+ All Categories
Home > Documents > Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven...

Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven...

Date post: 24-Jul-2020
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
22
Clinical Science (2011) 120, 99–120 (Printed in Great Britain) doi:10.1042/CS20090603 99 R E V I E W Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa, 737 Parkdale Avenue, Ottawa, Ontario, Canada K1Y 1J8 A B S T R A C T The acquired depigmenting disorder of vitiligo affects an estimated 1% of the world population and constitutes one of the commonest dermatoses. Although essentially asymptomatic, the psychosocial impact of vitiligo can be severe. The cause of vitiligo remains enigmatic, hampering efforts at successful therapy. The underlying pathogenesis of the pigment loss has, however, been clarified to some extent in recent years, offering the prospect of effective treatment, accurate prognosis and rational preventative strategies. Vitiligo occurs when functioning melanocytes disappear from the epidermis. A single dominant pathway is unlikely to account for all cases of melanocyte loss in vitiligo; rather, it is the result of complex interactions of biochemical, environmental and immunological events, in a permissive genetic milieu. ROS (reactive oxygen species) and H 2 O 2 in excess can damage biological processes, and this situation has been documented in active vitiligo skin. Tyrosinase activity is impaired by excess H 2 O 2 through oxidation of methionine residues in this key melanogenic enzyme. Mechanisms for repairing this oxidant damage are also damaged by H 2 O 2 , compounding the effect. Numerous proteins and peptides, in addition to tyrosinase, are similarly affected. It is possible that oxidant stress is the principal cause of vitiligo. However, there is also ample evidence of immunological phenomena in vitiligo, particularly in established chronic and progressive disease. Both innate and adaptive arms of the immune system are involved, with a dominant role for T-cells. Sensitized CD8 + T-cells are targeted to melanocyte differentiation antigens and destroy melanocytes either as the primary event in vitiligo or as a secondary promotive consequence. There is speculation on the interplay, if any, between ROS and the immune system in the pathogenesis of vitiligo. The present review focuses on the scientific evidence linking alterations in ROS and/or T-cells to vitiligo. INTRODUCTION Vitiligo is a common skin disorder characterized by the acquired loss of constitutional pigmentation manifesting as white macules and patches caused by a loss of functioning epidermal melanocytes. Extent of involvement is highly variable, ranging from focal to generalized, and the onset can be abrupt or gradual. Although essentially asymptomatic, the psychosocial impact of vitiligo can be devastating, and affected Key words: immune system, melanocyte, reactive oxygen species (ROS), skin, T-cell, vitiligo. Abbreviations: α-MSH, α-melanocyte-stimulating hormone; 4-TBP, 4-tertiary butyl phenol; 6BH 4 , (6R)-l-erythro-5,6,7, 8,-tetrahydrobiopterin; AchE, acetylcholinesterase; AIS, autoimmune susceptibility loci; BchE, butyrylcholinesterase; BH 4 , tetrahydrobiopterin; CLA, cutaneous lymphocyte-associated antigen; CTL, cytotoxic T-lymphocyte; CTLA-4, CTL antigen-4; CYP, cytochrome P450; DC, dendritic cell; ET-1, endothelin-1; FT, Fourier Transform; GSH-Px, glutathione peroxidase; GST, glutathione transferase; HLA, human leucocyte antigen; ICAM, intercellular adhesion molecule; IFN, interferon; IL, interleukin; iNOS, inducible NO synthase; MBEH, monobenzyl ether of hydroquinone; MDA, malondialdehyde; MITF-M, melanocyte- specific microphthalmia-associated transcription factor; MSR, methionine sulfoxide reductase; ONOO −• , peroxynitrite; POMC, pro-opiomelanocortin; ROS, reactive oxygen species; SCF, stem cell factor; SNP, single nucleotide polymorphism; SOD, superoxide dismutase; TGF-β, transforming growth factor-β; TNF-α, tumour necrosis factor-α; TRP-1, tyrosinase-related protein-1; XO, xanthine oxidase. Correspondence: Dr Steven J. Glassman (email [email protected]). C The Authors Journal compilation C 2011 Biochemical Society
Transcript
Page 1: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

Clinical Science (2011) 120, 99–120 (Printed in Great Britain) doi:10.1042/CS20090603 99

R E V I E W

Vitiligo, reactive oxygen species and T-cells

Steven J. GLASSMANDivision of Dermatology, Department of Medicine, University of Ottawa, 737 Parkdale Avenue, Ottawa, Ontario,Canada K1Y 1J8

A B S T R A C T

The acquired depigmenting disorder of vitiligo affects an estimated 1% of the world populationand constitutes one of the commonest dermatoses. Although essentially asymptomatic, thepsychosocial impact of vitiligo can be severe. The cause of vitiligo remains enigmatic, hamperingefforts at successful therapy. The underlying pathogenesis of the pigment loss has, however, beenclarified to some extent in recent years, offering the prospect of effective treatment, accurateprognosis and rational preventative strategies. Vitiligo occurs when functioning melanocytesdisappear from the epidermis. A single dominant pathway is unlikely to account for all casesof melanocyte loss in vitiligo; rather, it is the result of complex interactions of biochemical,environmental and immunological events, in a permissive genetic milieu. ROS (reactive oxygenspecies) and H2O2 in excess can damage biological processes, and this situation has beendocumented in active vitiligo skin. Tyrosinase activity is impaired by excess H2O2 through oxidationof methionine residues in this key melanogenic enzyme. Mechanisms for repairing this oxidantdamage are also damaged by H2O2, compounding the effect. Numerous proteins and peptides,in addition to tyrosinase, are similarly affected. It is possible that oxidant stress is the principalcause of vitiligo. However, there is also ample evidence of immunological phenomena in vitiligo,particularly in established chronic and progressive disease. Both innate and adaptive arms ofthe immune system are involved, with a dominant role for T-cells. Sensitized CD8+ T-cells aretargeted to melanocyte differentiation antigens and destroy melanocytes either as the primaryevent in vitiligo or as a secondary promotive consequence. There is speculation on the interplay,if any, between ROS and the immune system in the pathogenesis of vitiligo. The present reviewfocuses on the scientific evidence linking alterations in ROS and/or T-cells to vitiligo.

INTRODUCTION

Vitiligo is a common skin disorder characterizedby the acquired loss of constitutional pigmentationmanifesting as white macules and patches caused by a

loss of functioning epidermal melanocytes. Extent ofinvolvement is highly variable, ranging from focal togeneralized, and the onset can be abrupt or gradual.Although essentially asymptomatic, the psychosocialimpact of vitiligo can be devastating, and affected

Key words: immune system, melanocyte, reactive oxygen species (ROS), skin, T-cell, vitiligo.Abbreviations: α-MSH, α-melanocyte-stimulating hormone; 4-TBP, 4-tertiary butyl phenol; 6BH4, (6R)-l-erythro-5,6,7,8,-tetrahydrobiopterin; AchE, acetylcholinesterase; AIS, autoimmune susceptibility loci; BchE, butyrylcholinesterase; BH4,tetrahydrobiopterin; CLA, cutaneous lymphocyte-associated antigen; CTL, cytotoxic T-lymphocyte; CTLA-4, CTL antigen-4;CYP, cytochrome P450; DC, dendritic cell; ET-1, endothelin-1; FT, Fourier Transform; GSH-Px, glutathione peroxidase; GST,glutathione transferase; HLA, human leucocyte antigen; ICAM, intercellular adhesion molecule; IFN, interferon; IL, interleukin;iNOS, inducible NO synthase; MBEH, monobenzyl ether of hydroquinone; MDA, malondialdehyde; MITF-M, melanocyte-specific microphthalmia-associated transcription factor; MSR, methionine sulfoxide reductase; ONOO− •, peroxynitrite; POMC,pro-opiomelanocortin; ROS, reactive oxygen species; SCF, stem cell factor; SNP, single nucleotide polymorphism; SOD, superoxidedismutase; TGF-β, transforming growth factor-β; TNF-α, tumour necrosis factor-α; TRP-1, tyrosinase-related protein-1; XO,xanthine oxidase.Correspondence: Dr Steven J. Glassman (email [email protected]).

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 2: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

100 S.J. Glassman

persons are often desperate for effective therapy [1].As of 2010, this goal has not yet been reached, asthe underlying pathomechanisms in vitiligo are stillincompletely understood, despite intense scrutiny. Threerecurring themes emanate from clinical and scientificanalysis of melanocyte loss in vitiligo: genetic, immuneand biochemical. The dominant theorem describesan autoimmune process occurring in a geneticallysusceptible host and triggered by a variety of host andenvironmental factors [2]. The aim of the present reviewis to summarize the role of ROS (reactive oxygen species)and T-cells in the pathogenesis of vitiligo.

Vitiligo occurs worldwide, with an estimated preval-ence rate of about 1 %. A large population-based surveyfound a prevalence rate of 0.38 % in Denmark [3], anda clinic-based survey in Martinique found a prevalencerate of 0.34 % [4]. Other surveys have reported ratesfrom 0.15 % to 8.8 %, with the highest rates occurringin Indian locales [5]. Onset is before the age of 20 yearsin about half the cases, and three quarters have occurredby the age of 30 years. Sexes are equally affected, butthere might be a female preponderance owing to reportingbias [6]. Two major subtypes of vitiligo are described,segmental and non-segmental. Segmental vitiligo tends tohave an earlier onset, with the rapid evolution of unilateraldepigmentation in the distribution of a dermatome,suggesting neural involvement, or corresponding toBlaschko’s lines, suggesting a form of genetic mosaicism.Segmental vitiligo mostly affects the face and tends to bestable once fully developed. The area of depigmentationis sometimes not as well demarcated as in othertypes of vitiligo [7]. Non-segmental vitiligo is, by far, thecommoner type and is also known as generalized vitiligoor vitiligo vulgaris. Most work relating to pathogenesishas focused on this form of vitiligo exclusively. Onsetis usually gradual with depigmented macules evolvinginto well-demarcated patches with convex or scallopedborders, in a symmetrical distribution in sites ofpredilection. These include regions which are normallyslightly hyperpigmented, such as the face, groin, axillae,areolae and genitalia, or those prone to mechanicalinjury and repeated friction, such as the ankles, kneesand elbows. Macules can have indistinct borders,and other morphological variants include trichromevitiligo, where three shades in concentric zones arenoted: normal, intermediate depigmentation and fulldepigmentation, and inflammatory vitiligo, where thereis an erythematous, raised border to the lesion, withassociated itching. Ordinary vitiligo can also present withpruritus. Mixed patterns, including segmental and non-segmental types can occur. A hyperpigmented rim cansometimes be seen at the outer margin of a vitiligo lesion,especially after sun exposure. A loss of hair pigmentin affected areas is considered a late sign of vitiligo,with some prognostic significance. Therefore, it appearsthat epidermal melanocytes are lost before those in the

hair bulb [6]. Non-segmental vitiligo is a slow andprogressive disease, marked by occasional remissions andexacerbations, corresponding presumably to triggeringfactors. Full resolution of generalized vitiligo has neverbeen reported, emphasizing the relentless chronicityof the condition. Depigmented areas are surprisinglytolerant of sun exposure and relatively resistant tophotoaging and skin cancer. Up-regulation of wild-type p53 could be the reason [8]. The diagnosis isclinical, but occasionally, skin biopsy is required forconfirmation: melanocytes and melanin are absent.Examination of the affected areas with Wood’s light canhelp distinguish active vitiligo from several disorderscharacterized by varying degrees of hypopigmentationby showing fluorescence owing to the presence ofoxidized pteridines [9]. The degree of fluorescencedepends on the skin phototype, and fluorescence is notseen in postinflammatory hypopigmentation. The dif-ferential diagnosis of vitiligo includes piebaldism, nevusdepigmentosus, hypomelanosis of Ito, lichen sclerosus,pityriasis versicolor, idiopathic guttate hypomelanosis,progressive macular hypomelanosis, hypopigmentedmycosis fungoides, indeterminate leprosy, pityriasisalba and many other forms of postinflammatoryhypopigmentation, such as that following resolutionof psoriasis. Particularly challenging can be the totaldepigmentation which can follow repeated excoriationof the skin from atopic dermatitis and psychogenicexcoriations, and inflammation from connective tissuediseases like lupus erythematosus and systemic sclerosis.Depigmentation due to exposure to certain toxins canmimic vitiligo exactly and is termed chemical leukoderma.There can be shared pathomechanisms with ordinaryvitiligo. Depigmentation around melanocytic nevi (halonevi) can resemble vitiligo, and the two conditions canbe associated [10]. A regressing melanoma can alsodemonstrate vitiligo-like depigmentation. An accuratehistory is often the only way to distinguish pseudovitiligofrom the real entity. To improve the accuracy of diagnosisof vitiligo, and to monitor the response to treatment, theVitiligo European Task Force has created an assessmenttool, which incorporates a staging system [11]. Thetreatment of vitiligo encompasses preventative measures,camouflage makeup, medical and physical agents andsurgical procedures. Prevention focuses on avoidingtrauma, friction, sunburn and emotional stress. Thefriction from everyday activities like bathing and dryingthe skin has probably been overlooked. Tight clothingand sports apparel are also to be avoided when practical.Stable, inactive vitiligo, especially segmental vitiligoresponds quite well to surgical repigmentation [12].Evidence-based scrutiny supports treatment of vitiligowith UV light, especially narrow-band UVB, topicalcorticosteroids, topical calcineurin inhibitors and surgicaltechniques [13–15]. Repigmentation takes two mainforms, follicular and diffuse, and is usually incomplete. In

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 3: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

Vitiligo, ROS and T-cells 101

the former, tiny islands of pigmentation, correspondingto hair follicles, appear in the patch and graduallyincrease in diameter and coalesce. In the latter, there ishomogeneous repigmentation of the patch, starting witha pale hue and becoming darker. Certain sites, such as thehands and feet, are very resistant to therapy. Followingsuccessful treatment, relapse is very common at all sites.Very extensive vitiligo is sometimes best managed withdepigmentation of remaining islands of normal skin.Psychological interventions should be offered to helppatients with vitiligo cope with their disease. Patients canhave significant co-morbidities like autoimmune thyroiddisease, and ophthalmological and auditory effects fromthe loss of pigment have been reported [16]. The clinicalfeatures and management of vitiligo have been reviewedrecently [17–20].

MELANIN

It is interesting to reflect that one’s skin colour isnot a ‘permanent coat’ but the result of regularreplenishing or ‘repainting’ according to the turnoverof the epidermis. Melanin pigments in the form ofeumelanin (brown-black shades) and phaeomelanin (red-yellow shades) are transferred from melanocytes in thebasal layer to associated keratinocytes via the so-calledepidermal melanin unit. One melanocyte is in contactwith approximately 36 surrounding keratinocytes [21].Granules of melanin are transferred to keratinocytesvia melanosomes in a process of phagocytosis. Becausemelanocytes represent only 8–10 % of all epidermal cells[22], most of the skin’s pigmented colour is determinedby melanin in the keratinocytes, and only at localizedcollections of melanocytes, as occurs in melanocyticnevi or ‘moles’, is the colour due to the melanocytesthemselves. Dark skin has a higher content of eumelanin,with larger melanosomes, and lighter skin has a higherproportion of phaeomelanin, with smaller melanosomes.Skin colour is not determined by the number or sizeof melanocytes. Melanin is thought to filter out UVradiation and scavenge ROS, thereby limiting UV damageto other cutaneous cells. The supranuclear melanincap structure in keratinocytes presumably minimizesphotodamage to the nucleus [23,24]. Melanocytes arealso thought to play a role in the skin immune system,secreting a wide range of signal molecules and respondingto growth factors and cytokines. Melanocytes canphagocytize and eliminate exogenous antigens, whichhave penetrated the skin barrier [25], and they can processand present antigen in the form of peptides with HLA(human leucocyte antigen) class II molecules to T-cells,triggering an adaptive immune response. Melanosomalproteins are involved in this antigen processing [26].Activation of T-cells by melanocytes is shown bythe secretion of co-stimulatory molecules like ICAM

(intercellular adhesion molecule)-1 and LFA (leucocytefusion-associated molecule)-3 [25,27]. When melanocytesdie, migrate or stop functioning, skin reverts to itsunpigmented form. This default colour is best exemplifiedin the single gene disorder of albinism, where melaninproduction is severely curtailed or completely absent.An acquired, patchy form of melanocyte failure occursin vitiligo, with a fully depigmented area having thesame colour seen in albinism. Onset is usually gradual,as several epidermal turnovers are required before allremaining pigment in keratinocytes is lost in the processof keratinization and desquamation. It seems reasonableto conclude that the skin colour disappears in a processof fading, taking several weeks to months. The specificdynamics of this process are difficult to study in vivo.Melanocytes in hair follicles seem to be more robust, sothat normal hair colour often persists in the white patch[28]. Greying and eventual whitening of scalp and bodyhair in the condition of poliosis is not considered a form ofvitiligo, but there may be some shared pathomechanisms;early greying of the hair can be seen in some vitiligolesions [29]. New studies on the control of melanogenesisand constitutional skin colour point to roles for cytokineslike SCF (stem cell factor), ET-1 (endothelin-1) andgranulocyte macrophage colony stimulating factor [30].

VITILIGO AETIOPATHOGENESIS:HYPOTHESES

The subject of vitiligo aetiopathogenesis has beenreviewed by several groups recently [31–35], but despitetremendous progress in molecular biology and genetics,there is still no universally accepted hypothesis. Itcould well be that vitiligo represents a ‘syndrome’rather than one disease, with numerous different butnot mutually exclusive pathways leading to melanocytefailure or disappearance. Genetic factors probablydetermine which particular pathway predominates ina specific patient. This was first proposed in the‘convergence’ theory of Le Poole et al. [36] in 1993 andlater refined by Schallreuter et al. [34] in 2008. Oneproposed schema is as follows: increased endogenousor exogenous phenol/catechol concentrations aroundthe melanocyte compete with tyrosine for tyrosinasebinding sites in the melanocyte, generating aberrantsubstrates and curtailing melanin production. Theseaberrant substrates generate reactive quinones, especiallyin the presence of ROS and disturbed redox balance,which impair cell functions and bind covalently to thecatalytic centre of tyrosinase, impairing or inactivatingthe enzyme and further reducing melanogenesis [31].Similar detrimental effects from excessive ROS occurwith other peptides and enzymes. Another proposalemphasizes the immunogenicity of melanocytes andmelanosomal proteins, with aberrant T-cell attack

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 4: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

102 S.J. Glassman

resulting in melanocyte destruction by apoptosis. Vitiligorepresents an autoimmune disease under this proposal[24]. Within these polar views of vitiligo pathogenesis,there is speculation that ROS and the immune systemmight somehow interact synergistically, so that bothmechanisms might be relevant. Further hypothesesfocus on neural dysregulation, hormones, cytokines,infections, calcium imbalance and melanocytorrhagy inthe causation of vitiligo [37,38]. The molecular controlof melanogenesis has been more accurately definedin the past decade: new hypotheses regarding vitiligohave emerged from this work, in particular, the roleof SCF/KIT protein interactions, and the downstreameffector, MITF-M (melanocyte-specific microphthalmia-associated transcription factor) [30,39,40].

VITILIGO GENETICS

The genetics of generalized vitiligo was reviewed bySpritz in 2008 [41]. Genes are thought to play a role inall aspects of vitiligo pathogenesis. There is a positivefamily history in about 20 % of cases and similarconcordance in identical twins. Vitiligo is considereda complex trait, involving the interaction of multiplegenes with non-genetic factors, both environmentaland host. At present, there is strong support forrelatively few susceptibility genes, including certainHLA genes, PTPN22, NALP1 and perhaps CTLA-4[CTL (cytotoxic T-lymphocyte) antigen-4]. These areall associated with a tendency to autoimmunity. HLAmolecules present peptides to T-cells, and it has beenproposed that certain HLA haplotypes confer moreefficient presentation of cognate autoantigen, therebypredisposing to autoimmunity; an example is HLA-DQB1*0301 [28]. NALP1 is involved in the innateimmune response to pathogens. Recent fine-mappingstudies showed associations with chromosomes 7 and 9[42]. Numerous other candidate genes and susceptibilityloci bear ongoing scrutiny, including CAT, GST, COMT,ACE, mannose-binding lectin 2 and XBP1 [43–46]. Arecent genome-wide association study by Jin et al. [47]using European white subjects and controls showedsignificant associations of generalized vitiligo with thefollowing loci, which have been previously linked withautoimmune diseases: HLA class I and II molecules,PTPN22, LPP, IL2RA, UBASH3A and C1QTNF6. Twoadditional immune-related loci identified were REREand GZMB. The HLA class I association occurred inthe regions between HLA-A and HCG9, consistentwith previous reports of strong associations with theHLA-A*02 allele, and the HLA class II gene associationoccurred in the region between HLA-DRB1 and HLA-DQA1, in keeping with known associations to theHLA-DRB1*04 allele. With the exception of PTPN22,the associations were similar whether patients had

vitiligo alone or vitiligo as well as another autoimmunedisease. An important association with a non-immune-related gene was identified: SNPs (single nucleotidepolymorphisms) in the gene encoding tyrosinase, TYR.Tyrosinase is a melanocyte enzyme that catalyzes the rate-limiting step in melanin biosynthesis and is a putativetarget autoantigen in vitiligo. Interestingly, certain TYRSNPs are associated with melanoma risk, and some ofthese are in linkage disequilibrium with vitiligo TYRSNPs shown in this study. Vitiligo TYR SNPs couldbe more antigenic than melanoma TYR SNPs, therebyconferring protection from melanoma through immunesurveillance [47,48].

MELANOCYTE LOSS

It is generally accepted that vitiligo results from lossof melanocytes rather than reduced functioning alone.This is supported by histological, ultrastructural andimmunohistochemical techniques showing that vitiligoskin is devoid of melanocytes, and by the fact thatmelanocytes are almost never able to be culturedfrom affected skin. Whether melanocytes in vitiligoare eliminated by necrosis or apoptosis has beendebated. Histological and ultrastructural studies supportapoptosis by revealing nuclear shrinkage, vacuolization,loss of dendrites and detachment. Known cytotoxinslike phenolic compounds, which can induce leukodermasimilar to that seen in vitiligo, cause melanocyte loss withplasma membrane blebbing and DNA fragmentationsuggestive of apoptosis; similar mechanisms might occurin vitiligo. Cytokine changes and immune reactions caninitiate apoptosis too, and these are thought to be relevantin melanocyte loss in vitiligo [49].

ROS AND AUTOIMMUNITY

The potential role of oxygen free radicals in humanautoimmune disease was reviewed by Ahsan et al. [50]in 2003. Free radicals are atoms or molecules which canexist independently despite having one or more unpairedelectrons. This makes them more reactive than thecorresponding non-radical forms. Free radicals derivedfrom oxygen include four moieties: O2

• − (superoxideanion radical), 1O2 (singlet oxygen), •OH (hydroxylradical) and HO2

• (perhydroxyl radical). Together,these are termed ROS. ROS are routinely generatedduring many cellular biochemical and metabolic chemicalreactions. Superoxide anion is thought to be the first freeradical generated, primarily via the electron transportchain in mitochondria. Hydroxyl and perhydroxylradicals are formed directly from superoxide. H2O2 isnot as reactive as free radical-derived ROS, but it is animportant oxidant, which can cross biological membranesand generate highly reactive hydroxyl radicals throughan interaction with transition metal ions like Fe2+ and

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 5: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

Vitiligo, ROS and T-cells 103

Cu+ in the Fenton reaction. H2O2 can, therefore, beconsidered a marker of potential ROS. H2O2 is formedwhen superoxide anion undergoes a dismutation reactioncatalysed by SOD (superoxide dismutase). Hydroxylradical can also be generated from the interaction ofsuperoxide anion radical and hydrogen peroxide in theHaber–Weiss reaction. Hydroxyl radical seems to bethe most potent and damaging radical in biologicalsystems, capable of interacting with all macromoleculeslike lipids, proteins, nucleic acids and carbohydrates.Polyunsaturated fatty acids are particularly susceptibleto hydroxyl radicals, whereby removal of a hydrogenatom from the fatty acid starts the process of lipidperoxidation in a type of chain reaction. Aldehydesof lipid peroxidation can react with amino acids,altering their biological activity. Membrane structureand function are affected. Protein oxidation results incross-linking, fragmentation and the addition of aldehydegroups, all affecting structure and function of the protein.Nucleic acids are damaged by single-strand breaks, cross-linking and changes to individual nucleotide bases. DNAis damaged by single-strand breaks, base modifications,conformational changes and DNA–protein cross-links.Thymine and guanine are more susceptible to damagethan cytosine and adenine. The reaction product betweensuperoxide anion and NO is OONO− • (peroxynitrite),which is also a strong oxidant for proteins, lipids andnucleic acids, causing cell damage [51]. ROS can also begenerated by exogenous stimuli like UV radiation andenvironmental chemicals. ROS have beneficial effects too,notably in the ‘respiratory burst’ of neutrophils duringthe phagocytosis of bacteria. Host cells have developedprotective mechanisms to counter the deleterious effectsof ROS and other free radicals in the form of free radicalscavengers and antioxidant chemicals and reactions.An imbalance between production and removal ofROS leads to oxidant stress or redox imbalance, andthis phenomenon has been implicated in numerousdisease states as well as the process of normal aging.ROS can modulate the expression of a variety ofimmune and inflammatory molecules, leading to tissuedamage. Aberrant immune reactions have been related tooxidative imbalance, and antioxidant functions have beenlinked to anti-inflammatory and immunosuppressiveproperties. If ROS-induced damage to cells is severeenough, programmed cell death or apoptosis occurs.Apoptotic debris can be highly immunogenic, andefficient removal by proteasomal degradation is necessaryto prevent autoimmune reactions [52–56]. Antioxidantsystems have evolved to control the redox balance.In the skin, these include antioxidant enzymes likecatalase, thioredoxin reductase, glutathione peroxidaseand superoxide dismutase, and small molecules likemethionine, glutathione, vitamin C and vitamin E.Oxidatively damaged proteins are repaired by MSRs(methionine sulfoxide reductases) [57,58,122].

ROS IN VITILIGO

Human skin serves as an interface between theenvironment and the body. It is constantly exposedto a broad array of physical, chemical and biologicalagents, many of which are either inherent oxidants orcatalyse the generation of ROS. ROS can denatureproteins, alter apoptotic pathways, damage nuclearand mitochondrial DNA and mediate release ofproinflammatory cytokines [59,60]. ROS are believedto be involved in the pathogenesis of inflammatoryskin diseases, carcinogenesis, photoaging and hairgreying [61,62]. Mitochondria are the most importantendogenous source of ROS, but they are also a targetof ROS-mediated damage. Thus, ROS can lead tomitochondrial dysfunction, reduced efficiency and moreROS in a vicious cycle of oxidant imbalance [59]. Severalcompelling lines of research have shown evidence ofoxidative stress throughout the epidermis of patientswith vitiligo, attributed to massive amounts of H2O2

in the 10− 3-M range [63]. Impetus for this researchcame from the finding of low catalase levels in theepidermis of patients with vitiligo, first reported in1991 [64]. Catalase removes H2O2 by converting itinto water and oxygen. Generation of H2O2 is aphysiological reaction in all cells via several metabolicpathways. There are also numerous exogenous directand indirect sources of epidermal H2O2. While lowconcentrations, of the order of 10− 6 M, are necessaryfor cell signalling and transcription, high concentrationscan have deleterious effects. Ultrastructural changessuggestive of lipid peroxidation have been demonstratedin melanocytes, keratinocytes and Langerhans cells inthe skin of patients with vitiligo, both in affected andperilesional areas [65–68]. High levels of epidermal H2O2

as well as the methionine oxidation product, methioninesulfoxide, have been demonstrated in vivo in vitiligousing FT (Fourier Transform) Raman spectroscopy[34,63]. This augmented previous findings of increasedH2O2, which were in vitro, based on cell cultureand skin biopsies [63]. FT Raman spectroscopy alsorevealed oxidation of l-tryptophan in epidermal albumin,and HPLC showed the presence of allantoin in theepidermis, confirming the presence of oxidative stress invitiligo [69,70]. Oxidative destruction of polyunsaturatedfatty acids of phospholipids is referred to as lipidperoxidation. It is one of the hallmarks of oxidativestress. MDA (malondialdehyde) is an end-product oflipid peroxidation, and elevated serum levels of MDAhave been documented in patients with vitiligo [71–73].

Sources of epidermal H2O2There are numerous sources of H2O2 in the normalepidermis. NADPH oxidase activity in neutrophils andmacrophages generates H2O2 [74]. TNF-α (tumour nec-rosis factor-α) leads to the formation of H2O2 indirectly,

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 6: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

104 S.J. Glassman

by inducing manganese superoxide dismutase [39]. Othercytokines have been reported to generate H2O2: TGF-β(transforming growth factor-β), EGF (epidermal growthfactor) and PDGF (platelet-derived growth factor) [75].Monoamine oxidase A activity in the epidermis generatesH2O2 [76]. Nitric oxide synthases create H2O2 in anenvironment deficient in l-arginine [77]. XO (xanthineoxidase) catalyses the conversion of purine bases into uricacid, and generates H2O2 as a by-product [70]. Oxidationof aromatic phenols like 17β-oestradiol to catechols byan NADPH-dependent CYP (cytochrome P450) yieldssuperoxide anion, which disproportionates to H2O2;this diffuses into the epidermis [78]. Photo-oxidation ofepidermal 6-biopterin and sepiapterin yields H2O2 [80].The enzymes tyrosine hydroxylase and phenylalaninehydroxylase also produce H2O2 as by-products. Externalphenols, ortho- and para-quinols, UVA and UVB, andX-rays also generate H2O2 in the mM range [63,77].Metabolism of xenobiotics, principally by CYP enzymes,generates ROS and H2O2, through toxic quinone andsemiquinone intermediates [56].

Several of these epidermal sources of H2O2 areshown to be augmented in vitiligo, providing thepresumed source for the elevated levels, which havebeen documented, both in affected and normal skin inpatients with vitiligo (Table 1). Increased epidermal TNF-α levels have been shown [39]. Increased photo-oxidationof epidermal 6-biopterin and sepiapterin has been demon-strated [80]. Impaired recycling of the essential cofactor6BH4 [(6R)-l-erythro-5,6,7,8,-tetrahydrobiopterin] byelevated H2O2 causes accumulation of H2O2 in theepidermis and affects all cofactor-dependent mechanisms.6BH4 is an essential electron donor in the hydroxylationof the aromatic amino acids, l-phenylalanine, l-tyrosineand l-tryptophan. These amino acids are substratesfor melanogenesis, and thus, 6BH4 is an essentialcomponent of the pigmentary system [79]. Induciblenitric oxide synthase levels in vitiligo epidermis areelevated, producing both H2O2 and peroxynitrite [8].Homocysteine oxidation also causes elaboration of ROS,and elevated serum homocysteine levels have beenreported in vitiligo patients [81]. Increased monoamineoxidase A activity has been found in the epidermis ofpatients with vitiligo, elaborating H2O2 [76]. ElevatedSOD activity would seem a likely source of H2O2

in vitiligo, but results have been contradictory. Bothnormal and elevated serum and tissue SOD activityhas been shown [71,82–85]. XO catalyses the oxidativehydroxylation of hypoxanthine to xanthine, and xanthineto uric acid as part of purine degradation. These reactionsgenerate H2O2, and XO is considered a major biologicalsource of ROS, leading to oxidative stress in many organs.XO contains two non-haem iron atoms in its structure,which can react with H2O2 to produce OH− •, the mostpotent ROS with respect to DNA damage. Because H2O2

also oxidizes uric acid to allantoin, this metabolite is a

Table 1 ROS in vitiligo: sources and effects

Level Reference(s)

SourcesH2O2 ↑ [63]Peroxynitrite ↑ [8]NADPH oxidase ↑ [74]TNF-α ↑ [39]Oxidized pterins ↑ [80]6BH4 recycling ↓ [79,80]iNOS ↑ [8]Homocysteine ↑ [81]Monoamine oxidase A ↑ [76]SOD n/↑ [71,82–85]Thioredoxin reductase ↓ [106]Xanthine oxidase ↑ [70]Catecholamines ↑ [33]GTP-cyclohydrolase I ↑ [98,99]Catalase ↓ [64,85]GSH-Px n/↓ [73,83–85]Vitamin E ↓ [72,73]

EffectsTyrosinase ↓ [121]TRP-1 ↓ [122]MSR ↓ [57,125]Catalase ↓ [64,85,106]Thioredoxin reductase ↓ [106]Tyrosine hydroxylase ↓ [79,80]POMC peptides ↓ [109–111]L-Phenylalanine ↑ [103]Acetylcholine ↑ [106,107]6BH4 recycling ↓ [79,102]Calmodulin, furin ↓ [103,113,114]Albumin (epidermal) ↓ [112]Malondialdehyde ↑ [71–73]Methionine sulfoxide ↑ [34,57,125]Allantoin ↑ [70]

useful marker of oxidative stress. XO activity has beenshown in skin, and elevated plasma levels have beenmeasured in patients with vitiligo. Recently, XO activityin melanocytes and keratinocytes was confirmed, withH2O2 regulating enzyme activity in a concentration-dependent fashion: low levels (10− 6 M) up-regulatedactivity, whereas high levels were suppressive. Oxidationby H2O2 of tryptophan and methionine residues in XOis thought to be the mechanism for this effect. Allantoinwas detected in the epidermis of acute vitiligo but not incontrol skin, further supporting a role for ROS in vitiligo[70].

Exogenous ROSThe role of exogenous oxidants in vitiligo is highlightedby the conditions of chemical leukoderma and contactvitiligo, as shared mechanisms might elucidate trigger

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 7: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

Vitiligo, ROS and T-cells 105

factors and reasons for progression and chronicityin idiopathic vitiligo. Chemical leukoderma refers toacquired depigmentation at sites of contact with certainchemicals; contact vitiligo starts in the same manner, butdepigmentation then spreads to distant sites, in the sameway as generalized idiopathic vitiligo. Depigmentationin both cases occurs from loss of melanocytes in theepidermis. Chemicals involved are mostly phenolic andcatecholic derivatives, which resemble tyrosine and canoccupy the catalytic centre of tyrosinase as surrogatesubstrates in the melanin synthesis pathway [86]. Theyinclude hydroquinone, MBEH (monobenzyl ether ofhydroquinone) and 4-TBP (4-tertiary butyl phenol).These are oxidized by tyrosinase or tyrosinase-relatedprotein to more reactive o-quinones, with the generationof ROS, which contribute to oxidative stress [87]. Inthe presence of excess H2O2, this process is accelerated.Melanin synthesis is reduced because the intermediatedopaquinone is not synthesized, and melanocyte viabilitycan be compromised. 4-TBP, a potent cause of contactvitiligo, is cytotoxic to cultured human melanocytesin a dose-dependent manner, with melanocyte lossattributed to apoptosis. There is marked variation inindividual susceptibility to chemical leukoderma andcontact vitiligo, emphasizing the key role of geneticfactors in determining melanocyte sensitivity to thesestimuli. Highly reactive o-quinones can react withnucleophilic groups on proteins to create antigensand stimulate an immune response. Phenols in well-known contact allergens, like poison ivy, might also beoxidized by similar mechanisms to form antigens onkeratinocytes. This could be the link between ROS andan altered immune response in vitiligo. Interestingly,the clinical picture of occupational contact vitiligois similar to allergic contact dermatitis, with itching,redness and scaling. When MBEH is used to removeremaining pigment in patients with vitiligo universalis,a similar reaction is seen, but only in the pigmentedareas, suggesting involvement of melanocytes ratherthan keratinocytes. Patients with established generalizedidiopathic vitiligo can be exquisitely sensitive toexogenous phenols and catechols [38,88–91]. Generationof a reactive o-quinone from MBEH via tyrosinase wasconfirmed recently in vitro, with isolation of several by-products with potential relevance to melanocyte toxicity[92]. Cytotoxic experiments have also confirmed recentlythat both 4-TBP and MBEH induce melanocyte death,but by different pathways: 4-TBP activates the caspasecascade and causes DNA fragmentation with apoptosis,while MBEH induces release of Mobility Group Box-1protein, which causes necrosis rather. This is confirmedby ultrastructural studies of MBEH-treated melanocytes[93]. Many drugs are potential exogenous sources ofROS, especially when metabolized by CYP enzymes,as they produce reactive quinones and semiquinones[56]. Interestingly, proton pump inhibitors were recently

shown to reactivate vitiligo, and the mechanism mightalso be through generation of free radicals [94]; pHchanges relating to melanogenic enzymes could also playa role [95].

Endogenous ROS: sources and effects

CatecholsEndogenous catechols are a source of ROS: elevatedplasma and urine catecholamines like norepinephrine,epinephrine and dopamine and their metabolites havebeen documented in vitiligo patients [96]. Keratinocytespossess β-2 adrenoceptors and synthesize and degradecatecholamines, and melanocytes synthesize norepineph-rine. Patients with vitiligo have markedly elevated GTP-cyclohydrolase I activity, which leads to excessive de novoproduction of 6BH4, leading to increased synthesis ofcatecholamines in the epidermis [96–99]. Catecholaminesalso compete preferentially with tyrosine for tyrosinaseactive binding sites, becoming hydrolysed in the processand generating H2O2 [33]. Norepinephrine-inducedvasoconstriction in vitiligo skin could cause hypoxiaand predispose to oxidative stress by this mechanism[38]. These phenomena, among others, are the basis forthe ‘neural’ theory of vitiligo aetiopathogenesis. Patientsoften report increased emotional stress prior to onsetof vitiligo or concurrent with a flare of disease activity[100]. Estrogens and progesterone can generate H2O2 invitiligo, contributing to quinone-mediated DNA damagein peripheral blood lymphocytes [78].

BH4 (tetrahydrobiopterin)BH4 cofactor is essential for various enzyme activitiesand is present in probably all cells. Six and sevenisoforms of BH4 are synthesized de novo from GTP,but regeneration is crucial to adequate functioning,and requires two enzymes, pterin-4a-carbinolaminedehydratase and dihydropteridine reductase. The latterare deactivated by H2O2 by oxidation of active sitetryptophan and methionine residues, and H2O2 alsooxidizes both 6- and 7-BH4 to 6- and 7-biopterin.This is the reason for fluorescence, which can beseen in vitiligo patches under Wood’s light. Thus, thehomoeostasis of this important cofactor is compromised[79,101,102]. Some of the enzymes that depend on BH4

are phenylalanine hydroxylase, tyrosine hydroxylaseand tryptophan hydroxylase, as well as all nitricoxide synthase isoforms. Thus, BH4 deficiency affectsmelanin, catecholamine, serotonin and NO synthesis.l-phenylalanine levels would be expected to rise in thissetting, and in fact, increased epidermal phenylalaninelevels have been documented in patients with vitiligo byin vivo FT Raman spectroscopy [103]. Increased de novosynthesis of 6-BH4 in vitiligo contributes to elevatednorepinephrine levels and up-regulates monoamineoxidase A and catechol-O-methyl transferase in the

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 8: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

106 S.J. Glassman

epidermis. These result in increased epidermal H2O2

[74,104,105].

AcetylcholineHigh epidermal levels of acetylcholine have beenreported in vitiligo, and this is attributed to almostabsent epidermal AchE (acetylcholinesterase) and BchE(butyrylcholinesterase) activities due to the effect of highlevels of H2O2. While low levels of H2O2 (10− 6 M)activate AchE, high concentrations (10− 3 M) deactivatethe enzyme. This regulation of enzyme activity byH2O2 is seen with several other enzymes. Molecularmodelling of AchE suggests that the inhibition is due toH2O2-mediated oxidation of tryptophan and methionineresidues in the protein, causing disorientation of theactive-site histidine residue. The tetramerization domainand calcium-binding domains in BchE are also affectedby high levels of H2O2 [106–108].

POMC (pro-opiomelanocortin)Oxidative stress via H2O2 directly affects POMCpeptides in the epidermis of patients with vitiligo. POMCpeptides have a key role in melanogenesis. Prohormoneconvertases 1 and 2, which cleave POMC, are oxidized byH2O2, and POMC-derived peptides like ACTH, α-MSH(α-melanocyte-stimulating hormone) and β-endorphinare also altered directly by oxidation of their methionineresidues. Reduced epidermal levels of α-MSH andβ-endorphin have been demonstrated in vitiligo bothin vitro and in vivo. These changes are reversed bythe reduction of H2O2 levels with narrow-band UVB-activated pseudocatalase PC-KUS. Oxidation of α-MSHcan also be ameliorated in the presence of abundant6BH4; this cofactor becomes deficient in the presenceof abnormal levels of H2O2. Regulation of tyrosinase,the key enzyme for pigmentation, by α- and β-MSH via6BH4 and 7BH4 has also been proposed, another linkbetween ROS and dyspigmentation [109–111].

TryptophanEpidermal l-tryptophan is oxidized by H2O2 in vitiligo,as shown in vivo by FT Raman spectroscopy. Albumincontains a tryptophan residue in its sequence, andoxidation could explain low levels of epidermal albumin,which have been reported in vitiligo. Several otheramino acid residues in albumin, such as methionine, arealso prone to oxidation. Some oxidation products oftryptophan like 5-OH-Trp are generated from H2O2

by Fenton chemistry. Albumin plays a key role incalcium homoeostasis, and reduced albumin levels mightaccount, in part, for impaired calcium uptake, which hasbeen described in vitiligo. H2O2 also affects all fourcalcium EF-hand-binding domains of calmodulin, andcalmodulin-ATPase activity is low in vitiligo skin. Theuptake of l-phenylalanine, which is defective in vitiligo,is also a calcium-dependent process. Epidermal furin is a

calcium-dependent prohormone convertase, which playsa role in the cleavage of POMC. Loss of a calcium-bindingsite in furin because of oxidation from H2O2 has beenshown in progressive vitiligo skin [103,112–114].

CytokinesSCF, a paracrine cytokine produced by keratinocytes,has a major role in promoting melanogenesis and inmelanocyte survival. SCF interacts with its receptor onmelanocytes, KIT protein, produced by the c-kit gene.KIT protein interacts with MITF-M, which serves as atranscription factor regulating expression of tyrosinasemRNA. In response to SCF/KIT protein signalling,MITF-M is activated by phosphorylation through a MAPkinase, whereafter it is translocated to the nucleus toactivate several genes. MITF-M also interacts with Bcl-2 to prevent apoptosis of the melanocyte [115]. ET-1 isalso an important regulator of melanin production, via itsETBR receptor [116]. SCF and ET-1 were not shown tobe deficient in vitiligo lesions, suggesting that abnormalparacrine secretion by keratinocytes is not the cause of thehypopigmentation. At the edge of a vitiligo lesion, thereare still melanocytes expressing tyrosinase, ETBR andS100α, albeit at slightly lower levels than unaffected skin,but KIT protein and MITF-M are markedly reduced.Melanocytes expressing tyrosinase, S100α, ETBR, KITprotein and MITF-M protein were absent at the centre ofthe vitiligo lesion. This suggests that reduced expressionof KIT protein, and its downstream effectors like MITF-M, could explain melanocyte loss and/or dysfunction invitiligo [30]. It is interesting that oxidative stress likeexcessive H2O2 leads to down-regulation of MITF-Mexpression in cultured human melanocytes. Thus, ROScould be the cause of the cytokine abnormality seen invitiligo [117].

TraumaTrauma to the skin, UV radiation and other sourcesof inflammation probably contribute to the epidermalpool of H2O2 in a non-specific manner via NADPHoxidase stimulation. This could explain the prominentKoebner phenomenon seen in vitiligo, especially in activestages of the disease [118,119]. In particular, UVB inducesgeneration of free radicals, which damage DNA. Thiscan be ameliorated by vitamin D, which up-regulatesexpression of metallothionein, a free radical scavengerprotein with photoprotective properties [120].

TyrosinaseHigh levels of H2O2 (0.5–5.0 × 10− 3 M) have been shownto deactivate tyrosinase, and this effect is compounded byincreased 6BH4 [121]. Recent work on the mechanism ofsenile hair greying has provided a possible explanationfor the inhibition of tyrosinase by H2O2. It was shownthat methionine 374, at the enzyme active site, is oxidizedby H2O2, disrupting enzyme activity, especially when

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 9: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

Vitiligo, ROS and T-cells 107

methionine sulfoxide repair by MSRA and MSRB isalso affected by the same ROS. A similar scenarioevidently occurs in vitiligo [62]. Abnormal expression ofTRP-1 (tyrosinase-related protein-1) has been reportedfollowing oxidative stress to cultured melanocytes fromthe advancing border of vitiligo lesions. This leads to earlycell death, possibly through an interaction with calnexin[122].

PeroxinitriteRecently, significantly elevated levels of 8-oxoguaninewere reported in plasma and skin of patients withvitiligo, an indication of DNA damage. Together withthis, there was up-regulation of epidermal wild-typep53 and enhanced short-patch base-excision repair. Inaddition, high epidermal levels of iNOS (inducible nitricoxide synthase) were demonstrated, with correspondingelevations of 3-nitrotyrosine and nitrated p53. Thisimplies increased epidermal ONOO− •, a reactivenitrogen species, which can be added to the list ofradicals involved in the pathogenesis of vitiligo. H2O2

was shown to enhance the DNA binding capacity ofp53, while ONOO− • completely inhibited this binding.Interestingly, H2O2 at a concentration of 10− 3 Mabolished this deleterious effect of ONOO− •. Thus,H2O2 appears to be protective in the sense of improvingDNA repair via enhanced p53. This could partly explainthe relative absence of photoaging and skin cancer inchronic lesions of vitiligo [8].

AntioxidantsElevated levels of H2O2 and other oxidants wouldordinarily be countered by antioxidant defenses and freeradical scavengers. However, several of these processesare actually inhibited by H2O2 itself. H2O2 in the mMrange deactivates catalase by oxidizing its porphyrin ringand methionine and tryptophan residues in the structureof the enzyme-active site and the cofactor NADPH-binding site. This explains why catalase levels are lowin vitiligo, despite unchanged mRNA expression for theenzyme [106]. Low tissue levels of catalase in patientswith vitiligo were recently confirmed, with lower levelsin active rather than stable disease [85,123]. Severalheterogeneous SNPs in the catalase gene, observed invitiligo patients, might render these catalase variantsparticularly susceptible to deactivation by H2O2, therebypredisposing to vitiligo [43,124]. Another antioxidantenzyme system, thioredoxin reductase, is impaired byhigh H2O2 levels through oxidation of the enzyme-active site and the NADPH cofactor-binding site. Lowactivities of this enzyme have been shown in vitiligo[99]. Oxidation of MSRA and MSRB by H2O2 leadsto reduced functioning of this important protein-repairmechanism. Low levels of MSR have been shown invitiligo. Furthermore, in vitro down-regulation of MSRA

in normal cultured melanocytes rendered them verysusceptible to oxidative stress [57,125].

GSH-Px (glutathione peroxidase) converts H2O2 andother peroxides into water and oxygen, utilizing gluta-thione, glutathione reductase and NADPH. Seleniumis a cofactor for some isoforms of GSH-Px. Variablelevels of GSH-Px and selenium have been found in tissueand blood of patients with active vitiligo [73,83–85,126–128]. GST (glutathione transferase) is a superfamily ofbroadly expressed isoenzymes involved in defence againstoxidative stress. Polymorphisms in the GST gene havebeen identified, which confer risk of vitiligo [45]. VitaminE levels were reported as low in vitiligo patients, who alsoshowed elevated blood levels of MDA, as evidence of lipidperoxidation [72,73]. Total blood antioxidant status usingthe Randox kit was increased in vitiligo patients in onestudy [128].

Melatonin has a strong antioxidant effect, and anaberration of melatonin receptors was once proposedas a mechanism for the development of vitiligo[129]. Melatonin is synthesized in the skin yieldingpotent free radical scavengers like N1-acetyl-N2-formyl-5-methoxykynuramine, which also stabilize themitochondrial electron transport chain [130].

Antioxidants react in tandem with proteins like Bcl-2and caspases to regulate apoptosis, which can result fromoxidative challenge [131]. For example, Bcl-2 expressionprevents cell death from H2O2 and menadione [132]. Bcl-2 is expressed by melanocytes and keratinocytes and iscrucial to their survival. Melanocytes from individualswith vitiligo have lower levels of Bcl-2 than normal,presumably contributing to melanocyte fragility [91].Elevated levels of Bcl-2 have also been reported invitiligo [8], as well as normal levels [133], and the roleof melanocyte apoptosis in vitiligo is still unclear.

Further proof of the role of H2O2 and ROS requiresevidence of improvement in lesions with antioxidanttherapies. Topical pseudocatalase (PC-KUS) is a narrow-band UVB-activated bis-MnIII-(EDTA)2-(HCO3

− )2

complex that is applied to the entire skin as a lotion,followed by low-dose UVB exposure. It functions as asynthetic catalyst to oxidize H2O2 into O2 and H2O, thusmimicking natural catalase. This regimen causes rapidrepigmentation of even long-standing vitiligo [134,135].Consequent with this is a marked reduction in epidermalH2O2 as measured in vivo with FT Raman spectroscopy.Bathing in Dead Sea water also had a pseudocatalaseeffect, though milder, probably from its high content oftransition metals like manganese [135]. A randomizedcontrolled trial comparing narrow-band UVB withor without a pseudocatalase cream did not confirmadditional benefit from the cream, but the methodologywas different, and the pseudocatalase was not thesame as PC-KUS [136]. A catalase of vegetable origin,combined with SOD in a microsphere formulation,showed equivalent efficacy to topical betamethasone,

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 10: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

108 S.J. Glassman

Figure 1 Summary of sources, effects and interactions of ROS/H2O2 in vitiligo

when combined with sunlight exposure in the treatmentof vitiligo [137]. Oral vitamin E appeared to confersome additional benefit to narrow-band UVB therapyof vitiligo, with a corresponding decrease in plasmaMDA in the vitamin E group versus the control [138].ROS/H2O2 sources, effects and interactions in vitiligoare summarized in Figure 1.

VITILIGO AND AUTOIMMUNITY

Generalized vitiligo is widely considered an autoimmunedisease, with involvement of humoral and cellularcomponents of the innate and adaptive immune system.The belief is supported by the following lines ofepidemiological, clinical and investigational research: anassociation with other autoimmune disorders; chronicrelapsing and remitting course so typical of autoimmunedisorders; possible response to immunosuppressivetherapies like UV phototherapy, topical and oralcorticosteroids, and topical calcineurin inhibitors;circulating anti-melanocyte antibodies; T-cell infiltratesin perilesional skin; anti-melanocyte cytotoxic T-cells inthe skin and circulation and proinflammatory cytokinepatterns of a Th-1 type response. Autoimmunity mightbe the triggering event in vitiligo, but it could functioninstead as a promoter of disease progression andchronicity [26,139].

Autoimmune conditions associated with vitiligoinclude autoimmune polyendocrine syndrome types 1and 2, pernicious anaemia, Type 1 diabetes, Addison’sdisease, Graves’ disease, alopecia areata, systemic lupus

erythematosus, rheumatoid arthritis, psoriasis and myas-thenia gravis. A recent survey of 2600 vitiligo patientsshowed increased frequencies of autoimmune thyroiddisease, Addison’s disease, systemic lupus erythematosusand pernicious anaemia, with about 30 % of patientshaving at least one of these disorders. In addition,family members who did not have vitiligo still had atendency to the same autoimmune conditions, pointing toa genetic risk for a specific cluster of autoimmune diseases.Other studies report true associations only with thyroiddysfunction and thyroid antibodies, regarding the otherconditions as random concomitant events. Psoriasis orlichen planus occurring in vitiligo lesions has also beenreported, with a shared pathogenesis postulated. Organ-specific autoantibodies are recorded with increasedfrequency in vitiligo patients, often in the absence ofclinical symptoms. There is probably an increased riskof developing clinical or subclinical disease later [139–142]. Several immunogenetic factors predispose patientsto autoimmune diseases, and some of these are associatedwith vitiligo, adding to the evidence that vitiligo mayhave an autoimmune basis. Various HLA class II alleleshave been associated with vitiligo, in particular, HLA-DR4 [143]. The particular haplotype association variesaccording to ethnic origin. Genes involved in antigenpresentation and processing have been associated withautoimmune diseases and in some cases with vitiligo.These include LMP-1 and -7 (low-molecular-masspolypeptide-2 and -7) and TAP-1 and -2 (transporterassociated with antigen processing protein-1 and-2) [144]. Homozygous or heterozygous complement

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 11: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

Vitiligo, ROS and T-cells 109

2 and 4 deficiency is associated with autoimmunity, andthis has been described in vitiligo [145]. The CTLA-4gene product down-regulates T-cell activation andcontrols T-cell apoptosis. Certain CTLA-4 polymorph-isms predispose to vitiligo in patients who alreadyhave other autoimmune conditions [146]. Autoimmunepolyendocrine syndrome type 1, which often includesvitiligo, is due to mutations in the autoimmune regulatorgene, AIRE [147]. A missense mutation in the PTPN22gene, which encodes LYP (lymphoid protein tyrosinephosphatase), has been linked to several autoimmunediseases including vitiligo [148]. Loci on chromosomes1, 7 and 8 have been linked with autoimmune diseasesand termed AIS (autoimmune susceptibility loci) 1, 2 and3, respectively. AIS2 and -3, in particular, are associatedwith vitiligo. A locus designated SLEV1 on chromosome17p13 has also been linked to vitiligo [149].

Animal models of vitiligo show prominent roles foranti-melanocyte antibodies. Some of these cross-reactwith mammalian TRP-1 [150]. Antibodies to melanocyteshave been found in the circulation of patients with vitiligo[151]. These antibodies correlate with disease activityand extent [152]. Targets of these antibodies include avariety of melanocyte and melanosomal antigens. Thefrequency of each antibody in the vitiligo populationis relatively low, and a dominant antigen has notbeen found. Antibodies might trigger vitiligo as aprimary event, but they could also arise secondary tomelanocyte damage or serve to perpetuate the disease.Whatever their role in vitiligo, these antibodies have thecapacity to injure pigment cells in vivo and in vitro.Even if they are not pathogenic, study of melanocyteantibodies and target antigens might refine the diagnosticand prognostic testing of vitiligo, reveal putativeT-cell targets and add to the therapeutic armamentarium[153]. Circulating anti-parietal cell, thyroid and adrenalantibodies have been detected in vitiligo patients,as well as antinuclear antibodies and rheumatoidfactor, again suggesting an autoimmune pathomechanismfor the disease [151]. Autoimmune diseases are theresult of complex interactions between T and B cellsubpopulations. A recent flow cytometric study of thesecells in vitiligo did not show a pathological distributionof B cells, suggesting that T-cells might have a moredominant role. Circulating immune complexes were notelevated either [154]. Cases of generalized vitiligo inrecipients of bone marrow transplants from donors withvitiligo have been reported, illustrating the role of bonemarrow-derived cells in disease pathogenesis [155].

T-CELLS IN VITILIGO

Peripheral bloodLymphocyte analysis in peripheral blood of patients withvitiligo has yielded variable results (Table 2). Earlier

Table 2 T-cell changes in vitiligo

Level

PhenotypePeripheral [156–163]

Total T-cell n/↓CD4+/CD8+ n/↓/↑CD45RA+ ↓CD45RO+ ↑Total NK T-cell ↓

Tissue [179–193]Total T-cell ↑CD8+ ↑CD45RO+ ↑IL-2 receptor ↑CLA+ ↑

CytokinePeripheral [164–174]

IL-1 ↑IL-6 ↑IL-8 n/↑TNF-α n/↑/↓IFN-γ ↓TGF-β ↓IL-2 receptor ↑/↓TNF receptor n

Tissue [165–174]IL-2 receptor ↑IL-6 ↑TNF-α ↑IFN-γ ↑IL-10 ↑

studies found normal total levels of T-cells but a decreasedCD4+/CD8+ ratio and normal or increased total NKcells. There was no particular correlation noted withdisease activity [156–160]. Abdel-Naser et al. [161] foundnormal levels of total T-cells, CD4+ T-cells and totalNK cells, but a decreased proportion of CD45RA+

(naıve CD4+) T-cells and increased HLA-DR expression,suggesting the presence of activated T-cells. Mahmoud etal. [162] found decreased total T-cells, CD45RA+ cellsand NK T-cells, with increased CD45RO+ (memoryphenotype) T-cells in severe disease. Basak et al. [163]found normal total T-cells with a decreased CD4+/CD8+

ratio, as well as increased monocytes, CD45RO+ T-cells and total NK cells. Total T-cells were relativelyhigher in generalized than in acral/acrofacial vitiligo,and CD45RO+ T-cells were higher in acral/acrofacialdisease. Unlike former studies, Pichler et al. [154]found an increased CD4+/CD8+ ratio in peripheralblood, but noted that the ratio in tissues could bedifferent.

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 12: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

110 S.J. Glassman

CytokinesCytokine studies of peripheral blood and skin in patientswith vitiligo have also yielded variable results, butwith a trend to proinflammatory T-cell patterns. Earlierwork showed increased IL (interleukin)-6 and IL-8 butdecreased TNF-α and IFN (interferon)-γ in serum,with elevated soluble IL-2 receptor in blood and tissue[164,165]. Moretti et al. [166] found increased IL-6, TNF-α and minimal TGF-β in tissue, and Tu et al. [167]found increased IL-6 but normal IL-1β, IL-8 and TNF-α in blood. Franczuk et al. [168] reported decreasedsoluble IL-2 receptor in blood. Grimes et al. [169] notedincreased tissue TNF-α, IFN-γ and IL-10, and Zailaie[170] detected increased IL-1, IL-6, IL-8 and TNF-αin blood, while Birol et al. [171] found elevated tissueTNF-α but normal blood levels of this cytokine. Itwas recently noted that imiquimod often causes vitiligo-like depigmentation when used to treat superficial basalcell carcinoma. Imiquimod binds Toll-like receptors 7and 8 and evokes a Th-1 response with the productionof IFN-α, TNF-α and IL-12. Imiquimod also causesincreased IL-6, IL-8 and IL-10. Similar cytokines mightbe involved in vitiligo [172]. Taher et al. [173] notedthat topical tacrolimus, used successfully to treat vitiligo,increases tissue IL-10, which is an immunosuppressiveTh-2 cytokine. This suggests that vitiligo might be aTh-1 type of autoimmune disease. Pichler et al. [154]recently found normal blood levels of TNF receptor withslightly elevated IL-6, while Basak et al. [174] reportedsignificantly decreased serum levels of TGF-β in vitiligo,with potential inhibition of regulatory T-cell function.

InfiltratesEarlier histopathological studies of vitiligo focused ondegenerative changes in melanocytes and keratinocytes,particularly at the perilesional location. Vacuolaralteration of the basal layer was noted, with scatterednecrotic keratinocytes reminiscent of a lichenoid reaction.Electron microscopy showed intracellular oedema,cytoplasmic vacuolation and dilation of organelles withindegenerating melanocytes and keratinocytes in normal-appearing skin adjacent to amelanotic skin. In somecases, degeneration was limited to melanocytes, sparingkeratinocytes. Little attention was paid to mononuclearcell infiltrates, but these were, in fact, noted, being in aperivascular distribution and also within the epidermis.The infiltrate was considered to comprise lymphocytesand histiocytes, and while generally of a mild intensity,denser infiltrates were observed in vitiligo of morerecent onset and in active rather than stable disease[66,175–178]. Lichenoid-type inflammation implies a T-cell-mediated autoimmune reaction [179], and in fact,many of the mononuclear cells in the perilesional infiltrateof ordinary generalized vitiligo were shown to be T-cells by immunoperoxidase techniques. CD8+ T-cellspredominated in the epidermis, while in the dermis,

there was a normal ratio of CD4+/CD8+ T-cells [180].Activation of cell-mediated immunity was also suggestedby the early finding of abnormal expression of HLA classII molecules and ICAM-1 on perilesional melanocytes incases of vitiligo [181]. Highly significant overall increasesin CD3+, CD4+ and CD8+ T-cells were also notedby Badri et al. [182] in the margins of depigmentedskin. Many of the T-cells also expressed HLA class IImolecules and IFN-γ . CD45RO+ memory phenotypepredominated, with many T-cells expressing the skin-homing marker CLA (cutaneous lymphocyte-associatedantigen), which binds to E-selectin on dermal endothelialcells [182]. Mononuclear infiltrates in perilesional vitiligoskin were noted to be IL-2 receptor- and IFN-γ -positive,suggesting the presence of activated T-cells [178]. CLA+

T-cells clustered around disappearing melanocytes weredescribed by van den Wijngaard et al. [183]: mostof the T-cells were CD8+, and those interacting withmelanocytes were perforin and granzyme B-positive,suggesting a cytotoxic phenotype. There was focalincreased expression of HLA-DR and ICAM-1 in theepidermis at sites of interaction with T-cells [183].Melanocytes are sensitive to the granule exocytosispathway of apoptosis (perforin/granzyme) but resistantto the Fas ligand-mediated pathway [184]. Melanocytedeath in vitiligo has generally been attributed to apoptosisrather than necrosis, and cytokines like IL-1, IFN-γor TNF-α, released by lymphocytes, keratinocytes andmelanocytes, can initiate apoptosis of both melanocytesand keratinocytes. Fas/Fas ligand interactions may alsobe involved in the apoptosis of keratinocytes in vitiligo[185,186].

Cellular infiltrates were particularly noted in rare casesof ‘inflammatory’ vitiligo, characterized by elevated,reddish, scaly borders. Histopathological analysis of theborder often showed a lichenoid pattern of lymphocyticinfiltration, even a few centimeters away towards normalskin [187,188]. Lymphocytes were often juxtaposedto remaining melanocytes, suggesting a role in theirdestruction. Dense perivascular infiltrates were notedin the upper dermis, with exocytosis of lymphocytesinto the epidermis. Vacuolar degeneration of the basallayer, with necrotic keratinocytes and colloid bodies werenoted. Immunophenotyping of inflammatory vitiligoalso confirmed the presence of many T-cells, particularlywithin the epidermis. Epidermal T-cells were eitherCD8+ or CD4+, while dermal T-cells were mainly CD4+.T-cells showed a memory subtype, being CD45RO+.Keratinocytes from the inflamed border were HLA DR+,and ICAM-1 staining was increased in one study. T-cellsshowed increased IL-2 receptor expression, suggestingactivation, and many T-cells were CLA+ [189,190]. Thus,both ordinary and inflammatory vitiligo had similarpatterns of T-cell infiltration.

In vitro characteristics of infiltrating T-cells inordinary vitiligo were reported for the first time by

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 13: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

Vitiligo, ROS and T-cells 111

Wankowicz-Kalinska et al. [191] in 2003: biopsies ofperilesional skin in four patients with active and onewith stable vitiligo confirmed the presence of T-cellinfiltrates, mainly CD8+ T-cells, but with many CD4+

T-cells as well. These were often noted in appositionto residual melanocytes. Normal skin usually shows anequal distribution of CD4+ to CD8+ subsets, mostly in adermal perivascular location. The predominance of CD8+

over CD4+ T-cells in vitiligo, as well as their proximityto melanocytes, could represent the effector phase of ananti-melanocyte response, as seen in the Smyth chickenmodel of vitiligo [192]. CD8+ T-cells are known toinfiltrate organs like the pancreas and thyroid gland inother autoimmune disorders like diabetes mellitus andHashimoto’s thyroiditis, respectively [193,194]. A novelfinding by Wankowicz-Kalinska et al. [191] in the studywas the presence of T-cell infiltrates in normal-appearingskin remote from the site of vitiligo, with microscopic lossof melanocytes termed ‘microdepigmentation’. Thesecould well represent sites of future vitiligo. T-cells weregenerated by mitogenic expansion and tested for cytokineelaboration and antigen specificity. Phenotypes generatedwere similar to those observed in situ. In all four patientswith active vitiligo, a high proportion of both CD8+ andCD4+ cells produced IFN-γ and TNF-α, with relativelylittle IL-4, IL-5 and IL-13. This pattern was not seenin T-cells derived from normal skin in these patients.Thus, the T-cells in vitiligo are polarized to a Th-1-likeprofile. TNF-α, and IFN-γ , might act synergisticallyto up-regulate HLA Class II molecules and ICAM-1,as noted previously [191]. Melanocytes can process andpresent antigenic peptides to antigen-specific CD4+ cellsafter pretreatment with IFN-γ ; therefore, elevated IFN-γ and TNF-α could predispose melanocytes to immunesurveillance [195]. The patient with stable vitiligo had adifferent cytokine profile, more Th-2-like. It is interestingthat multiple sclerosis shows a Th-1 type response inactive disease and a Th-2-dominant response in quiescentstages; perhaps a similar phenomenon occurs in vitiligo[196]. Several CD8+ T-cell clones were able to lyseautologous cultured melanocytes in an HLA-restrictedmanner, the first time that T-cells from tissue rather thanblood of patients with vitiligo have shown this effect.Blocking with anti-HLA class I antibody preventedthe lysis, confirming a specific, HLA-restricted CD8+

cytotoxic T-cell response rather than an NK-like response[191].

Antigen-specific T-cellsThe study of antigen-specific T-cells in vitiligo benefitedfrom work on the cell-mediated immune response tomelanoma, shared melanocyte differentiation antigenswith melanoma and a simpler technique to detectantigen-specific T-cells, known as soluble tetramerstaining. Vitiligo is more frequent in patients withmetastatic melanoma than the general population [197–

200] and is associated with increased survival. Vitiligo-like depigmentation has been reported after successfulimmunotherapy of melanoma, including high dose IL-2therapy, infusions of peptide-pulsed DCs (dendritic cells)and Melan-A/MART-1-specific cytotoxic T-cell clones[201–206]. During adoptive transfer therapy, infusedmelanocyte-specific T-cells accumulated in the peripheryof sites of incipient depigmentation. Depigmentationwas not observed in non-responding patients orthose with unrelated malignancy [203]. Cytotoxic T-cells generated from melanoma recognize melanocytedifferentiation antigens expressed by normal melanocytesas well [203,207,208]. Clonally expanded T-cells withidentical T-cell receptor Vβ regions were simultaneouslydemonstrated in the depigmented halo around aregressing melanoma and within the melanoma [209].The possibility that melanocyte differentiation antigensrepresent the targets of cell-mediated autoimmunity invitiligo has been investigated recently. These antigensinclude melanosomal proteins like Melan-A/MART-1, tyrosinase and gp100, involved in the synthesisof melanin [197]. High numbers of Melan-A-specificcytotoxic T-cells have been observed in the peripheralblood of melanoma patients with concurrent vitiligo,but not in those without vitiligo [210]. Regression ofmetastatic melanoma and the simultaneous developmentof vitiligo have occurred after immunization with Melan-A peptide, associated with oligoclonal expansion ofMelan-A-reactive cytotoxic T-cells [211]. Utilizing thetechnique described by Altman et al. [212], multimericsoluble complexes of known peptides with HLA-A2molecules could be used to identify peptide–antigen-specific T-cells in HLA matched hosts. These peptide–HLA-A2 complexes bind more than one T-cell receptoron a specific T-cell, allowing a slower dissociation rateand enabling successful immunological staining [212].Utilizing this technique, Ogg et al. [213] showed forthe first time a high frequency of Melan-A-specificCD8+ T-cells in the circulation of HLA-A2+ patientswith vitiligo. These cells were able to lyse A2-matchedmelanoma cells in vitro. They were not observed inA2-negative vitiligo patients or in A2-positive controlpatients who did not have vitiligo. The T-cells alsoexpressed high levels of skin-homing CLA [213]. Lang etal. [214] confirmed these findings, with levels of Melan-A-specific CD8+ T-cells correlating with disease activity.Four peptides from Melan-A were used. Gp100 andtyrosinase-specific CD8+ T-cells were also demonstrated,using five peptides from gp100 and two from tyrosinase.Intracellular IFN-γ was elevated in the CD8+ T-cells,confirming the active state shown by the enzyme-linkedimmunospot technique [214]. Wankowicz-Kalinska etal. [191] also showed Melan-A specificity of clonedT-cells from perilesional skin by the tetramer technique,revealing a high proportion of Melan-A-specific CD8+

T-cells in two of five HLA-A2.01+ patients with vitiligo.

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 14: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

112 S.J. Glassman

Interestingly, these two patients failed to show similarCD8+ cells in the peripheral circulation, emphasizing theimportance of testing both peripheral and tissue sites,if possible [191]. Mandelcorn-Monson et al. [215] inan ex vivo study utilized peripheral blood of patientswith vitiligo to isolate antigen-specific CD8+ T-cells,without additional in vitro culture, and revealed a highproportion of gp100-positive CD8+ T-cells, unlike theother studies. Two epitopes of gp100 were used to increasesensitivity. Utilizing recognized epitopes from Melan-A and tyrosinase, specific T-cells were not isolated.One patient showed Melan-A and tyrosinase specificityafter in vitro re-stimulation [215]. Palermo et al. [216]utilized the tetramer technique with peripheral bloodof patients with vitiligo or melanoma and showed thepresence of Melan-A-specific cytotoxic T-cells at similarlevels in both HLA-A2-matched groups. However, onlyvitiligo cells showed T-cell-receptor down-regulationand IFN-γ production after exposure to HLA-matchedmelanoma cells, suggesting a qualitative difference inreceptor affinity in the two groups. Vitiligo has beenconsidered the effective variant of melanoma immunity[216,217]. Van den Boorn et al. [218] expanded onall these findings in 2009: their study showed aprominent role for T-cells in vitiligo pathogenesis,being causative rather than just a consequence of otherstimuli. Perilesional T-cells were easily identified in nineHLA-A2+ patients with progressive vitiligo, culturedand subjected to flow cytometric analysis using HLA-A2/peptide tetramers for tyrosinase-(369–377), gp100-(280–288), gp100-(209–217) and MART-1-(26–35). Acontrol antigen in the form of influenza virus-(58–66)was employed. Compared with healthy donor skin-residing cells, significantly increased levels of T-cellsrecognizing all the melanocyte antigens were found inthe perilesional T-cell population. Similar proportionsof these T-cells were also found in the peripheralblood, showing that the presence of melanocyte antigen-specific T-cells in the skin usually coincides with similarincreased levels of these cells in the blood. Functionalactivation of cultured perilesional T-cells upon exposureto melanocyte differentiation antigens was tested forthe first time. T-cells were stimulated with the poolof HLA-A2-binding peptides in five patients and threecontrols. In order to exclude bias from prolonged invitro culture, only those samples that grew sufficientcells within 14 days were analysed. Activation of T-cells was measured by CD-69 expression for earlyT-cell activation, CD137 for CD8+ T-cell activationand CD154 for CD4+ T-cell activation. Cytolyticaction was measured by granzyme-B production andcytotoxic degranulation by CD107a expression. IL-4, ahumoral response promoter, and the proinflammatorycytokines IL-17, TNF-α and IFN-γ were also measured.Results showed that after melanocyte antigen exposure,perilesional CD8+ T-cells became activated (CD69+,

CD137+, granzyme-B+ and CD107a+); they were notactivated by influenza controls. The magnitude of theCD8+ T-cell response was proportional to the extentof vitiligo or the presence of poliosis. PerilesionalCD4+ T-cells showed bystander activation, with CD69and CD154 expressed but not granzyme-B. Thisdemonstrates an antigen-independent activation of theseCD4+ T-cells in this HLA class-I-restricted stimulationsetup. There was a variable increase in TNF-α andIFN-γ by both perilesional CD4+ and CD8+ T-cells.One patient, who also had halo nevi, demonstratedelevated IL-17. Donor control skin-residing T-cells didnot show a cytotoxic response to the melanocyte antigens.Therefore, perilesional T-cells in vitiligo are primed toreact against melanocyte antigens unlike healthy skin-residing T-cells. To examine the functional capacityof these T-cells to kill melanocytes, a skin explantmodel, originally developed to predict GVHD (graftversus host disease) in bone marrow transplantation, wasused. Perilesional T-cells were co-cultured for 2 dayswith non-lesional, normally pigmented autologous skin;CD8+-enriched and -depleted populations were alsotested. Explants were subsequently analysed for thepresence of infiltrated T-cells, melanocytes and apoptosis.Results in all cases showed infiltration of explants, withinduction of apoptosis of melanocytes as indicated bycytoplasmic active caspase-3 staining. Other epidermalcells in the suprabasal layers, probably keratinocytes, alsounderwent apoptosis. CD8+-depleted samples showedinfiltration but minimal apoptosis, while CD8+ enrichedsamples had the most apoptosis of melanocytes andkeratinocytes, corresponding to sites of epidermalT-cell infiltration. Keratinocyte apoptosis did not occurin lesional skin devoid of melanocytes, and is thusconsidered a bystander effect from proinflammatorycytokines rather than a specific cytotoxic effect of T-cellson keratinocytes. Finally, the same explant experimentwas performed, but utilizing purified melanocyteantigen-specific T-cell populations. Gp100-(280–288)-specific CTLs were co-cultured with autologous skin anddemonstrated infiltration of the explant, with extensivedisruption of the skin tissue. CLSM (confocal laserscanning microscopy) showed CTLs infiltrating thedermis and epidermis, causing damage to the dermo-epidermal junction and basal melanocytes. When theseCTLs were stimulated in vitro with the same gp100-(280–288) antigen, they produced large amounts of IFN-γ .The same cells did not produce IFN-γ after stimulationwith irrelevant tyrosinase epitopes, or if there was noadditional peptide stimulation, indicating the antigen-specific nature of the activation. Tyrosinase-(369–377)-and MART-1-(26–35)-specific CD8+ T-cell clones werealso co-cultured with explant skin, also demonstratinginfiltration and apoptosis of melanocytes. Keratinocyteapoptosis was not observed in these samples. Noapoptosis was found when these T-cells were incubated

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 15: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

Vitiligo, ROS and T-cells 113

with lesional (melanocyte-free) skin explants, showingthat activation was antigen-specific and dependent on thepresence of melanocytes [218].

With respect to tyrosinase as a potential autoimmunetarget in vitiligo, it is interesting that the recentgenomewide association study confirmed a significantassociation with several TYR SNPs, in particularrs1393350. Significant epistasis was observed between thisSNP and the HLA-A SNP rs12206499, and it is postulatedthat this combination presents the TYR polypeptide moreefficiently to the immune system by HLA-A*02 [47].

Regulatory T-cellsAlthough T-cells are considered integral to the onsetof vitiligo, it is acknowledged that anti-melanocyteantibodies could play an important role in maintainingthe disease [219]. CTLs alone are probably necessarybut not sufficient for perpetuating generalized vitiligo,as regulatory mechanisms that prevent autoaggressionby the immune system need to be overcome first [220].Regulatory T-cells play a key role in removing self-reactive T-cells that have escaped the process of clonaldeletion by the thymus [221]. Some regulatory T-cellshave been shown in vitro to suppress activated T-cells in acontact-dependent process, utilizing CD25 and forkheadand winged-helix family factor FOXP3 (forkhead box P3)[33]. Regulatory T-cells may be stimulated by immatureDCs in a Tol–DC regulatory loop [222]. DCs alsohave the capacity to augment melanocyte apoptosis incertain circumstances: melanocytes exposed to 4-TBPgenerate HSP-70 (heat shock protein-70), which althoughpartly protective, also induces TRAIL (TNF-relatedapoptosis-inducing ligand) receptor expression on theircell surface. This activates DC effector functions andresults in melanocyte death [223]. Tumour and self-directed CD8+ T-cells are augmented by CD4+ T-cellsand suppressed by regulatory T-cells [224]. RegulatoryT-cells can inhibit CD8+ and CD4+ T-cells and NKcells, via the cytokine TGF-β [219]; decreased TGF-βlevels in peripheral blood of vitiligo patients were, in fact,recently reported, pointing towards impaired regulatoryT-cell functioning [174]. Regulatory T-cells also controlconversion of naıve CD4+ T-cells (CD45RA+) intoTh-17 T-cells, producing IL-17. In the absence of regulat-ory T-cells, naıve CD4+ T-cells become Th-1 cells [225].IL-17 is proinflammatory, acting synergistically with IL-6, IL-1β and TNF-α. IL-17 is increased in several chronicautoimmune diseases including psoriasis and systemicsclerosis [226]. Increased levels of IL-17-producing T-cells were reported in one patient with vitiligo [218],but serum IL-17 was not elevated in another studyof vitiligo patients [174]. Dysregulation of CTLA-4can also predispose to autoreactive T-cells, as thismolecule decreases T-cell responsiveness and raises thethreshold for T-cell activation. Increased CD8+ self-reactive function has been reported in the presence of

defective CTLA-4, and CD4+ T-cells are also involved inthis process [227]. CTLA-4 polymorphisms predispose tovitiligo in patients who already have other autoimmuneconditions [146].

CONCLUSIONS

A complex interplay of genetic, immunological, envir-onmental and biochemical factors occurs in the leadup to generalized vitiligo. The hierarchy of events isstill unknown, but evidence points to a major rolefor ROS and T-cells, probably acting in concert todestroy or incapacitate melanocytes in a vicious cycleof cellular fatigue, cytokine imbalance, immune attackand apoptosis. Melanocytes produce melanin, which hasantioxidant properties, but its synthesis predisposes toROS generation, and melanocytes, therefore, operate ina potentially hostile environment. Melanocytes seem tobe uniquely fragile in people with a tendency to vitiligo.Numerous endogenous and exogenous sources of H2O2

and other ROS have been described in vitiligo, which aredeleterious to a variety of melanocytic cellular processes,particularly in the context of impaired cellular antioxidantdefence. The resultant protein and lipid damage couldbe sufficient, on its own, to initiate melanocyte failure,but another effect of oxidization could be to initiatemelanocyte failure and apoptosis leading to uptake byLangerhans cells or DCs. If these Langerhans cells orDCs become activated, they may trigger a melanocyte-reactive immune response that can eradicate melanocytesin the skin, leading to depigmentation. This immuneresponse principally involves cytotoxic T-cells. Failureof regulatory T-cell mechanisms allows the processto continue indefinitely, in keeping with the chronic,relentless course of generalized vitiligo. Further researchinto the pathogenesis of this common and distressingdisorder will offer the potential for better therapiesand might even facilitate effective strategies for primaryprevention in genetically susceptible individuals.

REFERENCES

1 Linthorst Homan, M. W., Spuls, P. I., de Korte, J., Bos, J.D., Sprangers, M. A. and van der Veen, M. A. (2009) Theburden of vitiligo: patient characteristics associated withquality of life. J. Am. Acad. Dermatol. 61, 411–420

2 Millington, G. W. and Levell, N. J. (2007) Vitiligo: thehistorical curse of depigmentation. Int. J. Dermatol. 46,990–995

3 Howitz, J., Brodthagen, H., Schwartz, M. and Thomsen,K. (1977) Prevalence of vitiligo. Epidemiological surveyon the Isle of Bornholm, Denmark. Arch. Dermatol. 113,47–52

4 Boisseau-Garsaud, A. M., Garsaud, P., Cales-Quist, D.,Helenon, R., Qeneherve, C. and Claire, R. C. (2000)Epidemiology of vitiligo in the French West Indies (Isle ofMartinique). Int. J. Dermatol. 39, 18–20

5 Handa, S. and Kaur, I. (1999) Vitiligo: clinical findings in1436 patients. J. Dermatol. 26, 653–657

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 16: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

114 S.J. Glassman

6 Le Poole, C. and Boissy, R. E. (1997) Vitiligo. Semin.Cutan. Med. Surg. 16, 3–14

7 Hann, S. K. and Lee, H. J. (1996) Segmental vitiligo:clinical findings in 208 patients. J. Am. Acad. Dermatol.35, 671–674

8 Salem, M. M., Shalbaf, M., Gibbons, N. C. J., Chavan, B.,Thornton, J. M. and Schallreuter, K. U. (2009) EnhancedDNA binding capacity on up-regulated epidermalwild-type p53 in vitiligo by H2O2-mediated oxidation: apossible repair mechanism for DNA damage. FASEB J.23, 3790–3807

9 Schallreuter, K. U., Wood, J. M., Pittelkow, M. R.,Gutlich, M., Lemke, R., Rodl, W., Swanson, N. N.,Hitzemann, K. and Ziegler, I. (1994) Regulation ofmelanin biosynthesis in the human epidermis bytetrahydrobiopterin. Science 263, 1444–1446

10 De Vijlder, H. C., Westerhof, W., Schreuder, G. M. Th.,De Lange, P. and Claas, F. H. (2004) Difference inpathogenesis between vitiligo vulgaris and halo neviassociated with vitiligo is supported by an HLAassociation study. Pigment Cell Res. 17, 270–274

11 Taıeb, A and Picardo, M. (2007) The definition andassessment of vitiligo: a consensus report of the VitiligoEuropean Task Force. Pigment Cell Res. 20, 27–35

12 Falabella, R. (1988) Treatment of localized vitiligo byautologous minigrafting. Arch. Dermatol. 124, 1649–1655

13 Forschner, T., Buchholtz, S. and Stockfleth, E. (2007)Current state of vitiligo therapy-evidence-based analysisof the literature. J. Dtsch. Dermatol. Ges. 5, 467–475

14 Westerhof, W and Nieuweboer-Krobotova, L. (1997)Treatment of vitiligo with UV-B radiation vs topicalpsoralen plus UV-A. Arch. Dermatol. 133, 1525–1528

15 Kanwar, A. J., Dogra, S., Parsad, D. and Kumar, B. (2005)Narrow-band UVB for the treatment of vitiligo: anemerging effective and well-tolerated therapy. Int. J.Dermatol. 44, 57–60

16 Angrisani, R. M., Azevedo, M. F., Pereira, L. D., Lopes,C. and Garcia, M. V. (2009) A study on otoacousticemissions and suppression effects in patients with vitiligo.Braz. J. Otorhinolaryngol. 75, 111–115

17 Sehgal, V. N. and Srivastava, G. (2007) Vitiligo:Compendium of clinico-epidemiological features. IndianJ. Dermatol. Venereol. Leprol. 73, 149–156

18 Halder, R. M. and Chappell, J. L. (2009) Vitiligo update.Semin. Cutan. Med. Surg. 28, 86–92

19 Taıeb, A. and Picardo, M. (2009) Vitiligo. N. Engl. J. Med.360, 10–169

20 Gawkrodger, D. J., Ormerod, A. D., Shaw, L., Mauri-Sole,I., Whitton, M. E., Watts, M. J., Anstey, A. V., Ingham, J.and Young, K. (2008) Guideline for the diagnosis andmanagement of vitiligo. Br. J. Dermatol. 159, 1051–1076

21 Fitzpatrick, T. B., Miyomato, M and Ishikawa, K. (1967)The evolution of concepts of melanin biology. Arch.Dermatol. 96, 305–323

22 Ivanova, K., Block, I., Das, P. and Gerzer, R. (2006) Roleof cyclic GMP signaling in the melanocyte response tohypergravity. Signal Transduct. 6, 406–413

23 Kollias, N., Sayre, R. M., Zeise, L. and Chedekel, M. R.(1991) Photoprotection by melanin. J. Photochem.Photobiol. B. 9, 135–160

24 Kobayashi, N., Nakagawa, A., Muramatsu, T.,Yamashina, Y., Shirai, T., Hashimoto, M. W., Ishigaki, Y.,Ohnishi, T. and Mori, T. (1998) Supranuclear melanincaps reduce ultraviolet induced DNA photoproducts inhuman epidermis. J. Invest. Dermatol. 110, 806–810

25 Le Poole, I. C., Mutis, T., van den Wijngaard, R. M.,Westerhof, W., Ottenhoff, T., de Vries, R. R. and Das, P.K. (1993) A novel, antigen-presenting function ofmelanocytes and its possible relationship tohypopigmentary disorders. J. Immunol. 151, 7284–7292

26 Le Poole, I. C. and Luiten, R. M. (2008) Autoimmuneetiology of generalized vitiligo. Curr. Dir. Autoimmun.10, 227–243

27 Das, P. K., van den Wijngaard, R. M., Wankowicz-Kalinska, A. and Le Poole, I. C. (2001) A symbioticconcept of autoimmunity and tumor immunity: lessonsfrom vitiligo. Trends Immunol. 22, 130–136

28 Gilhar, A., Paus, R. and Kalish, R. S. (2007) Lymphocytes,neuropeptides, and genes involved in alopecia areata.J. Clin. Invest. 117, 2019–2027

29 Anbar, T. S., Abdel-Raouf, H., Awad, S. S., Ragaie, M. H.and Abdel-Rahman, A. T. (2009) The hair folliclemelanocytes in vitiligo in relation to disease duration.J. Eur. Acad. Dermatol. Venereol. 23, 934–939

30 Kitamura, R., Tsukamoto, K., Harada, K., Shimizu, A.,Shimada, S., Kobayashi, T. and Imokawa, G. (2004)Mechanisms underlying the dysfunction of melanocytesin vitiligo epidermis: role of SCF/KIT proteininteractions and the downstream effector, MITF-M.J. Pathol. 202, 463–475

31 Passerson, T. and Ortonne, J. -P. (2005) Physiopathologyand genetics of vitiligo. J. Autoimmun. 25, 63–68

32 Dell’Anna, M. L. and Picardo, M. (2006) A review and anew hypothesis for non-immunological pathogeneticmechanisms in vitiligo. Pigment Cell Res. 19, 406–411

33 Westerhof, W. and d’Ischia, M. (2007) Vitiligo puzzle: thepieces fall in place. Pigment Cell Res. 20, 345–359

34 Schallreuter, K. U., Bahadoran, P., Picardo, M., Slominski,A., Elassiuty, E. H., Kemp, C., Giachino, C., Liu, J. B.,Luiten, R. M., Lambe, T. et al. (2008) Vitiligopathogenesis: autoimmune disease, genetic defect,excessive reactive oxygen species, calcium imbalance, orwhat else? Exp. Dermatol. 17, 139–160

35 Boissy, R. E. and Spritz, R. A. (2009) Frontiers andcontroversies in the pathobiology of vitiligo: separatingthe wheat from the chaff. Exp. Dermatol. 18, 583–585

36 Le Poole, I. C., Das, P. K., van den Wijngaard, R. M., Bos,J. D. and Westerhof, W. (1993) Review of theetiopathomechanism of vitiligo: a convergence theory.Exp. Dermatol. 2, 145–153

37 Gauthier, Y., Cario-Andre, M., Lepreux, S., Pain, C. andTaıeb, A. (2003) Melanocyte detachment after skinfriction in non lesional skin of patients with generalizedvitiligo. Br. J. Dermatol. 148, 95–101

38 Namazi, M. R. (2007) Neurogenic dysregulation,oxidative stress, autoimmunity, and melanocytorrhagy invitiligo: can they be interconnected? Pigment Cell Res. 20,360–363

39 Moretti, S., Spallanzani, A., Amato, L., Hautmann, G.,Gallerani, I., Fabiani, M. and Fabbri, P. (2002) Newinsights into the pathogenesis of vitiligo: imbalance ofepidermal cytokines at sites of lesions. Pigment Cell Res.15, 87–92

40 Moretti, S., Fabbri, P., Baroni, G., Berti, S., Bani, D.,Berti, E., Nassini, R., Lotti, T. and Massi, D. (2009)Keratinocyte dysfunction in vitiligo epidermis: cytokinemicroenvironment and correlation to keratinocyteapoptosis. Histol. Histopathol. 24, 849–857

41 Spritz, R. A. (2008) The genetics of generalized vitiligo.Curr. Opin. Autoimmun. 10, 244–257

42 Jin, Y., Riccardi, S. L., Gowan, K., Fain, P. R. and Spritz,R. A. (2009) Fine-mapping of vitiligo susceptibility locion chromosomes 7 and 9 and interactions with NLRP1(NALP1). J. Invest. Dermatol. 130, 774–783

43 Casp, C. B., She, J. -X. and McCormack, W. T. (2002)Genetic association of the Catalase gene (CAT) withvitiligo susceptibility. Pigment Cell Res. 15, 62–66

44 Onay, H., Pehlivan, M., Alper, S., Ozkinay, F. andPehlivan, S. (2007) Might there be a link betweenmannose-binding lectin and vitiligo. Eur. J. Dermatol. 17,146–148

45 Liu, L., Li, C., Gao, J., Li, K., Gao, L. and Gao, T. (2009)Genetic polymorphisms of glutathione S-transferase andrisk of vitiligo in the Chinese population. J. Invest.Dermatol. 129, 2646–2652

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 17: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

Vitiligo, ROS and T-cells 115

46 Ren, Y., Yang, S., Xu, S., Gao, M., Huang, W., Gao, T.,Fang, Q., Quan, C., Zhang, C., Sun, L. et al. (2009)Genetic variation of promoter sequence modulates XBP1expression and genetic risk for vitiligo. PLoS Genet. 5,e1000523

47 Jin, Y., Birlea, S. A., Fain, P. R., Gowan, K., Riccardi, S.L., Holland, P. J., Mailloux, B. S., Sufit, A. J. D., Hutton,S. M., Amadi-Myers, A. et al. (2010) Variant of TYR andautoimmunity susceptibility loci in generalized vitiligo.N. Eng. J. Med. 362, 1686–1697

48 Bishop, T. D., Demenais, F., Iles, M. M., Harland, M.,Taylor, J. C., Corda, E., Randerson-Moor, J., Aitken, J. F.,Avril, M. F., Azizi, E. et al. (2009) Genome-wideassociation study identifies three loci associated withmelanoma risk. Nat. Genet. 41, 920–925

49 Huang, C. L., Nordlund, J. J. and Boissy, R. (2002)Vitiligo: a manifestation of apoptosis? Am. J. Clin.Dermatol. 3, 301–308

50 Ahsan, H., Ali, A. and Ali, R. (2003) Oxygen free radicalsand systemic autoimmunity. Clin. Exp. Imunol. 131,398–404

51 Beckman, J. S. and Koppenol, W. H. (1996) Nitric oxide,superoxide, and peroxynitrite: the good, the bad, andugly. Am. J. Physiol. 271, C1424–C1437

52 Halliwell, B. and Gutteridge, J. M. (1984) Oxygentoxicity, oxygen radicals, transition metals and disease.Biochem. J. 219, 1–14

53 Marx, J. L. (1987) Oxygen free radicals linked to manydiseases. Science 235, 529–531

54 Imlay, J. A. and Linn, S. (1988) DNA damage and oxygenradical toxicity. Science 240, 1302–1309

55 Linn, S. (1998) DNA damage by iron and hydrogenperoxide in vitro and in vivo. Drug Metab. Rev. 30,313–326

56 Namazi, M. R. (2009) Cytochrome-P450 enzymes andautoimmunity: expansion of the relationship andintroduction of free radicals as the link. J. Autoimmun.Dis. 6, 4

57 Zhou, Z., Li, C. Y., Wang, T., Zhang, B. and Gao, T. W.(2009) Decreased methionine sulfoxide reductase Aexpression renders melanocytes more sensitive tooxidative stress: a possible cause for melanocyte loss invitiligo. Br. J. Dermatol. 161, 504–509

58 Ogawa, F., Sander, C. S., Hansel, A., Oehrl, W.,Kasperczyk, H., Elsner, P., Shimizu, K., Heinemann, S. H.and Thiele, J. J. (2006) The repair enzyme peptidemethionine-S-sulfoxide reductase is expressed in humanepidermis and upregulated by UVA radiation. J. Invest.Dermatol. 126, 947–949

59 Shigenaga, M. K., Hagen, T. M. and Ames, B. N. (1994)Oxidative damage and mitochondrial decay in aging.Proc. Natl. Acad. Sci. U. S. A. 91, 10771–10778

60 Briganti, S. and Picardo, M. (2003) Antioxidant activity,lipid peroxidation and skin diseases. What’s new. J. Eur.Acad. Dermatol. Venereol. 17, 663–669

61 Bickers, D. R. and Athar, M. (2006) Oxidative stress in thepathogenesis of skin disease. J. Invest. Dermatol. 126,2565–2575

62 Wood, J. M., Decker, H., Hartmann, H., Chavan, B.,Rokos, H., Spencer, J. D., Hasse, S., Thornton, M. J.,Shalbaf, M., Paus, R. and Schallreuter, K. U. (2009) Senilehair graying: H2O2-mediated oxidative stress affectshuman hair color by blunting methionine sulfoxide repair.FASEB J. 23, 2065–2075

63 Schallreuter, K. U., Moore, J., Wood, J. M., Beazley, W.D., Gaze, D. C., Tobin, D. J., Marshall, H. S., Panske, A.,Panzig, E. and Hibberts, N. A. (1999) In vivo and in vitroevidence for hydrogen peroxide (H2O2) accumulation inthe epidermis of patients with vitiligo and its successfulremoval by a UVB-activated pseudocatalase. J. Invest.Dermatol. Symp. Proc. 4, 91–96

64 Schallreuter, K. U., Wood, J. M. and Berger, J. (1991) Lowcatalase levels in the epidermis of patients with vitiligo. J.Invest. Dermatol. 97, 1081–1085

65 Moellmann, G., Klein-Angerer, S., Scollay, D. A.,Nordlund, J. J. and Lerner, A. B. (1982) Extracellulargranular material and degeneration of keratinocytes in thenormally pigmented epidermis of patients with vitiligo. J.Invest. Dermatol. 79, 321–330

66 Bhawan, J. and Bhutani, L. K. (1983) Keratinocytedamage in vitiligo. J. Cutan. Pathol. 10, 207–212

67 Boissy, R. E., Liu, Y. Y., Medrano, E. E. and Nordlund, J.J. (1991) Structural aberration of the rough endoplasmicreticulum and melanosome compartmentalization inlong-term cultures of melanocytes from vitiligo patients.J. Invest. Dermatol. 97, 395–404

68 Tobin, D. J., Swanson, N. N., Pittelkow, M. R., Peters, E.M. and Schallreuter, K. U. (2000) Melanocytes are notabsent in lesional skin of long duration vitiligo. J. Pathol.191, 407–416

69 Rokos, H., Wood, J. M., Hasse, S. and Schallreuter, K. U.(2008) Identification of epidermal L-tryptophan and itsoxidation products by in vivo FT-Raman spectroscopyfurther supports oxidative stress in patients with vitiligo.J. Raman. Spectrosc. 39, 1214–1218

70 Shalbaf, M., Gibbons, N. C., Wood, J. M., Maitland, D. J.,Rokos, H., Elwary, S. M., Marles, L. K. and Schallreuter,K. U. (2008) Presence of epidermal allantoin furthersupports oxidative stress in vitiligo. Exp. Dermatol. 17,761–770

71 Koca, R., Armutcu, F., Altinyazar, H. C. and Gurel, A.(2004) Oxidant-antioxidant enzymes and lipidperoxidation in generalized vitiligo. Clin. Exp. Dermatol.29, 406–409

72 Jain, D., Misra, R., Kumar, A. and Jaiswal, G. (2008)Levels of malondialdehyde and antioxidants in the bloodof patients with vitiligo of age group 11–20 years. IndianJ. Physiol. Pharmacol. 52, 297–301

73 Khan, R., Satyam, A., Gupta, S., Sharma, V. K. andSharma, A. (2009) Circulatory levels of antioxidants andlipid peroxidation in Indian patients with generalized andlocalized vitiligo. Arch. Dermatol. Res. 301, 731–737

74 Darr, D. and Fridovich, I. (1994) Free radicals incutaneous biology. J. Invest. Dermatol. 102, 671–675

75 Thannickal, V. J., Day, R. M., Klinz, S. G., Bastien, M. C.,Larios, J. M. and Fanburg, B. L. (2000) Ras-dependentand -independent regulation of reactive oxygen species bymitogenic growth factors and TGF-β1. FASEB J. 14,1741–1748

76 Schallreuter, K. U., Wood, J. M., Pittelkow, M. R.,Buttner, G., Swanson, N., Korner, C. and Ehrke, C. (1996)Increased monoamine oxidase A activity in the epidermisof patients with vitiligo. Arch. Dermatol. Res. 288, 14–18

77 Schallreuter, K. U. (1999) A review of recent advances onthe regulation of pigmentation in the human epidermis.Cell Mol. Biol. 45, 943–949

78 Schallreuter, K. U., Chiuchiarelli, G., Cemeli, E., Elwary,S. M., Gillbro, J. M., Spencer, J. D., Rokos, H., Panske,A., Chavan, B., Wood, J. M. and Anderson, D. (2006)Estrogens can contribute to hydrogen peroxidegeneration and quinone-mediated DNA damage inperipheral blood lymphocytes from patients with vitiligo.J. Invest. Dermatol. 126, 1036–1042

79 Schallreuter, K. U., Moore, J., Wood, J. M., Beazley, W.D., Peters, E. M., Marles, L. K., Behrens-Williams, S. C.,Dummer, R., Blau, N and Thony, B. (2001) EpidermalH2O2 accumulation alters tetrahydrobiopterin (6BH4)recycling in vitiligo: identification of a general mechanismin regulation of all 6BH4-dependent processes? J. Invest.Dermatol. 116, 167–174

80 Rokos, H., Beazley, W. D. and Schallreuter, K. U. (2002)Oxidative stress in vitiligo: photo-oxidation of pterinsproduces H2O2 and pterin-6-carboxylic acid. Biochem.Biophys. Res. Commun. 292, 805–811

81 Shaker, O. G. and El-Tahlawi, S. M. (2008) Is there arelationship between homocysteine and vitiligo? A pilotstudy. Br. J. Dermatol. 159, 720–724

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 18: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

116 S.J. Glassman

82 Picardo, M., Passi, S., Morrone, A., Grandinetti, M., DiCarlo, A. and Ippolito, F. (1994) Antioxidant status in theblood of patients with active vitiligo. Pigment Cell Res. 7,110–115

83 Yildirim, M., Baysal, V., Inaloz, H. S. and Can, M. (2004)The role of oxidants and antioxidants in generalizedvitiligo at tissue level. J. Eur. Acad. Dermatol. Venereol.18, 683–686

84 Hazneci, E., Karabulut, A. B., Ozturk, C., Batcioglu, K.,Dogan, G., Karaca, S. and Esrefoglu, M. (2005) Acomparative study of superoxide dismutase, catalase, andglutathione peroxidase activities and nitrate levels invitiligo patients. Int. J. Dermatol. 44, 636–640

85 Dammak, I., Boudaya, S., Ben Abdallah, F., Turki, H.,Attia, H. and Hentati, B. (2009) Antioxidant enzymes andlipid peroxidation at the tissue level in patients with stableand active vitiligo. Int. J. Dermatol. 48, 476–480

86 Miyamoto, L. and Taylor, J. S. (2000) Chemicalleukoderma. In Vitiligo: a Comprehensive Monograph onBasic and Clinical Science (Hann, S.-K. and Nordlund,J. J., eds), pp. 269–280, Blackwell Science Ltd. Oxford

87 Thorneby-Andresson, K., Sterner, O. and Hansson, C.(2000) Tyrosinase-mediated formation of a reactivequinone from the depigmenting agents, 4-tert-butylphenol and 4-tert-butylcatechol. Pigment Cell Res.13, 33–38

88 Jimbow, K., Obata, H., Pathak, M. A. and Fitzpatrick, T.B. (1974) Mechanism of depigmentation byhydroquinone. J. Invest. Dermatol. 62, 436–449

89 Cummings, M. P. and Nordlund, J. J. (1995) Chemicalleukoderma: fact or fancy. Am. J. Contact Dermatitis 6,122–127

90 Taıeb, A. (2000) Intrinsic and extrinsic pathomechanismsin vitiligo. Pigment Cell Res. 13, 41–47

91 Boissy, R. E. and Manga, P. (2004) On the etiology ofcontact/occupational vitiligo. Pigment Cell Res. 17,208–214

92 Manini, P., Napolitano, A., Westerhof, W., Riley, P. A. andd’Ischia, M. (2009) A reactive ortho-quinone generated bytyrosinase-catalyzed oxidation of the skin depigmentingagent monobenzone: self-coupling and thiol-conjugationreactions and possible implications for melanocytetoxicity. Chem. Res. Toxicol. 22, 1398–1405

93 Hariharan, V., Klarquist, J., Reust, M. J., Koshoffer, A.,McKee, M. D., Boissy, R. E. and Le Poole, I. C. (2010)Monobenzyl ether of hydroquinone and 4-tertiary butylphenol activate markedly different physiologicalresponses in melanocytes: relevance to skindepigmentation. J. Invest. Dermatol. 130, 211–220

94 Namazi, M. R. (2008) Proton pump inhibitors maytrigger vitiligo by rendering melanocytes prone toapoptosis. Br. J. Dermatol. 158, 844–845

95 Schallreuter, K. U. and Rokos, H. (2007) From the benchto the bedside: proton pump inhibitors can worsenvitiligo. Br. J. Dermatol. 156, 1371–1373

96 Morrone, A., Picardo, M., de Luca, C., Terminali, O.,Passi, S. and Ippolito, F. (1992) Catecholamines andvitiligo. Pigment Cell Res. 5, 65–69

97 Schallreuter, K. U., Wood, J. M., Pittelkow, M. R.,Swanson, N. N. and Steinkraus, V. (1993) Increased invitro expression of β2-adrenoceptors in differentiatinglesional keratinocytes of vitiligo patients. Arch. Dermatol.Res. 285, 216–220

98 Schallreuter, K. U., Wood, J. M., Ziegler, I., Lemke, K. R.,Pittelkow, M. R., Lindsey, N. J. and Gutlich, M. (1994)Defective tetrahydrobiopterin and catecholaminebiosynthesis in the depigmentation disorder vitiligo.Biochim. Biophys. Acta 1226, 181–192

99 Chavan, B., Beazley, W., Wood, J. M., Rokos, H.,Ichinose, H. and Schallreuter, K. U. (2009) H2O2increases de novo synthesis of (6R)-L-erythro-5,6,7,8-tetrahydrobiopterin via GTP cyclohydrolase I andits feedback regulatory protein in vitiligo. J. Inherit.Metab. Dis. 32, 86–94

100 Papadopoulos, L., Bor, R., Legg, C. and Hawk, J. L.(1998) Impact of life events on the onset of vitiligo inadults: preliminary evidence for a psychologicaldimension in aetiology. Clin. Exp. Dermatol. 23, 243–248

101 Thony, B., Auerbach, G. and Blau, N. (2000)Tetrahydrobiopterin biosynthesis, regeneration andfunctions. Biochem. J. 347, 1–16

102 Hasse, S., Gibbons, N. C., Rokos, H., Marles, L. K. andSchallreuter, K. U. (2004) Perturbed 6-tetrahydro-biopterin recycling via decreased dihydropteridinereductase in vitiligo: more evidence for H2O2 stress. J.Invest. Dermatol. 122, 307–313

103 Schallreuter, K. U., Zschiesche, M., Moore, J., Panske, A.,Hibberts, N. A., Herrmann, F. H., Metelmann, H. R. andSawatzki, J. (1998) In vivo evidence for compromisedphenylalanine metabolism in vitiligo. Biochem. Biophys.Res. Commun. 243, 395–399

104 Le Poole, C., Van den Wijngaard, R., Smit, N. P. M.,Oosting, J., Westerhof, W. and Pavel, S. (1994)Catechol-O-methyl transferase in vitiligo. Arch.Dermatol. Res. 286, 81–86

105 Namazi, M. R. (2005) Phenytoin as a novel anti-vitiligoweapon. J. Autoimmun. Dis. 2, 11–13

106 Gibbons, N. C., Wood, J. M., Rokos, H. and Schallreuter,K. U. (2006) Computer simulation of native epidermalstructures in the presence and absence of hydrogenperoxide (H2O2): potential and pitfalls. J. Invest.Dermatol. 126, 2576–2582

107 Schallreuter, K. U., Elwary, S. M., Gibbons, N. C., Rokos,H. and Wood, J. M. (2004) Activation/deactivation ofacetylcholinesterase by H2O2: more evidence foroxidative stress in vitiligo. Biochem. Biophys. Res.Commun. 315, 502–508

108 Schallreuter, K. U., Gibbons, N. C., Elwary, S. M., Parkin,S. M. and Wood, J. M. (2007) Calcium-activatedbutyrylcholinesterase in human skin protectsacetylcholinesterase against suicide inhibition byneurotoxic organophosphates. Biochem. Biophys. Res.Commun. 355, 1069–1074

109 Graham, A., Westerhof, W. and Thody, A. J. (1999) Theexpression of alpha-MSH by melanocytes is reduced invitiligo. Ann. N. Y. Acad. Sci. 885, 470–473

110 Spencer, J. D., Chavan, B., Marles, L. K., Kauser, S.,Rokos, H. and Schallreuter, K. U. (2005) A novelmechanism in control of human pigmentation byβ-melanocyte-stimulating hormone and7-tetrahydrobiopterin. J. Endocrinol. 187, 293–302

111 Spencer, J. D., Gibbons, N. C., Rokos, H., Peters, E. M.,Wood, J. M. and Schallreuter, K. U. (2007) Oxidativestress via hydrogen peroxide affects proopiomelanocortinpeptides directly in the epidermis of patients with vitiligo.J. Invest. Dermatol. 127, 411–420

112 Rokos, H., Moore, J., Hasse, S., Gillbro, J. M., Wood,J. M. and Schallreuter, K. U. (2004) In vivo fluorescenceexcitation spectroscopy and in vivo Fourier-transformRaman spectroscopy in human skin: evidence of H2O2oxidation of epidermal albumin in patients with vitiligo.J. Raman Spectrosc. 35, 125–130

113 Schallreuter, K. U., Gibbons, N. C., Zothner, C., AbouElloof, M. M. and Wood, J. M. (2007) Hydrogenperoxide-mediated oxidative stress disrupts calciumbinding on calmodulin: more evidence for oxidative stressin vitiligo. Biochem. Biophys. Res. Commun. 360, 70–75

114 Spencer, J. D., Gibbons, N. C. J., Bohm, M. andSchallreuter, K. U. (2008) The Ca2+-binding capacity ofepidermal furin is disrupted by H2O2-medaited oxidationin vitiligo. Endocrinology 149, 1638–1645

115 Hachiya, A., Kobayashi, A., Ohuchi, A., Takema, Y. andImokawa, G. (2001) The paracrine role of stem cellfactor/c-kit signaling in the activation of humanmelanocytes in ultraviolet B-induced pigmentation.J. Invest. Dermatol. 116, 578–586

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 19: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

Vitiligo, ROS and T-cells 117

116 Imokawa, G., Yada, Y. and Kimura, M. (1996) Signalingmechanisms of endothelin-induced mitogenesis andmelanogenesis in human melanocytes. Biochem. J. 314,305–312

117 Jimenez-Cervantes, C., Martinez-Esparza, M., Perez, C.,Daum, N., Solano, F. and Garcia-Borron, J. C. (2001)Inhibition of melanogenesis in response to oxidativestress: transient downregulation of melanocytedifferentiation markers and possible involvement ofmicrophthalmia transcription factor. J. Cell Sci. 114,2335–2344

118 Gauthier, Y. (1996) The importance of Koebner’sphenomenon in the induction of vitiligo lesions. Eur. J.Dermatol. 5, 704–708

119 Jezek, P and Hlavata, L. (2005) Mitochondria inhomeostasis of reactive oxygen species in cell, tissues andorganism. Int. J. Biochem. Cell Biol. 37, 2478–2503

120 Birlea, S. A., Costin, G. -E. and Norris, D. A. (2009) Newinsights on therapy with vitamin D analogs targeting theintracellular pathways that control repigmentation inhuman vitiligo. Medicinal Res. Rev. 29, 514–546

121 Wood, J. M., Chavan, B., Hafeez, I. and Schallreuter, K.U. (2004) Regulation of tyrosinase by tetrahydro-pteridines and H2O2. Biochem. Biophys. Res. Commun.325, 1412–1417

122 Jimbow, K., Chen, H., Park, J. -S. and Thomas, P. D.(2001) Increased sensitivity of melanocytes to oxidativestress and abnormal expression of tyrosinase-relatedprotein in vitiligo. Br. J. Dermatol. 144, 55–65

123 Sravani, P. V., Babu, N. K., Gopal, K. V., Rao, A. R.,Moorthy, B. and Rao, T. R. (2009) Determination ofoxidative stress in vitiligo by measuring superoxidedismutase and catalase levels in vitiliginous andnon-vitiliginous skin. Indian J. Dermatol. Venereol.Leprol. 75, 268–271

124 Wood, J. M., Gibbons, N. C., Chavan, B. and Schallreuter,K. U. (2008) Computer simulation of heterogeneoussingle nucleotide polymorphisms in the catalase geneindicates structural changes in the enzyme active site,NADPH-binding and tetramerization domains: a geneticpredisposition for an altered catalase in patients withvitiligo? Exp. Dermatol. 17, 366–371

125 Schallreuter, K. U., Rubsam, K., Gibbons, N. C.,Maitland, D. J., Chavan, B., Zothner, C., Rokos, H. andWood, J. M. (2008) Methionine sulfoxide reductases Aand B are deactivated by hydrogen peroxide (H2O2) inthe epidermis of patients with vitiligo. J. Invest. Dermatol.128, 808–815

126 Passi, S., Grandinetti, M., Maggio, F., Stancato, A. and DeLuca, C. (1998) Epidermal oxidative stress in vitiligo.Pigment Cell Res. 11, 81–85

127 Beazley, W. D., Gaze, D., Panske, A., Panzig, E. andSchallreuter, K. U. (1999) Serum selenium levels andblood glutathione peroxidase activities in vitiligo. Br. J.Dermatol. 141, 301–303

128 Boisseau-Garsaud, A. -M., Garsaud, P., Lejoly-Boisseau,H., Robert, M., Quist, D. and Arveiler, B. (2002) Increasein total blood antioxidant status and selenium levels inblack patients with active vitiligo. Int. J. Dermatol. 41,640–642

129 Slominski, A., Paus, R. and Bomirski, A. (1989)Hypothesis: possible role for the melatonin receptor invitiligo: discussion paper. J. R. Soc. Med. 82, 539–541

130 Reiter, R. J., Tan, D. X., Terron, M. P., Flores, L. J. andCzarnocki, Z. (2007) Melatonin and its metabolites: newfindings regarding their production and their radicalscavenging actions. Acta Biochim. Pol. 54, 1–9

131 Simon, H. U., Haj-Yehia, A. and Levi-Schaffer, F. (2000)Role of reactive oxygen species (ROS) in apoptosisinduction. Apoptosis 5, 415–418

132 Lee, Y. J., Chen, J. C., Amoscato, A. A., Bennouna, J.,Spitz, D. R., Suntharalingham, M. and Rhee, J. G. (2001)Protective role of Bcl-2 in metabolic oxidativestress-induced cell death. J. Cell Sci. 114, 677–684

133 Van den Wijngaard, R. M., Aten, J., Scheepmaker, A., LePoole, I. C., Tigges, A. J., Westerhof, W. and Das, P. K.(2000) Expression and modulation of apoptosisregulatory molecules in human melanocytes: significancein vitiligo. Br. J. Dermatol. 143, 573–581

134 Schallreuter, K. U., Kruger, C., Wurfel, B. A., Panske, A.and Wood, J. M. (2008) From basic research to thebedside: efficacy of topical treatment with pseudocatalasePC-KUS in 71 children with vitiligo. Int. J. Dermatol. 47,743–753

135 Schallreuter, K. U., Moore, J., Behrens-Williams, S.,Panske, A. and Harari, M. (2002) Rapid initiation ofrepigmentation in vitiligo with Dead Sea climatotherapyin combination with pseudocatalase (PC-KUS). Int. J.Dermatol. 41, 482–487

136 Bakis-Petsoglou, S., Le guay, J. L. and Wittal, R. (2009) Arandomized double-blinded placebo-controlled trial ofpseudocatalase cream and narrowband ultraviolet B in thetreatment of vitiligo. Br. J. Dermatol. 161, 721–722

137 Sanclemente, G., Garcia, J. J., Zuleta, J. J., Diehl, C.,Correa, C. and Falabella, R. (2008) A double-blind,randomized trial of 005 % betamethasone vs topicalcatalase/dismutase superoxide in vitiligo. J. Eur. Acad.Dermatol. Venereol. 22, 1359–1364

138 Elgoweini, M. and Nour El Din, N. (2009) Response ofvitiligo to narrowband ultraviolet B and oral antioxidants.J. Clin. Pharmacol. 49, 852–855

139 Ongenae, K., Van Geel, N. and Naeyaert, J. -M. (2003)Evidence for an autoimmune pathogenesis of vitiligo.Pigment Cell Res. 16, 90–100

140 Schallreuter, K. U., Lemke, R., Brandt, O., Schwartz, R.,Westhofen, M., Montz, R. and Berger, J. (1994) Vitiligoand other diseases: coexistence or true association.Dermatology 188, 269–275

141 Alkhateeb, A., Fain, P. R., Thody, A., Bennett, D. C. andSpritz, R. A. (2003) Epidemiology of vitiligo andassociated autoimmune diseases in Caucasian probandsand their relatives. Pigment Cell Res. 16, 208–214

142 Jandus, C., Bioley, G., Rivals, J. -P., Dudler, J., Speiser, D.and Romero, P. (2008) Increased numbers of circulatingpolyfunctional Th17 memory cells in patients withseronegative spondyloarthritides. Arthritis Rheum. 58,2307–2317

143 Foley, L. M., Lowe, N. J., Miseloff, E. and Tiwari, J. L.(1983) Association of HLA-DR4 with vitiligo. J. Am.Acad. Dermatol. 38, 39–40

144 Casp, C. B., She, J. X. and McCormack, W. T. (2003)Genes of the LMP/TAP cluster are associated with thehuman autoimmune disease vitiligo. Genes Immun. 4,492–499

145 Venneker, G. T., Westerhof, W., de Vries, I. J., Drayer, N.M., Wolthers, B. G., de Waal, L. P., Bos, J. D. and Asghar,S. S. (1992) Molecular heterogeneity of the fourthcomponent of complement (C4) and its genes in vitiligo. J.Invest. Dermatol. 99, 853–858

146 Blomhoff, A., Kemp, E. H., Gawkrodger, D. J., Weetman,A. P., Husebye, E. S., Akselsen, H. E., Lie, B. A. andUndlien, D. E. (2005) CTLA-4 polymorphisms areassociated with vitiligo in patients with concomitantautoimmune diseases. Pigment Cell Res. 18, 55–58

147 Collins, S. M., Dominguez, M., Ilmarinen, T., Costigan, C.and Irvine, A. D. (2006) Dermatological manifestations ofautoimmune polyendocrinopathy-candidiasis-ectodermaldystrophy syndrome. Br. J. Dermatol. 154, 1088–1093

148 Canton, I., Akhtar, S., Gavalas, N. G., Gawkrodger, D. J.,Blomhoff, A., Watson, P. F., Weetman, A. P. and Kemp, E.H. (2005) A single nucleotide polymorphism in the geneencoding lymphoid protein tyrosine phosphatase(PTPN22) confers susceptibility to generalized vitiligo.Genes Immun. 6, 584–587

149 Spritz, R. A., Gowan, K., Bennett, D. C. and Fain, P. R.(2004) Novel susceptibility loci on chromosomes 7 (AIS2)and 8 (AIS3), confirmation of SLEV1 on chromosome 17,and their roles in an autoimmune diathesis. Am. J. Hum.Genet. 74, 188–191

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 20: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

118 S.J. Glassman

150 Austin, L. M. and Boissy, R. E. (1995) Mammaliantyrosinase-related protein-1 is recognized byautoantibodies from vitiliginous Smyth chickens. Am. J.Pathol. 146, 1529–1541

151 Farrokhi, S., Hojjat-Farsangi, M., Noohpisheh, M. K.,Tahmasbi, R. and Rezaei, N. (2005) Assessment of theimmune system in 55 Iranian patients with vitiligo. J. Eur.Acad. Dermatol. Venereol. 19, 706–711

152 Harning, R., Cui, J. and Bystryn, J. -C. (1991) Relationbetween the incidence and level of pigment cell antibodiesand disease activity in vitiligo. J. Invest. Dermatol. 97,1078–1080

153 Oyarbide-Valencia, K., van den Boorn, J. G., Denman, C.J., Li, M., Carlson, J. M., Hernandez, C., Nishimura, M.I., Das, P. K., Luiten, R. M. and Le Poole, I. C. (2006)Therapeutic implications of autoimmune vitiligo cells.Autoimmun Rev. 5, 486–492

154 Pichler, R., Sfetsos, K., Badics, B., Gutenbrunner, S., Berg,J. and Aubock, J. (2009) Lymphocyte imbalance in vitiligopatients indicated by elevated CD4+/CD8+ T-cell ratio.Wien. Med. Wochenschr. 159, 337–341

155 Mellouli, F., Ksouri, H., Dhouib, N., Torjmen, L.,Abdelkefi, A., Ladeb, S., Ben Othman, T., Hmida, S., BenHassen, A. and Bejaoui, M. (2009) Possible transfer ofvitiligo by allogeneic bone marrow transplantation: a casereport. Pediatr. Transplant. 13, 1058–1061

156 Ortonne, J. P. and Alario, A. (1978) T and B lymphocytesin vitiligo. Arch. Dermatol. 261, 147–151

157 Grimes, P. E., Ghoneum, M., Stockton, T., Payne, C.,Kelly, A. P. and Alfred, L. (1986) T cell profiles in vitiligo.J. Am. Acad. Dermatol. 14, 196–201

158 Soubiran, P., Benzaken, S., Bellet, C., Lacour, J. P. andOrtonne, J. P. (1985) Vitiligo: peripheral T-cell subsetimbalance as defined by monoclonal antibodies. Br J.Dermatol. 113, 124–127

159 Halder, R. M., Walters, C. S., Johnson, B. A., Chakrabarti,S. G. and Kenney, Jr, J. A. (1986) Aberrations in Tlymphocytes and natural killer cells in vitiligo: a flowcytometric study. J. Am. Acad. Dermatol. 14, 733–737

160 Hann, S. K., Park, Y. K., Chung, K. Y., Kim, H. I., Im, S.and Won, J. H. (1993) Peripheral blood lymphocyteimbalance in Koreans with active vitiligo. Int. J.Dermatol. 32, 286–289

161 Abdel-Naser, M. B., Ludwig, W. D., Gollnick, H. andOrfanos, C. E. (1992) Nonsegmental vitiligo: decrease ofthe CD45RA+ T-cell subset and evidence for peripheralT-cell activation. Int. J. Dermatol. 31, 321–326

162 Mahmoud, F., Abdul, H., Haines, D., Al-Saleh, C.,Khajeji, M. and Whaley, K. (2002) Decreased totalnumbers of peripheral blood lymphocytes with elevatedpercentages of CD4+, CD45RO+ and CD4+ CD25+T-helper cells in non-segmental vitiligo. J. Dermatol. 29,68–73

163 Basak, P. Y., Adiloglu, A. K., Koc, I. G., Tas, T. andAkkaya, V. B. (2008) Evaluation of activatory andinhibitory natural killer cell receptors in non-segmentalvitiligo: a flow cytometric study. J. Eur. Acad. Dermatol.Venereol. 22, 970–976

164 Yu, H. S., Chang, K. L., Yu, C. L., Wu, M. T., Wu, C. S.and Wu, C. S. (1997) Alterations in IL-6, IL-8, GM-CSF,TNF-α and IFN-γ release by peripheral mononuclearcells in patients with active vitiligo. J. Invest. Dermatol.108, 527–529

165 Tu, C. X., Fu, H. W. and Lin, X. R. (1999) Levels ofsoluble interleukin-2 receptor in the sera and skin tissuefluids of patients with vitiligo. J. Dermatol. Sci. 21, 59–62

166 Moretti, S., Spallanzani, A., Amato, L., Hautmann, G.,Gallerani, I., Fabiani, M. and Fabbri, P. (2002) Newinsights into the pathogenesis of vitiligo: imbalance ofepidermal cytokines at sites of lesions. Pigment Cell Res.15, 87–92

167 Tu, C. X., Gu, J. S. and Lin, X. R. (2003) Increasedinterleukin-6 and granulocyte macrophage colonystimulating factor levels in the sera of patients withnon-segmental vitiligo. J. Dermatol. Sci. 31, 73–78

168 Franczuk, A., Szepietowski, J. C. and Noworolska, A.(2004) Serum concentrations of interleukin-2 solublereceptor (IL-2 sR) in patients with vitiligo: relationshipwith type and extent of the disease. ActaDermatovenereol. Croat. 12, 71–76

169 Grimes, P. E., Morris, R., Avaniss-Aghajani, E., Soriano,T., Meraz, M. and Metzger, A. (2004) Topical tacrolimustherapy for vitiligo: therapeutic responses and skinmessenger RNA expression of proinflammatorycytokines. J. Am. Acad. Dermatol. 51, 52–61

170 Zailaie, M. Z. (2005) Decreased proinflammatorycytokine production by peripheral blood mononuclearcells from vitiligo patients following aspirin treatment.Saudi Med. J. 26, 799–805

171 Birol, A., Kisa, U., Kurtipek, G. S., Kara, F., Kocak, M.,Erkek, E. and Caglayan, O. (2006) Increased tumornecrosis factor α (TNF-α) and interleukin 1α (IL1-α)levels in the lesional skin of patients with nonsegmentalvitiligo. Int. J. Dermatol. 45, 992–993

172 Mashiah, J and Brenner, S. (2007) Possible mechanisms inthe induction of vitiligo-like hypopigmentation by topicalimiquimod. Clin. Exp. Dermatol. 33, 74–76

173 Taher, Z. A., Lauzon, G., Maguiness, S. and Dytoc, M. T.(2009) Analysis of interleukin-10 levels in lesions ofvitiligo following treatment with topical tacrolimus. Br. J.Dermatol. 161, 654–659

174 Basak, P. Y., Adiloglou, A. K., Ceyhan, A. M., Tas, T. andAkkaya, V. B. (2009) The role of helper and regulatory Tcells in the pathogenesis of vitiligo. J. Am. Acad.Dermatol. 60, 256–260

175 Gokhale, B. B. and Mehta, L. N. (1983) Histopathologyof vitiliginous skin. Int. J. Dermatol. 22, 477–480

176 Hann, S. K., Park, Y. K., Lee, K. G., Choi, E. H. and Im,S. (1992) Epidermal changes in active vitiligo. J. Dermatol.19, 217–222

177 Galadari, E., Mehregan, A. H. and Hashimoto, K. (1993)Ultrastructural study of vitiligo. Int. J. Dermatol. 32,269–271

178 Abdel-Naser, M. B., Kruger-Krasagakes, S., Krasagakis,K., Gollnick, H., Abdel-Fattah, A. and Orfanos, C. E.(1994) Further evidence for involvement of both cellmediated and humoral immunity in generalized vitiligo.Pigment Cell Res. 7, 1–8

179 Shiohara, T., Noriko Moria, B. S. and Nagashima, M.(1988) The lichenoid tissue reaction. Int. J. Dermatol. 27,365–374

180 Gross, A., Tapia, F. J., Mosca, W., Perez, R. M., Briceno,L., Henriquez, J. J. and Convit, J. (1987) Mononuclear cellsubpopulations and infiltrating lymphocytes in erythemadyschromicum perstans and vitiligo. Histol. Histopathol.2, 277–283

181 Al Badri, A. M., Foulis, A. K., Todd, P. M., Gariouch, J. J.,Stewart, D. G., Gracie, J. A. and Goudie, R. B. (1993)Abnormal expression of MHC class II and ICAM-1 bymelanocytes in vitiligo. J. Pathol. 169, 203–206

182 Badri, A. M., Todd, P. M., Garioch, J. J., Gudgeon, J. E.,Stewart, D. G. and Goudie, R. B. (1993) Animmunohistological study of cutaneous lymphocytes invitiligo. J. Pathol. 170, 149–155

183 van den Wijngaard, R., Wankowicz-Kalinska, A., LePoole, C., Tigges, B., Westerhof, W. and Das, P. (2000)Local immune response in skin of generalized vitiligopatients. Destruction of melanocytes is associated withthe prominent presence of CLA+ T cells at theperilesional site. Lab. Invest. 80, 1299–1309

184 Rivoltini, L., Radrizzani, M., Accornero, P., Squarcina, P.,Chiodoni, C., Mazzocchi, A., Castelli, C., Tarsini, P.,Viggiano, V., Belli, F. et al. (1998) Human melanoma-reactive CD4+ and CD8+ CTL clones resist Fasligand-induced apoptosis and use Fas/Fas ligand-independent mechanisms for tumor killing. J. Immunol.161, 1220–1230

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 21: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

Vitiligo, ROS and T-cells 119

185 Moretti, S., Fabbri, P., Baroni, G., Berti, S., Bani, D.,Berti, E., Nassini, R., Lotti, T. and Massi, D. (2009)Keratinocyte dysfunction in vitiligo epidermis: cytokinemicroenvironment and correlation to keratinocyteapoptosis. Histol. Histopathol. 24, 849–857

186 Arnold, R., Seifert, M., Asadulla, K. and Volk, H. D.(1999) Crosstalk between keratinocytes and Tlymphocytes via Fas/Fas ligand interaction: modulationby cytokines. J. Immunol. 162, 7140–7147

187 Michaelsson, G. (1968) Vitiligo with raised borders. ActaDerm. Venereol. 48, 158–161

188 Attili, V. R. and Attili, S. K. (2008) Lichenoidinflammation in vitiligo – a clinical and histopathologicreview of 210 cases. Int. J. Dermatol. 47, 663–669

189 Le Poole, I. C., van den Wijngaard, R. M., Westerhof, W.and Das, P. K. (1996) Presence of T cells and macrophagesin inflammatory vitiligo skin parallels melanocytedisappearance. Am. J. Pathol. 148, 1219–1228

190 Yagi, H., Tokura, Y., Furukawa, F. and Takigawa, M.(1997) Vitiligo with raised inflammatory borders:involvement of T cell immunity and keratinocytesexpressing MHC class II and ICAM-1 molecules. Eur. J.Dermatol. 7, 19–22

191 Wankowicz-Kalinska, A., van den Wijngaard, R. M.,Tigges, B. J., Westerhof, W., Ogg, G. S., Cerundolo, V.,Storkus, W. J. and Das, P. K. (2003) Immunopolarizationof CD4+ and CD8+ T cells to Type-1-like is associatedwith melanocyte loss in human vitiligo. Lab. Invest. 83,683–695

192 Shresta, S., Smyth, Jr, J. R. and Erf, G. F. (1997) Profiles ofpulp infiltrating lymphocytes at various times throughoutfeather regeneration in Smyth line chickens with vitiligo.Autoimmunity 25, 193–201

193 Del Prete, G. F., Vercelli, D., Tiri, A., Maggi, E., Mariotti,S., Pinchera, A., Ricci, M. and Romagnani, S. (1986) Invivo activated cytotoxic T cells in the thyroid infiltrate ofpatients with Hashimoto’s thyroiditis. Clin. Exp.Immunol. 65, 140–147

194 Itoh, N., Hanafusa, T., Miyazaki, A., Miyagawa, J.,Yamagata, K., Yamamoto, K., Waguri, M., Imagawa, A.,Tamura, S., Inada, M. et al. (1993) Mononuclear cellinfiltration and its relation to the expression of majorhistocompatibility complex antigens and adhesionmolecules in pancreas biopsy specimens from newlydiagnosed insulin-dependent diabetes mellitus patients. J.Clin. Invest. 92, 2313–2322

195 Le Poole, I. C., Mutis, T., van den Wijngaard, R. M.,Westerhof, W., Ottenhoff, T., de Vries, R. R. and Das, P.K. (1993) A novel, antigen-presenting function ofmelanocytes and its possible relationship tohypopigmentary disorders. J. Immunol. 151, 7284–7292

196 Comabella, M., Balashov, K., Issazadeh, S., Smith, D.,Weiner, H. L. and Khoury, S. J. (1998) Elevatedinterleukin-12 in progressive multiple sclerosis correlateswith disease activity and is normalized by pulsecyclophosphamide therapy. J. Clin. Invest. 102, 671–678

197 Nordlund, J. J., Kirkwood, J. M., Forget, B. M., Milton,G., Albert, D. M. and Lerner, A. B. (1983) Vitiligo inpatients with metastatic melanoma: a good prognosticsign. J. Am. Acad. Dermatol. 9, 689–696

198 Bystryn, J. C., Rigel, D., Friedman, R. J. and Kopf, A.(1987) Prognostic significance of hypopigmentation inmalignant melanoma. Arch. Dermatol. 123, 1053–1055

199 Cui, J. and Bystryn, J. C. (1995) Melanoma and vitiligoare associated with antibody responses to similar antigenson pigment cells. Arch. Dermatol. 131, 314–318

200 Cavallari, V., Cannavo, S. P., Ussia, A. F., Moretti, G. andAlbanese, A. (1996) Vitiligo associated with metastaticmalignant melanoma. Int. J. Dermatol. 35, 738–740

201 Rosenberg, S. A. and White, D. E. (1996) Vitiligo inpatients with melanoma: Normal tissue antigens can betargets for cancer immunotherapy. J. Immunother.Emphasis Tumor Immunol. 19, 81–84

202 Nestle, F. O., Alijagic, S., Gilliet, M., Sun, Y., Grabbe, S.,Dummer, R., Burg, G. and Schadendorf, D. (1998)Vaccination of melanoma patients with peptide- ortumor-lysate pulsed dendritic cells. Nat. Med. 4, 328–332

203 Yee, C., Thompson, J. A., Roche, P., Byrd, D. R., Lee, P.P., Piepkan, M., Kenyon, K., Davis, M. M., Riddell, S. R.and Greenberg, P. D. (2000) Melanocyte destruction afterantigen-specific immunotherapy of melanoma: directevidence of T cell-mediated vitiligo. J. Exp. Med. 192,1637–1644

204 Dudley, M. E., Wunderlich, J. R., Robbins, P. F., Yang, J.C., Hwu, P., Schwartzentruber, D. J., Topalian, S. L.,Sherry, R., Restifo, N. P., Hubicki, A. M. et al. (2002)Cancer regression and autoimmunity in patients afterclonal repopulation with antitumor lymphocytes. Science298, 850–854

205 Phan, G. Q., Yang, J. C., Sherry, R. M., Hwu, P., Topalian,S. L., Schwartzentruber, D. J., Restifo, N. P., Haworth, L.R., Seipp, C. A., Freezer, L. J. et al. (2003) Cancerregression and autoimmunity induced by cytotoxic Tlymphocyte-associated antigen 4 blockade in patientswith metastatic melanoma. Proc. Natl. Acad. Sci. U.S.A.100, 8372–8377

206 Luiten, R. M., Kueter, E. W., Mooi, W., Gallee, M. P.,Rankin, E. M., Gerritsen, W. R., Clift, S. M., Nooijen, W.J., Weder, P., van de Kasteele, W. F. et al. (2005)Immunogenicity, including vitiligo, and feasibility ofvaccination with autologous GM-CSF-transduced tumorcells in metastatic melanoma patients. J. Clin. Oncol. 23,8978–8991

207 Anichini, A., Maccalli, C., Mortarini, R., Salvi, S.,Mazzocchi, A., Squarcina, P., Herlyn, M. and Parmiani,G. (1993) Melanoma cells and normal melanocytes shareantigens recognized by HLA-A2-restricted cytotoxic Tcell clones from melanoma patients. J. Exp. Med. 177,989–998

208 Kawakami, Y., Dang, N., Wang, X., Tupesis, J., Robbins,P. F., Wang, R. F., Wunderlich, J. R., Yannelli, J. R. andRosenberg, S. A. (2000) Recognition of shared melanomaantigens in association with major HLA-A alleles bytumor infiltrating T lymphocytes from 123 patients withmelanoma. J. Immunother. 23, 17–27

209 Becker, J. C., Guldberg, P., Zeuthen, J., Brecker, E. B. andStraten, P. T. (1999) Accumulation of identical T cells inmelanoma and vitiligo-like leukoderma. J. Invest.Dermatol. 113, 1033–1038

210 Pittet, M. J., Valmori, D., Dunbar, P. R., Speiser, D. E.,Lienard, D., Lejeune, F., Fleischhauer, K., Cerundolo, V.,Cerottini, J. C. and Romero, P. (1999) High frequencies ofnaıve Melan-A/MART-1-specific CD8+ T cells in a largeproportion of human histocompatibility leukocyteantigen (HLA)-A2 individuals. J. Exp. Med. 190, 705–715

211 Jager, E., Maeurer, M., Hohn, H., Karbach, J., Jager, D.,Zidianakis, Z., Bakhshandeh-Bath, A., Orth, J., Neukirch,C., Necker, A. et al. (2000) Clonal expansion of MelanA-specific cytotoxic T lymphocytes in a melanomapatient responding to continued immunization withmelanoma-associated peptides. Int. J. Cancer 86, 538–547

212 Altman, J. D., Moss, P. A., Goulder, P. J. R., Barouch, D.H., McHeyzer-Williams, M. G., Bell, J. I., McMichael, A.J. and Davis, M. M. (1996) Phenotypic analysis ofantigen-specific T lymphocytes. Science 274, 94–96

213 Ogg, G. S., Dunbar, P. R., Romero, P., Chen, J. andCerundolo, V. (1998) High frequency of skin-homing melanocyte-specific cytotoxic T lymphocytes inautoimmune vitiligo. J. Exp. Med. 188, 1203–1208

214 Lang, K. S., Caroli, C. C., Muhm, A., Wernet, D., Moris,A., Schittek, B., Knauss-Scherwitz, E., Stevanovic, S.,Rammensee, H. G. and Garbe, C. (2001) HLA-A2restricted, melanocyte-specific CD8+ T lymphocytesdetected in vitiligo patients are related to disease activityand are predominantly directed against MelanA/MART1.J. Invest. Dermatol. 116, 891–897

C© The Authors Journal compilation C© 2011 Biochemical Society

Page 22: Vitiligo, reactive oxygen species and T-cells...Vitiligo, reactive oxygen species and T-cells Steven J. GLASSMAN Division of Dermatology, Department of Medicine, University of Ottawa,

120 S.J. Glassman

215 Mandelcorn-Monson, R. L, Shear, N. H., Yau, E.,Sambhara, S., Barber, B. H., Spaner, D. and DeBenedette,M. A. (2003) Cytotoxic T lymphocyte reactivity to gp100,MelanA/MART-1, and tyrosinase, in HLA-A2-positivevitiligo patients. J. Invest. Dermatol. 121, 550–556

216 Palermo, B., Garbelli, S., Mantovani, S., Scoccia, E., DaPrada, G. A., Bernabei, P., Avanzini, M. A., Brazzelli, V.,Borroni, G. and Giachino, C. (2005) Qualitativedifference between the cytotoxic T lymphocyte responsesto melanocyte antigens in melanoma and vitiligo. Eur. J.Immunol. 35, 3153–3162

217 Garbelli, S., Mantovani, S., Palermo, B. and Giachino, C.(2005) Melanocyte-specific, cytotoxic T cell responses invitiligo: the effective variant of melanoma immunity?Pigment Cell Res. 18, 234–242

218 Van den Boorn, J. G., Konijnenberg, D., Dellemijn, T. A.,Wietze van der Veen, J. P., Bos, J. D., Melief, C. J.,Vyth-Dreese, F. A. and Luiten, R. M. (2009)Autoimmune destruction of skin melanocytes byperilesional T cells from vitiligo patients. J. Invest.Dermatol. 129, 2220–2232

219 Rezaei, N., Gavalas, N. G., Weetman, A. P. and Kemp, E.H. (2007) Autoimmunity as an aetiological factor invitiligo. J. Eur. Acad. Dermatol. Venereol. 21, 865–876

220 Degaco, C., Duftner, C., Grubeck-Loebenstein, B. andSchirmer, M. (2006) Imbalance of regulatory T cells inhuman autoimmune diseases. Immunology 117, 289–300

221 Sakaguchi, S., Sakaguchi, N., Shimizu, J., Yamazaki, S.,Sakihama, T., Itoh, M., Kuniyasu, Y., Nomura, T., Toda,M. and Takahashi, T. (2001) Immunologic tolerancemaintained by CD25+ CD4+ regulatory T cells: theircommon role in controlling autoimmunity, tumorimmunity, and transplantation tolerance. Immunol. Rev.182, 18–32

222 Min, W. P., Zhou, D., Ichim, T. E., Strejan, G. H., Xia, X.,Yang, J., Huang, X., Garcia, B., White, D., Dutarte, P.et al. (2003) Inhibitory feedback loop between tolerogenicdendritic cells and regulatory T cells in transplanttolerance. J. Immunol. 170, 1304–1312

223 Kroll, T. M., Bommiasamy, H., Boissy, R. E.,Hernandez, C., Nickoloff, B. J., Mestril, R. and Le Poole,I. C. (2005) 4-Tertiary butyl phenol exposure sensitizeshuman melanocytes to dendritic cell-mediated killing:relevance to vitiligo. J. Invest. Dermatol. 124,798–806

224 Antony, P. A., Piccirillo, C. A., Akpinarli, A., Finkelstein,S. E., Speiss, P. J., Surman, D. R., Palmer, D. C., Chan,C. -C., Klebanoff, C. A., Overwijk, W. W. et al. (2005)CD8+ T cell immunity against a tumor/self-antigen isaugmented by CD4 T helper cells and hindered bynaturally occurring T regulatory cells. J. Immunol. 174,2591–2601

225 Veldhoen, M., Hocking, R. J., Atkins, C. J., Locksley, R.M. and Stockinger, B. (2006) TGFβ in the context of aninflammatory cytokine milieu supports de novodifferentiation of IL-17-producing T cells. Immunity 24,179–189

226 Aflazi, B., Lombardi, G., Lechler, R. I. andLord, G. M. (2007) The role of T helper 17 (Th17) andregulatory T cells (Treg) in human organ transplantationand autoimmune disease. Clin. Exp. Immunol. 148,32–46

227 Gattinoni, L., Ranganathan, A., Surman, D. R., Palmer, D.C., Antony, P. A., Theoret, M. R., Heimann, D. M.,Rosenberg, S. A. and Restifo, N. P. (2006) CTLA-4dysregulation of self/tumor-reactive CD8+ T-cellfunction is CD4+ T-cell dependent. Blood 108,3818–3823

Received 23 November 2009/26 August 2010; accepted 26 August 2010Published on the Internet 20 October 2010, doi:10.1042/CS20090603

C© The Authors Journal compilation C© 2011 Biochemical Society


Recommended