Información de la revista
Visitas
448
Review
Acceso a texto completo
Pruebas no corregidas. Disponible online el 3 de septiembre de 2025
New Insights on the Etiopathogenesis of Vitiligo
Actualización en etiopatogenia del vitíligo
Visitas
448
B. Clemente Hernándeza,b,
, T. Gracia-Cazañaa,b, J. Cirizac,d,e,f, Y. Gilabertea,b
a Servicio de Dermatología, Hospital Miguel Servet, IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
b Grupo de Investigación del Gobierno de Aragón B59_23D Dermatología y Fotobiología, Zaragoza, Spain
c Tissue Microenvironment (TME) Lab, Instituto de Investigación en Ingeniería de Aragón (I3A), Universidad de Zaragoza, Zaragoza, Spain
d Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain
e Departamento de Anatomía e Histología, Facultad de Medicina, Universidad de Zaragoza, Zaragoza, Spain
f Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
Contenido relacionado
B. Clemente Hernández, T. Gracia-Cazaña, J. Ciriza, Y. Gilaberte
Este artículo ha recibido
Información del artículo
Resumen
Texto completo
Bibliografía
Descargar PDF
Estadísticas
Figuras (2)
Tablas (5)
Table 1. HLA genes associated with increased risk of developing vitiligo. These studies include genome-wide association studies (GWAS) and linkage analyses in affected families.51–60
Tablas
Table 2. Main genes involved in the etiopathogenesis of vitiligo.
Tablas
Table 3. Immunoregulatory genes involved in T cell development, activation, signaling, and innate immune response associated with vitiligo.4,28,49,50
Tablas
Table 4. Genes related to melanocyte function and survival.4,28,49,50
Tablas
Table 5. Environmental triggers of vitiligo.76–86,88–91
Tablas
Mostrar másMostrar menos
Abstract

Vitiligo is an acquired skin disorder characterised by progressive depigmentation of the skin due to selective loss of melanocytes. Its aetiopathogenesis is complex and multifactorial, involving an interaction between genetic, immunological and environmental factors. Recent evidence leans towards an integrated model in which autoimmunity, mediated mainly by cytotoxic T lymphocytes and inflammatory cytokines, plays a central role. In addition, oxidative stress contributes significantly to melanocyte dysfunction and apoptosis. Genetic studies have identified numerous loci associated with vitiligo, most notably the involvement of genes related to adaptive and innate immunity as well as cellular metabolism. Environmental factors such as trauma, chemical exposure and psychosocial stress may also act as triggers in predisposed individuals. This review synthesises the most recent advances in the aetiopathogenesis of vitiligo, providing a comprehensive overview of the mechanisms involved and opening up new possibilities for the development of therapeutic approaches based on this knowledge.

Keywords:
Vitiligo
Aetiopathogenesis
Melanocytes
Autoimmunity
Oxidative stress
Genetics
Resumen

El vitíligo es un trastorno cutáneo adquirido caracterizado por la despigmentación progresiva de la piel debido a la pérdida selectiva de melanocitos. Su etiopatogenia es compleja y multifactorial, involucrando una interacción entre factores genéticos, inmunológicos y ambientales. Las evidencias recientes se inclinan hacia un modelo integrado en el que la autoinmunidad, mediada principalmente por linfocitos T citotóxicos y citoquinas inflamatorias, desempeña un papel central. Además, el estrés oxidativo contribuye significativamente a la disfunción y apoptosis de los melanocitos. Estudios genéticos han identificado numerosos loci asociados con el vitíligo, destacando la participación de genes relacionados con la inmunidad adaptativa e innata, así como con el metabolismo celular. Asimismo, factores ambientales como el trauma, la exposición a sustancias químicas y el estrés psicosocial pueden actuar como desencadenantes en individuos predispuestos. Esta revisión sintetiza los avances más recientes en la etiopatogenia del vitíligo, ofreciendo una visión completa de los mecanismos implicados, y abriendo nuevas posibilidades para el desarrollo de enfoques terapéuticos basados en este conocimiento.

Palabras clave:
Vitíligo
Etiopatogenia
Melanocitos
Autoinmunidad
Estrés oxidativo
Genética
Resumen gráfico
Texto completo
Introduction

Vitiligo is a depigmenting skin disorder characterized by the selective loss of melanocytes. Its prevalence ranges from 0.5% to 2%, according to an extensive review of prevalence data from over 50 studies worldwide, with no predilection for gender or race.1

For years, the etiopathogenesis of vitiligo has been a subject of study, and although various theories have been proposed, many questions still remain about its precise causes and underlying mechanisms. In this article, we will explore the most recent research that sheds light on the complex interaction of genetic, autoimmune, neurogenic, and environmental factors contributing to the development and progression of vitiligo. Through a comprehensive review of current literature, we aim to offer an updated view of advances in understanding this enigmatic disease, as well as new therapeutic perspectives emerging from this knowledge.

Immunological mechanisms involved in the etiopathogenesis of vitiligoInnate immunity

The innate immune system is considered the crucial link connecting oxidative stress with the adaptive immune response in the development of vitiligo. In lesional and perilesional skin of patients, dendritic cells, macrophages, activated NK cells, and IFN-γ-producing cells are found. Melanocytes, through exosomes, communicate stress to the innate immune system, especially to dendritic cells that present antigens to T lymphocytes. In vitiligo patients, an increase in pro-inflammatory cytokines typical of innate immunity, IL IL-1α, IL-1β, IL-6, IL-8, IL-12, IL-15, and TNF-α, is observed in both serum and skin.2,3

Damage-associated molecular patterns

The Koebner phenomenon is considered an initial trigger for vitiligo. Attempts have been made to identify factors released during stress response and melanocyte damage in this disease. Damage-associated molecular patterns (DAMPs) are of particular interest, as they can trigger the inflammatory response observed in the active stage of vitiligo, with a series of DAMP-associated proteins existing in vitiligo.4

The DAMP with the most evidence of association with vitiligo is heat shock protein 70 (HSP70), which belongs to the family of intracellular chaperones whose function is to prevent incorrect protein folding. The overexpression of HSP70 in active vitiligo lesions indicates a possible role of this protein in the immune response and inflammation associated with the disease. Furthermore, HSP70 could be considered a potential marker to evaluate disease activity.3,5,6

On the other hand, MxA protein (human mixovirus resistance protein 1), inducible by IFN-α, shows strong expression in active perilesional vitiligo skin, suggesting a contributing effect of IFN-α in disease progression.7 Similarly, S100B is another DAMP released by damaged melanocytes, whose levels are increased in active vitiligo and can stimulate inflammatory responses.8 Also, high-mobility group box 1 (HMGB1) proteins can induce the production of chemokine ligands, such as CXCL1 or IL-8 by keratinocytes, acting in the recruitment of immune cells.9

Calreticulin (CRT) is another of the most studied molecules in vitiligo, as it induces melanocyte apoptosis and the release of membrane degradation products important for immunogenicity.10

Melanocyte adhesion deficiency

Several research groups have demonstrated that melanocytes in vitiligo exhibit reduced adhesive properties. Thus, altered levels of E-cadherin expression have been observed in melanocytes from vitiligo-affected skin before depigmentation develops. Deficient E-cadherin expression leads to the loss of epidermal melanocyte adhesion during situations of oxidative or mechanical stress.11

Furthermore, a relevant role of integrins and the Melanoma Inhibitory Activity (MIA) protein has been identified. Integrins are involved in the interaction of melanocytes with the extracellular matrix, and their dysfunction can contribute to melanocyte loss. On the other hand, MIA protein, originally studied in melanoma, has shown a pro-apoptotic effect on melanocytes, favoring their disappearance in vitiligo-affected areas. These findings reinforce the idea that cell adhesion plays a crucial role in the pathogenesis of the disease.12

Inflammasomes

Inflammasomes are multiprotein complexes that, when activated, trigger a cascade of events resulting in the activation of an enzyme called caspase-1. Inflammasome activation could play a crucial role in the inflammation and destruction of melanocytes in non-segmental vitiligo. Inflammasome activation has been found in vitiligo lesions, associated with increased expression of inflammatory cytokines such as IL-1β and IL-18. Therefore, IL-1β inhibition can be seen as a potential therapeutic target in vitiligo.

Polymorphisms in IL-1β and in NOD-like receptor family pyrin domain containing 1 (NLRP1) inflammasome are associated with an increased risk of developing vitiligo. By recognizing DAMPs, this receptor activates the inflammasome, which, via the caspase-1 pathway, induces the processing of pro-IL-1β into active IL-1β. Although the specific connection between NRLP1 and vitiligo is not completely defined, it is likely that inflammasomes, including NRLP1, play an important role in the inflammatory and cell stress processes that characterize this disease.13,14

Type 1 interferon pathway (IFN type 1)

The IFN-1 pathway is an early and transient link in disease progression and marks the junction point between the innate and adaptive immune response. Vitiligo is associated with type 1 interferon activation. Variants of the IFN-induced helicase C domain 1 (IFIH1) gene are related to protection vs vitiligo by inhibiting IFN-α production. This interferon induces chemokines that recruit autoreactive T cells such as CXCL10 and CXCL16. Plasmacytoid dendritic cells (pDC) are responsible for the production of IFN-α in vitiligo skin, stimulated by HSP70.15,36

Adaptive immunity

Damage to melanocytes caused by oxidative stress activates innate immunity, with consequent cytokine secretion and antigen presentation. This triggers the activation of the adaptive immune system, where autoreactive T lymphocytes exacerbate melanocyte damage in vitiligo-affected skin through the secretion of pro-inflammatory cytokines (Fig. 1).3,4,16,17

Figure 1.

Etiopathogenic mechanisms of vitiligo. This figure illustrates all the important keys of how a molecular cascade is triggered, starting with increased melanocyte susceptibility to oxidative stress, which, in the presence of a susceptible genetic basis, leads to immune system activation. CD8+ T cells produce various cytokines such as IFN-γ. The binding of IFN-γ to its receptor activates the JAK-STAT pathway and causes the secretion of CXCL9 and CXCL10. CCL5: chemokine ligand 5; CXCL9: CXC chemokine ligand 9; CXCL10: CXC chemokine ligand 10; CXCL12: CXC chemokine ligand 12; CXCL16: CXC chemokine ligand 16; CXCR3: chemokine receptor type 3; DAMP: damage-associated molecular pattern; IFN-γ: interferon-γ; JAK: janus kinase; MAPK: mitogen-activated protein kinase; NF-kB: nuclear factor kappa light chain enhancer of activated B cells; NLRP1: NOD-like receptor family pyrin domain containing 1 (NLRP1); TNF-α: tumor necrosis factor alpha; ROS: reactive oxygen species; STAT1: signal transducer and activator of transcription 1.

Cytotoxic T cells

Cytotoxic T lymphocytes (CD8+) are the main immune cells involved in the pathogenesis of the disease, primarily in initial or active phases. The mechanism of action is based on the production of inflammatory cytokines such as TNF-α and IFN-γ, in addition to the release of granzymes and perforins. IFN-γ locally increases levels of CXCL9 and CXCL10 in the skin and serum of vitiligo patients, which in turn attracts pathogenic T cells expressing the CXCR3A receptor.18 A predominant lymphocytic infiltrate of CD8+ T cells has been observed in perilesional skin, correlating with disease activity.3

In vitiligo patients, CD8+ T lymphocytes recognize specific melanocyte antigens (MelanA, tyrosinase, gp100, tyrosinase-related proteins 1 and 2, Mart-1). These antigens are found in significantly higher amounts in the peripheral blood of patients vs healthy individuals. Although this phenomenon is considered the primary pathway of melanocyte destruction in vitiligo, the presence of melanocyte-specific T cells is not sufficient, as they have also been found in healthy individuals, showing an anergic phenotype.19,20

Regulatory T cells

The imbalance between pro- and anti-inflammatory signaling is crucial in the pathogenesis of vitiligo. The number of regulatory T cells (Tregs), capable of attenuating the immune response, is decreased in patients. Furthermore, their suppressive capacity is also compromised.21

Differences in the immune response in patients with non-segmental vitiligo and healthy individuals have been explored by examining Treg levels and LRP1 receptor expression in monocytes. Treg and LRP1/CD91 levels before and after treatment in vitiligo patients, vs healthy individuals, show higher levels in vitiligo patients, demonstrating that abnormalities in the immune response contribute to the pathogenesis of vitiligo.22

Cytokines and chemokines

Cytokines, especially IFN-γ and TNF-α, play a crucial role in melanocyte loss and vitiligo progression. IFN-γ activates the JAK/STAT pathway, and TNF-α activates the MAPK and NF-kB pathways, both contributing to inflammation and cell damage. IFN-γ induces the production of chemokines CXCL9 and CXCL10, which amplify inflammation and immune cell recruitment. In murine models, CXCL10 inhibition prevented the disease and promoted repigmentation. Moreover, CXCL12 and CCL5 play an important role in immune cell recruitment in affected skin, and serum CXCL12 levels are associated with disease activity.23 Additionally, oxidative stress induces the secretion of chemokines like CXCL16.24

Other cytokines elevated in vitiligo include IL-1β, IL-2, IL-17, IL-22, IL-23, and IL-33. However, therapies targeting these molecules have shown contradictory results, underscoring the need for further research to find effective treatments.25–27

Autoimmunity

Genetic evidence supports the autoimmune hypothesis as the main mechanism of vitiligo, with approximately 85% of susceptibility genes involved in innate, adaptive immunity, and apoptosis.4,28 This theory is also reinforced by the relationship of vitiligo with other autoimmune disorders, the presence of specific antibodies in patients, and the use of immunomodulatory therapies.4,28

Furthermore, it has been observed that treatments with immune checkpoint inhibitors in cancer patients can induce the development of vitiligo, suggesting a link between autoimmunity and this disease.29

Autoimmune diseases are the main comorbidity associated with vitiligo, including thyroid disorders, pernicious anemia, alopecia areata, connective tissue diseases, among others.30,31

Zombie cells, senescent cells in vitiligo

Cellular senescence is induced in response to cellular stressors. These cells remain metabolically active and secrete a series of proteins and bioactive factors. It has recently been accepted that the presence of senescent cells contributes to the progression of various diseases, including vitiligo. Thus, a significant subset of melanocytes in vitiligo lesions was found to be positive for p16INK4A vs unaffected skin. This finding suggests that p16INK4A could be involved in regulating melanocyte function in non-segmental vitiligo, possibly as a response to oxidative or inflammatory stressors in the skin microenvironment.32

Trained immunity and immune tolerance

A long-term increase in innate memory function, described as trained immunity after vaccination and in other inflammatory diseases, could play a role as an enhancer and continuous trigger in the pathogenesis of vitiligo. After exposure to certain stimuli, the innate immune system is capable of showing an enhanced immunological response to a second stimulus, indicating a memory function of the innate immune system, a concept known as trained immunity. Trained immunity is regulated by epigenetic reprogramming, which includes chemical changes to histones and chromatin accessibility causing sustained changes in the transcription of specific genes.33

Resident memory T cells

The recurrence of vitiligo in previously affected areas demonstrates a memory response involved in the etiopathogenesis of this disease. This relapse risk could be due to the persistence of tissue-resident memory T (TRM) cells, whose maintenance and function are promoted by IL-15. These TRM cells exhibit high levels of CD122, the IL-15 receptor subunit.34–37 This antibody has demonstrated disease reversal in mice, making it a promising therapeutic target.38 Recently, it has been shown that TRM survival depends on the uptake and metabolism of exogenous fatty acids, suggesting that a possible treatment could be the regulation of lipid metabolism, focused on the elimination of TRMs in peripheral tissues.39

Intrinsic melanocyte abnormalities and epidermal and dermal alterations in vitiligo skin

Several in vitro and in vivo studies have demonstrated the presence of intrinsic abnormalities in vitiligo melanocytes characterized by increased susceptibility to pro-oxidant agents. In the epidermis of vitiligo lesions, a marked decrease in the expression of c-kit, the stem cell factor receptor, and the MITF transcription factor (microphthalmia-associated transcription factor) has been detected, along with a reduction in endothelin B receptor.40 A marked reduction in microRNA-211 expression has also been observed, associated with a lower oxygen consumption rate, aberrant mitochondrial complexes, alterations in lipid metabolism, and an increase in reactive oxygen species. This microRNA-211 could serve in the future as a biomarker to evaluate the therapeutic response in this disease.41

The role of endoplasmic reticulum (ER) stress in the pathogenesis of vitiligo has been examined due to its connection with the accumulation of misfolded proteins, which activates the unfolded protein response (UPR). This response tries to restore cellular homeostasis, but if it fails, it can contribute to the development of autoimmune diseases. In vitiligo, ER stress could link oxidative stress with autoimmunity. Oxidative stress alters cellular redox potential, extending to the ER and causing accumulation of misfolded proteins. The UPR, crucial for innate and adaptive immunity, is activated in this situation, suggesting its role in regulating and maintaining the immune response in vitiligo.42

Another recent study analyzed the role of mitochondrial energy metabolism in melanocytes from vitiligo patients, finding reduced ATP production, increased proton leakage, altered expression of glycolytic enzymes, and hyperactivity of the PGC1α axis. Furthermore, it was demonstrated that pharmacological stabilization of cardiolipin, a key lipid in the mitochondrial membrane, can reverse the energy dysfunction observed in vitiligo melanocytes, which suggests that manipulating cardiolipin could be a promising new therapeutic strategy for treating vitiligo.43

Vitiligo skin shows morphological changes in the epithelium and in the upper dermis. Histologically, reduced pigmentation in the basal layer is observed, along with an absence of melanocytes in the lesions, which can be confirmed by specific immunohistochemical techniques, such as staining for HMB-45 or S100 proteins. Occasionally, a pattern of associated lichenoid dermatitis is observed, with CD3+, CD8+ T lymphocytes, more evident in perilesional skin. On the other hand, Langerhans cells are increased in the epidermis, the basement membrane is thickened, and cytoplasmic vacuolization phenomena are observed.44,45 It should be noted that Dickkopf 1 (DKK1) protein, involved in reducing melanogenesis, is overexpressed in fibroblasts in lesional skin.46 There is also increased fibronectin expression and reduced elastin in lesional dermis, with no changes observed in collagen fibers.47,48

Genetic changes involved in the etiopathogenesis of vitiligo

Vitiligo is a complex disease with a significant genetic component, estimated between 75% and 83%. This theory involves multiple genes, most of which correspond to the immune system, with a smaller number involving melanocytes. There is a notable overlap between genes involved in vitiligo and those that have been associated with autoimmune disorders. First-degree relatives of vitiligo patients have a 6–8% risk of developing the disease, and the concordance rate between monozygotic twins is approximately 23%.28,34

Five genome-wide association studies (GWAS) have been conducted on vitiligo in European and Asian populations, identifying at least 54 vitiligo susceptibility loci.49,50

Human leukocyte antigen region

Several genes within the human leukocyte antigen (HLA) region, specifically in class I and II regions, are associated with vitiligo. These studies include GWAS association analyses and linkage studies in affected families. The pathogenesis of vitiligo has been associated with the XBP1 gene, a transcription factor that plays a crucial role in endoplasmic reticulum stress response and regulates HLA class II gene expression.51 In the Chinese population, HLA-DQB1 and HLA-B have been identified as associated risk factors; in European populations, 3 specific loci; and 1 in Asians (linked respectively to FOXD3 and PDGFRA).52–54 On the other hand, some studies correlate HLA-A09 and HLA-Aw19 with a lower risk for the disease55 (Table 1).

Table 1.

HLA genes associated with increased risk of developing vitiligo. These studies include genome-wide association studies (GWAS) and linkage analyses in affected families.51–60

HLA genes 
HLA-A02 
HLA-Aw31 
HLA-A32 
HLA-A33 
HLA-DQB106 
HLA-DQB10303 
HLA-DR4 
HLA-DRB1*07 
HLA-DR7 

HLA: human leukocyte antigen.

Several genes linked to vitiligo susceptibility related to immunomodulation of innate and adaptive immunity have also been identified (Tables 2 and 3).

Table 2.

Main genes involved in the etiopathogenesis of vitiligo.

Gene  Chromosome and region  Function 
XBP151  Chr 22q12  Transcription factor involved in HLA II expression, cellular stress response 
FOXD353  Chr 1p31.3-p32.2  Transcription factor involved in cell differentiation, immune regulation 
PDGFRA52  Chr 4q13-q21  Transcription factor involved in cell proliferation, and angiogenesis 
NLRP164  Chr 17p13  Pathogen sensing, inflammasome formation, caspase-1 activation, cytokine secretion, and pyroptosis induction 
PTPN2261  Chr 1p13.2  It codes for a tyrosine phosphatase involved in T-cell signaling, which is linked to various autoimmune diseases. 
IKZF462  Chr 12q13.2  FOXP3-mediated gene silencing in Tregs 
FOXP362  Chr X  Gene silencing in Tregs 
DDR165  Chr 6p21.3  It codes for a tyrosine kinase receptor involved in the adhesion of melanocytes to the basement membrane. 
VEGF66  Chr 6p21.1  Regulator of angiogenesis involved in chronic diseases and neoplasms 

HLA: human leukocyte antigen.

Table 3.

Immunoregulatory genes involved in T cell development, activation, signaling, and innate immune response associated with vitiligo.4,28,49,50

T cell development  T cell receptor signaling  T cell activation  Innate immune response  Chemokine and cytokine receptors 
CD44  SLA  BTNL2  IFIH1  CXCR5 
CD80  PTPN22  FOXP3  TICAM1  CCR6 
  UBASH3A  IKZF4  SH2B3   
  CLNK  IL2RA     
    CTLA4     

Variants of the PTPN22 gene have been associated with several autoimmune diseases, such as rheumatoid arthritis or systemic lupus erythematosus, making it a gene with a relevant role in the context of autoimmune diseases.61 The IKZF4 and FOXP3 genes, which play a role in gene silencing in Tregs, have also been linked.62

Several genes related to melanocyte function and survival have been invovled in vitiligo (Table 4). The TYR gene, involved in melanin biosynthesis in melanocytes, has been significantly associated with vitiligo in the European population.63 The roles of many susceptibility loci remain unknown, highlighting the lack of understanding in the etiopathogenesis of vitiligo, which continues to be a challenge today. This genetic knowledge highlights the multifactorial nature of vitiligo with a polygenic inheritance pattern, involving a complex interaction between immune system dysfunction and melanocyte dysfunction.

Table 4.

Genes related to melanocyte function and survival.4,28,49,50

Autoantigen formation  Melanocyte development and survival  Melanocyte adhesion to basement membrane  Cell death during oxidative stress  Immune-induced apoptosis  Apoptosis regulation  Inflammasome activation 
PMEL  ZMIZ1  DDR1  RNASET2  GZMB  FGFR1OP  NLRP1 
MC1R  ASIP  CDH1    FASLG  CASP7   
OCA2          BCL2L12   
TYR          RERE   
          NEK6   
          SERPINB9   
          BAD   
Role of oxidative stress

Oxidative stress plays a fundamental role in the pathogenesis of vitiligo, contributing to the damage and destruction of melanocytes. Oxidative stress refers to an imbalance between the production of reactive oxygen species (ROS) and the body's antioxidant capacity to neutralize them. In vitiligo patients, an excessive accumulation of ROS has been observed in melanocytes, which can damage crucial cellular components, including lipids, proteins, and DNA, leading to their dysfunction and death. Cellular damage caused by oxidative stress can release autoantigens and activate an autoimmune response, contributing to melanocyte destruction.4,28,67,68 Former studies have shown elevated levels of oxidative stress biomarkers, such as malondialdehyde (MDA), and decreased antioxidants, such as superoxide dismutase (SOD) and catalase, in both affected skin and blood of patients. Additionally, certain variants in genes related to antioxidant response, such as the catalase (CAT) gene and the glutathione peroxidase (GPX) gene, have been associated with increased susceptibility to vitiligo.67,69,70

Furthermore, vitiligo has been associated with defective recycling of tetrahydrobiopterin (6BH4), essential for melanin formation, leading to an excess of 7BH4 and inhibition of phenylalanine hydroxylase (PAH). This produces hydrogen peroxide (H2O2), which decreases the activity of the dihydropteridine reductase (DHPR) enzyme.71,72 Furthermore, H2O2 negatively affects acetylcholinesterase (AchE) and xanthine oxidase (XO).73,74

Repair mechanisms and immune system activation in vitiligo patients could reduce the risk of skin cancer, which is attributed to immune activation capable of identifying and eliminating damaged cells, including those with potentially precancerous mutations. Additionally, oxidative stress and autoimmunity in vitiligo create a less favorable microenvironment for the development of skin cancer, including melanoma.75

Environmental agents as disease activators

Environmental factors influence the onset or worsening of vitiligo in susceptible individuals (Table 5). Physical trauma, known as the Koebner phenomenon, can induce the appearance of vitiligo patches in areas of repeated friction, wounds, scars, and pre-existing lesions of contact eczema or psoriasis.76 Exposure to chemicals, such as phenolic derivatives, generates a melanotoxic effect in vitiligo by inducing oxidative stress and causing direct damage to melanocytes. Additionally, tattoos, vaccines, and certain cosmetics can also precipitate vitiligo by altering melanocytes. Heat, derived from procedures such as laser hair removal or pulsed light, can damage the skin and trigger depigmentation.77,78 On the other hand, smoking, with its detrimental effects on skin health and the immune system, can exacerbate the disease.79 Furthermore, numerous drugs, including some immunomodulators and biological agents, can induce or worsen vitiligo.80–83 Moreover, viral infections, such as HCV, HIV, varicella-zoster virus (VZV), and COVID-19, and exposure to allergens suchas dust mites, have been associated with vitiligo outbreaks.84–86 Finally, psychological stress is a known factor that can aggravate the condition, possibly due to its impact on the immune system.87 However, there are no studies that prove a direct association between vitiligo and pollution, specific diets, or alcohol consumption.88–90

Table 5.

Environmental triggers of vitiligo.76–86,88–91

Environmental agents
Chemicals  Phenolic derivatives (monobenzone, rhododendrol), tattoos, vaccines, cosmetic dyes (paraphenylenediamine) 
Heat  Laser hair removal, intense pulsed light 
Trauma  Koebner phenomenon in areas of repeated trauma, friction, wounds, use of face masks, residual lesions from contact eczema, psoriasis, etc. 
Drugs  Paracetamol (acetaminophen), drugs used in hematologic patients (mogamulizumab, TKIs), dermatologic patients (anti-TNF, dupilumab, secukinumab, imiquimod, diphencyprone), neurologic patients (alemtuzumab, carbamazepine, tolcapone, levodopa), antibiotics (chloroquine, clofazimine), beta-blockers, anti-PD1 drugs (pembrolizumab, nivolumab), anti-CTLA-4 drugs (ipilimumab) 
Infections  HIV, HCV, herpesviruses (varicella-zoster virus, herpes simplex virus), COVID-19 
Smoking  – 
Psychological stress  – 

TKI: tyrosine kinase inhibitors; HCV: hepatitis C virus; HIV: human immunodeficiency virus.

Neural hypothesis and genetic mosaicism in segmental vitiligo

The neural hypothesis suggests that segmental vitiligo is related to nervous system dysfunction. It is postulated that neurotransmitters or neurochemicals released by nerve endings, specifically substance P, can be toxic to melanocytes, causing their destruction in specific areas.92,93 The segmental distribution of vitiligo patches corresponds to the innervation of certain nerves, supporting this hypothesis.92,93 A study analyzing the presence of VZV in segmental vitiligo skin suggests a possible relationship between the 2. The results indicate the presence of viral particles and related changes in the affected skin, suggesting that VZV could contribute to the development or progression of vitiligo.86

Sometimes, however, segmental vitiligo doesn’t follow a dermatomal distribution, suggesting that neuronal mechanisms aren’t the sole cause. For this reason, genetic mosaicism is currently the most accepted theory to explain this condition, though it has not yet been genetically confirmed.94

New pathogenic pathways and potential new therapeutic targets

The pathogenesis of vitiligo is multifactorial, involving a complex interaction between genetic predisposition and various environmental factors, oxidative stress, and autoimmune responses. The main challenge in creating pathophysiological models is to integrate these diverse theories and elements.

The discovery of new pathogenic pathways raises potential new therapeutic targets to act upon. Emerging therapeutic strategies include the reduction of reactive oxygen species (ROS), the inhibition of IFN-γ and IL-15 signaling through the JAK-STAT pathway, and the use of anti-IL-15 and anti-CD122. Additionally, the reduction of resident memory T cells and intervention in the NRLP1 pathway by acting on the inflammasome are also promising approaches to treat this disease.

CRediT authorship contribution statement

All authors contributed to the preparation and critical modification of the manuscript and approved the submitted version.

Conflicts of interest

None declared.

References
[1]
N. Mastacouris, A. Strunk, A. Garg.
Incidence and prevalence of diagnosed vitiligo according to race and ethnicity, age, and sex in the US.
JAMA Dermatol, 159 (2023), pp. 986-990
[2]
R. Yu, R. Broady, Y. Huang, Y. Wang, J. Yu, M. Gao, et al.
Transcriptome analysis reveals markers of aberrantly activated innate immunity in vitiligo lesional and non-lesional skin.
PLoS One, 7 (2012),
[3]
H.Z. Marchioro, C.C. Silva de Castro, V.M. Fava, P.H. Sakiyama, G. Dellatorre, H.A. Miot.
Update on the pathogenesis of vitiligo.
An Bras Dermatol, 97 (2022), pp. 478-490
[4]
R. Speeckaert, N. van Geel.
Vitiligo: an update on pathophysiology and treatment options.
Am J Clin Dermatol, 18 (2017), pp. 733-744
[5]
R. Doss, A.A. El-Rifaie, A. Abdel-Wahab, Y. Gohary, L. Rashed.
Heat shock protein-70 expression in vitiligo and its relation to the disease activity.
Indian J Dermatol, 61 (2016), pp. 408
[6]
C. Jacquemin, J. Rambert, S. Guillet, D. Thiolat, N. Boukhedouni, M.S. Doutre, et al.
Heat shock protein 70 potentiates interferon alpha production by plasmacytoid dendritic cells: relevance for cutaneous lupus and vitiligo pathogenesis.
Br J Dermatol, 177 (2017), pp. 1367-1375
[7]
A. Bertolotti, K. Boniface, B. Vergier, D. Mossalayi, A. Taieb, K. Ezzedine, et al.
Type I interferon signature in the initiation of the immune response in vitiligo.
Pigment Cell Melanoma Res, 27 (2014), pp. 398-407
[8]
R. Speeckaert, S. Voet, E. Hoste, N. van Geel.
S100B is a potential disease activity marker in nonsegmental vitiligo.
J Invest Dermatol, 137 (2017), pp. 1445-1453
[9]
J.Y. Kim, E.J. Lee, J. Seo, S.H. Oh.
Impact of high-mobility group box 1 on melanocytic survival and its involvement in the pathogenesis of vitiligo.
Br J Dermatol, 176 (2017), pp. 1558-1568
[10]
Y. Zhang, L. Liu, L. Jin, X. Yi, E. Dang, Y. Yang, et al.
Oxidative stress-induced calreticulin expression and translocation: new insights into the destruction of melanocytes.
J Invest Dermatol, 134 (2014), pp. 183-191
[11]
R.Y. Wagner, F. Luciani, M. Cario-André, I. Rubera, V. Petit, L. Benzekri, et al.
Altered E-cadherin levels and distribution in melanocytes precede clinical manifestations of vitiligo.
J Invest Dermatol, 135 (2015), pp. 1810-1819
[12]
M. Bordignon, C. Castellani, M. Fedrigo, G. Thiene, A. Peserico, M. Alaibac, et al.
Role of alpha5beta1 integrin and MIA (melanoma inhibitory activity) in the pathogenesis of vitiligo.
J Dermatol Sci, 71 (2013), pp. 142-145
[13]
J. Marie, D. Kovacs, C. Pain, T. Jouary, C. Cota, B. Vergier, et al.
Inflammasome activation and vitiligo/nonsegmental vitiligo progression.
Br J Dermatol, 170 (2014), pp. 816-823
[14]
C.B. Levandowski, C.M. Mailloux, T.M. Ferrara, K. Gowan, S. Ben, Y. Jin, et al.
NLRP1 haplotypes associated with vitiligo and autoimmunity increase interleukin-1β processing via the NLRP1 inflammasome.
Proc Natl Acad Sci U S A, 110 (2013), pp. 2952-2956
[15]
Y. Jin, G.H.L. Andersen, S.A. Santorico, R.A. Spritz.
Multiple functional variants of IFIH1, a gene involved in triggering innate immune responses, protect against vitiligo.
J Invest Dermatol, 137 (2017), pp. 522-524
[16]
L. Migayron, K. Boniface, J. Seneschal.
Vitiligo from physiopathology to emerging treatments: a review.
Dermatol Ther (Heidelb), 10 (2020), pp. 1185-1198
[17]
K. Boniface, J. Seneschal, M. Picardo, A. Taïeb.
Vitiligo: focus on clinical aspects, immunopathogenesis, and therapy.
Clin Rev Allergy Immunol, 54 (2018), pp. 52-67
[18]
M. Rashighi, P. Agarwal, J.M. Richmond, T.H. Harris, K. Dresser, M.W. Su, et al.
CXCL10 is critical for the progression and maintenance of depigmentation in a mouse model of vitiligo.
Sci Transl Med, 6 (2014),
[19]
J.G. Van den Boorn, D. Konijnenberg, T.A. Dellemijn, J.P. van der Veen, J.D. Bos, C.J. Melief, et al.
Autoimmune destruction of skin melanocytes by perilesional T cells from vitiligo patients.
J Invest Dermatol, 129 (2009), pp. 2220-2232
[20]
B. Palermo, R. Campanelli, S. Garbelli, S. Mantovani, E. Lantelme, V. Brazzelli, et al.
Specific cytotoxic T lymphocyte responses against Melan-A/MART1, tyrosinase and gp100 in vitiligo by the use of major histocompatibility complex/peptide tetramers: the role of cellular immunity in the etiopathogenesis of vitiligo.
J Invest Dermatol, 117 (2001), pp. 326-332
[21]
Y. Maeda, H. Nishikawa, D. Sugiyama, D. Ha, M. Hamaguchi, T. Saito, et al.
Detection of self-reactive CD8+ T cells with an anergic phenotype in healthy individuals.
Science, 346 (2014), pp. 1536-1540
[22]
J. Piquero-Casals, L. Martínez-Martínez, A. Alomar, E. Rozas-Muñoz, C. Juarez, L. Puig.
LRP1/CD91 is highly expressed in monocytes from patients with vitiligo – even after repigmentation.
Exp Dermatol, 30 (2021), pp. 390-395
[23]
A.F. Rezk, D.M. Kemp, M. El-Domyati, E.A. El-Harith, J.B. Lee, J. Uitto, et al.
Misbalanced CXCL12 and CCL5 chemotactic signals in vitiligo onset and progression.
J Invest Dermatol, 137 (2017), pp. 1126-1134
[24]
S. Li, G. Zhu, Y. Yang, Z. Jian, S. Guo, W. Dai, et al.
Oxidative stress drives CD8+ T-cell skin trafficking in patients with vitiligo through CXCL16 upregulation by activating the unfolded protein response in keratinocytes.
J Allergy Clin Immunol, 140 (2017),
[25]
M. Vaccaro, F. Cicero, C. Mannucci, G. Calapai, G. Spatari, O. Barbuzza, et al.
IL-33 circulating serum levels are increased in patients with non-segmental generalized vitiligo.
Arch Dermatol Res, 308 (2016), pp. 527-530
[26]
M. Vaccaro, S.P. Cannavò, S. Imbesi, M. Cristani, O. Barbuzza, V. Tigano, et al.
Increased serum levels of interleukin-23 circulating in patients with non-segmental generalized vitiligo.
Int J Dermatol, 54 (2015), pp. 672-674
[27]
R.K. Singh, K.M. Lee, I. Vujkovic-Cvijin, D. Ucmak, B. Farahnik, M. Abrouk, et al.
The role of IL-17 in vitiligo: a review.
Autoimmun Rev, 15 (2016), pp. 397-404
[28]
F. Diotallevi, H. Gioacchini, E. De Simoni, A. Marani, M. Candelora, M. Paolinelli, et al.
Vitiligo, from pathogenesis to therapeutic advances: state of the art.
Int J Mol Sci, 24 (2023),
[29]
J.E. Lommerts, M.W. Bekkenk, R.M. Luiten.
Vitiligo induced by immune checkpoint inhibitors in melanoma patients: an expert opinion.
Expert Opin Drug Saf, 20 (2021), pp. 883-888
[30]
J.H. Lee, H.J. Ju, J.M. Seo, A. Almurayshid, G.M. Kim, K. Ezzedine, et al.
Comorbidities in patients with vitiligo: a systematic review and meta-analysis.
J Invest Dermatol, 143 (2023),
[31]
A.M. Dahir, S.F. Thomsen.
Comorbidities in vitiligo: comprehensive review.
Int J Dermatol, 57 (2018), pp. 1157-1164
[32]
J.W. Lee, T.H. Kim, T.J. Park, H.Y. Kang.
p16ink4a positivity of melanocytes in non-segmental vitiligo.
Diagnostics (Basel), 10 (2020),
[33]
N.F. Post, G. Ginski, R. Peters, N.O.P. Van Uden, M.W. Bekkenk, A. Wolkerstorfer, et al.
Trained immunity in the pathogenesis of vitiligo.
Pigment Cell Melanoma Res, 36 (2023), pp. 348-354
[34]
R.A. Clark.
Resident memory T cells in human health and disease.
Sci Transl Med, 7 (2015),
[35]
L. Migayron, R. Merhi, J. Seneschal, K. Boniface.
Resident memory T cells in nonlesional skin and healed lesions of patients with chronic inflammatory diseases: appearances can be deceptive.
J Allergy Clin Immunol, 153 (2024), pp. 606-614
[36]
K. Boniface, C. Jacquemin, A.S. Darrigade, B. Dessarthe, C. Martins, N. Boukhedouni, et al.
Vitiligo skin is imprinted with resident memory CD8 T cells expressing CXCR3.
J Invest Dermatol, 138 (2018), pp. 355-364
[37]
M. Willemsen, R. Linkutė, R.M. Luiten, T.R. Matos.
Skin-resident memory T cells as a potential new therapeutic target in vitiligo and melanoma.
Pigment Cell Melanoma Res, 32 (2019), pp. 612-622
[38]
J.M. Richmond, J.P. Strassner, L. Zapata Jr., M. Garg, R.L. Riding, M.A. Refat, et al.
Antibody blockade of IL-15 signaling has the potential to durably reverse vitiligo.
Sci Transl Med, 10 (2018),
[39]
Y. Pan, T.S. Kupper.
Metabolic reprogramming and longevity of tissue-resident memory T cells.
Front Immunol, 9 (2018),
[40]
R. Kitamura, K. Tsukamoto, K. Harada, A. Shimizu, S. Shimada, T. Kobayashi, et al.
Mechanisms underlying the dysfunction of melanocytes in vitiligo epidermis: role of SCF/KIT protein interactions and the downstream effector, MITF-M.
J Pathol, 202 (2004), pp. 463-475
[41]
A. Sahoo, B. Lee, K. Boniface, J. Seneschal, S.K. Sahoo, T. Seki, et al.
MicroRNA-211 regulates oxidative phosphorylation and energy metabolism in human vitiligo.
J Invest Dermatol, 137 (2017), pp. 1965-1974
[42]
S.D. Jadeja, J.M. Mayatra, J. Vaishnav, N. Shukla, R. Begum.
A concise review on the role of endoplasmic reticulum stress in the development of autoimmunity in vitiligo pathogenesis.
Front Immunol, 11 (2021),
[43]
M.L. Dell’Anna, M. Ottaviani, D. Kovacs, S. Mirabilii, D.A. Brown, C. Cota, et al.
Energetic mitochondrial failing in vitiligo and possible rescue by cardiolipin.
Sci Rep, 7 (2017),
[44]
Y.C. Kim, Y.J. Kim, H.Y. Kang, S. Sohn, E.S. Lee.
Histopathologic features in vitiligo.
Am J Dermatopathol, 30 (2008), pp. 112-116
[45]
L.F. Montes, J. Abulafia, W.H. Wilborn, B.M. Hyde, C.M. Montes.
Value of histopathology in vitiligo.
Int J Dermatol, 42 (2003), pp. 57-61
[46]
S. Rani, R. Chauhan, D. Parsad, R. Kumar.
Effect of Dickkopf1 on the senescence of melanocytes: in vitro study.
Arch Dermatol Res, 310 (2018), pp. 343-350
[47]
T. Hirobe, H. Enami, A. Nakayama.
Elastin fiber but not collagen fiber is decreased dramatically in the dermis of vitiligo patients.
Int J Dermatol, 59 (2020), pp. e369-e372
[48]
D. Kovacs, E. Bastonini, M. Ottaviani, C. Cota, E. Migliano, M.L. Dell’Anna, et al.
Vitiligo skin: exploring the dermal compartment.
J Invest Dermatol, 138 (2018), pp. 394-404
[49]
S.L. Said-Fernandez, C.N. Sanchez-Domínguez, M.A. Salinas-Santander, H.G. Martinez-Rodriguez, D.E. Kubelis-Lopez, N.A. Zapata-Salazar, et al.
Novel immunological and genetic factors associated with vitiligo: a review.
Exp Ther Med, 21 (2021),
[50]
R.A. Spritz, G.H.L. Andersen.
Genetics of vitiligo.
Dermatol Clin, 35 (2017), pp. 245-255
[51]
Y. Liang, S. Yang, Y. Zhou, J. Gui, Y. Ren, J. Chen, et al.
Evidence for two susceptibility loci on chromosomes 22q12 and 6p21-p22 in Chinese generalized vitiligo families.
J Invest Dermatol, 127 (2007), pp. 2552-2557
[52]
J.J. Chen, W. Huang, J.P. Gui, S. Yang, F.S. Zhou, Q.G. Xiong, et al.
A novel linkage to generalized vitiligo on 4q13-q21 identified in a genomewide linkage analysis of Chinese families.
Am J Hum Genet, 76 (2005), pp. 1057
[53]
P.R. Fain, K. Gowan, G.S. LaBerge, A. Alkhateeb, G.L. Stetler, J. Talbert, et al.
A genomewide screen for generalized vitiligo: confirmation of AIS1 on chromosome 1p31 and evidence for additional susceptibility loci.
Am J Hum Genet, 72 (2003), pp. 1560
[54]
C. Yang, J. Wu, X. Zhang, L. Wen, J. Sun, Y. Cheng, et al.
Fine-mapping analysis of the MHC region for vitiligo based on a new Han-MHC reference panel.
[55]
Z. Li, J. Ren, X. Niu, Q. Xu, X. Wang, Y. Liu, et al.
Meta-analysis of the association between vitiligo and human leukocyte antigen-A.
Biomed Res Int, 2016 (2016),
[56]
M. Arcos-Burgos, E. Parodi, M. Salgar, E. Bedoya, J. Builes, D. Jaramillo, et al.
Vitiligo: complex segregation and linkage disequilibrium analyses with respect to microsatellite loci spanning the HLA.
Hum Genet, 110 (2002), pp. 334-342
[57]
S.A. Birlea, Y. Jin, D.C. Bennett, D.M. Herbstman, M.R. Wallace, W.T. McCormack, et al.
Comprehensive association analysis of candidate genes for generalized vitiligo supports XBP1, FOXP3, and TSLP.
J Invest Dermatol, 131 (2011), pp. 371-381
[58]
A. Singh, P. Sharma, H.K. Kar, V.K. Sharma, M.K. Tembhre, S. Gupta, et al.
HLA alleles and amino-acid signatures of the peptide-binding pockets of HLA molecules in vitiligo.
J Invest Dermatol, 132 (2012), pp. 124-134
[59]
M. Hayashi, Y. Jin, D. Yorgov, S.A. Santorico, J. Hagman, T.M. Ferrara, et al.
Autoimmune vitiligo is associated with gain-of-function by a transcriptional regulator that elevates expression of HLA-A*02:01 in vivo.
Proc Natl Acad Sci U S A, 113 (2016), pp. 1357-1362
[60]
L.D. Ramire, E.V.C. Marcos, D.A.S. Godoy, F.C. de Souza-Santana.
Association of class I and II HLA alleles and haplotypes with susceptibility to vitiligo: a study of patients with vitiligo from southeast Brazil.
Int J Dermatol, 55 (2016), pp. e347-e355
[61]
S.A. Chung, L.A. Criswell.
PTPN22: its role in SLE and autoimmunity.
Autoimmunity, 40 (2007), pp. 582
[62]
F. Pan, H. Yu, E.V. Dang, J. Barbi, X. Pan, J.F. Grosso, et al.
Eos mediates Foxp3-dependent gene silencing in CD4+ regulatory T cells.
Science, 325 (2009), pp. 1142-1146
[63]
Y. Jin, S.A. Birlea, P.R. Fain, K. Gowan, S.L. Riccardi, P.J. Holland, et al.
Variant of TYR and autoimmunity susceptibility loci in generalized vitiligo.
N Engl J Med, 362 (2010), pp. 1686-1697
[64]
Y. Jin, C.M. Mailloux, K. Gowan, S.L. Riccardi, G. LaBerge, D.C. Bennett, et al.
NALP1 in vitiligo-associated multiple autoimmune disease.
N Engl J Med, 356 (2007), pp. 1216-1225
[65]
A. Reichert-Faria, J.E. Jung, V. Moreschi Neto, C.C. Silva De Castro, M.T. Mira, L. Noronha.
Reduced immunohistochemical expression of Discoidin Domain Receptor 1 (DDR1) in vitiligo skin.
J Eur Acad Dermatol Venereol, 27 (2013), pp. 1057-1059
[66]
M. Almasi-Nasrabadi, M.M. Amoli, R.M. Robati, F. Rajabi, S. Parichehreh Dizaji.
Is the +405 G/C single nucleotide polymorphism of the vascular endothelial growth factor (VEGF) gene associated with late-onset vitiligo?.
Int J Immunogenet, 46 (2019), pp. 241-246
[67]
R. Speeckaert, J. Dugardin, J. Lambert, H. Lapeere, K. Vermaelen, M.M. Speeckaert, et al.
Critical appraisal of the oxidative stress pathway in vitiligo: a systematic review and meta-analysis.
J Eur Acad Dermatol Venereol, 32 (2018), pp. 1089-1098
[68]
Z.A. Abdel-Malek, C. Jordan, T. Ho, P.R. Upadhyay, A. Fleischer, I. Hamzavi.
The enigma and challenges of vitiligo pathophysiology and treatment.
Pigment Cell Melanoma Res, 33 (2020), pp. 778-787
[69]
G. Akoglu, S. Emre, A. Metin, A. Akbas, A. Yorulmaz, S. Isikoglu, et al.
Evaluation of total oxidant and antioxidant status in localized and generalized vitiligo.
Clin Exp Dermatol, 38 (2013), pp. 701-706
[70]
H. Zedan, A.A. Abdel-Motaleb, N.M.A. Kassem, H.A.A. Hafeez, M.R.A. Hussein.
Low glutathione peroxidase activity levels in patients with vitiligo.
J Cutan Med Surg, 19 (2015), pp. 144-148
[71]
D. Kowlessur, B.A. Citron, S. Kaufman.
Recombinant human phenylalanine hydroxylase: novel regulatory and structural properties.
Arch Biochem Biophys, 333 (1996), pp. 85-95
[72]
K.U. Schallreuter, J.M. Wood, M.R. Pittelkow, M. Gütlich, K.R. Lemke, W. Rödl, et al.
Regulation of melanin biosynthesis in the human epidermis by tetrahydrobiopterin.
Science, 263 (1994), pp. 1444-1446
[73]
M. Shalbaf, N.C. Gibbons, J.M. Wood, D.J. Maitland, H. Rokos, S.M. Elwary, et al.
Presence of epidermal allantoin further supports oxidative stress in vitiligo.
Exp Dermatol, 17 (2008), pp. 761-770
[74]
K.U. Schallreuter, S.M.A. Elwary, N.C.J. Gibbons, H. Rokos, J.M. Wood.
Activation/deactivation of acetylcholinesterase by H2O2: more evidence for oxidative stress in vitiligo.
Biochem Biophys Res Commun, 315 (2004), pp. 502-508
[75]
I. Gupta, S. Shankrit, K. Narta, M. Ghazi, R. Grover, R. Pandey, et al.
Whole-exome sequencing of vitiligo lesions indicates lower burden of somatic variations: implications in risk for nonmelanoma skin cancers.
J Invest Dermatol, 143 (2023), pp. 1111-1114
[76]
Y. Gauthier, M. Cario-Andre, S. Lepreux, C. Pain, A. Taïeb.
Melanocyte detachment after skin friction in non lesional skin of patients with generalized vitiligo.
Br J Dermatol, 148 (2003), pp. 95-101
[77]
J.E. Harris.
Chemical-induced vitiligo.
Dermatol Clin, 35 (2017), pp. 151
[78]
N. Chivukula, K. Ramesh, A. Subbaroyan, A.K. Sahoo, G.B. Dhanakoti, J. Ravichandran, et al.
ViCEKb: vitiligo-linked chemical exposome knowledgebase.
Sci Total Environ, (2024), pp. 913
[79]
Y. Enomoto, Y. Kanayama, K. Ikumi, M. Sakurai, A. Yamamoto, A. Morita.
Cigarette smoking is an independent risk factor for developing vitiligo on the hands.
Photodermatol Photoimmunol Photomed, 40 (2024),
[80]
L.M. Assaedi, H.M. Alshamrani, R.A. Abbas, F.A. Alghamdi.
Carbamazepine-induced reversible vitiligo.
JAAD Case Rep, 26 (2022),
[81]
E. Selvaag.
Chloroquine-induced vitiligo. A case report and review of the literature.
Acta Derm Venereol, 76 (1996), pp. 166-167
[82]
J.M. Bae, M. Kim, H.H. Lee, K.J. Kim, H. Shin, H.J. Ju, et al.
Increased risk of vitiligo following anti-tumor necrosis factor therapy: a 10-year population-based cohort study.
J Invest Dermatol, 138 (2018), pp. 768-774
[83]
M. Sachar, B.M. Lin, V. Wong, W. Li, V. Huang, J. Harris, et al.
Association between acetaminophen use and vitiligo in US women and men.
Australas J Dermatol, 64 (2023), pp. e348-e351
[84]
M.M. Fawzy, N.M. Hammad, A.L. Sharaf, F. Khattab, C. Hepatitis.
virus infection could be a risk factor for adult-onset vitiligo in Egyptian patients: a cross-sectional study.
J Cosmet Dermatol, 21 (2022), pp. 4983-4989
[85]
L. Macca, L. Peterle, M. Ceccarelli, Y. Ingrasciotta, G. Nunnari, C. Guarneri.
Vitiligo-like lesions and COVID-19: case report and review of vaccination- and infection-associated vitiligo.
Vaccines (Basel), 10 (2022),
[86]
Y. Gauthier, S. Lepreux, M. Cario-Andre, J. Rambert, A. Dakdaki, M.E. Lafon, et al.
Varicella-zoster virus in actively spreading segmental vitiligo skin: pathological, immunochemical, and ultrastructural findings (a first and preliminary study).
Pigment Cell Melanoma Res, 36 (2023), pp. 78-85
[87]
N. Pondeljak, L. Lugović-Mihić.
Stress-induced interaction of skin immune cells, hormones, and neurotransmitters.
[88]
M.A. Lefebvre, D.M. Pham, B. Boussouira, D. Bernard, C. Camus, Q.L. Nguyen.
Evaluation of the impact of urban pollution on the quality of skin: a multicentre study in Mexico.
Int J Cosmet Sci, 37 (2015), pp. 329-338
[89]
M. Mansilla-Polo, J. Piquero-Casals, D. Morgado-Carrasco.
[Translated article] Popular diets and skin effects: a narrative review.
Actas Dermosifiliogr, 115 (2024), pp. T374-T386
[90]
R.K. Singh, H.W. Chang, D. Yan, K.M. Lee, D. Ucmak, K. Wong, et al.
Influence of diet on the gut microbiome and implications for human health.
J Transl Med, 15 (2017),
[91]
N. Hermann, L.V. Maul, M. Ameri, S. Traidl, R. Ziadlou, K. Papageorgiou, et al.
Clinical presentation and prognostic features in patients with immunotherapy-induced vitiligo-like depigmentation: a monocentric prospective observational study.
Cancers (Basel), 14 (2022),
[92]
M. Gf, G. Ah, A.-D. Ms.
Highlights in pathogenesis of vitiligo.
World J Clin Cases, 3 (2015), pp. 221
[93]
C. Tu, D. Zhao, X. Lin.
Levels of neuropeptide-Y in the plasma and skin tissue fluids of patients with vitiligo.
J Dermatol Sci, 27 (2001), pp. 178-182
[94]
N. van Geel, R. Speeckaert, E. Melsens, S.P. Toelle, M. Speeckaert, et al.
The distribution pattern of segmental vitiligo: clues for somatic mosaicism.
Br J Dermatol, 168 (2013), pp. 56-64
Descargar PDF
Idiomas
Actas Dermo-Sifiliográficas
Opciones de artículo
Herramientas
es en

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?