You are on page 1of 9

Series

Psoriasis 1
Pathogenesis and clinical features of psoriasis
Christopher E M Griffiths, Jonathan N W N Barker

Psoriasis, a papulosquamous skin disease, was originally thought of as a disorder primarily of epidermal keratinocytes, Lancet 2007; 370: 263–71
but is now recognised as one of the commonest immune-mediated disorders. Tumour necrosis factor α, dendritic See Perspectives page 213
cells, and T-cells all contribute substantially to its pathogenesis. In early-onset psoriasis (beginning before age This is the first in a Series of two
40 years), carriage of HLA-Cw6 and environmental triggers, such as β-haemolytic streptococcal infections, are major papers about psoriasis
determinants of disease expression. Moreover, at least nine chromosomal psoriasis susceptibility loci have been Dermatology Centre, Hope
identified. Several clinical phenotypes of psoriasis are recognised, with chronic plaque (psoriasis vulgaris) accounting Hospital, University of
Manchester, Manchester
for 90% of cases. Comorbidities of psoriasis are attracting interest, and include impairment of quality of life and M6 8HD, UK
associated depressive illness, cardiovascular disease, and a seronegative arthritis known as psoriatic arthritis. A more (C E M Griffiths MD); and St
complete understanding of underlying pathomechanisms is leading to new treatments, which will be discussed in John’s Institute of
Dermatology, Guy’s Hospital
the second part of this Series.
Campus, King’s College
London, London, UK
Introduction Site-specific variants of psoriasis vulgaris exist. (J N W N Barker MD)
“I think it also necessary…to express the scaly psora by a Flexural (inverse) psoriasis in intertriginous sites is Correspondence to:
distinctive appellation; for this purpose, the term shiny, red, and typically devoid of scales (figure 3); Professor C E M Griffiths
psoriasis…” So wrote Robert Willan in his treatise On sebopsoriasis, which can be confused with seborrhoeic Dermatology Centre, Hope
Hospital, University of
Cutaneous Diseases,1 published in 1808. Willan, a British dermatitis, has greasy scales and occurs in eyebrows, Manchester, Manchester
dermatologist, is credited with the first accurate description nasolabial folds, and postauricular and presternal sites. M6 8HD, UK
of psoriasis and thereby helping to distinguish the disorder Psoriasis vulgaris will probably prove to be several christopher.griffiths
from leprosy. Psoriasis, one of the few skin diseases closely related but phenotypically and genotypically @manchester.ac.uk

common and distinctive enough to be recognised by distinct conditions,4 which might account for the
medical students (and most doctors) remains something variability of response to therapy, particularly with the
of an enigma. Relatively little attention has been paid to T-cell targeted biological agents.
identifying clinical phenotypes of psoriasis, or Children and adolescents can develop an acute form of
understanding the natural history and prognosis of the psoriasis known as guttate psoriasis (from the Latin
disease. In the past quarter century, substantial advances gutta meaning droplet), in which papules less than 1 cm
have been made in our understanding of the genetics and in diameter erupt on the trunk about 2 weeks after a
pathomechanisms of psoriasis. Debate continues as to β-haemolytic streptococcal infection such as tonsillitis or
whether psoriasis is an autoimmune disorder. Belatedly, pharyngitis, or a viral infection. Guttate psoriasis is
researchers and clinicians have started to recognise and self-limiting, resolving within 3–4 months of onset,
document the substantial impairment to quality of life although its long-term prognosis is unknown. One study
caused by psoriasis, resulting in recognition that the indicated that only a third of individuals with guttate
disease leads to marked loss of productivity2 in those it psoriasis develop classic plaque disease.5
afflicts. In this review, we will detail and put into clinical Generalised pustular psoriasis (von Zumbusch psoriasis)
context recent advances in the understanding of psoriasis. is an acute form in which small, monomorphic sterile
pustules develop in painful inflamed skin. The patient has
Clinical features
The commonest type of psoriasis, accounting for 90% of
all cases, is psoriasis vulgaris, in which papulosquamous Search strategy and selection criteria
plaques are well-delineated from surrounding normal We identified publications relating to psoriasis pathogenesis
skin. The plaques are red or salmon pink in colour, and psoriasis in general by searching Medline, Ovid, and the
covered by white or silvery scales (figure 1), and may be Cochrane Library databases with the term “psoriasis” alone or
thick, thin, large or small. They are most active at the edge: in combination with the terms “pathogenesis”, “genetics”,
rapidly progressing lesions may be annular, with normal “psychosocial”, “history”, “immunology”, “comorbidity”,
skin in the centre. Plaques are usually distributed sym- “angiogenesis”, “prevalence”, “epidemiology”, “innate
metrically, and occur most commonly on the extensor immunity”, “adaptive immunity”, “keratinocyte”, “animal
aspects of elbows and knees; scalp (where they rarely models”, and “phenotype”. We preferentially selected the
encroach beyond the hairline),3 lumbosacral region, and most recent papers and authoritative articles. Additional
umbilicus (figure 2). Active inflammatory psoriasis is articles known to the authors were also included. Date and
characterised by the Koebner phenomenon, in which new language were not limited.
lesions develop at sites of trauma or pressure.

www.thelancet.com Vol 370 July 21, 2007 263


Series

demographics to psoriasis vulgaris in that patients are


predominantly women (9:1 female:male ratio), and
either current or previous smokers (95%), and onset
occurs in the 4th or 5th decades of life. Moreover,
genetic analysis has implied that palmoplantar
pustulosis and psoriasis vulgaris have different causes.6
The condition may therefore be a comorbidity rather
than a form of psoriasis.
In erythroderma, the whole body surface is affected by
psoriasis—which can lead to hypothermia, hypo-
albuminaemia, and high output cardiac failure. Erythro-
derma can be a life-threatening condition and can be
caused by other diseases, including atopic dermatitis,
drug eruptions, and cutaneous T-cell lymphoma.
Figure 1: Single plaque of psoriasis, well demarcated and heavily scaled About 50% of patients with psoriasis have distinctive
nail changes related to the disease: the commonest is
pitting, which is best seen under oblique lighting
conditions; onycholysis (nail plate separation); oil spots
(orange-yellow sub-ungual discolouration), and dystrophy
(figure 4) similar to that observed in onychomycosis.
Psoriatic nail disease occurs most commonly in patients
with psoriatic arthritis.
Psoriatic arthritis, as originally defined by Moll and
Wright,7 is a seronegative inflammatory arthritis that
occurs in the presence of psoriasis (figure 5). Five types
of psoriatic arthritis have been proposed: distal
interphalangeal joint only; asymmetrical oligoarthritis;
polyarthritis; spondylitis; and arthritis mutilans. Classic
psoriatic arthritis consists of oligoarthritis, distal
interphalangeal joint involvement, dactylitis, and
calcaneal enthesitis.8 Recently presented data indicate
that its prevalence has been greatly underestimated, and
may be as high as 25% in people with psoriasis.9 In about
10% of people with psoriatic arthritis, the arthritis
appears before skin manifestations of psoriasis. The
prevailing view that the skin condition and arthritis
represent different manifestations of the same disease is
under challenge. Genetic evidence dissociates the two,
as do immunological studies and responses to various
treatments.10,11
Despite advances in our understanding of the
pathogenesis of psoriasis, relatively little is known about
the natural history of the disease, particularly predictors
of disease severity. The few longitudinal studies that have
been done suggest that spontaneous remission (for as
long as 54 years) might occur in about a third of patients.12
Figure 2: Chronic plaque psoriasis The determinants of spontaneous remission, and the
roles that comorbidities and age play in the prognosis
a fever and is systemically unwell. Precipitants of and activity of psoriasis are poorly understood.
generalised pustular psoriasis include intercurrent The underlying activity of psoriasis can be different
infection, and the abrupt withdrawal of systemic and, on from its expression, particularly when a patient is
occasion, ultrapotent topical corticosteroids. receiving treatment. No biomarkers are known to be
Palmoplantar pustulosis, consisting of yellow-brown, representative or predictive of psoriasis activity—
sterile pustules on palms and soles, is still described in C-reactive protein, soluble adhesion molecules, and
textbooks of dermatology as a subtype of psoriasis. soluble cytokine receptors have all been studied, but
About 25% of people with palmoplantar pustulosis also found to be unreliable. If a patient appears to have been
have chronic plaque psoriasis. The disease has different cleared of psoriasis by systemic therapy, there is no

264 www.thelancet.com Vol 370 July 21, 2007


Series

rare or absent in some isolated populations. Latitude also


seems to affect prevalence, probably because of the
beneficial effect of sunlight on the disease.13 However, the
prevalence of psoriasis is estimated to be between 1·5%
and 3% of the general population of Northern Europe
and Scandinavia,3 with men and women equally affected.
The incidence in white individuals is estimated to be
60 cases per 100 000 head of population per year.3
Psoriasis can appear at any time of life. However, the
mean age of onset for psoriasis vulgaris was recently
estimated at 33 years, with 75% of cases occurring before
46 years of age.14 Some studies suggest that onset is
bimodal, with peaks between 16 years and 22 years and
later at 57–60 years.15 The age of onset seems to be slightly
earlier in women than in men.

Comorbidity
Awareness is increasing that psoriasis as a disease is
more than skin deep and that it is associated with
systemic disorders,16 including Crohn’s disease, diabetes
mellitus (notably type 2),17 metabolic syndrome,18
depression,19 and cancer. It is unclear whether cancers,
particularly lymphoma20 and skin cancer,21 are related to
psoriasis or to its treatment. For example, the risk of
developing non-melanoma skin cancer is increased by
the excessive use of photochemotherapy—and can be
compounded by the subsequent use of ciclosporin.
Figure 3: Inverse psoriasis Of emerging concern is the relation between psoriasis
and cardiovascular disease.22 Although no excess risk
reliable way to ascertain whether control is absolute seems to exist for patients with mild psoriasis, moderate
other than reducing the dose of, or withdrawing, therapy. and severe disease is associated with a relative risk of
Clinical assessment relies on the classical skills of almost three.23 In part, this association is due to the
history and examination. over-representation of Framingham risk factors in the
psoriatic population, but evidence indicates that psoriasis
Epidemiology per se is an independent risk factor.24 Potential
Accurate figures for the prevalence of psoriasis are mechanisms could therefore include the presence of
difficult to obtain because of an absence of validated circulating proinflammatory factors and endothelial
diagnostic criteria. Moreover, rates vary greatly between activation,24 analogous to the situation noted in
people of different ethnic backgrounds: psoriasis is most rheumatoid arthritis. If confirmed, such mechanisms
common in white people, but is estimated to affect only would have major implications for future therapeutic
0·3% of the general population in China,3 and is very strategies.

Figure 4: Dystrophic nail psoriasis Figure 5: Psoriatic arthritis

www.thelancet.com Vol 370 July 21, 2007 265


Series

Histological features the disease.28 The interaction between VEGF and the
Psoriasis has three principal histological features: angiopoietin/Tie signalling pathway is modulated by
epidermal hyperplasia; dilated, prominent blood vessels tumour necrosis factor α (TNFα):29,30 results of a recent
in the dermis; and an inflammatory infiltrate of leucocytes, clinical study31 suggests that infliximab, a TNF-blocking
predominantly into the dermis (figure 6). Histology of chimeric monoclonal antibody, exerts part of its benefit in
uninvolved, clinically symptomless areas of skin is psoriasis by inhibition of this key VEGF/angiopoietin/Tie
normal. pathway.
The hyperplastic epidermal changes are associated with
an underexpression of markers of keratinocyte differ- The immune response in psoriasis
entiation, including keratins K1 and K10; loss of the Until the early 1980s, psoriasis was believed to be a disease
granular cell layer; parakeratosis (retention of nuclei in primarily of epidermal keratinocyte proliferation, and the
cells of the stratum corneum); elongation of rete ridges; cutaneous inflammatory infiltrate to be a secondary
and the presence of micropustules of Kogoj and event.32 However, strong evidence now exists that the
microabscesses of Munro. Keratinocytes of the hair follicle cell-mediated adaptive immune response is crucial in
are unaffected.25 psoriasis. The leucocyte infiltrate in psoriasis consists
Of the three main histological features of psoriasis, predominantly of CD4-positive and CD8-positive T-cells
increased vascularity in the dermis is probably the most (figure 7), and may precede epidermal hyperplasia.33 Some
overlooked. Indeed, relatively little research has been adhesion molecules which promote leucocyte adherence
devoted to this area since Braverman’s descriptions more are highly expressed in psoriatic skin: intercellular
than 30 years ago.26 Angiogenic factors produced by adhesion molecule-1 is expressed on epidermal
epidermal keratinocytes are now recognised as drivers of keratinocytes and along with E-selectin on dermal
abnormal dermal vascular proliferation and angiogenesis. capillaries.34
Levels of one such factor—vascular endothelial growth Cytokines of the Th1 pathway—interferon-γ,
factor (VEGF), also known as vascular permeability interleukin 2, and interleukin 12—predominate in plaques.
factor—are significantly raised in plaques of psoriasis;27 its Psoriasis is classified as a Th1 disease,35 which is consistent
serum concentration correlates with the clinical severity of with the relative under-representation of Th2 diseases,
such as atopic dermatitis, in patients with psoriasis.16,36
T-cell-targeted immunosuppressants such as ciclosporin
are efficacious in psoriasis,37 as is denileukin diftitox, an
interleukin-2 diphtheria fusion toxin cytolytic for activated
T cells.38 Moreover, bone marrow transplantation can
appear to transmit39 or clear40 psoriasis.
T cells in the cutaneous infiltrate are predominantly of
the memory effector CD45 RO+ designation41 and most
are positive for cutaneous lymphocyte-associated antigen,
a marker for skin-homing leucocytes.42 There is evidence
that these cells form clones in the epidermis of psoriatic
plaques.43 However, no definitive autoantigen or
immunogen has yet been identified to which the
inflammatory response is directed. If psoriasis is indeed
an autoimmune disease, an epidermal component,
perhaps keratin, might be the most likely candidate for
such an antigen.
In recent years, clinical and basic science observations
have shown that innate as well as adaptive immunity is
crucial in the initiation and maintenance of psoriatic
plaques. Indeed, since the epidermis is the body’s main
barrier to environmental insult, epidermal hyperplasia
forms a key component of the innate immune response.
Natural killer cells and natural killer T cells are part of the
cutaneous inflammation in psoriasis;44 at times,
neutrophils form a large proportion of the leucocytic
infiltrate, particularly in generalised pustular disease.45
Recent studies have considered the role of dendritic cells
Figure 6: Histology of psoriasis showing epidermal acanthosis, elongation of
and endogenous antimicrobial peptides. Three types of
rete ridges and inflammatory infiltrate (haematoxylin and eosin) dendritic cells appear likely to be involved in the
Scale bar=200 μm. development of psoriasis: Langerhans cells in the

266 www.thelancet.com Vol 370 July 21, 2007


Series

of TNFα, a key pro-inflammatory cytokine of the innate


immune response, is substantially increased in psoriasis.
The central role of this cytokine in psoriasis came to light
through observations of the efficacy of anti-TNF biological
therapies for the disease.50,51

Genetic contributions to psoriasis


Population studies show that the incidence of psoriasis
vulgaris is greater in first and second degree relatives of
patients than in the general population.52 About 30% of
individuals with psoriasis vulgaris have an affected first
degree relative.53 If both parents and a sibling are affected,
a further child has a 50% chance of developing psoriasis
vulgaris; if the sibling is affected but not the parents, the
risk drops to 8%.53 The risk of psoriasis vulgaris is two to
three times greater in monozygotic than in dizygotic
twins.52
There appears to be more than one pathway to the
development of psoriasis vulgaris. In a small minority of
families, the disease seems to follow Mendelian patterns
(autosomal dominant and recessive), implying a defect in a
single gene. However, in general psoriasis vulgaris appears
to arise through multiple genetic risk factors interacting
with each other and with environmental factors such as
β-haemolytic streptococcal infection, HIV, stress, and
drugs, including β blockers and lithium (figure 8).54 This
concept is further supported by substantial genetic
Figure 7: CD4-positive T cells in a plaque of psoriasis heterogeneity.
Scale bar=100 μm.
Several genome-wide scans have now been reported in
extended and nuclear families. At least nine chromosomal
epidermis; dermal factor XIIIa-positive dendritic cells; and loci have been identified for which statistically significant
a subset of dendritic cells, known as plasmacytoid dendritic evidence for linkage to psoriasis has been observed
cells, which are found in involved psoriatic skin but not in (nomenclature PSORS1–9).55 Of particular interest are:
normal skin. Langerhans cells are the outermost sentinels PSORS2, a replicated locus on chromosome 17q, at which
of the immune system: they recognise and capture a polymorphism associated with psoriasis has been
antigens, and migrate to local draining lymph nodes, identified, the polymorphism causing loss of binding to
where they present them to T cells.46 The migration of the RUNX1 transcription factor;56 PSORS4,57 which is
epidermal Langerhans cells in response to cytokine and located within the epidermal differentiation complex on
allergic stimuli is impaired in patients with early-onset chromosome 1q; and PSORS8,58 which overlaps with the
psoriasis, implying that these cells may be acting in a Crohn’s disease locus on chromosome 16q. PSORS8 is
regulatory manner to maintain cutaneous immune notable because psoriasis and Crohn’s disease are
homoeostasis.47 Plasmacytoid dendritic cells are distinct observed together more frequently than would be expected
from dermal factor XIIIa-positive dendritic cells and by chance.59 Despite this observation, several studies have
Langerhans cells in that they express CD123 and HLA-DR, failed to find an association between psoriasis and the
but lack CD11c.48 After activation via surface Toll-like CARD15/NOD2 mutation linked to Crohn’s disease.
receptors, plasmacytoid dendritic cells produce interferon By far the major genetic determinant of psoriasis is
α—which, hypothetically, leads to the formation of plaques PSORS1, which probably accounts for 35–50% of the
in predisposed individuals by driving Th1 responses, heritability of the disease,60 and has been replicated in
forming a further link between the innate and adaptive virtually all linkage studies of psoriasis.61 PSORS1 is
arms of the immune response. located in the MHC on chromosome 6p, spanning a
Endogenous anti-microbial peptides—cathelicidins segment of about 300 kb in the class I region telomeric of
and β defensins—are overexpressed in psoriatic skin, by HLA-B. The region contains less than ten genes, of which
contrast with atopic dermatitis, in which such peptides three have been the major focus of attention for
are underexpressed.49 This observation is congruent with investigators, because of strong associations between
the clinical observation that psoriatic skin is rarely polymorphic coding sequence variants of these genes
secondarily infected, whereas in atopic dermatitis and psoriasis vulgaris. HLA-C (associated variant
secondary infection is a substantial problem. The activity HLA-Cw6) encodes a class I MHC protein. CCHCR1

www.thelancet.com Vol 370 July 21, 2007 267


Series

are not.65 The implications for disease management are


1 2 Modifier 3 4
yet to be ascertained.
genes

Potential contribution of genetic studies to


Comorbidities
Genetic
Disease mechanisms Psoriasis including
treatment
Innate Adaptive
susceptibility (clinical cardiovascular and Without definitive identification of a major susceptibility
disease) psoriatic arthritis
Vascular gene for psoriasis, prediction of how genetic discovery
effects might inform disease pathogenesis is difficult at present.
Environmental Epidermal/ Nevertheless, several recent genetic observations show
factors barrier effects how pathogenic models can integrate perturbations in
the epidermal barrier and inflammation. For example, in
mice, disruption in keratinocyte signalling, either by
Disease Disease Outcome Concomitant abrogation of activation protein 1 (AP1) pathways66 or by
prevention measures, therapy,
measures
treatment
biomarkers preventable?
upregulation of signal transduction and activation of
transcription 3 (STAT3) pathways, leads to psoriasiform
Phenotypic classification hyperplasia.67 Mutations in filaggrin, a key protein that
facilitates terminal differentiation and formation of the
Pharmacogenetics skin barrier, are strongly associated with atopic dermatitis
Figure 8: The march of psoriasis (eczema),68 another common inflammatory skin disease,
This schematic combines information that is currently accepted and under active investigation. Boxes under dotted which shares many phenotypic features with psoriasis
line refer to clinical issues and intervention strategies; the latter would be better informed by use of validated (erythema, induration, and scale formation with a
pharmacogenetic and phenotypic classification tools. Step 1—PSORS1 harbours the main genetic determinant for
symmetrical distribution). These findings suggest that
psoriasis. With or without (depending on pedigree) involvement of environmental factors, genetic mutations or
polymorphisms drive disease-specific pathogenic processes. Future disease prevention measures will target these primary defects in the epidermis lead to T-cell mediated
aspects as well as disease mechanisms. Step 2—evidence indicates key roles for innate and adaptive immune immune events, which are at present the target for most
responses; changes in epidermis are also important. Vascular changes are prominent. Genes controlling degree of therapeutic intervention. Furthermore, findings of
expression or function of key molecules in these pathways determine disease severity; these might include
several studies show an association of psoriasis vulgaris
polymorphisms in VEGF and TNFα. Present treatments target key elements of these pathways, including TNFα
(etanercept, infliximab), T cells (ciclosporin, efalizumab), and epidermal changes (vitamin D₃ analogues). Step 3—the with functional polymorphisms in genes for factors that
consequence of activation of these processes is disease expression, including distribution and area of expression, control inflammation (eg, TNFα69) and vascular growth
phenotype, and duration of disease. Improvements in outcome measures including identification of biomarkers (eg, VEGF70). These data suggest the existence of modifier
would significantly improve our ability to predict disease responses and natural history. Step 4—substantial
comorbidities, particularly cardiovascular disease, are likely to result from chronic inflammation. Up to 25% of
genes that can regulate the severity of disease in
psoriasis is associated with arthritis; whether as a comorbidity or a direct consequence of the psoriatic process susceptible individuals. Finally, and most compelling, is
remains to be ascertained. Treatment approaches need to take account of these issues to minimise morbidity and the recent observation that missense mutations in the
mortality, but can themselves also lead to comorbidity. interleukin-23 receptor gene and SNPs in the untranslated
region of interleukin-12B identify psoriasis risk genes.71
(variant WWCC) encodes the coiled-coil x-helical rod The importance of these genetic observations is
protein 1, a ubiquitously expressed protein that is emphasised by two key facts. First, a human antibody to
overexpressed in psoriatic epidermis.62 CDSN p40, a component of interleukin 12 and of interleukin 23,
(variant=allele 5) encodes corneodesmosin, a protein that when administered to individuals with psoriasis, leads to
is uniquely expressed in the granular and cornified layers a rapid and sustained improvement in the disease.72
of the epidermis and is upregulated in psoriasis.63 Second, intradermal injection of interleukin 23 into
Absolute identification of the causative gene at this locus mouse skin stimulates epidermal hyperplasia,73 a
has been hampered by extensive linkage disequilibrium hallmark of psoriasis. Interleukin 23 is important for the
within the MHC. Although evidence suggests a genetic production of Th17 cells, which secrete interleukin 17.
event close to (or within) HLA-Cw6 as the susceptibility Together, these findings suggest that products of these
factor at PSORS1,64 no mutations have been identified, genes have a fundamental role in the pathogenesis of
and variants in regulatory sequences potentially affecting psoriasis.
several downstream genes cannot be excluded. Gene studies might allow the identification of molecules
Nevertheless, HLA-Cw6 is a strong marker for early-onset that contribute to pathogenesis, and can be targeted by
psoriasis. In one study, 85% of patients with onset before therapies. In addition, modifier genes may exist that can
age 40 years had at least one HLA-Cw6 allele, compared regulate the severity of disease in susceptible individuals.
with 15% of those with onset after this age.15 Studies of such mechanisms in validated models are
Investigations have shown that the phenotypic variants urgently needed.
of psoriasis are genetically heterogeneous at least at the
level of PSORS1. Thus guttate psoriasis (an acute onset Animal models
form, usually occurring in adolescents) is strongly Psoriasis vulgaris is unique to human beings. However,
associated with PSORS1,6 whereas palmoplantar transgenic knockout/deletion animal models of psoriasis
pustulosis and late onset (age >50 years) psoriasis vulgaris have been proposed as surrogates for the disease.

268 www.thelancet.com Vol 370 July 21, 2007


Series

Epidermal overexpression of VEGF in mice produces a behaviours are similar to those used by patients with
phenotype of chronic inflammation, psoriasiform cancer or chronic pain.87 Concerns about chronicity of
hyperplasia, and vascular proliferation reminiscent of disease and the absence of a cure are an important
psoriasis.74 Mice that are transgenic for epidermal undercurrent: even patients in whom systemic therapy
keratinocyte expression of STAT367 have a psoriasiform causes remission maintain high levels of anxiety because
appearance. A recently described mouse model in which of the fear of relapse.88 Psychological sequelae of the
epidermal expression of the c-Jun component of AP1 is disease can impair response to treatment: patients with
deleted is characterised by persistent epidermal hyperplasia pathological levels of anxiety are less likely to respond to
and an accompanying inflammatory arthritis.66 This photochemotherapy.89 Even patients’ partners and close
finding implies that changes in the epidermis can initiate relatives are less attuned to the psychosocial sequelae of
extracutaneous pathology. The most faithful replicants of the disease than might be expected.90 Doctors experienced
psoriasis itself are chimeras comprising biopsies of in the management of psoriasis remain fairly poor at
uninvolved, symptomless skin from patients with psoriasis identifying and subsequently addressing depression and
grafted onto the flank of immunodeficient mice. This anxiety in their patients.91 An understanding of the
approach takes two main forms. First, use of the severe psychosocial difficulties encountered by patients with
combined immunodeficient (SCID) mouse, in which a psoriasis and other chronic skin diseases, and how a
graft of uninvolved skin from a psoriasis patient can be biopsychosocial model could be used in the management
transformed into psoriasis by virtue of injections with of such conditions, are unmet needs.
activated T cells or natural killer T cells.75,76 The resulting
graft provides clinical, histological, and immunological Conclusion
features consistent with psoriasis. Second, the AGR129 The past 20 years, and in particular the past 5 years, have
mouse,77 which is more immunosuppressed than its SCID witnessed great advances in our knowledge of the
counterpart because it lacks T and B cells, has impaired pathogenesis of psoriasis, courtesy of genetic and
natural killer cell function and is deficient in types I and II immunological techniques. It is now accepted that
interferon receptors and recombination activating gene-2. psoriasis is a chronic, immune-mediated inflammatory
A graft of uninvolved psoriatic skin onto the AGR129 disease that can act as a paradigm for other diseases of
mouse spontaneously transforms into psoriasis, with the this genre. These basic science observations have catalysed
implication that the grafting process can stimulate the development of targeted biological treatments that
expansion of resident immune cells in the graft. Anti-CD3 will revolutionise the management of psoriasis. There are
and anti-TNFα agents injected into the graft can prevent still gaps in our basic understanding of psoriasis,
conversion of uninvolved to involved skin. A recently specifically clinical observational work on phenotypes and
described mouse model has emphasised the previously the natural history of the disease; the role of angiogenesis;
underappreciated role of macrophages in psoriasis.78,79 the association with metabolic syndrome; and an in-depth
analysis of psychosocial factors relating to the disease.
Psychosocial aspects However, slowly but surely psoriasis vulgaris is giving up
Psoriasis is rarely life-threatening; however, it is its secrets to clinicians and cutaneous biologists.
life-ruining for the majority of patients. Dennis Potter80 Conflict of interest statement
and John Updike81 have written eloquently and movingly CEMG has received research support or has acted as a consultant or
about the despair and social isolation that accompany lecturer for Abbott, Amgen, Biogen-IDEC, Centocor, Essex Pharma,
Galderma, Leo Pharma, Novartis, Novo Nordisk, Schering-Plough,
psoriasis. Although Willan1 separated psoriasis from Serono, Stiefel, UCB Pharma, and Wyeth. JNWNB has acted as a
leprosy, the stigma of psoriasis persists. These difficulties consultant to Abbott, Centocor, Janssen Cilag, Novartis, Schering-Plough,
manifest as significant impairment of quality of life82 and Serono, and Wyeth, and has lectured at sponsored symposia for the
profound psychosocial disability.83 Studies have shown above companies. He has no shares in any of these companies.
reliably that psoriasis produces a decrease in quality of Acknowledgments
life at least equal to and often greater than that of We thank Sarah Smith and Val Hill for help with preparation of the
manuscript and Thomas Brenn for supplying figure 6.
conditions such as diabetes, ischaemic heart disease, and
chronic obstructive pulmonary disease.84 The dermatology References
1 Willan R. On cutaneous diseases. London: J Johnson, St Paul’s
life quality index is a questionnaire used increasingly to Churchyard, 1808.
assess improvements in quality of life resulting from 2 Finlay AY, Coles EC. The effect of severe psoriasis on the quality of
therapeutic intervention.85 The stress caused by living life of 369 patients. Br J Dermatol 1995; 132: 236–44.
3 Griffiths CEM, Camp RDR, Barker JNWN. Psoriasis. In: Burns DA,
with psoriasis can manifest as avoidance behaviours; for Breathnach SM, Cox NH, Griffiths CEM, eds. Rook’s textbook of
example, unwillingness to attend local swimming pools dermatology, 7th edn. Oxford: Blackwell, 2005: 35.1–35.69.
for fear of the ignominy of being asked to leave by a 4 Griffiths CEM, Christophers E, Barker JNWN, et al. A new
classification of psoriasis according to clinical phenotype.
member of the public with unwarranted concerns about Br J Dermatol 2007; 156: 258–62.
the contagiousness of the condition. Objective studies 5 Martin BA, Chalmers RJG, Telfer NR. How great is the risk of
have emphasised the role of automatic vigilance86 for further psoriasis following a single episode of a guttate psoriasis?
Arch Dermatol 1996; 132: 717–18.
threat in patients with psoriasis, and indeed their coping

www.thelancet.com Vol 370 July 21, 2007 269


Series

6 Asumalahti K, Ameen M, Suomela S, et al. Genetic analysis of 32 Bos JD, de Rie MA, Teunissen MB, Piskin G. Psoriasis:
PSORS1 distinguishes guttate psoriasis and palmoplantar dysregulation of innate immunity. Br J Dermatol 2005; 152: 1098–107.
pustulosis. J Invest Dermatol 2003; 120: 627–32. 33 Valdimarsson H, Baker BS, Jonsdottir I, Powles A, Fry L. Psoriasis:
7 Moll JMH, Wright V. Psoriatic arthritis. Semin Arthritis Rheum 1973; a T-cell-mediated autoimmune disease induced by streptococcal
3: 55–78. superantigens? Immunol Today 1995; 16: 145–49.
8 Helliwell PS, Taylor WJ. Classification and diagnostic criteria for 34 Robert C, Kupper T. Inflammatory skin diseases, T cells and
psoriatic arthritis. Ann Rheum Dis 2005; 64: ii3-ii8. immune surveillance. N Engl J Med 1999; 341: 1817–28.
9 Zachariae H. Prevalence of joint disease in patients with psoriasis: 35 Austin LM, Ozawa M, Kikuchi T, Walters IB, Krueger JG. The
implications for therapy. Am J Clin Dermatol 2003; 4: 441–47. majority of epidermal T cells in Psoriasis vulgaris lesions can
10 Ho P, Bruce IN, Silman A, et al. Evidence for common genetic produce type 1 cytokines, interferon-gamma, interleukin-2, and
control in pathways of inflammation for Crohn’s disease and tumor necrosis factor-alpha, defining TC1 (cytotoxic T lymphocyte)
psoriatic arthritis. Arthritis Rheum 2005; 52: 3596–602. and TH1 effector populations: a type 1 differentiation bias is also
11 Pitzalis C, Cauli A, Pipitone N, et al. Cutaneous lymphocyte measured in circulating blood T cells in psoriatic patients.
antigen-positive T lymphocytes preferentially migrate to the skin J Invest Dermatol 1999; 113: 752–59.
but not to the joint in psoriatic arthritis. Arthritis Rheum 1996; 39: 36 Landgren E, Bråbäck L, Hedlin U, Hjern A, Rasmussen F. Psoriasis
137–45. in Swedish conscripts: time trend and association with T-helper 2-
12 Farber EM, Nall ML. The natural history of psoriasis in mediated disorders. Br J Dermatol 2006; 154: 332–36.
5600 patients. Dermatologica 1974; 148: 1–18. 37 Mueller W, Herrmann B. Cyclosporin A for psoriasis. N Engl J Med
13 Braathen LR, Botten G, Bjerkedak T. Prevalence of psoriasis in 1979; 301: 555.
Norway. Acta Derm Venereol (Stockh) 1989; 142: 5–8. 38 Gottlieb SL, Gilleaudeau P, Johnson R, et al. Response of psoriasis
14 Nevitt GJ, Hutchinson PE. Psoriasis in the community: prevalence, to a lymphocyte-selective toxin (DAB389IL-2) suggests a primary
severity and patients’ beliefs and attitudes towards the disease. immune, but not keratinocytes, pathogenic basis. Nat Med 1995; 1:
Br J Dermatol 1996; 135: 533–37. 442–47.
15 Henseler T, Christophers E. Psoriasis of early and late onset: 39 Wahie S, Alexandruff A, Reynolds NJ, Meggitt SJ. Psoriasis
characterization of two types of psoriasis vulgaris. occurring after myeloablative therapy and autologous stem cell
J Am Acad Dermatol 1985; 13: 450–56. transplantation. Br J Dermatol 2006; 154: 194–95.
16 Henseler T, Christophers E. Disease concomitance in psoriasis. 40 Eedy DJ, Burrows D, Bridges JM, Jones FG. Clearance of severe
J Am Acad Dermatol 1995; 32: 982–86. psoriasis after allogenic bone marrow transplantation. BMJ 1990;
300: 908.
17 Neimann AL, Shin DB, Wang X, Margolis DJ, Troxel AB,
Gelfand JM. Prevalence of cardiovascular risk factors in patients 41 Vissers WH, Arndtz CH, Muys L, Van Erp PE, de Jong EM,
with psoriasis. J Am Acad Dermatol 2006; 55: 829–35. van de Kerkhof PC. Memory effector (CD45RO+) and
cytotoxic (CD8+) T cells appear early in the margin zone of
18 Sommer DM, Jenisch S, Suchan M, Christophers E,
spreading psoriatic lesions in contrast to cells expressing natural
Weichenthal M. Increased prevalence of the metabolic syndrome in
killer receptors, which appear late. Br J Dermatol 2004; 150: 852–59.
patients with moderate to severe psoriasis. Arch Dermatol Res 2006;
298: 321–28. 42 Davison SC, Ballsdon A, Allen MH, Barker JNWN. Early
migration of cutaneous lymphocyte-associated antigen (CLA)
19 Esposito M, Saraceno R, Giunta A, Maccarone M, Chimenti S. An
positive cells into evolving psoriatic plaques. Exp Dermatol 2001;
Italian study on psoriasis and depression. Dermatol 2006; 212:
10: 280–85.
123–27.
43 Prinz JC, Vollmer S, Boehncke WH, Menssen A, Laisney I,
20 The risk of lymphoma in patients with psoriasis. J Invest Dermatol
Trommler P. Selection of conserved TCR VDJ rearrangements in
2006; 126: 2194–201.
chronic psoriatic plaques indicates a common antigen in psoriasis
21 Lindelof B, Eklund G, Liden S, Stern RS. The prevalence of vulgaris. Eur J Immunol 1999; 29: 3360–68.
malignant tumors in patients with psoriasis. J Am Acad Dermatol
44 Gaspari AA. Innate and adaptive immunity and the pathophysiology
1990; 22: 1056–60.
of psoriasis. J Am Acad Dermatol 2006; 54 (suppl 2): S67–80.
22 Gelfand JM, Neimann AL, Shin DB, Wang X, Margolis DJ,
45 Nickoloff BJ. Skin innate immune system in psoriasis: friend or
Troxel AB. Risk of myocardial infarction in patients with psoriasis.
foe? J Clin Invest 1999; 104: 1161–64.
JAMA 2006; 296: 1735–41.
46 Griffiths CEM, Dearman RJ, Cumberbatch M, Kimber I. Cytokines
23 Mallbris L, Akre O, Granath F, et al. Increased risk for
and Langerhans cell mobilisation in mouse and man. Cytokine
cardiovascular mortality in psoriasis inpatients but not in
2005; 32: 67–70.
outpatients. Eur J Epidemiol 2004; 19: 225–30.
47 Cumberbatch M, Singh M, Dearman RJ, Young HS, Kimber I,
24 Mrowietz U, Elder JT, Barker J. The importance of disease
Griffiths CEM. Impaired Langerhans cell migration in psoriasis.
associations and concomitant therapy for the long-term
J Exp Med 2006; 203: 953–60.
management of psoriasis patients. Arch Dermatol Res 2006; 298:
309–18. 48 Nestle FO, Conrad C, Tun-Kyi A, et al. Plasmacytoid predendritic
cells initiate psoriasis through interferon-alpha production.
25 Krueger JG, Bowcock A. Psoriasis pathophysiology: current
J Exp Med 2005; 202: 135–43.
concepts of pathogenesis. Ann Rheum Dis 2005; 64 (suppl II):
ii30–36. 49 Ong PY, Ohtake T, Brandt C, et al. Endogenous antimicrobial
peptides and skin infections in atopic dermatitis. N Engl J Med
26 Braverman IM, Yen A. Microcirculation in psoriatic skin.
2002; 347: 1151–60.
J Invest Dermatol 1974; 62: 493–502.
50 Leonardi CL, Powers JL, Matheson RT, et al. Etanercept as
27 Detmar M, Brown LF, Claffey KP, et al. Overexpression of vascular
monotherapy in patients with psoriasis. N Engl J Med 2003; 349:
permeability factor/vascular endothelial growth factor and its
2014–22.
receptors in psoriasis. J Exp Med 1994; 180: 1141–46.
51 Reich K, Nestle FO, Papp K, et al. Infliximab induction and
28 Creamer D, Allen MH, Groves RW, Barker JNWN. Circulating
maintenance therapy for moderate-to-severe psoriasis: a phase III,
vascular permeability factor/vascular endothelial growth factor
multicentre, double-blind trial. Lancet 2005; 366: 1367–74.
erythroderma. Lancet 1996; 348: 1101.
52 Farber EM, Nall NL, Watson W. Natural history of psoriasis in 61
29 Holash J, Maisonpierre PC, Compton D, et al. Vessel cooption,
twin pairs. Arch Dermatol 1974; 109: 207–11.
regression and growth in tumors mediated by angiopoientins and
VEGF. Science 1999; 284: 1994–98. 53 Andressen C, Henseler T. Inheritance of psoriasis. Analysis of
2035 family histories. Hautarzt 1982; 33: 214–17.
30 Kuroda K, Sapadin A, Shoji T, Fleischmajer R, Lebwohl M. Altered
expression of angiopoietins and Tie2 endothelium receptor in 54 Barker JNWN. Pathophysiology of psoriasis. Lancet 1991; 338: 227–30.
psoriasis. J Invest Dermatol 2001; 116: 713–20. 55 Capon F, Trembath RC, Barker JNWN. An update on the genetics of
31 Markham T, Mullan R, Golden-Mason L, et al. Resolution of psoriasis. Dermatol Clin 2004; 22: 339–47.
endothelial activation and down-regulation of Tie2 receptor in 56 Helms C, Cao L, Krueger JG, et al. A putative RUNX1 binding site
psoriatic skin after infliximab therapy. J Am Acad Dermatol 2006; 54: variant between SLC9A3R1 and NAT9 is associated with susceptibility
1003–12. to psoriasis. Nat Genet 2003; 35: 349–56.

270 www.thelancet.com Vol 370 July 21, 2007


Series

57 Capon F, Semprini S, Chimenti S, et al. Fine mapping of the PSORS4 74 Xia YP, Li B, Hylton D, Detmar M, Yancopoulos GD, Rudge JS.
psoriasis susceptibility region on chromosome 1q21. J Invest Dermatol Transgenic delivery of VEGF to mouse skin leads to an
2001; 116: 728–30. inflammatory condition resembling human psoriasis. Blood 2003;
58 Karason A, Gudjonsson JE, Upmanyu R, et al. A susceptibility gene 102: 161–68.
for psoriatic arthritis maps to chromosome 16q: evidence for 75 Wrone-Smith T, Nickoloff BJ. Dermal injection of immunocytes
imprinting. Am J Hum Genet 2003; 72: 125–31. induces psoriasis. J Clin Invest 1996; 98: 1878–87.
59 Lee FI, Bellary SV, Francis C. Increased occurrence of psoriasis in 76 Nickoloff BJ, Bonish B, Huang BB, Porcelli SA. Characterization of
patients with Crohn’s disease and their relatives. Am J Gastroenterol a T cell line bearing natural killer receptors and capable of creating
1990; 85: 962–63. psoriasis in a SCID mouse model system. J Dermatol Sci 2000; 24:
60 Trembath RC, Clough RL, Rosbotham JL, et al. Identification of a 212–25.
major susceptibility gene locus on chromosome 6p and evidence for 77 Boyman O, Hefti HP, Conrad C, Nickoloff BJ, Suter M, Nestle FO.
further disease loci revealed by a two stage genome-wide search in Spontaneous development of psoriasis in a new models shows an
psoriasis. Human Mol Genet 1997; 6: 813–20. essential role for resident T cells and tumor necrosis factor-alpha.
61 Capon F, Munro M, Trembath R, Barker J. Searching for the MHC J Exp Med 2004; 199: 731–36.
psoriasis susceptibility gene. J Invest Dermatol 2002; 118: 745–51. 78 Wang H, Peters T, Kess D, et al. Activated macrophages are
62 Suomela S, Elomaa O, Asumalahti K, et al. HCR, a candidate gene for essential in a murine model for T cell-mediated chronic
psoriasis, is expressed differently in psoriasis and other psoriasiform skin inflammation. J Clin Invest 2006; 116: 2105–14.
hyperproliferative skin disorders and is downregulated by 79 Stratis A, Pasparakis M, Rupec RA, et al. Pathogenic role for skin
interferon-gamma in keratinocytes. J Invest Dermatol 2003; 121: macrophages in a mouse model of keratinocytes induced
1360–64. psoriasis-like skin inflammation. J Clin Invest 2006; 116: 2094–104.
63 Allen M, Ishida-Yamamoto A, McGrath J, et al. Corneodesmosin 80 Potter D. The singing detective. London: Faber and Faber, 1986.
expression in psoriasis vulgaris differs from normal skin and other 81 Updike J. Self-consciousness—memoirs. London: Deutsch, 1989.
inflammatory skin disorders. Lab Invest 2001; 81: 969–76. 82 Finlay AY, Kelly SE. Psoriasis—an index of disability.
64 Nair RP, Stuart PE, Nistor I, et al. Sequence and haplotype analysis Clin Exp Dermatol 1987; 12: 8–11.
supports HLA-C as the psoriasis susceptibility 1 gene. 83 Gupta MA, Gupta AK. Psychiatric and psychological co-morbidity in
Am J Hum Genet 2006; 78: 827–51. patients with dermatologic disorders: epidemiology and
65 Allen MH, Ameen M, Veal C, et al. The major psoriasis susceptibility management. Am J Clin Dermatol 2003; 4: 833–42.
locus PSORS1 is not a risk factor for late-onset psoriasis. J Invest Derm 84 Rapp SR, Feldman SR, Exum ML, Fleischer AB Jr, Reboussin DM.
2005; 124: 103–07. Psoriasis causes as much disability as other major medical diseases.
66 Zenz R, Eferl R, Kenner L, et al. Psoriasis-like skin disease and J Am Acad Dermatol 1999; 41: 401–07.
arthritis caused by inducible epidermal deletion of Jun proteins. 85 Finlay AY, Khan GK. Dermatology Life Quality Index (DLQI)—a
Nature 2005; 437: 369–75. single practical measure for clinical use. Clin Exp Dermatol 1994; 19:
67 Sano S, Chan KS, Carbajal S, et al. Stat3 links activated keratinocytes 210–16.
and immunocytes required for development of psoriasis in a novel 86 Fortune DG, Richards HL, Corrin A, Taylor RJ, Griffiths CEM,
transgenic mouse model. Nat Med 2005; 11: 43–49. Main CJ. Attentional bias for psoriasis-specific and psychosocial
68 Palmer CN, Irvine AD, Terron-Kwiatkowski A, et al. Common threat in patients with psoriasis. J Behav Med 2003; 26: 211–24.
loss-of-function variants of the epidermal barrier protein filaggrin are 87 Fortune DG, Richards HL, Main CJ, Griffiths CEM. Patients’
a major predisposing factor for atopic dermatitis. Nat Genet 2006; 38: strategies for coping with psoriasis. Clin Exp Dermatol 2002; 27:
441–46. 177–84.
69 Mössner R, Kingo K, Kleensang A, et al. Association of TNF-238 and 88 Fortune DG, Richards HL, Kirby B, McElhone K, Main CJ,
-308 promoter polymorphisms with psoriasis vulgaris and psoriatic Griffiths CEM. Successful treatment of psoriasis improves
arthritis but not with pustulosis palmoplantaris. J Inv Dermatol 2005; psoriasis-specific but not more general aspects of patients’
124: 282–84. well-being. Br J Dermatol 2004; 151: 1219–26.
70 Young HS, Summers AM, Bhushan M, Brenchley PEC, 89 Fortune DG, Richards HL, Kirby B, et al. Psychological distress
Griffiths CEM. Single nucleotide polymorphisms of vascular impairs clearance of psoriasis in patients treated with
endothelial growth factor (VEGF) in psoriasis of early onset. photochemotherapy. Arch Dermatol 2003; 139: 752–56.
J Invest Dermatol 2004; 122: 209–15.
90 Richards HL, Fortune DG, Chong SL, et al. Divergent beliefs about
71 Cargill M, Schrodi SJ, Chang M, et al. A large scale genetic psoriasis are associated with increased psychological distress.
association study confirms IL-12B and leads to IL-23R as J Invest Dermatol 3004; 123: 49–56.
psoriasis-risk genes. Am J Hum Genet 2007; 80: 273–90.
91 Richards HL, Fortune DG, Weidmann A, Sweeney SK,
72 Krueger GG, Langley RG, Leonardi C, et al. A human interleukin- Griffiths CEM. Detection of psychological distress in patients with
12/23 monoclonal antibody for the treatment of psoriasis. psoriasis: low consensus between dermatologist and patient.
N Engl J Med 2007; 356: 580–92. Br J Dermatol 2004; 151: 1227–33.
73 Chan JR, Blumenschein W, Murphy E, et al. IL-23 stimulates
epidermal hyperplasia via TNF and IL-20R2 – dependent
mechanisms with implications for psoriasis pathogenesis.
J Exp Med 2006; 203: 2577–87.

www.thelancet.com Vol 370 July 21, 2007 271

You might also like