You are on page 1of 10

doi: 10.1111/1346-8138.

13806 Journal of Dermatology 2017; 44: 863872

REVIEW ARTICLE
Critical role of environmental factors in the pathogenesis of
psoriasis
Jinrong ZENG, Shuaihantian LUO, Yumeng HUANG, Qianjin LU
Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital of Central South University,
Changsha, China

ABSTRACT
Psoriasis is a common cutaneous disease with multifactorial etiology including genetic and non-genetic factors,
such as drugs, smoking, drinking, diet, infection and mental stress. Now, the role of the interaction between envi-
ronmental factors and genetics are considered to be a main factor in the pathogenesis of psoriasis. However, it is
a challenge to explore the mechanisms how the environmental factors break the body balance to affect the onset
and development of psoriasis. In this article, we review the pathogenesis of psoriasis and summarize numerous
clinical data to reveal the association between environmental factors and psoriasis. In addition, we focus on the
mechanisms of environmental risk factors impact on psoriasis and provide a series of potential treatments
against environmental risk factors.

Key words: environmental factors, epigenetics, genetics, pathogenesis, psoriasis.

INTRODUCTION influenced by environmental factors.68 T cells have been


believed to be a key mediator in psoriasis. Environmental risk
Psoriasis is a chronic and recurrent inflammatory skin disease factors trigger the immune response of the body, where naive
which is characterized by hyperproliferative keratinocytes and T cells are activated by antigen-presenting cells (APC) in the
infiltrating immune cells, including T cells, dendritic cells, epidermis, especially Langerhans cells. APC can also release
macrophages and neutrophils. The main clinical features of cytokines such as interleukin (IL)-12 and IL-23, which promote
psoriasis are a thick epidermis that expresses keratin 16 (K16) naive T cells to differentiate into Th1 and Th17 cells.9 Then, the
in cells above the basal layer and consistent T-lymphocyte activated T cells migrate from lymph nodes to skin, where they
(positive by CD3 staining) infiltration into both the epidermis are stimulated to produce plenty of cytokines. Therefore, these
and dermis of lesions. The prevalence in adults ranges 0.51 cytokines interact with the resident epidermal and dermal cells
11.43%, and in children 01.37% depending on age, geo- and then cause changes, including keratinocyte proliferation
graphic location and genetic background.13 Initially, psoriasis and epidermal thickness. All the changes involved in the two
was just considered a disease with hyperproliferation and main pathways around T cells include: IL-2/Th1/interferon
abnormal differentiation of keratinocytes, while current (IFN)-c and IL-23/Th17/IL-17. In the first pathway, IFN-c pro-
researches confirm that psoriasis is an autoimmune skin dis- duced by activated Th1 cells induces the expression of co-sti-
ease characterized by T-cell-mediated responses. The imbal- mulatory molecules on dendritic cells and intercellular adhesion
ance among CD4+ T-cell subsets contributes to hyperkeratosis molecule (ICAM)-1 on keratinocytes, stimulates tumor necrosis
and parakeratosis.4,5 Various T-cell subsets, such as T-helper factor (TNF)-a release from macrophages, thus inhibiting ker-
(Th)1, Th2, Th17 and regulatory T cells (Treg), are involved in atinocyte apoptosis.10 In the second pathway, Th17 cells
the immunopathogenesis and chronic inflammation response release the main effector cytokines IL-17 and IL- 22. IL-17 can
of psoriasis. The fact that Th1 and Th17 cells increase while influence the expression of cytokines such as TNF-a and
Th2 and Treg decrease has been found in patients with psoria- ICAM-1, increase keratinocyte expression of chemokines and
sis. So, blockade of pathogenic T-cell activation contributes to reduce expression of cell adhesion molecules.11 IL-22 also pro-
the improvement of both clinical and histopathological disease motes the proliferation of keratinocytes.12 Both of them result
condition, including abnormal K16 expression and excessive in the disruption of the skin barrier (Fig. 1).
T-cell infiltration. Unfortunately, there is no cure for psoriasis at present, and
Currently, psoriasis is believed to be triggered by the combi- most of the current treatments just manage the symptoms by
nation of genetic, epigenetic and environmental influences, and drugs while omitting the importance of avoiding risk factors,
its genetic diversity and epigenetic modifications can be such as excess ultraviolet (UV) exposure, specific drugs,

Correspondence: Qianjin Lu, Ph.D., Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, Hunan 410011,
China. Email: qianlu5860@gmail.com
Received 20 December 2016; accepted 22 January 2017.

2017 Japanese Dermatological Association 863


J. Zeng et al.

Table 1. Environmental risk factors in psoriasis

Main risk Critical role in the pathogenesis


factors of psoriasis References
UV exposure Immunoregulatory and 48
pro-apoptotic effects
Downregulates Th17/IL-23
response
Medications Interfering individual immune 56,57
system
Including signaling pathway
and cytokine expression
Smoking Toxins in cigarettes; interfere 65
Diet and with normal nervous system 7
obesity signaling and immune
cell signaling
Overexpression of
pro-inflammatory and
anti-inflammatory cytokines
Alcohol Sensitive to gluten; immunological 67,71
intake disorders
Infections Inducing autoimmunity response; 83,99
breakdown of immune tolerance
Stress Disturbing immune system 108
regulation and
abnormal T-cell activation

IL, interleukin; Th17, T-helper 17; UV, ultraviolet.


Figure 1. Psoriasis pathogenesis. Psoriasis is believed to be
triggered by a combination of genetic, disordered immune sys-
tem and environmental influences, and its genetics and associated with psoriasis can generally be divided into two
immune system can be affected by environmental factors. categories: immune-specific and skin-specific genes.16 Among
Naive T cells are activated by antigen-presenting cells (APC) in them, it is worth mentioning that mutations of caspase recruit-
the epidermis, which promote naive T cells to differentiate into ment domain-containing protein 14 (CARD14) and IL36RN
T-helper (Th)1 and Th17, so that Th1 and Th17 cells increase genes have been recognized as responsible genetic factors of
while Th2 and regulatory T cells (Treg) decrease. Then, the generalized pustular psoriasis.17,18 The CARD14 gene encodes
activated T cells migrate from lymph nodes to skin, where they a protein that promotes the activation of nuclear factor (NF)-jB
are stimulated to produce plenty of cytokines. So these cytoki-
and upregulates the expression of pro-inflammatory pathway
nes interact with the resident epidermal and dermal cells and
genes.19 IL36RN encodes the IL-36 receptor antagonist (IL-
then cause changes, including keratinocyte proliferation and
epidermal thickness. DC, dendritic cells; IFN, interferon; IL, 36Ra) which counteracts the inflammatory effect of IL-36
interleukin; TNF, tumor necrosis factor; UV, ultraviolet. cytokines (IL-36a, IL-36b and IL-36c) by binding their receptor
(IL-1RL2) and prevents the downstream activation of NF-jB
signaling.20 Recent studies identified more than dozens of
infections, smoking, alcohol abusing and stress, and the critical gene locus mutations of CARD14 and IL36RN, which differed
role of them on psoriasis are summarized in Table 1. depending on geography and race. Now, psoriasis is com-
monly described as a T-cell-mediated autoimmune disease, in
which IFN-c and TNF-a act as key pro-inflammatory players
PATHOGENESIS OF PSORIASIS influenced by environmental factors and susceptibility genes.
Genetic polymorphism and epigenetic modification Several biologics targeting cytokines and T cells have been
in psoriasis approved by the US Food and Drug Administration (FDA)
The pathogenesis of psoriasis mainly relates to the combined applied in clinics to treat psoriasis. Psoriasis is a multigene dis-
effect of multiple gene susceptibility loci, the disordered order whose manifestation and severity are likely to depend on
immune system together with pervasive environmental risk fac- the patients genetic background.21,22 The incidence of psoria-
tors. Genome-wide association studies (GWAS) have been sis increased significantly (P < 0.05) in the patients relatives
applied successfully in psoriasis to explore the genetic archi- and the concordance rate was much higher between monozy-
tecture. Large amounts of data indicate psoriasis-susceptibility gotic twins (6572%) than dizygotic twins (1530%), which
loci or candidate genes (showed in Table 2) among different indicated that genetic linkage played the key role in psoria-
populations, such as PSORS1 that could account for 3540% sis.2,2325 Family lineage studies suggest that psoriasis has two
in psoriasis in heritability.1315 In brief, the gene variants different genetic subpopulations, one with multifactorial

864 2017 Japanese Dermatological Association


Environmental factors on psoriasis

Table 2. Several susceptibility loci associated with psoriasis

Susceptibility loci SNP Function References


HLA-C rs10484554c Involve in the immune reactions 115
IL-12B rs3213094 Encode IL-12-p40 subunit and promote Th1 development 116
IL-23R rs11209026 Initiate a tumor necrosis factor-dependent epidermal hyperplasia 117
LCE3D rs4112788 Control epidermal terminal differentiation 117
TNFAIP3 rs610604 Act downstream of TNF-a and regulate NF-jB signaling 115
IL13 rs20541 Involve in the modulation of Th2 immune responses 115
TNIP1 rs1024995b Act downstream of TNF-a and regulate NF-jB signaling 115
IL23A rs2066808 Promote inflammation response 115
ZNF313 rs2235617d Regulate T-cell activation 118

Superscript indicates alleles. NF, nuclear factor; SNP, single nucleotide polymorphisms; Th1, T-helper 1; T-helper 2; TNF, tumor necrosis factor.

determination and one with multigenic inheritance, which also leukocyte antigen (HLA)-A26 had significant positive correlation
strongly suggests the contribution of epigenetic or environmen- with psoriasis of no environmental exposure (P < 0.0001). In
tal factors in the pathogenesis of disease.26,27 Among these their research, they illustrated that environmental factors had
researches there are even conflicting results obtained in differ- significant interaction with HLA-A26. Jin et al. discovered the
ent populations. Recent studies indicate that epigenetic-regu- joint effect of HLA-Cw6 and smoking, a wonderful explanation
lated networks may be the key causative elements in some to identify the susceptibly variant related to psoriasis. The risk
cases to explain the clinical manifestations. Epigenetic modifi- of psoriasis is 11-fold less for non-smokers without the HLA-
cation is often seen in the psoriasis, which can change the Cw6 gene in comparison with smokers carrying the HLA-Cw6
manifestations instead of altering DNA sequence but can be gene in the Chinese Han population.35 GWAS have verified
passed on to offspring. Studies showed the fact that the gen- amounts of genes correlated with environmental risks of psori-
ome-wide methylation level of peripheral blood mononuclear asis. As we all know, obesity is a common risk factor for psori-
cells (PBMC) in psoriatic skin lesions and peripheral blood asis, and the roles of genetic factors may be modified by
increased significantly; more interestingly, the expression of obesity in psoriasis development.3537 Li et al.38 demonstrated
DNMT1 increased while MBD2 and MeCP2 decreased in the epigenetic modifying action of obesity in the relevance of
PBMC and the acetylation level of histone H4 decreased signif- psoriasis and genetic variants, and they even affirmed the
icantly because of the downregulated expression of P300, CBP increased risk of psoriasis related to the single nucleotide poly-
and Sirt1, and the upregulated expression of HDAC1, morphism in IL-12B. Although little is known about the mecha-
SUV39H1 and EZH2.28,29 However, it is still a challenge to nism of geneenvironment interactions in psoriasis, we have
know the exact mechanism how the environmental factors the opportunity to evaluate geneenvironment interactions in
influence epigenetic changes in psoriasis. cohort studies because plenty of published GWAS applied to
psoriasis since 2007.
Interaction between environmental factors and
genetics in psoriasis Interaction between environmental factors and
Environmental factors appear to induce inflammatory diseases epigenetics in psoriasis
in individuals with latent genetic susceptibility. Many alterations Epidemiological studies have showed that environmental risk
could be triggered by environmental factors such as diet, factors may result in the susceptibility and severity of psoriasis
microbial infections (from bacteria, fungus and virus), chemical among isogenic lines by epigenetic changes.39 Recently, epi-
irritants or UV radiation exposure,30 and bad habits (such as genetic modification changes including DNA methylation, his-
smoking and drinking).31,32 Now, it is widely accepted that the tone modification and non-coding RNA have been found to be
interplay between environmental and genetic factors con- involved in many diseases, such as psoriasis. These epigenetic
tributes to the onset, development and present clinical symp- changes can often been seen in the pathogenesis of psoriasis.
toms of psoriasis. Parisi et al.33 suggested that the prevalence Secreted frizzled-related protein (SFRP)4, a negative regulator
of psoriasis distribution differed between Europe (0.732.9%) of the Wnt signaling pathway, can directly inhibit pathological
and the USA (0.72.6%), indicating that different races had keratinocyte proliferation induced by pro-inflammatory cytoki-
various genetic backgrounds. The author also discussed the nes, and the high methylation level of the promoter has led to
variability in psoriasis prevalence among distinct geographic epigenetic downregulation of SFRP4 in inflamed skin of psori-
positions. Several studies of psoriasis and psoriatic arthritis atic patients and imiquimod mouse models, which contributed
have demonstrated the interaction of genes and environment. to the hyperplasia of epidermis in psoriasis.40 In a study direc-
Wei et al.34 had verified that environmental elements (including ted by Zhou et al.,41 the researchers found nine instances of
mental stress, infection, trauma, damp, drugs smoking and skin-specific DNA methylation of differentially methylated sites
drinking) were all the risk factors in the course of occurrence, associated with psoriasis by a three-stage epigenome-wide
aggravation and recurrence of psoriasis and found that human association study, which were not obviously influenced by

2017 Japanese Dermatological Association 865


J. Zeng et al.

genetic variations. Among them, the expression levels of


CYP2S1, ECE1, EIF2C2, MAN1C1 and DLGAP4 were nega-
tively related to DNA methylation. Recently, the technology of
using methylated DNA immunoprecipitation sequencing sug-
gested that DNA methylation may play an important role in the
epigenetic dysregulation of biological pathways in psoriasis.28
miRNA-31 induced by NF-jB is one of the most dynamic
miRNA identified in the skin of psoriatic patients and mouse
models, whose aberrant rise in keratinocytes will promote their
abnormal proliferation, increase acanthosis and inhibit the dis-
ease recovery in IL-23 mouse models. Study has confirmed
that the protein phosphatase 6 (ppp6c) acting as a targeted
molecular is a negative regulator that restricts the G1 to S
phase progression. The rise of miR-31 directly inhibits ppp6c
expression, thus enhancing keratinocyte proliferation.42
Recently, the emergence of long non-coding RNA (lncRNA) has
attracted our attention, defined as non-protein coding RNA
transcripts longer than 200 nucleotides, are acting as key regu-
lators of diverse cellular processes. Moreover, the expression
levels of various lncRNA are closely related to epidermal differ-
entiation and immunoregulation,43 such as PRINS, which is ele-
vated in non-lesional skin areas in patients with psoriasis while
deceased in the lesion. GIP3, which is increased in the lesional
skin areas, as its potential target gene, can reduce the sensitiv-
ity of keratinocytes to spontaneous apoptosis.44 All these find-
ings will benefit us in better understanding the molecular
mechanism of psoriasis (Fig. 2).

MECHANISMS OF ENVIRONMENTAL RISK


FACTORS IMPACT ON PSORIASIS
UV radiation exposure Figure 2. Epigenetic regulation in the pathogenesis of psoria-
Ultraviolet radiation from natural sunlight or artificial sources has sis. Environmental risk factors may lead to the high susceptibil-
been actually exploited for treating psoriasis in recent decades. ity and severity of psoriasis by epigenetic changes. Epigenetic
modification changes including DNA methylation, histone modi-
The incidence of psoriasis may differ among various populations
fication and non-coding RNA. The activation of nuclear factor
owing to regional disparity with distinct UV distribution. A case
(NF)-jB and Wnt signaling pathways have been found to play
control study explored absolute latitude suggesting that there a vital role in the epigenetic mechanisms of the epidermal
was a wide variation in prevalence between 0% (in Samoa, aver- hyperplasia in psoriasis. They activate the inflammation
age absolute latitude 13.35) and 3.3% (in Tanzania, average response by targeting key molecular such as ppp6c, following
absolute latitude 6).45 Moreover, sunscreen habits, altitude and exposure to certain environmental triggers. M, methylation; IL,
ground reflection of UV light may also result in differences in interleukin; SFRP4, secreted frizzled-related protein 4. [Correc-
average UV exposure. The mechanism of UV therapy mainly tion added on 6 June 2017, after first online publication: The
includes three aspects. First, more vitamin D can be produced figure has been modified to remove the mention of STAT6.]
by exposure to UV-B. Vitamin D is a hormone which plays a criti-
cal role in the treatment of psoriasis. Data shows that the optimal psoriasis.49 Studies showed that APC and autoreactive dermal T
concentration of vitamin D (25[OH]D) in serum circulating level cells with intense stimulation were considered to be primary initi-
for maximum effect should be between 30 and 50 ng/mL (75 ating factors contributing to the pathogenesis of this condi-
125 nmol/L).35. Therefore, if vitamin D is lower than 30 ng/mL, tion.45,50 UV-B is an excellent therapeutic option for moderate
UV-B exposure will benefit patients a lot. 25(OH)D plays a vital psoriasis because of its high efficacy, relative safety and afford-
role in the regulation of dendritic cells, the modulation of ker- ability. Weatherhead et al.51 investigated whether UV-B has
atinocytes and the proliferation of T cells.46,47 Second, UV-B has higher efficacy in clearing psoriatic lesions and inducing remis-
pro-apoptotic effects leading to production of various pro- and sion. They also pointed out that only 311-nm UV-B could con-
anti-inflammatory cytokines. It simultaneously suppresses the tribute to fatal apoptosis in lesional epidermis, and the main
differentiation of Th17 cells and inhibits IL-23/IL-17 expression.48 apoptotic cells were keratinocytes. What is more, they provided
In addition, UV exposure can reduce immune cells migration to a new insight into the use of keratinocyte apoptosis as a promis-
skin, and lower psoriasis-related antimicrobial peptides, particu- ing biomarker to further explore which wavelengths of UV-B
larly LL-37 and psoriasin which are visibly increased in were most effective in curing psoriasis.52

866 2017 Japanese Dermatological Association


Environmental factors on psoriasis

Medications disruptions of normal nervous system signaling. The nAChR-


Studies have showed that many drugs, such as antiproliferative mediated transduction pathway can regulate keratinocyte
agents (imiquimod), antivirals and antidepressants (lithium), anti- function. Also, many immune cells including T cells, B cells
hypertensives (beta-blockers), TNF-a, and many novel biologics leukemic cell lines and thymocyte, have active nAChR and
including anticytokine therapies used in the treatment of psoria- nicotine binding to these receptors interferes with immune cell
sis (anti-TNF antibodies), have all been clinically associated with signaling.65 What is more, it is believed that smoking is posi-
initiation and exacerbation of psoriasis.53 The most widely tively related to an increased risk of psoriatic comorbidity
accepted hypothesis suggests there is an interaction between including cardiovascular diseases.66
TNF-a and IFN-a. IFN-a is an important cytokine that controls
natural immunity and is mainly produced from plasmacytoid Alcohol intake
dendritic cells through stimuli such as infection or trauma. IFN-a Even though the hypothesis that alcohol consumption is asso-
is also identified as a key element in the early phase of psoriatic ciated with the increased risk of psoriasis onset and worsening
induction. TNF-a negatively regulates IFN-a production. There- is not credible enough, studies showed that alcohol could
fore, anti-TNF-a therapy could increase IFN-a expression at the directly and indirectly enhance the production of pro-inflamma-
tissue level and induce psoriasis.54 Vinter et al.55 induced a pso- tory cytokines from many cell types, leading to persistent sys-
riasis-like inflammatory skin condition through applying Aldara temic inflammation and promoting lymphocyte proliferation.
(Aldara 5%; Meda, Solna, Sweden) cream containing 5% imiqui- Moreover, alcohol and acetone could directly stimulate ker-
mod, which may be because of its defective recruitment of atinocyte proliferation and increase the mRNA levels of genes
adaptive immune system and the strong stimulation of IL-10 characteristic of proliferating keratinocytes, such as a5 integrin,
counter-regulation. Imiquimod is a Toll-like receptor 7/8 agonist cyclin D1 and keratinocyte growth factor receptor. In addition,
used to treat genital warts and non-melanoma skin tumors. It acetylcholine may also trigger psoriasis.67 Qureshi et al.31 sug-
can activate the type I IFN-signaling pathway and has been con- gested that there was an obviously positive correlation
sidered as one of the best investigated triggers of psoriasis.56 between heavy beer intake and risk of developing psoriasis
Dalkilic et al.57 conducted a research involving 514 patients trea- among women, whereas another survey showed that alcohol
ted with anti-TNF-a and matched healthy controls in Turkey. The intake may exacerbate psoriasis severity.68 The relationship
incidence of anti-TNF-a-induced psoriasis was 1.16% (6/514 between psoriasis and alcohol intake is complicated, and dif-
patients) which suggested that biological agents should be ferent studies reached various conclusions. However, the con-
restricted to an appropriate application field and physicians sensus is that patients with moderate to severe psoriasis have
should take full account of their indications. Sahuquillo-Torralba a higher incidence and mortality of alcohol-associated dis-
et al.58 found that pembrolizumab, a new drug approved by the eases, including cardiovascular diseases and tristimania.69 Bre-
FDA for cancer immunotherapy, could worsen the symptoms of naut et al. conducted a systematic published work review in
psoriasis. These drugs mainly aim at interfering with the individ- 2013 using the Medline, Embase and Cochrane databases. He
uals immune system, resulting in psoriasis induction. The mech- found four of five studies showing that alcohol was a risk factor
anisms by which the drugs trigger psoriasis are complicated, so for psoriasis.70 The possible mechanism is that beer and some
the clinical application of those drugs inevitably encounters alcoholic beverages are made with wheat or other products
some difficulties. containing gluten, so only those people who are sensitive to
gluten frequently have symptoms. In conclusion, smoking and
Smoking alcohol may induce the onset of psoriasis by various mecha-
Smoking or secondhand smoke exposure is a possible risk nisms, including immunological disorders such as keratinocyte
factor for psoriasis. However, controversial results are also hyperproliferation and overexpression of pro-inflammatory
observed.59,60 The exact impact of smoking on disease activity cytokines.71 However, the exact mechanisms by which
of psoriasis is not clearly known yet. Studies have showed that tobacco and alcohol induce psoriasis still require further inves-
smoking can increase the activity of spondyloarthritis and tigation to verify the relation between tobacco and alcohol con-
rheumatoid arthritis, thus reducing quality of life.6163 Hojgaard sumption and psoriasis.
et al.64 conducted an observational research of 1388 psoriatic
patients to study the correlation between smoking and treat- Diet and obesity
ment effect of TNF-a inhibitors. The result showed that heavy Patients often wonder what they cannot eat and whether diet-
smoking could decrease the treatment effect of TNF-a-targeted ary changes can improve their symptoms or prevent recur-
drugs, and the impact seemed partially reversible. Naldi et al.36 rence. Of course, this refers to whether weight loss can
directed an interesting comparative study and showed that contribute to their skin condition in this discussion. Association
smoking expedited the occurrence of psoriatic arthritis in of psoriasis severity and high body mass index (BMI) score
patients without psoriasis, while it slowed down the recurrence has been identified in several recent researches which suggest
of psoriatic arthritis in patients with psoriasis. The reasons why that bodyweight loss can improve Psoriasis Area and Severity
smoking is a possible risk factor of psoriasis are highly com- Index score of the patients.72 In addition, it is certain that
plex due to a variety of unknown toxins contained in cigarettes. healthy eating patterns would benefit psoriatic patients, includ-
However, one critical reason is the binding between nicotine ing the use of oral vitamin B12, vitamin D, selenium and
and nicotinic acetylcholine receptors (nAChR) leading to omega-3 fatty acids in fish oils.73 As stated earlier, the optimal

2017 Japanese Dermatological Association 867


J. Zeng et al.

concentration of vitamin D (25[OH]D) in serum circulating level environmental risk factors. The composition of the microbiome
is 3050 ng/mL; if obese and lean subjects take the same is specific to each individual. Usually, the microbes and their
dose, the circulating level of vitamin D in the obese subject host interact with each other in a harmonious way.82 However,
would be insufficient, leaving gradually less 25(OH)D in the disordered immune responses can cause inflammatory dis-
organism. However, more data about nutritional interventions is eases, and the exact etiology remains unknown. Hajishengallis
needed for clinical physicians to manage psoriatic patients. A et al.83 assumed that microbes could induce autoimmunity in
meta-analysis performed by Millsop et al.73 found that fish oil those persons with a potential genetic susceptibility. Micro-
seemed to be the most useful to improve the condition of ecosystem disturbance induces infection when the host is in
patients in a randomized controlled trial. Recently, epidemio- poor condition because of drug abuse, complications and
logical and clinical studies74 have provided a possible curative undernutrition, together with other environmental factors such
effect of a gluten-free diet for psoriasis. It was reported that a as diet, stress, and tobacco and alcohol intake.84 Without the
gluten-free diet seemed to be in certain psoriatic patients impact of environmental factors, the composition of the micro-
favor according to some early evidence, but it also needed biome would remain relatively stable over a whole lifetime but
more trials in different populations. Obesity has been proved change a little with aging.85 Th17 cells have been identified as
correlative with a pro-inflammatory state and numbers of study major constituents of psoriasis pathogenesis and many other
results have showed that being overweight could worsen psori- autoimmune diseases. Cutaneous microbiota was revealed
asis. Wolk et al.75 conducted a casecontrol study composed 100 years ago and accumulated evidences confirmed the rela-
of 373 psoriatic patients and matched healthy controls in 2009. tion between skin microorganisms and psoriatic lesions, such
They showed that obese individuals had a twofold increased as streptococcal b-hemolytic group A throat infections linked
risk to develop psoriasis in comparison with those of normal to guttate psoriasis.85 Drago et al.86 also suggested that the
bodyweight. Furthermore, they found that each unit increase in type of microorganisms resides at different skin locations could
BMI was associated with a 9% increase in the risk of psoriasis influence the onset and the progression of cutaneous diseases.
onset and a 7% increase in the risk of psoriasis severity. How- Studies showed that antibiotics targeting Gram-negative and
ever, it is unknown whether psoriasis outcome was influenced Gram-positive bacteria could ameliorate psoriasiform dermatitis
by weight loss interventions, and still now none of any studies induced by imiquimod, and it inhibited the differentiation of T
can expound the problem clearly. Several studies7678 have cells and the production of pro-inflammatory IL-17 and IL-22.87
sought to establish the possible impact of diet change and Importantly, T cells detected from psoriatic lesions react to
weight loss on the therapeutic effect of psoriasis. They also streptococcal and staphylococcal peptidoglycan in an antigen-
showed a condition improvement in the process of psoriasis specific way.88 Surprisingly, the deterioration of the skin inflam-
treatment and disease severity index that declined obviously mation is not related to the rise in IL-17-producing b+ or cd+ T
after following a gluten-free diet for those anti-gliadin antibody cells. Instead, the severity of skin inflammation was mainly
(AGA)-positive patients. There is an agreement that overex- ascribed to prominent increase in skin T-cell-derived IL-22,
pression levels of pro-inflammatory and anti-inflammatory especially T-cell receptor cd+ IL-22+ cells. So, they suggested
cytokines is most important in the pathogenesis of psoriasis. that IL-22-producing T cells, particularly cd+ T cells, play the
Interestingly, obese individuals bear more chronic inflamma- most vital role in the aggravation of skin inflammation.87 Other
tion, which increases the risk for psoriasis onset.79 A study microorganisms including Staphylococcus aureus, Malassezia
conducted by Edson-Heredia et al.80 explored which clinical and Candida are also involved in the pathogenesis of psoria-
factors in psoriasis correlated with the response to biologic sis.89,90 Staphylococcus and Streptococcus are commonly
therapy, and they found that BMI had the most intense seen in all levels of skin.90 Propionibacterium acnes are the
response to biologics in the whole studies. Ustekinumab was main microorganisms located in a normal individuals skin but
mainly used for treating psoriasis and it was proved to be a not in those influenced by psoriasis,86,91,92 which may be a
promising biologic agent. Its phase III clinical trial showed that predictor of a disordered microbial system that induces the
higher bodyweight was a potential predictor of worse response onset of psoriasis. A disordered micro-ecosystem probably
to it at the 28th week (P < 0.0001).81 Based on the above stud- plays a critical role in the pathogenesis of psoriasis or may be
ies, proper diet adjustment and healthy bodyweight may bene- engaged in the disease exacerbation.
fit psoriatic patients in the process of treatment but more
clinical data is needed to confirm this conclusion. Intestinal microbiota and psoriasis
The intestinal microbiome plays a vital role in maintaining our
intestinal mucosal barrier and normal immune function which
Microbiota and psoriasis
exerts extra-intestinal regulation function at remote individual
Cutaneous microbiota and psoriasis body sites.93 There is a well-known correlation between
Skin and intestinal microbiota have been the focus of much Crohns disease (CD) and psoriasis. Patients with CD have a
attention in all fields recently, especially immunology. A sum- fivefold higher risk of developing psoriasis than those without
mary of studies of microorganisms in psoriasis is displayed in CD. On the contrary, patients with psoriasis are more likely to
Table 3. There is a peaceful coexistence among bacteria, fungi suffer from CD.94 More importantly, it has been reported that
and virus in a healthy body, which can develop an effective the innate immune system is active in psoriasis. Recently,
first-line barrier defense against pathogens and other studies have suggested that the mechanism of intestinal tract

868 2017 Japanese Dermatological Association


Environmental factors on psoriasis

Table 3. Main microorganisms in psoriasis

Species Most common genus in psoriasis Main distribution levels References


Bacteria Streptococcus and Staphylococcus Epithelial and 90
Dermal layers Corynebacterium Epithelial layer
Pseudomonas and Escherichia Outer epithelial layer
Bacteroides and Clostridium Subdermal layer
Proteus Dermal layer
Fungi Malassezia Epithelial layer 119,120
Candida Epithelial layer

infection was similar to skin infection. Bacteroides fragilis can cell activation.84 The most common psychiatric troubles related
promote Treg differentiation via producing polysaccharide A to dermatological conditions in general are depression, anxiety
and regulating the balance of Th1/Th2,95,96 while segmented and suicidal thoughts. The biological links between psoriasis
filamentous bacteria direct Th17 cell differentiation,97 and and depression are now well-established, and the psychiatric
Clostridium species promote Treg development.98 Danese comorbidity is estimated to be approximately 30%.110 Cha-
et al. proposed that a breakdown of immune tolerance affected moun et al.110 suggested that psoriasis has an important
by disordered microbiota of the intestinal tract may contribute impact on the formation of a patients personality in their child-
to the onset of psoriasis.99 However, the exact etiology of the hood or adolescence. Conversely, personality will have an
activation has not been entirely clear. In fact, the presence of influence on the morbidity of psoriasis. Martn-Brufau et al.111
intestinal inflammation in psoriatic arthritis has been showed in evaluated the relationship between emotional condition and
multiple studies.100102 An obvious correlation is apparent skin lesions in a study of 823 psoriatic patients. Their data sup-
between psoriasis and inflammatory bowel disease (IBD), and ported the association and also explained the association
they have similarities in genetic factors and pathogene- between the specific kind of emotions and psoriatic lesions.
sis.103,104 Evidence showed that guttate psoriasis was com- Therefore, it is essential to take care of the emotion of psoriatic
monly triggered by streptococcal infection, which even patients. Sometimes, disease prevention is more important
presented in the blood of psoriasis.105 What is more, emotion- than the treatment of the disease in the clinic.
induced skin inflammation is related to dysbiosis in the intesti-
nal microbiome. A hypothesis of the presence of the gut
ENVIRONMENTAL FACTORS IN TREATMENT
brainskin axis in normal individuals may be possible.106 Stud-
OF PSORIASIS
ies have suggested that Streptococcus in the tonsils may con-
tribute to the onset of chronic plaque psoriasis,107 and bacteria Treatment of psoriasis has been a long process, from the very
in the intestinal tract would initiate and maintain psoriasis.107 first using coal tar, until now the classic treatment methods
The microbiome may be the director, or a mediator at least, emerge, which involve methotrexate, cyclosporin, vitamin D
often involved in the common inflammatory pathways in derivatives, calcineurin inhibitors and newer biologic agents.
autoimmune diseases. These explanations may also refer to However, most psoriatic patients are increasingly taking medi-
other extra-intestinal clinical manifestations such as IBD and cine to manage their symptoms clinically; some patients even
extracutaneous clinical manifestations such as psoriasis.84 have to take long-term drugs to maintain the condition while
However, not all clinical data is consistent. This phenomenon ignoring the important role of environmental factors in the
may be concerned with susceptibility genes, which embody development and prognosis of their diseases. Once their con-
the interaction between environmental factors and genetics. dition gets better, they continue to smoke, drink and eat
The relationship between organism infection and psoriasis extravagantly. What is worse, some of them are so ashamed
seems to be a vital and potentially promising field for future of their bad appearance that they fall into depression, leading
research. to recurrence. Therefore, psychological interventions including
psychological counseling and relaxation therapy combined
Psychiatric factors with related medications can offer benefits to most patients
Psoriatic patients often feel nervous and great pressure with psoriasis. Physicians should pay more attention to psy-
because of their appearance. So, they are not willing to take chological comfort of their patients, encourage them to acquire
daily physical activities or join in social activities, even fall into confidence and teach them how to prevent psoriasis. Above
depression, the prevalence of which varies 1062% according all, prospective and casecontrol studies have certified that
to different studies. In turn, depression can worsen psoriasis, higher BMI, alcohol intake, smoking, drugs and infection are
forming a vicious circle. Stress in daily life is related to the new connected with a high risk of psoriasis. So, high BMI or AGA-
onset of psoriasis and will worsen the symptoms of psoria- positive patients should adopt dietotherapy, which is eating
sis.107 Park and Youn109 also found that mental stress played more food containing rich vitamin D, losing weight and avoid-
a critical role in the occurrence, development and aggravation ing gluten in food, especially AGA-positive patients. Recently,
of psoriasis (P < 0.01). Psychological stress is known to impact studies have found the link between microbiota and psoriasis,
psoriasis through immune system regulation and abnormal T- so topical treatment takes anti-infection into consideration.

2017 Japanese Dermatological Association 869


J. Zeng et al.

More interestingly, a womens health study found that women psoriasis: identification of epigenetically dysregulated genes. PLoS
who exercised regularly were less likely to have psoriasis than Genet 2012; 8: e1002454.
3 Michalek IM, Loring B, John SM. A systematic review of worldwide
women who did not like to participate in regular physical activ-
epidemiology of psoriasis. J Eur Acad Dermatol Venereol 2017; 31:
ities.112 Frankel et al.113 also carried out a large cohort study 205212.
to demonstrate that running and performing aerobic exercise 4 Lowes MA, Russell CB, Martin D et al. The IL-23/T17 pathogenic
or calisthenics could reduce risk of psoriasis, and the real axis in psoriasis is amplified by keratinocyte responses. Trends
Immunol 2013; 34: 174181.
effectiveness of physical activity would be impacted by the
5 Mak RK, Hundhausen C, Nestle FO. Progress in understanding the
patients mood. Exercise can relieve stress, reduce anxiety immunopathogenesis of psoriasis. Actas Dermosifiliogr 2009; 100
and tension, improve emotional well-being and help maintain (Suppl 2): 213.
an ideal weight. So, it may be a potentially promising treat- 6 Seldin MF. The genetics of human autoimmune disease: a per-
ment option for depression.114 Moreover, outdoor sports spective on progress in the field and future directions. J Autoim-
mun 2015; 64: 112.
encouraging optimal UV radiation exposure is an effective
7 Yang CA, Chiang BL. Inflammasomes and human autoimmunity: a
treatment for psoriasis plaque, which can replenish more vita- comprehensive review. J Autoimmun 2015; 61: 18.
min D made in the skin regularly. Above all, the physicians 8 Chang C. Autoimmunity: from black water fever to regulatory func-
final goal is to supervise patients maintaining a healthy, body tion. J Autoimmun 2014; 4849: 19.
9 Boehncke WH. Etiology and pathogenesis of psoriasis. Rheum Dis
free of infection, depression, obesity, alcohol and cigarettes.
Clin North Am 2015; 41: 665675.
All the above environmental factors are interrelated and func- 10 Myers WA, Gottlieb AB, Mease P. Psoriasis and psoriatic arthritis:
tion together in the onset, development and recurrence of pso- clinical features and disease mechanisms. Clin Dermatol 2006; 24:
riasis. Therefore, the management of psoriasis should 438447.
emphasize avoiding recurrence and improving the prognosis of 11 Fitzgerald O, Winchester R. Editorial: emerging evidence for critical
involvement of the interleukin-17 pathway in both psoriasis and
psoriasis instead of only depending on drug therapy, especially
psoriatic arthritis. Arthritis Rheumatol 2014; 66: 10771080.
for those who have high risk of family history or susceptibility 12 Fujita H. The role of IL-22 and Th22 cells in human skin diseases.
genes. J Dermatol Sci 2013; 72: 38.
13 Gandhi G, Buttar BS, Albert L et al. Psoriasis-associated genetic
polymorphism in North Indian population in the CCHCR1 gene and
CONCLUSION in a genomic segment flanking the HLA-C region. Dis Markers
2011; 31: 361370.
Psoriasis is believed to be triggered by a combination of 14 Ammar M, Bouchlaka-Souissi C, Helms CA et al. Genome-wide
genetic, disordered immune system and environmental influ- linkage scan for psoriasis susceptibility loci in multiplex Tunisian
ences. Recently, numbers of studies showed that the environ- families. Br J Dermatol 2013; 168: 583587.
15 Shaiq PA, Stuart PE, Latif A et al. Genetic associations of psoriasis
mental risk factors include UV, drugs, smoking and alcohol,
in a Pakistani population. Br J Dermatol 2013; 169: 406411.
infection and mental stress, all of which play a critical role in 16 Alwan W, Nestle FO. Pathogenesis and treatment of psoriasis:
the pathogenesis of psoriasis. Currently, studies have showed exploiting pathophysiological pathways for precision medicine. Clin
that environmental risk factors affect the occurrence, progres- Exp Rheumatol 2015; 33: S2S6.
17 Bertin J, Wang L, Guo Y et al. CARD11 and CARD14 are novel
sion and recurrence of psoriasis mainly via disturbing its genet-
caspase recruitment domain (CARD)/membrane-associated
ics and immune system; as part of this process, the guanylate kinase (MAGUK) family members that interact with
mechanisms of epigenetic modifications cannot be ignored. BCL10 and activate NF-kappa B. J Biol Chem 2001; 276: 11877
However, the exact evidence needs to be investigated more 11882.
deeply and more risk factors wait to be discovered. Therefore, 18 Marrakchi S, Guigue P, Renshaw BR et al. Interleukin-36-receptor
antagonist deficiency and generalized pustular psoriasis. N Engl J
this review suggests that doctors should provide patients more
Med 2011; 365: 620628.
recommendations on how to prevent the onset or recurrence 19 Jordan CT, Cao L, Roberson ED et al. PSORS2 is due to muta-
of psoriasis as part of their daily lives. tions in CARD14. Am J Hum Genet 2012; 90: 784795.
20 Sims JE, Smith DE. The IL-1 family: regulators of immunity. Nat
Rev Immunol 2010; 10: 89102.
ACKNOWLEDGMENTS: This work is supported by the Key 21 Feng BJ, Sun LD, Soltani-Arabshahi R et al. Multiple Loci within
Project of National Natural Science Foundation of China (no. 81430074) the major histocompatibility complex confer risk of psoriasis. PLoS
and Major International Cooperation Project of National Natural Science Genet 2009; 5: e1000606.
Foundation of China (no. 81220108017). 22 Lu Q. The critical importance of epigenetics in autoimmunity. J
Autoimmun 2013; 41: 15.
23 Lonnberg AS, Skov L, Skytthe A et al. Heritability of psoriasis in a
large twin sample. Br J Dermatol 2013; 169: 412416.
CONFLICT OF INTEREST: None declared.
24 Pedersen OB, Svendsen AJ, Ejstrup L et al. On the heritability of
psoriatic arthritis. Disease concordance among monozygotic and
dizygotic twins. Ann Rheum Dis 2008; 67: 14171421.
REFERENCES 25 Pietrzyk JJ, Turowski G, Kapinska-Mrowka M et al. Family studies
in psoriasis. I. Complex segregation analysis. Arch Dermatol Res
1 Chandran V, Raychaudhuri SP. Geoepidemiology and environmen- 1982; 273: 287294.
tal factors of psoriasis and psoriatic arthritis. J Autoimmun 2010; 26 Speckman RA, Wright DJ, Helms C et al. Novel immunoglobulin
34: J314J321. superfamily gene cluster, mapping to a region of human chromo-
2 Gervin K, Vigeland MD, Mattingsdal M et al. DNA methylation and some 17q25, linked to psoriasis susceptibility. Hum Genet 2003;
gene expression changes in monozygotic twins discordant for 112: 3441.

870 2017 Japanese Dermatological Association


Environmental factors on psoriasis

27 Chandra A, Ray A, Senapati S et al. Genetic and epigenetic basis 51 Weatherhead SC, Farr PM, Jamieson D et al. Keratinocyte apopto-
of psoriasis pathogenesis. Mol Immunol 2015; 64: 313323. sis in epidermal remodeling and clearance of psoriasis induced by
28 Zhang P, Zhao M, Liang G et al. Whole-genome DNA methylation UV radiation. J Invest Dermatol 2011; 131: 19161926.
in skin lesions from patients with psoriasis vulgaris. J Autoimmun 52 Weatherhead SC, Farr PM, Reynolds NJ. Spectral effects of UV on
2013; 41: 1724. psoriasis. Photochem Photobiol Sci 2013; 12: 4753.
29 Zhang P, Su Y, Zhao M et al. Abnormal histone modifications in 53 Kim GK, Del RJ. Drug-provoked psoriasis: is it drug induced or
PBMCs from patients with psoriasis vulgaris. Eur J Dermatol 2011; drug aggravated?: understanding pathophysiology and clinical rele-
21: 552557. vance. J Clin Aesthet Dermatol 2010; 3: 3238.
30 Di Meglio P, Villanova F, Nestle FO. Psoriasis. Cold Spring Harb 54 Ishii-Osai Y, Yoneta A, Mizugaki N et al. Infliximab treatment-
Perspect Med 2014; 4: a015354. induced paradoxical psoriasiform reaction in patient with psoriasis
31 Qureshi AA, Dominguez PL, Choi H et al. Alcohol intake and risk of vulgaris showing positive lymphocyte transportation test reaction.
incident psoriasis in US women: a prospective study. Arch Derma- JAAD Case Rep 2015; 1: 230233.
tol 2010; 146: 13641369. 55 Vinter H, Iversen L, Steiniche T et al. Aldara(R)-induced skin inflam-
32 Ho J, Antrum JH. Severity of psoriasis among adult males is asso- mation: studies of patients with psoriasis. Br J Dermatol 2015; 172:
ciated with smoking, not with alcohol use. Indian J Dermatol 2014; 345353.
59: 516. 56 Flutter B, Nestle FO. TLRs to cytokines: mechanistic insights from
33 Parisi R, Symmons DP, Griffiths C et al. Global epidemiology of the imiquimod mouse model of psoriasis. Eur J Immunol 2013; 43:
psoriasis: a systematic review of incidence and prevalence. J 31383146.
Invest Dermatol 2013; 133: 377385. 57 Dalkilic E, Bulbul BE, Alkis N et al. Tumor necrosis factor-alpha
34 Wei S, Gao M, Zhang X et al. [Study of the association between antagonist therapy-induced psoriasis in Turkey: analysis of 514
HLA-DQA1 alleles and environmental factors in psoriasis]. Zhon- patients. Mod Rheumatol 2012; 22: 738742.
ghua Liu Xing Bing Xue Za Zhi 2002; 23: 445448. 58 Sahuquillo-Torralba A, Ballester-Sanchez R, Pujol-Marco C et al.
35 Jin Y, Yang S, Zhang F et al. Combined effects of HLA-Cw6 and Pembrolizumab: a new drug that can induce exacerbations of pso-
cigarette smoking in psoriasis vulgaris: a hospital-based case-con- riasis. Actas Dermosifiliogr 2016; 107: 264266.
trol study in China. J Eur Acad Dermatol Venereol 2009; 23: 132 59 Li W, Han J, Qureshi AA. Smoking and risk of incident psoriatic
137. arthritis in US women. Ann Rheum Dis 2012; 71: 804808.
36 Naldi L, Mercuri SR. Smoking and psoriasis: from epidemiology to 60 Eder L, Shanmugarajah S, Thavaneswaran A et al. The association
pathomechanisms. J Invest Dermatol 2009; 129: 27412743. between smoking and the development of psoriatic arthritis among
37 Duffin KC, Freeny IC, Schrodi SJ et al. Association between IL13 psoriasis patients. Ann Rheum Dis 2012; 71: 219224.
polymorphisms and psoriatic arthritis is modified by smoking. J 61 Chung HY, Machado P, van der Heijde D et al. Smokers in early
Invest Dermatol 2009; 129: 27772783. axial spondyloarthritis have earlier disease onset, more disease
38 Li W-Q, Han JL, Zhang M et al. Interactions between adiposity and activity, inflammation and damage, and poorer function and health-
genetic polymorphisms on the risk of psoriasis. Br J Dermatol related quality of life: results from the DESIR cohort. Ann Rheum
2013; 168: 639642. Dis 2012; 71: 809816.
39 Quan C, Zhu KJ, Zhang C et al. Combined effects of the BDNF 62 Poddubnyy D, Haibel H, Listing J et al. Cigarette smoking has a
rs6265 (Val66Met) polymorphism and environment risk factors on dose-dependent impact on progression of structural damage in the
psoriasis vulgaris. Mol Biol Rep 2014; 41: 70157022. spine in patients with axial spondyloarthritis: results from the GEr-
40 Bai J, Liu Z, Xu Z et al. Epigenetic downregulation of SFRP4 con- man SPondyloarthritis Inception Cohort (GESPIC). Ann Rheum Dis
tributes to epidermal hyperplasia in psoriasis. J Immunol 2015; 2013; 72: 14301432.
194: 41854198. 63 Westhoff G, Rau R, Zink A. Rheumatoid arthritis patients who
41 Zhou F, Wang W, Shen C et al. Epigenome-wide association anal- smoke have a higher need for DMARDs and feel worse, but they
ysis identified nine skin DNA methylation loci for psoriasis. J Invest do not have more joint damage than non-smokers of the same
Dermatol 2016; 136: 779787. serological group. Rheumatology (Oxford) 2008; 47: 849854.
42 Yan S, Xu Z, Lou F et al. NF-kappaB-induced microRNA-31 pro- 64 Hojgaard P, Glintborg B, Hetland ML et al. Association between
motes epidermal hyperplasia by repressing protein phosphatase 6 tobacco smoking and response to tumour necrosis factor alpha
in psoriasis. Nat Commun 2015; 6: 7652. inhibitor treatment in psoriatic arthritis: results from the DANBIO
43 Gupta R, Ahn R, Lai K et al. Landscape of long noncoding RNAs registry. Ann Rheum Dis 2015; 74: 21302136.
in psoriatic and healthy skin. J Invest Dermatol 2016; 136: 603 65 Moriwaki Y, Takada K, Tsuji S et al. Transcriptional regulation of
609. SLURP2, a psoriasis-associated gene, is under control of IL-22 in
44 Szell M, Danis J, Bata-Csorgo Z et al. PRINS, a primate-specific the skin: a special reference to the nested gene LYNX1. Int
long non-coding RNA, plays a role in the keratinocyte stress Immunopharmacol 2015; 29: 7175.
response and psoriasis pathogenesis. Pflugers Arch 2016; 468: 66 Votrubova J, Juzlova K, Dzambova M et al. Cardiovascular comor-
935943. bidities in patients with psoriasis: risk profile including carotide
45 Enamandram M, Kimball AB. Psoriasis epidemiology: the interplay ultrasonography assessed in hospital-based case control study.
of genes and the environment. J Invest Dermatol 2013; 133: 287 Acta Dermatovenerol Croat 2016; 24: 187192.
289. 67 Farkas A, Kemeny L. Alcohol, liver, systemic inflammation and
46 LoPiccolo MC, Lim HW. Vitamin D in health and disease. Photo- skin: a focus on patients with psoriasis. Skin Pharmacol Physiol
dermatol Photoimmunol Photomed 2010; 26: 224229. 2013; 26: 119126.
47 Gniadecki R, Gajkowska B, Hansen M. 1,25-dihydroxyvitamin D3 68 Poikolainen K, Reunala T, Karvonen J et al. Alcohol intake: a risk
stimulates the assembly of adherens junctions in keratinocytes: factor for psoriasis in young and middle aged men? BMJ 1990;
involvement of protein kinase C. Endocrinology 1997; 138: 2241 300: 780783.
2248. 69 Adamzik K, McAleer MA, Kirby B. Alcohol and psoriasis: sobering
48 Wolf P, Weger W, Patra V et al. Desired response to phototherapy thoughts. Clin Exp Dermatol 2013; 38: 819822.
versus photo-aggravation in psoriasis: what makes the difference? 70 Brenaut E, Horreau C, Pouplard C et al. Alcohol consumption and
Exp Dermatol 2016; 25: 937944. psoriasis: a systematic literature review. J Eur Acad Dermatol
49 de Gruijl FR. For better or for worse, UV in psoriasis. Exp Dermatol Venereol 2013; 27(Suppl 3): 3035.
2016; 25: 945946. 71 Farkas A, Kemeny L, Szell M et al. Ethanol and acetone stimulate
50 Jacobson CC, Kumar S, Kimball AB. Latitude and psoriasis preva- the proliferation of HaCaT keratinocytes: the possible role of alco-
lence. J Am Acad Dermatol 2011; 65: 870873. hol in exacerbating psoriasis. Arch Dermatol Res 2003; 295: 5662.

2017 Japanese Dermatological Association 871


J. Zeng et al.

72 Bardazzi F, Balestri R, Baldi E et al. Correlation between BMI and 97 Ivanov II, Atarashi K, Manel N et al. Induction of intestinal Th17
PASI in patients affected by moderate to severe psoriasis undergo- cells by segmented filamentous bacteria. Cell 2009; 139: 485498.
ing biological therapy. Dermatol Ther 2010; 23(Suppl 1): S14S19. 98 Atarashi K, Tanoue T, Shima T et al. Induction of colonic regulatory
73 Millsop JW, Bhatia BK, Debbaneh M et al. Diet and psoriasis, part T cells by indigenous Clostridium species. Science 2011; 331:
III: role of nutritional supplements. J Am Acad Dermatol 2014; 71: 337341.
561569. 99 Danese S. New therapies for inflammatory bowel disease: from the
74 Bhatia BK, Millsop JW, Debbaneh M et al. Diet and psoriasis, part bench to the bedside. Gut 2012; 61: 918932.
II: celiac disease and role of a gluten-free diet. J Am Acad Derma- 100 Lindqvist U, Kristjansson G, Pihl-Lundin L et al. Patients with psori-
tol 2014; 71: 350358. atic arthritis have an increased number of lymphocytes in the duo-
75 Wolk K, Mallbris L, Larsson P et al. Excessive body weight and denal mucosa in comparison with patients with psoriasis vulgaris.
smoking associates with a high risk of onset of plaque psoriasis. J Rheumatol 2006; 33: 924927.
Acta Derm Venereol 2009; 89: 492497. 101 Scarpa R, Manguso F, DArienzo A et al. Microscopic inflammatory
76 Michaelsson G, Gerden B, Hagforsen E et al. Psoriasis patients changes in colon of patients with both active psoriasis and psori-
with antibodies to gliadin can be improved by a gluten-free diet. Br atic arthritis without bowel symptoms. J Rheumatol 2000; 27:
J Dermatol 2000; 142: 4451. 12411246.
77 Michaelsson G, Ahs S, Hammarstrom I et al. Gluten-free diet in 102 Van Praet L, Van den Bosch F, Mielants H et al. Mucosal inflam-
psoriasis patients with antibodies to gliadin results in decreased mation in spondylarthritides: past, present, and future. Curr
expression of tissue transglutaminase and fewer Ki67+ cells in the Rheumatol Rep 2011; 13: 409415.
dermis. Acta Derm Venereol 2003; 83: 425429. 103 Christophers E. Comorbidities in psoriasis. Clin Dermatol 2007; 25:
78 Frikha F, Snoussi M, Bahloul Z. Osteomalacia associated with 529534.
cutaneous psoriasis as the presenting feature of coeliac disease: a 104 Binus AM, Han J, Qamar AA et al. Associated comorbidities in
case report. Pan Afr Med J 2012; 11: 58. psoriasis and inflammatory bowel disease. J Eur Acad Dermatol
79 Debbaneh M, Millsop JW, Bhatia B et al. Diet and psoriasis, part I: Venereol 2012; 26: 644650.
impact of weight loss interventions. J Am Acad Dermatol 2014; 71: 105 Munz OH, Sela S, Baker BS et al. Evidence for the presence of
133140. bacteria in the blood of psoriasis patients. Arch Dermatol Res
80 Edson-Heredia E, Sterling KL, Alatorre C et al. Heterogeneity of 2010; 302: 495498.
response to biologic treatment: perspective for psoriasis. J Invest 106 Arck P, Handjiski B, Hagen E et al. Is there a gut-brain-skin axis?
Dermatol 2014; 134: 1823. Exp Dermatol 2010; 19: 401405.
81 Papp KA, Langley RG, Lebwohl M et al. Efficacy and safety of ustek- 107 Baker BS, Powles A, Fry L. Peptidoglycan: a major aetiological
inumab, a human interleukin-12/23 monoclonal antibody, in patients factor for psoriasis? Trends Immunol 2006; 27: 545551.
with psoriasis: 52-week results from a randomised, double-blind, 108 Naldi L, Chatenoud L, Linder D et al. Cigarette smoking, body
placebo-controlled trial (PHOENIX 2). Lancet 2008; 371: 16751684. mass index, and stressful life events as risk factors for psoriasis:
82 Ivanov II, Honda K. Intestinal commensal microbes as immune results from an Italian case-control study. J Invest Dermatol 2005;
modulators. Cell Host Microbe 2012; 12: 496508. 125: 6167.
83 Hajishengallis G, Darveau RP, Curtis MA. The keystone-pathogen 109 Park BS, Youn JI. Factors influencing psoriasis: an analysis based
hypothesis. Nat Rev Microbiol 2012; 10: 717725. upon the extent of involvement and clinical type. J Dermatol 1998;
84 Eppinga H, Konstantinov SR, Peppelenbosch MP et al. The micro- 25: 97102.
biome and psoriatic arthritis. Curr Rheumatol Rep 2014; 16: 407. 110 Chamoun A, Goudetsidis L, Poot F et al. [Psoriasis and depres-
85 Ottman N, Smidt H, de Vos WM et al. The function of our micro- sion]. Rev Med Brux 2015; 36: 2328.
biota: who is out there and what do they do? Front Cell Infect 111 Martin-Brufau R, Romero-Brufau S, Martin-Gorgojo A et al. Psoria-
Microbiol 2012; 2: 104. sis lesions are associated with specific types of emotions. Emo-
86 Drago L, De Grandi R, Altomare G et al. Skin microbiota of first tional profile in psoriasis. Eur J Dermatol 2015; 25: 329334.
cousins affected by psoriasis and atopic dermatitis. Clin Mol 112 Prizment AE, Alonso A, Folsom AR et al. Association between pso-
Allergy 2016; 14: 2. riasis and incident cancer: the Iowas Womens Health Study. Can-
87 Zanvit P, Konkel JE, Jiao X et al. Antibiotics in neonatal life cer Causes Control 2011; 22: 10031010.
increase murine susceptibility to experimental psoriasis. Nat Com- 113 Frankel HC, Han J, Li T et al. The association between physical
mun 2015; 6: 8424. activity and the risk of incident psoriasis. Arch Dermatol 2012; 148:
88 Baker BS, Laman JD, Powles A et al. Peptidoglycan and peptido- 918924.
glycan-specific Th1 cells in psoriatic skin lesions. J Pathol 2006; 114 Mead GE, Morley W, Campbell P et al. Exercise for depression.
209: 174181. Cochrane Database Syst Rev 2009; 3: D4366.
89 Fry L, Baker BS. Triggering psoriasis: the role of infections and 115 Nair RP, Duffin KC, Helms C et al. Genome-wide scan reveals
medications. Clin Dermatol 2007; 25: 606615. association of psoriasis with IL-23 and NF-kappaB pathways. Nat
90 Nakatsuji T, Chiang HI, Jiang SB et al. The microbiome extends to Genet 2009; 41: 199204.
subepidermal compartments of normal skin. Nat Commun 2013; 4: 116 Indhumathi S, Rajappa M, Chandrashekar L et al. Investigation of
1431. association of the IL-12B and IL-23R genetic variations with psori-
91 Blaser MJ, Kirschner D. The equilibria that allow bacterial persis- atic risk in a South Indian Tamil cohort. Hum Immunol 2016; 77:
tence in human hosts. Nature 2007; 449: 843849. 5462.
92 Gao Z, Tseng CH, Strober BE et al. Substantial alterations of the 117 Stuart PE, Nair RP, Tsoi LC et al. Genome-wide association analy-
cutaneous bacterial biota in psoriatic lesions. PLoS ONE 2008; 3: sis of psoriatic arthritis and cutaneous psoriasis reveals differences
e2719. in their genetic architecture. Am J Hum Genet 2015; 97: 816836.
93 Elson CO, Alexander KL. Host-microbiota interactions in the intes- 118 Capon F, Bijlmakers MJ, Wolf N et al. Identification of ZNF313/
tine. Dig Dis 2015; 33: 131136. RNF114 as a novel psoriasis susceptibility gene. Hum Mol Genet
94 Fry L, Baker BS, Powles AV et al. Is chronic plaque psoriasis trig- 2008; 17: 19381945.
gered by microbiota in the skin? Br J Dermatol 2013; 169: 4752. 119 Paulino LC, Tseng CH, Strober BE et al. Molecular analysis of fun-
95 Sommer F, Backhed F. The gut microbiotamasters of host devel- gal microbiota in samples from healthy human skin and psoriatic
opment and physiology. Nat Rev Microbiol 2013; 11: 227238. lesions. J Clin Microbiol 2006; 44: 29332941.
96 Round JL, Mazmanian SK. The gut microbiota shapes intestinal 120 Paulino LC, Tseng CH, Blaser MJ. Analysis of Malassezia micro-
immune responses during health and disease. Nat Rev Immunol biota in healthy superficial human skin and in psoriatic lesions by
2009; 9: 313323. multiplex real-time PCR. FEMS Yeast Res 2008; 8: 460471.

872 2017 Japanese Dermatological Association

You might also like