns.2013.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Huppert J, Closhen D, Croxford A, White R, Kulig P, Pietrowski E, et al. Cellular mechanisms of IL-17-induced blood-brain barrier disruption. FASEB J. 2010;24:1023–34. doi: 10.1096/fj.09-141978. [DOI] [PubMed] [Google Scholar]
  • 28.Alunno A, Carubbi F, Bartoloni E, Bistoni O, Caterbi S, Cipriani P, et al. Unmasking the pathogenic role of IL-17 axis in primary Sjögren’s syndrome: A new era for therapeutic targeting? Autoimmun Rev. 2014;13:1167–73. doi: 10.1016/j.autrev.2014.08.022. [DOI] [PubMed] [Google Scholar]
  • 29.Ogawa A, Andoh A, Araki Y, Bamba T, Fujiyama Y. Neutralization of interleukin-17 aggravates dextran sulfate sodium-induced colitis in mice. Clin Immunol. 2004;110:55–62. doi: 10.1016/j.clim.2003.09.013. [DOI] [PubMed] [Google Scholar]
  • 30.Cao AT, Yao S, Gong B, Elson CO, Cong Y. Th17 cells upregulate polymeric Ig receptor and intestinal IgA and contribute to intestinal homeostasis. J Immunol. 2012;189:4666–73. doi: 10.4049/jimmunol.1200955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Hueber W, Sands BE, Lewitzky S, Vandemeulebroecke M, Reinisch W, Higgins PDR, et al. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn’s disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut. 2012;61:1693–700. doi: 10.1136/gutjnl-2011-301668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Targan SR, Feagan BG, Vermeire S, Panaccione R, Melmed GY, Blosch C, et al. Mo2083 A Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Safety, Tolerability, and Efficacy of AMG 827 in Subjects With Moderate to Severe Crohn’s Disease. Gastroenterology. 2012;143:e26. doi: 10.1053/j.gastro.2012.07.084. [DOI] [Google Scholar]
  • 33.Fuss I. IL-17: intestinal effector or protector? Mucosal Immunol. 2011;4:366–7. doi: 10.1038/mi.2011.24. [DOI] [Google Scholar]
  • 34.Fitzpatrick LR. Inhibition of IL-17 as a Pharmacological Approach for IBD. Int Rev Immunol. 2013;32:544–55. doi: 10.3109/08830185.2013.821118. [DOI] [PubMed] [Google Scholar]
  • 35.Jin Y, Birlea SA, Fain PR, Gowan K, Riccardi SL, Holland PJ, et al. Variant of TYR and autoimmunity susceptibility loci in generalized vitiligo. N Engl J Med. 2010;362:1686–97. doi: 10.1056/NEJMoa0908547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Jin Y, Birlea SA, Fain PR, Gowan K, Riccardi SL, Holland PJ, et al. Genome-wide analysis identifies a quantitative trait locus in the MHC class II region associated with generalized vitiligo age of onset. J Invest Dermatol. 2011;131:1308–12. doi: 10.1038/jid.2011.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Quan C, Ren Y-Q, Xiang L-H, Sun L-D, Xu A-E, Gao X-H, et al. Genome-wide association study for vitiligo identifies susceptibility loci at 6q27 and the MHC. Nat Genet. 2010;42:614–8. doi: 10.1038/ng.603. [DOI] [PubMed] [Google Scholar]
  • 38.Cheong KA, Kim N-H, Noh M, Lee A-Y. Three new single nucleotide polymorphisms identified by a genome-wide association study in Korean patients with vitiligo. J Korean Med Sci. 2013;28:775–9. doi: 10.3346/jkms.2013.28.5.775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.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. doi: 10.1034/j.1600-0749.2003.00032.x. [DOI] [PubMed] [Google Scholar]
  • 40.Norris D. The Neurosensory System Controls Keratinocyte Release of Growth and Survival Factor Nerve Growth Factor. J Invest Dermatol. 2001;117:1025–6. doi: 10.1046/j.1523-1747.2001.00000.x. [DOI] [Google Scholar]
  • 41.Peters EMJ, Handjiski B, Kuhlmei A, Hagen E, Bielas H, Braun A, et al. Neurogenic Inflammation in Stress-Induced Termination of Murine Hair Growth Is Promoted by Nerve Growth Factor. Am J Pathol. 2004;165:259–71. doi: 10.1016/S0002-9440(10)63294-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.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. doi: 10.1111/1523-1747.ep12492607. [DOI] [PubMed] [Google Scholar]
  • 43.Kemp EH, Emhemad S, Akhtar S, Watson PF, Gawkrodger DJ, Weetman AP. Autoantibodies against tyrosine hydroxylase in patients with non-segmental (generalised) vitiligo. Exp Dermatol. 2011;20:35–40. doi: 10.1111/j.1600-0625.2010.01181.x. [DOI] [PubMed] [Google Scholar]
  • 44.You S, Cho Y-H, Byun J-S, Shin E-C. Melanocyte-specific CD8+ T cells are associated with epidermal depigmentation in a novel mouse model of vitiligo. Clin Exp Immunol. 2013;174:38–44. doi: 10.1111/cei.12146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Yang L, Wei Y, Sun Y, Shi W, Yang J, Zhu L, et al. Interferon-gamma Inhibits Melanogenesis and Induces Apoptosis in Melanocytes: A Pivotal Role of CD8+ Cytotoxic T Lymphocytes in Vitiligo. Acta Derm Venereol. 2015;95:664–70. doi: 10.2340/00015555-2080. [DOI] [PubMed] [Google Scholar]
  • 46.Kolls JK, Lindén A. Interleukin-17 family members and inflammation. Immunity. 2004;21:467–76. doi: 10.1016/j.immuni.2004.08.018. [DOI] [PubMed] [Google Scholar]
  • 47.Taher ZA, Lauzon G, Maguiness S, Dytoc MT. Analysis of interleukin-10 levels in lesions of vitiligo following treatment with topical tacrolimus. Br J Dermatol. 2009;161:654–9. doi: 10.1111/j.1365-2133.2009.09217.x. [DOI] [PubMed] [Google Scholar]
  • 48.Khan R, Satyam A, Gupta S, Sharma VK, Sharma A. Circulatory levels of antioxidants and lipid peroxidation in Indian patients with generalized and localized vitiligo. Arch Dermatol Res. 2009;301:731–7. doi: 10.1007/s00403-009-0964-4. [DOI] [PubMed] [Google Scholar]
  • 49.Maddur MS, Miossec P, Kaveri SV, Bayry J. Th17 cells: biology, pathogenesis of autoimmune and inflammatory diseases, and therapeutic strategies. Am J Pathol. 2012;181:8–18. doi: 10.1016/j.ajpath.2012.03.044. [DOI] [PubMed] [Google Scholar]
  • 50.Zhou L, Shi Y-L, Li K, Hamzavi I, Gao T-W, Huggins RH, et al. Increased circulating Th17 cells and elevated serum levels of TGF-beta and IL-21 are correlated with human non-segmental vitiligo development. Pigment Cell Melanoma Res. 2015;28:324–9. doi: 10.1111/pcmr.12355. [DOI] [PubMed] [Google Scholar]
  • 51.Jandus C, Bioley G, Rivals J-P, Dudler J, Speiser D, Romero P. Increased numbers of circulating polyfunctional Th17 memory cells in patients with seronegative spondylarthritides. Arthritis Rheum. 2008;58:2307–17. doi: 10.1002/art.23655. [DOI] [PubMed] [Google Scholar]
  • 52.Bassiouny DA, Shaker O. Role of interleukin-17 in the pathogenesis of vitiligo. Clin Exp Dermatol. 2011;36:292–7. doi: 10.1111/j.1365-2230.2010.03972.x. [DOI] [PubMed] [Google Scholar]
  • 53.Khan R, Gupta S, Sharma A. Circulatory levels of T-cell cytokines (interleukin [IL]-2, IL-4, IL-17, and transforming growth factor-β) in patients with vitiligo. J Am Acad Dermatol. 2012;66:510–1. doi: 10.1016/j.jaad.2011.07.018. [DOI] [PubMed] [Google Scholar]
  • 54.Basak PY, Adiloglu AK, Ceyhan AM, Tas T, Akkaya VB. The role of helper and regulatory T cells in the pathogenesis of vitiligo. J Am Acad Dermatol. 2009;60:256–60. doi: 10.1016/j.jaad.2008.09.048. [DOI] [PubMed] [Google Scholar]
  • 55.Elela MA, Hegazy RA, Fawzy MM, Rashed LA, Rasheed H. Interleukin 17, interleukin 22 and FoxP3 expression in tissue and serum of non-segmental vitiligo: a case- controlled study on eighty-four patients. Eur J Dermatol. n.d;23:350–5. doi: 10.1684/ejd.2013.2023. [DOI] [PubMed] [Google Scholar]
  • 56.Tembhre MK, Sharma VK, Sharma A, Chattopadhyay P, Gupta S. T helper and regulatory T cell cytokine profile in active, stable and narrow band ultraviolet B treated generalized vitiligo. Clin Chim Acta. 2013;424:27–32. doi: 10.1016/j.cca.2013.05.005. [DOI] [PubMed] [Google Scholar]
  • 57.Esmaeili B, Rezaee SAR, Layegh P, Tavakkol Afshari J, Dye P, Ghayoor Karimiani E, et al. Expression of IL-17 and COX2 gene in peripheral blood leukocytes of vitiligo patients. Iran J Allergy Asthma Immunol. 2011;10:81–9. doi:010.02/ijaai.8189. [PubMed] [Google Scholar]
  • 58.Habeb A. Expression of Interleukin-17 mRNA in Vitiligo Patients. Egypt J Dermatology Venerol. 2013;33:67–60. [Google Scholar]
  • 59.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. doi: 10.1002/path.1711700209. [DOI] [PubMed] [Google Scholar]
  • 60.van den Boorn JG, Konijnenberg D, Dellemijn TAM, van der Veen JPW, Bos JD, Melief CJM, et al. Autoimmune destruction of skin melanocytes by perilesional T cells from vitiligo patients. J Invest Dermatol. 2009;129:2220–32. doi: 10.1038/jid.2009.32. [DOI] [PubMed] [Google Scholar]
  • 61.Wang CQF, Cruz-Inigo AE, Fuentes-Duculan J, Moussai D, Gulati N, Sullivan-Whalen M, et al. Th17 cells and activated dendritic cells are increased in vitiligo lesions. PLoS One. 2011;6:e18907. doi: 10.1371/journal.pone.0018907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kotobuki Y, Tanemura A, Yang L, Itoi S, Wataya-Kaneda M, Murota H, et al. Dysregulation of melanocyte function by Th17-related cytokines: significance of Th17 cell infiltration in autoimmune vitiligo vulgaris. Pigment Cell Melanoma Res. 2012;25:219–30. doi: 10.1111/j.1755-148X.2011.00945.x. [DOI] [PubMed] [Google Scholar]
  • 63.Hegazy RA, Fawzy MM, Gawdat HI, Samir N, Rashed LA. T helper 17 and Tregs: a novel proposed mechanism for NB-UVB in vitiligo. Exp Dermatol. 2014;23:283–6. doi: 10.1111/exd.12369. [DOI] [PubMed] [Google Scholar]
  • 64.Nicolaidou E, Antoniou C, Stratigos A, Katsambas AD. Narrowband ultraviolet B phototherapy and 308-nm excimer laser in the treatment of vitiligo: a review. J Am Acad Dermatol. 2009;60:470–7. doi: 10.1016/j.jaad.2008.07.053. [DOI] [PubMed] [Google Scholar]
  • 65.Malerba M, Damiani G, Radaeli A, Ragnoli B, Olivini A, Calzavara-Pinton PG. Narrowband ultraviolet B phototherapy in psoriasis reduces proinflammatory cytokine levels and improves vitiligo and neutrophilic asthma. Br J Dermatol. 2015 doi: 10.1111/bjd.13988. [DOI] [PubMed] [Google Scholar]
  • 66.Zhan L, Xiong X, Wang L. Treatment of BCG polysaccharide nucleic acid combined with CO2 laser reduces Th17 cells and their related cytokines in cutaneous lesion of vitiligo patients. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2014;30:1300–3. [PubMed] [Google Scholar]
  • 67.Eby JM, Kang H-K, Klarquist J, Chatterjee S, Mosenson JA, Nishimura MI, et al. Immune responses in a mouse model of vitiligo with spontaneous epidermal de- and repigmentation. Pigment Cell Melanoma Res. 2014;27:1075–85. doi: 10.1111/pcmr.12284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Chatterjee S, Eby JM, Al-Khami AA, Soloshchenko M, Kang H-K, Kaur N, et al. A quantitative increase in regulatory T cells controls development of vitiligo. J Invest Dermatol. 2014;134:1285–94. doi: 10.1038/jid.2013.540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Nelson MH, Kundimi S, Bowers JS, Rogers CE, Huff LW, Schwartz KM, et al. The inducible costimulator augments Tc17 cell responses to self and tumor tissue. J Immunol. 2015;194:1737–47. doi: 10.4049/jimmunol.1401082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Annunziato F, Cosmi L, Santarlasci V, Maggi L, Liotta F, Mazzinghi B, et al. Phenotypic and functional features of human Th17 cells. J Exp Med. 2007;204:1849–61. doi: 10.1084/jem.20070663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Singh SP, Zhang HH, Foley JF, Hedrick MN, Farber JM. Human T cells that are able to produce IL-17 express the chemokine receptor CCR6. J Immunol. 2008;180:214–21. doi: 10.4049/jimmunol.180.1.214. [DOI] [PubMed] [Google Scholar]
  • 72.Martin-Orozco N, Muranski P, Chung Y, Yang XO, Yamazaki T, Lu S, et al. T helper 17 cells promote cytotoxic T cell activation in tumor immunity. Immunity. 2009;31:787–98. doi: 10.1016/j.immuni.2009.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Muranski P, Boni A, Antony PA, Cassard L, Irvine KR, Kaiser A, et al. Tumor-specific Th17-polarized cells eradicate large established melanoma. Blood. 2008;112:362–73. doi: 10.1182/blood-2007-11-120998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Zhang B-X, Lin M, Qi X-Y, Zhang R-X, Wei Z-D, Zhu J, et al. Characterization of circulating CD8+T cells expressing skin homing and cytotoxic molecules in active non-segmental vitiligo. Eur J Dermatol. n.d;23:331–8. doi: 10.1684/ejd.2013.2011. [DOI] [PubMed] [Google Scholar]
  • 75.Wu J, Zhou M, Wan Y, Xu A. CD8+ T cells from vitiligo perilesional margins induce autologous melanocyte apoptosis. Mol Med Rep. 2013;7:237–41. doi: 10.3892/mmr.2012.1117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.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. doi: 10.1111/ijd.12160. [DOI] [PubMed] [Google Scholar]
  • 77.Suzuki E, Mellins ED, Gershwin ME, Nestle FO, Adamopoulos IE. The IL-23/IL-17 axis in psoriatic arthritis. Autoimmun Rev. 2014;13:496–502. doi: 10.1016/j.autrev.2014.01.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Schröder JM, Gregory H, Young J, Christophers E. Neutrophil-activating proteins in psoriasis. J Invest Dermatol. 1992;98:241–7. doi: 10.1111/1523-1747.ep12556058. [DOI] [PubMed] [Google Scholar]
  • 79.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. [PMC free article] [PubMed] [Google Scholar]
  • 80.Gliński W, Barszcz D, Janczura E, Zarebska Z, Jabłońska S. Neutral proteinases and other neutrophil enzymes in psoriasis, and their relation to disease activity. Br J Dermatol. 1984;111:147–54. doi: 10.1111/j.1365-2133.1984.tb04037.x. [DOI] [PubMed] [Google Scholar]
  • 81.Karsli N, Akcali C, Ozgoztasi O, Kirtak N, Inaloz S. Role of oxidative stress in the pathogenesis of vitiligo with special emphasis on the antioxidant action of narrowband ultraviolet B phototherapy. J Int Med Res. 2014;42:799–805. doi: 10.1177/0300060513516294. [DOI] [PubMed] [Google Scholar]
  • 82.Sigmundsdottir H, Johnston A, Gudjonsson JE, Valdimarsson H. Narrowband-UVB irradiation decreases the production of pro-inflammatory cytokines by stimulated T cells. Arch Dermatol Res. 2005;297:39–42. doi: 10.1007/s00403-005-0565-9. [DOI] [PubMed] [Google Scholar]
  • 83.Coimbra S, Oliveira H, Reis F, Belo L, Rocha S, Quintanilha A, et al. Interleukin (IL)-22, IL-17, IL-23, IL-8, vascular endothelial growth factor and tumour necrosis factor-α levels in patients with psoriasis before, during and after psoralen-ultraviolet A and narrowband ultraviolet B therapy. Br J Dermatol. 2010;163:1282–90. doi: 10.1111/j.1365-2133.2010.09992.x. [DOI] [PubMed] [Google Scholar]
  • 84.Norris DA, Horikawa T, Morelli JG. Melanocyte destruction and repopulation in vitiligo. Pigment Cell Res. 1994;7:193–203. doi: 10.1111/j.1600-0749.1994.tb00049.x. [DOI] [PubMed] [Google Scholar]
  • 85.Pickens SR, Volin MV, Mandelin AM, Kolls JK, Pope RM, Shahrara S. IL-17 contributes to angiogenesis in rheumatoid arthritis. J Immunol. 2010;184:3233–41. doi: 10.4049/jimmunol.0903271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Silva-Santos B. Promoting angiogenesis within the tumor microenvironment: the secret life of murine lymphoid IL-17-producing gammadelta T cells. Eur J Immunol. 2010;40:1873–6. doi: 10.1002/eji.201040707. [DOI] [PubMed] [Google Scholar]
  • 87.Liu J, Duan Y, Cheng X, Chen X, Xie W, Long H, et al. IL-17 is associated with poor prognosis and promotes angiogenesis via stimulating VEGF production of cancer cells in colorectal carcinoma. Biochem Biophys Res Commun. 2011;407:348–54. doi: 10.1016/j.bbrc.2011.03.021. [DOI] [PubMed] [Google Scholar]
  • 88.Aroni K, Voudouris S, Ioannidis E, Grapsa A, Kavantzas N, Patsouris E. Increased angiogenesis and mast cells in the centre compared to the periphery of vitiligo lesions. Arch Dermatol Res. 2010;302:601–7. doi: 10.1007/s00403-010-1040-9. [DOI] [PubMed] [Google Scholar]
  • 89.Stylianos V, Eleftherios I, Nikolaos K, Kyriaki A. Correlating epidermal thickness and basement membrane length to angiogenesis in the centre and the periphery of vitiligo lesion. Indian J Dermatol Venereol Leprol. n.d;78:368–71. doi: 10.4103/0378-6323.95462. [DOI] [PubMed] [Google Scholar]

RESOURCES