You are on page 1of 7

Medicine in Microecology 4 (2020) 100016

Contents lists available at ScienceDirect

Medicine in Microecology
journal homepage: www.journals.elsevier.com/medicine-in-microecology

Role of skin and gut microbiota in the pathogenesis of psoriasis, an


inflammatory skin disease
Daniel K. Hsu a, Maxwell A. Fung a, Hung-Lin Chen b, *
a
Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, CA, USA
b
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan

A R T I C L E I N F O A B S T R A C T

Keywords: Psoriasis is a chronic inflammatory skin disease associated with epidermal keratinocyte hyperplasia and
Psoriasis epidermal immune cell over-activation. Compositions of both local skin and gut microbiome are linked to
Microbiota modulation of inflammation and disease severity in psoriasis. Owing to the situation that different bacteria may
Microbiome
elicit differential immune or inflammatory responses from epidermal immune cells and keratinocytes, and to-date
no single pathogen was highlighted to be responsible for psoriasis, disruption of homeostasis (dysbiosis) in the
original microbial ecosystems may create a disease-promoting environment, and as a whole may be a primary
causal factor. Several studies have provided evidence that the dominant IL-23/IL-17 pathogenesis pathway is
regulated by metabolites produced by gut and skin microbiota. This review summarizes the approaches
commonly used for functional characterization of the microbiome compositions associated with development of
clinical phenotypes of psoriasis. The underlying mechanisms by which microbiota modulate immune cells and
keratinocytes are also proposed.

1. Introduction infection and responds to antibiotic therapy [18,19]. However, antibi-


otics are atypical first-line therapies for psoriasis [20].
Psoriasis has been reported to affect 0.09%–11.4% of the general As the largest organ of the body, the skin provides a physical barrier
population worldwide [1]. It is among the most common chronic skin to injury and microbial insults. Keratinocytes and indigenous immune
inflammatory diseases associated with metabolic disorders, including cells play indispensable roles in cell renewal and antimicrobial function,
metabolic syndrome [2–4]. Psoriasis is characterized by epidermal ker- in general [21]. Epidermal keratinocytes secrete antimicrobial peptides
atinocyte hyperproliferation (associated with skin thickening) and [12,22,23], form tight junctions [24], and construct a physical barrier
inflammation (associated with skin redness) in the epidermis and dermis. [25] to control microbial infections through cornification [25–27].
Pruritus causes psychological stress in some patients. In addition, psori- Therefore, commensal bacteria may develop special mechanisms that
asis is associated with comorbidities such as obesity, hypertension, characterize their adaptation as microbiota. The microbial ecology of the
atherogenic dyslipidemia, type 2 diabetes, and inflammatory bowel skin is mostly composed of bacteria from Firmicutes, Actinobacteria, Bac-
disease [2,5–9]. Therefore, psoriasis represents a substantial cumulative teroidetes, and Proteobacteria phyla. Skin from different body regions each
disease burden on patient quality of life, including psychological health. have their unique microbiome ecology [28] with differences associated
The etiology of psoriasis is multifactorial and involves the interplay of with variations in temperature, moisture, and pH value [29]. This pro-
genetic and environmental factors that exacerbate innate and adaptive vides a unique colonization environment which favors the survival of
immune responses [10–12]. Although the clinical manifestations of some bacteria over others. Staphylococcus and Corynebacterium spp. are
psoriasis are in the skin, associated inflammation develops throughout typically the most abundant bacteria in moist sites (such as axillary vault,
the body [13]. Psoriasis is considered a multifactorial condition, and antecubital fossa and inguinal crease), whereas sebaceous sites such as
current therapies include antibodies to key cytokines associated with this glabella, alar crease and external auditory canal are enriched with Pro-
disease, such as Interleukin-23 (IL-23) [14,15], Interleukin-17 (IL-17) pionibacterium spp., and dry sites such as volar forearm and buttock are
[15,16], and tumor necrosis factor alpha (TNFα) [17]. One form of enriched with some Gram-negative bacilli [25,29–32]. In lesional pso-
psoriasis, guttate psoriasis, is associated with bacterial upper respiratory riasis, unlike normal skin, microbial colonization is mostly confined to

* Corresponding author. Institute of Biomedical Sciences, Academia Sinica, Taiwan.


E-mail address: hunglinc@gate.sinica.edu.tw (H.-L. Chen).

https://doi.org/10.1016/j.medmic.2020.100016
Received 18 December 2019; Received in revised form 6 May 2020; Accepted 9 May 2020
Available online 20 May 2020
2590-0978/© 2020 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-
nc-nd/4.0/).
D.K. Hsu et al. Medicine in Microecology 4 (2020) 100016

Table 1
Summary of previous studies on changes in the skin microbiota of psoriasis patients.
Reference Samples Methods Bacteria enriched in psoriasis Bacteria decreased in
psoriasis

Gao et al., Samples collected from 6 patients, 19 Broad-range 16S rDNA PCR and Firmicutes and Streptococcus Actinobacteria and
2008 cutaneous samples (13 from diseased taxonomical assignment using Ribosomal Propionibacterium
skin and 6 from normal skin). Database Project II (RDP II).
Fahlen et al., Skin biopsies taken from 10 for pyrosequencing on the 454-FLX GS- Proteobacteria and Streptococci Actinobacteria,
2011 patientswith Psoriasis and 12 control 100 platformthe. Ribosomal Staphylococci and
subjectsfrom unmatched sites. databaseproject (RDP) sequence data were Propionibacteria
used for analysis.
Alekseyenko Psoriatic lesions (lesion Targeting 16 S rRNA gene V1–V3 Corynebacterium, Propionibacterium, Acidobacteria Gp4 and
et al., 2013 group).unaffected contralateral skin regionsfor pyrosequencing on the 454 Staphylococcus and Streptococcus Schlegelella
frompsoriatic patients (unaffected platform and RDP Classifier was used for
group), and similar skin loci in sequence analysis.
matched healthy controls (control
group) 51 matched triplets
Drago et al., Samples from three first cousin, two Targeting 16 S rRNA gene V2-4-8 and V3- Proteobacteria, Streptococcaceae, Firmicutes,
2015 lesional skin samples and two non- 6, 7–9 regions for sequecing use Ion Rhodobacteraceae, Staphylococcaceae,
lesional skin samples (for AD Torrent PGM platform. A Campylobacteraceaeand Moraxellaceae Propionibacteriaceae and
andpsoriatic subjects) were taken. curatedGreengenes databased was used Propionibacterium acnes
fortaxonomical analysis.
Assarsson Sample collected from 30 patients with Targeting 16 S rRNA gene V3–V4 for Psoriasis patients skin enrich Firmicutes and
et al., 2018 plaque type Psoriasis with indication MiSeq sequencing, and data was analyzed inConchiformibius, while nbUVB Staphylococcus
for nbUVB treatment by CLC Software map to Greengene treatement non-responders enrich in
V13⋅516 S rRNA sequencing (V1–V2) and Staphylococcus, Finegoldia, Anaerococcus,
Peptoniphilus, Gardnerella, Prevotella and
Clostridium.
Langan et al., Swabs from lesional and nonlesional traditional culture combined with Firmicutes, Prevotella, Staphylococcu and Actinobacteria,
2019 skin from 23 patients and 20 healthy massspectrometry (MALDI-TOF). RDP Corynebacterium Anaerococcus and
controls Gold database version 9 and SILVA Propionibacterium
database(version 123) was used as
reference database.
Quan et al., Sample colleced from lesional Targeting 16 S rRNA gene V3–V4 Coryne bacterium, Brevun dimonas and Cutibacterium,
2019 andunaffected skin of 16 patients and regionand library were made and Micrococcus Streptococcus and
6 healthy controls sequenced by Miseq-system.RDP Classifier Deinococcus
was used for sequnce analysis. Targeting
16 S rRNA gene sequencing use
Fyhrquist Sample collected from healthy 454, and Greengene C. simulans and C. kroppenstedtii as well Corynebacterium spp.,
et al., 2019 controls(n ¼ 115), AD (n ¼ 82) and database.Metagenomic shotgun asFinegoldia, Neisseriaceae species and Lactobacilli, Burkholderia
PSO (n ¼ 119). sequencing use illumina HiSeq and Coryn e bacteria spp. and P. acnes
analyzed by MetaPhlan2 and humann2.

AD-atopic dermatitis; nbUVB-narrow band ultraviolet B; PSO-psoriasis.

the epidermis. In lesional psoriatic skin, pruritus-induced scratching can reported to produce short chain fatty acid (SCFA) that exert
cause wounding to the skin; therefore some bacteria (epidermal colo- anti-inflammatory functions [50,51]. Dysbiosis of the gut microbiome
nizers and other bacteria) can be found in the deep dermis [33] or even may also exacerbate psoriasis, wherein the interactions of different
peripheral blood [34] where they readily encounter immune cells and bacterial populations in the skin and gut microbiome play key roles in the
elicit innate and adaptive inflammation [35]. This causes dysbiosis of pathogenesis of psoriasis. The aim of this review is to identify a link
skin microbiota as observed in a significantly reduced population of between the microbial ecology of the gut and skin, and their relevance to
Corynebacterium spp., Lactobacillus spp., Burkholderis spp., and Propioni- psoriasis.
bacterium acnes in lesional psoriatic skin compared with healthy skin
[36]. 2. Methods
The gut microbiome in psoriasis is also enriched in Firmicutes and
Actinobacteria spp [37,38]. Dysbiosis may cause changes in metabolite We searched NCBI PubMed for articles having both “psoriasis” and
production and elicit anti-microbial signaling, and these may affect im- “microbiome” as keywords. The search yielded 174 articles, including 81
mune cell activation through the IL-23/IL-17 signaling pathway through review articles. Selected articles on skin microbiome and gut microbiome
IL-22 and interferon gamma (IFN-γ) production, leading to keratinocyte analyses in psoriatic patients are summarized in Table 1 and Table 2,
hyperproliferation. Besides genetic predisposition, dietary and life-style respectively.
factors are known key regulators of gut microbiome homeostasis [39,
40]. Gut dysbiosis also promotes other inflammatory diseases such as
inflammatory bowel disease (IBD) [41], including Crohn disease [42]. In 2.1. Genetic and environmental factors involved in psoriasis pathogenesis
addition, in psoriatic patients who also have IBD, the severity of symp-
toms of psoriasis correlates with that of IBD [9,43–46]. Psoriatic patients Psoriasis is a complex disease in which genetic susceptibility loci and
have 3-fold higher risk of developing Crohn disease, and Crohn disease environmental factors contribute to the disease phenotype. In the past
patients have a 7-fold higher risk of developing psoriatic lesions, decade, several psoriasis susceptibility loci (PSORSs) were identified [10,
compared to heathy individuals [47]. This is mainly because pathogenic 52,53]. Genes and allele within the PSORS1 locus (human leukocyte
T helper type 17 (Th17) cells and Th17 cytokines are associated with antigen (HLA-Cw6) [54], alpha-helix coiled-coil rod homolog
these chronic inflammatory diseases [48]. Reduction in the numbers of (HCR*WWCC) haplotype [55] and HLA-associated S genes [56]) were
specific bacteria (Faecalibacterium prausnitzii and Akkermansia mucini- also discovered. Furthermore, HLA-B, HLA-C, HLA-C12, HLA-DQAT, and
phila) have been reported in psoriasis [46,49], and these bacteria are endoplasmic reticulum aminopeptidase 1 (ERAP1), which are responsible
for antigen presentation and processing, were also identified as

2
D.K. Hsu et al. Medicine in Microecology 4 (2020) 100016

contributory. In addition to these factors, the following are all involved in fragment amplicon, and mass-spectrometry, data of different studies can
the pathogenesis of psoriasis: Nuclear factor kappa B (NF-ƘB) signaling, only be partially integrated [30,31,36,70–77]. Metagenomic shotgun
Janus kinases (JAKs) signaling, transforming growth factor beta (TGF-β) sequencing is an advanced and very informative sequencing technology
signaling, T cell regulation, interleukin 23–interleukin 17 (IL-23-IL-17) but also expensive; therefore, more limited cost-effective sequencing of
signaling, disruption of epithelial barrier function, and dysregulated 16 S rRNA V1-3 and/or V3–V4 regions are commonly used for micro-
apoptosis and autophagy, [10,57,58]. Environmental risk factors biome analyses. Some studies have suggested that V1–V3 is more
including ultraviolet exposure [59], medications, including [anti- appropriate for cutaneous microbiome profiling; however, V4 and V3–V4
proliferative agents (imiquimod), antivirals and antidepressants amplicons are suitable for fecal microbiome analysis. Apart from
(lithium), antihypertensives (beta-blockers) and TNF-α] [60,61], smok- sequencing technologies, the use of appropriate databases for the map-
ing [62], obesity [63] and alcohol intake [64], as well as stress [62] and ping of phylogenetic information is also critical. To that end, we sum-
infection [65], combine with the genetic burden to initiate and exacer- marize different studies and the associated technologies for psoriasis in
bate epidermal inflammation [11]. The innate immune response plays a Table 1.
critical role in an imiquimod (IMQ)-induced psoriasis mouse model [66]. In psoriasis, the relative abundance of Streptococcus pyogenes increases
The activation of TLR-7 by IMQ triggers IL-23 secretion from conven- in swabs compared with healthy controls and non-lesional skin [76,77].
tional dendritic cells (DCs). In addition, IMQ directly induces M protein of Streptococcus has sequence homology with the 50-kDa type I
interleukin-36 alpha (IL-36α) expression in bone-marrow derived Lang- keratin, suggesting that streptococcal super antigen-induced T cells
erhans cells, and in granulocyte-macrophage colony-stimulating factor target skin-specific antigens through molecular mimicry, which leads to
(GM–CSF)–induced DCs. IL-36α then acts on bone-marrow derived chronic inflammation [78,79]. Dysbiosis is commonly observed in the
Langerhans cells and keratinocytes to produce IL-23, interleukin 1 beta skin of psoriasis patients. Yan et al. reviewed the analysis of changes in
(IL-1β), C–C Motif Chemokine Ligand 20 (CCL20), and C-X-C Motif microbiome alpha-diversity [73]. A large cohort study reported 75 pso-
Chemokine Ligand 1, 2 (CXCL1 and CXCL2). Apart from IMQ, IL-36α can riasis cases (both lesional and non-lesional) and 124 controls in analyses
be induced in DCs by other microorganism ligands such as the β-glucan of skin swab microbiomes [31,73]. Significant decreases in richness and
from Candida albicans, suggesting that dysbiosis of the skin microbiome Shannon-index were reported in lesional samples compared with
may cause a psoriasis-like (psoriasiform) dermatitis [66]. In humans, the non-lesional and control samples. This study also included a longitudinal
critical cellular and molecular pathways in lesional psoriatic skin are analysis (17 psoriasis patients and 15 controls) that examined micro-
orchestrated by the activation of dermal DCs, which secrete IL-23 to biome changes in systematic therapies (i.e. methotrexate and TNF-α in-
promote IL-17 production by gamma delta (γδ) T cells and type 3 innate hibitors). At 12 weeks after systematic therapy, the richness and
lymphoid cells (ILC3), but not αβ T cells (Fig. 1). IL-17 then stimulates Shannon-index declined for both lesional and non-lesional samples, but
keratinocytes to produce chemokines such as CXCL1, CXCL2, CCL20, at 36 weeks, only the richness and Shannon-index of the control samples
interleukin 6 (IL-6), and interleukin 8 (IL-8), causing leukocyte infiltra- returned to the baseline, suggesting an interplay between the immune
tion. The ILC3 can respond to stimulating cytokines including IL-1β, system and microbiome ecology. In addition, anti-microbial substances
interleukin 18 (IL-18) and IL-23 by secreting IL-22 and IL-17 [67], which are secreted in the lesional site, and hence, lesional skin lacks adequate
stimulate keratinocyte hyperproliferation. Topical imiquimod applica- environmental support for microbiota colonization. Fyhrquist et al.
tion or intradermal IL-23 injection can induce skin inflammation and conducted a microbiome analysis of skin swabs in 115 healthy volunteers
psoriasiform plaques in a mouse psoriasis model. However, (HV), 82 atopic dermatitis (AD), and 119 psoriasis (PSO) subjects using
antibiotic-treated or germ-free mice failed to establish chronic skin 16 S rRNA V3–V4 sequencing platform and whole genome sequencing.
inflammation and exhibited reduced psoriasiform plaque formation [68]. They found that some bacterial distributions showed significant differ-
This suggests that the microbiome on lesional skin, and possibly other ential changes even after adjusting for confounding factors (e.g. Cory-
niches, play important roles in the triggering of prolonged inflammation nebacterium kroppenstedii was associated with age, Staphylococcus aureus
and the establishment of psoriasis. Recently, CD109, a glycoprotein with anatomical location, and Lactobacillus sp. with gender), [36]. In
expressed in keratinocytes, was found to suppress murine psoriasiform lesional psoriasis, the abundance of Corynebacterium simulans, Coryne-
dermatitis [69], occurring through regulation of the infiltration of γδT bacterium kroppenstedii, Finegoldia spp., and Neisseria spp. increased;
cells, and thus impact of IL23/17 signaling; a potential involvement of however, that of Lactobacilli, Burkholderia spp. and Propionibacterium
TGFβ in skin homeostasis may also exist. Keratinocytes, which are pri- acnes decreased, compared with that in healthy skin. Other studies also
marily responsible for the formation of the skin barrier, are known to reported that the abundance of Corynebacterium is associated with the
produce several antimicrobial peptides and proteins that assist in the severity of local lesions [70]; the abundance of Corynebacterium and
maintenance of the normal microbial ecosystem. Dysbiosis of the Staphylococcus significantly correlated with the psoriasis area severity
microbiome commonly occurs in skin diseases including atopic derma- index (PASI) scores. A co-occurrence relationship analysis reported that
titis and psoriasis. Therefore, the normal skin microbiome contains both among 24 microbes identified for psoriasis classification, 16 species
beneficial and harmful microbes, which regulate resident immune cells showed significant correlation as determined by an operational tax-
and keratinocytes, and these cells actively participate in the development onomical unit (OTUs) correlation analysis tool, SparCC [36]. Coryne-
of inflammation. bacterium simulans and Corynebacterium kroppenstedii displayed positive
correlation with Streptococcus spp., Peptostreptococcus anaerobius, Anae-
2.2. Dysbiosis of the skin microbiome in psoriasis rococcus spp., Neisseria spp., and Rothia dentocariosa. This suggests that
the microbiome ecosystem of psoriasis contains communities of both
In a previous study, the sampling procedure for cutaneous micro- pathogenic and non-pathogenic bacteria with potential mutualism. In
biome analysis (i.e., swab vs scrape vs biopsies) using matched controls addition, several studies have suggested that a single taxonomy is unable
(i.e., healthy individual vs non-lesional sites from patients) was noted to to discriminate psoriasis from healthy skin. The loss of homeostasis and
be more challenging compared to sampling the fecal microbiome [33]. the presence of specific bacterial genera such as Cutibacterium, Strepto-
The most abundant microbial phyla in the skin swabs were Firmicutes, coccus, Deinococcus, Actinobacteria, Staphylococci, Propionibacteria, Anae-
Actinobacteria, and Proteobacteria. It is worthy of note that the order rococcus, and Corynebacterium spp. also contribute to psoriasis.
Clostriadales and Bacteroides were more enriched in biopsy samples Corynebacterium spp. are negatively associated with the interferon
compared to swab samples. signaling pathway, and this may lead to changes in its symbiotic
In addition, due to the different analytic technologies available, for microbiome, contributing to the pathogenesis of psoriasis [34,36,70–73,
example, species-specific polymerase chain reaction (PCR), cultured- 77], (Fig. 1).
bacterial PCR, 16 S rRNA gene full length PCR and 16 S rRNA gene

3
D.K. Hsu et al.
Table 2
Summary of previous studies on gut microbiota changes in psoriasis patients.
Reference Samples Methods Bacteria enriched in psoriasis Bacteria decreased in psoriasis

Scheret al. Fecal samples from PsA (n ¼ 16), Patients with Targeting 16 S rRNA gene V1–V2 regionfor Bocteroidetes Akkermansia, Ruminococcus, Akkermansia,
2015 Psoriasis of the skin(n ¼ 15), and healthy, matched sequencing by 454 pyrosequencing method Coprococcus
control subjects (n ¼ 17). species and Coprobacillus
Tan et al., 2018 Healthy controls (n ¼ 14) and patients with vulgaris Targeting 16 S rRNA gene V4 region for sequencing Bocteroidaceae, Enterococcaceae, Akkermansia muciniphila, Verrucomicrobia,
Psoriasis (n ¼ 14) wererecruited. and analyze using RibosomalDatabase Project (RDP) Enterococcus, Bacteroides andClostridium Jenericutes, Mollicutes,
Naive Bayesian Classifier v.2.2. citroniae Verrucomicrobiae,Verrucomicrobiaes and
RF39
Codoner et al., 52 patients and over 300 healthyindividuals extracted The V3–V4 of the bacterial 16s rRNA genewas Streptococcus, Bifidobocterium, Bacteroides
2018 from the humanmicrobiome project(http://hmp amplified and sequenced with aMiSeq lllumina Akkermansia, Akkermonsia spp and
dacc.org/). Platform and analyzedusing NCBI 16s rRNA Faecalibacterium
database.
Chen et al., Psoriasis (n ¼ 32) and age-, gender- and body mass Targeting 16S rRNA gene V3-V4 region for Firmicutes, Ruminococcus and Megasphaera Bacteroides
2018 index (BMI)-matched non-psoriasis subjects (n ¼ 64). sequencing by lllumina MiSeq 2000 platform and
analyze using Greengenes 13.5 database.
Shapiro et al., psoriasis subjects (n ¼ 64)Fecal samples from 24 platform and analyze using Greengenes13_5 Firmicutes, Blautia, Faecalibacterium,Ruminococcus, Lachnospira, Collinsella, Sutterella and
4

2019 Psoriaticpatients and 22 Control participants databaseThe V4 region of the bacterial 16 S Prevotella,Coprococcus, Bifidobacterium, Dorea and Actinomyces
rRNAgene was amplifiedand sequenced using the Christensenella
lllumina MiSeq platform and analyzed using
Greengenedatabase.
Hidalgo- The faecal microbiota of 19 patientswith Psoriasis and Ion 16STMMetagenomics Kit targeting the V2–V3 Firmicutes, Actinobacteria, Bifidobacteriaceae, Bacteroidaceae, Barnesiellaceae,
Cantabrana 20 healthy individuals from the same hypervariable regions, and sequence wasmapped to Coriobacteriaceae, Lachnospiraceae,Clostridiales Prevotellaceae, Tannerellaceae,
et al., 2019 geographiclocation. SILVA database vl32 99. Family XIII, Eggerthellaceae,Peptostreptococcaceae, Burkholderiaceae, Rikenellaceae,
Ruminococcaceae andE rysipelotrichaceae Lactobacillaceae, Streptococcaceae,
Desulfovibrionaceae, Veillonellaceae,
Marinifilaceae, Victivallaceae and
Pasteurellaceae
Huang et al., 35 Psoriasis patients in this study.Among them, 16 The V4 and V5 regions of the bacterial 16 S ribosomal Bacteroidetes, Bacillus, Bacteroides,Sutterella, Firmicutes, Thermus, Streptococcus, Rothia,
2019 were diagnosed with Psoriasis vulgaris (VUL), eight RNA gene were amplifiedand sequenced by lllumina Lactococcus, Lachnospiraceae_ UCG004,Lachnospira, Granulicatella, Gordonibacter,
withPsoriasis pustulosa (PUS), seven withPsoriasis MiSeqplatform. Mitochondria_norank,Cyanobacteria_noran and Allobaculumand Carnobacterium
arthropathica (ART), and fourwith erythroderma Parabacteroides B1:F40D35B2:FB1:F40
psoriaticum (DER).

Medicine in Microecology 4 (2020) 100016


PsA-psoriatic arthritis.
D.K. Hsu et al. Medicine in Microecology 4 (2020) 100016

Fig. 1. Pathogenesis of psoriasis and interactions of the immune system, keratinocytes, and skin microbiome. In humans, dysbiosis of the microbiome ecosystem may
trigger the activation of conventional dendritic cells (cDC) in the dermis to secrete IL-23 ①. IL-23 consequently promotes γδ T cells and Th17 cells to produce IL-17 ②.
In addition, type 3 innate lymphoid cells (ILC3) can respond to stimulating cytokines including IL-1β, IL-18 and IL-23, and secrete IL-17, IL-22 and IFN-γ to act on
keratinocytes ③. IL-17 stimulates keratinocytes to produce chemokines such as CXCL1, CXCL2, CXL20, IL-6, and IL-8, resulting in leukocyte infiltration ④. Infiltrated
leukocytes can further produce IL-1β and IL-18 to stimulate ILC3 cells to produce more IL-22, IL-22 thus promoting keratinocyte hyperproliferation ⑤. During skin
inflammation, hyperproliferating keratinocytes can produce more antimicrobial peptides (AMPs) such as S100 calcium-binding protein (S100A), cathelicidin anti-
microbial peptides LL-37 and beta defensin (β-defensin). The effect of these antimicrobial peptides on the microbiome at the lesional site remains to be investigated.
However, the dysbiosis of the skin microbiome in lesional skin has been characterized by several studies that identified the enrichment of Streptococcus pyogenes,
Corynebacterium, Propionibacterium, Staphylococcus, Streptococcus, Corynebacterium simulans, Corynebacterium kroppenstedtii, Finegoldia, and Neisseria spp. These
bacteria may create a proinflammatory environment that promotes the development of psoriasis through keratinocytes and resident immune cells.

2.3. Dysbiosis of gut microbiome in psoriasis psoriasis; however, some studies also found an enrichment of Bacter-
oidetes and a decrease of Actinobacteria in the gut of psoriasis subjects
Gut microbiome dysbiosis is associated with psoriasis, as well as other (Table 2). At the species level, psoriasis has an increased abundance of
diseases (e.g., metabolic syndrome, abdominal obesity, hypertension, Ruminococcus gnavus, Dorea formicigenerans, and Collinsella aerofaciens
type 2 diabetes, insulin resistance, and non-alcoholic fatty liver disease), [82]. Surprisingly, Akkermansia spp. and Faecalibacterium increased in a
and typically involves an impaired gut epithelial barrier [39–42]. Apart cohort of psoriasis patients [80]. Other studies demonstrated the deple-
from that, a dysfunction of gut microbiome function including activation tion of Akkermansia muciniphila and Faecalibacterium prausnitzii in psori-
of regulatory T cells, bacterial chemotaxis, and carbohydrate transport asis patients [49,83], which has also been reported in IBD and Crohn
have been predicted to be associated with an increase in the Firmicu- disease patients [42,49,84]. This discrepancy between studies again
tes-to-Bacteroidetes ratio (F/B ratio) and loss of some bacterial genera indicated the importance of using standard methods for sample collec-
(Ruminococcus and Megasphaera) [38]. In a human microbiome project tion, DNA extraction, sequence library preparation, and/or data analysis.
involving 52 psoriasis patients and 300 healthy controls, Codoner et al. Increased abundance of A. muciniphila causes reduced body weight and
reported that microbiome diversity was higher in psoriasis patients than gut barrier dysfunction [85]. Faecalibacterium prausnitzii is a butyrate
in the healthy controls [80]. The study of Scher et al. demonstrated that producer that fuels enterocytes and has an anti-inflammatory function
in a small cohort of samples (16 psoriatic arthritis (PsA), 15 psoriasis [86,87]. All these reports suggest that gut microbiome homeostasis is
(PSO), and 17 controls), the microbiome of the PSO group was less important for the maintenance of epithelial barrier integrity. The
diverse than that of the controls [81]. The discrepancy in diversity be- dysfunction of the gut barrier is associated with increased blood intes-
tween these two studies may have resulted from differences in sample tinal fatty acid binding protein (I-FABP) and bacterial DNA levels [34,
collection, DNA extraction, sequence library preparation, and/or data 88]. This evidence suggests that the regulation of gut microbes could
analyses. In general, at the plylum level, Firmicutes and Actinobacteria as have a potential ameliorating effect on skin inflammation by regulating
well as the Firmicutes-to-Bacteroidetes ratio (F/B ratio) increases in the systemic immune system [34,89–91].

5
D.K. Hsu et al. Medicine in Microecology 4 (2020) 100016

2.4. Regulation of the gut-skin axis attenuates psoriasis [6] Singh S, Young P, Armstrong AW. Relationship between psoriasis and metabolic
syndrome: a systematic review, Giornale italiano di dermatologia e venereologia :
organo ufficiale. Societa italiana di dermatologia e sifilografia 2016;151:663–77.
There are studies indicating that a gut-skin axis exists. For example, [7] Ramessur R, Gill D. The effect of statins on severity of psoriasis: a systematic review.
probiotics such as Bifidobacterium infantis 35,624 and Lactobacillus pen- Indian J Dermatol, Venereol Leprol 2017;83:154–61. https://doi.org/10.4103/
tosus GMNL-77 have shown beneficial effects in psoriasis patients, and in 0378-6323.188655.
[8] Ogdie A, Gelfand JM. Clinical risk factors for the development of psoriatic arthritis
mice with imiquimod-induced psoriasis [90,91]. The levels of serum among patients with psoriasis: a review of available evidence. Curr Rheumatol Rep
proinflammatory cytokines (i.e. TNF-α and IL-6) and plasma C-reactive 2015;17:64. https://doi.org/10.1007/s11926-015-0540-1.
protein (CRP) levels were reduced by probiotics. Higher circulating levels [9] Alinaghi F, Tekin HG, Burisch J, Wu JJ, Thyssen JP, Egeberg A. Global prevalence
and bidirectional association between psoriasis and inflammatory bowel disease - a
of these cytokines are also associated with psoriasis [92,93], and other systematic review and meta-analysis. J Crohns Colitis 2019. https://doi.org/
co-morbid diseases [94]. This implies that the immune modulatory 10.1093/ecco-jcc/jjz152.
function of probiotics is not limited to the gut mucosa. These observa- [10] Schon MP, Boehncke WH, Psoriasis. N Engl J Med 2005;352:1899–912. https://
doi.org/10.1056/NEJMra041320.
tions provide an opportunity to investigate the interaction of the human [11] Zeng J, Luo S, Huang Y, Lu Q. Critical role of environmental factors in the
gut microbiome and the skin and systemic immune systems, in order to pathogenesis of psoriasis. J Dermatol 2017;44:863–72. https://doi.org/10.1111/
unravel the significance of the gut-skin axis in the pathogenesis of 1346-8138.13806.
[12] Ogawa E, Sato Y, Minagawa A, Okuyama R. Pathogenesis of psoriasis and
psoriasis. development of treatment. J Dermatol 2018;45:264–72. https://doi.org/10.1111/
1346-8138.14139.
3. Conclusion [13] Austin LM, Ozawa M, Kikuchi T, Walters IB, Krueger JG. The majority of epidermal
T cells in Psoriasis vulgaris lesions can produce type 1 cytokines, interferon-gamma,
interleukin-2, and tumor necrosis factor-alpha, defining TC1 (cytotoxic T
The gut and skin share many common features and functions. Each lymphocyte) and TH1 effector populations: a type 1 differentiation bias is also
site has a unique ecosystem of normal flora interacting with epithelial measured in circulating blood T cells in psoriatic patients. J Invest Dermatol 1999;
113:752–9. https://doi.org/10.1046/j.1523-1747.1999.00749.x.
and immune cells. The IL-23/IL-17 signaling pathway plays a critical role [14] Haugh IM, Preston AK, Kivelevitch DN, Menter AM. Risankizumab: an anti-IL-23
in the inflammation of both the gut and the skin. With newer and more antibody for the treatment of psoriasis. Drug Des Dev Ther 2018;12:3879–83.
advanced technologies such as culturomics, metagenomics, whole https://doi.org/10.2147/dddt.S167149.
[15] Dong J, Goldenberg G. New biologics in psoriasis: an update on IL-23 and IL-17
genome analysis, metabolite analysis, and germ-free animal models, re- inhibitors. Cutis 2017;99:123–7.
searchers will be able to identify critical microbiomes and unravel their [16] Kamata M, Tada Y. Safety of biologics in psoriasis. J Dermatol 2018;45:279–86.
mechanisms in human psoriasis. In addition, keratinocytes primarily https://doi.org/10.1111/1346-8138.14096.
[17] Molinelli E, Campanati A, Brisigotti V, Offidani A. Biologic therapy in psoriasis (Part
responsible for skin barrier integrity express several important genes II): efficacy and safety of new treatment targeting IL23/IL-17 pathways. Curr
(e.g., CD109, antimicrobial peptides, TLRs, C-type lectins, and galectins) [69, Pharmaceut Biotechnol 2017;18:964–78. https://doi.org/10.2174/
95–99], that may potentially regulate the interaction of the microbiome 1389201019666180103140643.
[18] Villeda-Gabriel G, Santamaria-Cogollos LC, Perez-Lorenzo R, Reyes-Maldonado E,
with immune cells. This network of microbiome interactions is an
Saul A, Jurado-Santacruz F, Jimenez-Zamudio L, Garcia-Latorre E. Recognition of
exciting and wide-open topic to researchers in diverse fields. Streptococcus pyogenes and skin autoantigens in guttate psoriasis. Arch Med Res
1998;29:143–8.
[19] Rasmussen JE. The relationship between infection with group A beta hemolytic
Author contribution streptococci and the development of psoriasis. Pediatr Infect Dis J 2000;19:153–4.
[20] Tsai Y-C, Tsai T-F. A review of antibiotics and psoriasis: induction, exacerbation,
Hung-Lin Chen: Conceptualization, Writing- Original draft manu- and amelioration. Expet Rev Clin Pharmacol 2019;12:981–9. https://doi.org/
10.1080/17512433.2019.1665027.
script, Figure and Table preparation. Daniel K. Hsu: Writing- Reviewing [21] Fuchs E, Nowak JA. Building epithelial tissues from skin stem cells. Cold Spring
and Editing manuscript. Maxwell A. Fung: Writing- Reviewing and Harbor Symp Quant Biol 2008;73:333–50. https://doi.org/10.1101/
Editing manuscript. sqb.2008.73.032.
[22] Morizane S, Gallo RL. Antimicrobial peptides in the pathogenesis of psoriasis.
J Dermatol 2012;39:225–30. https://doi.org/10.1111/j.1346-8138.2011.01483.x.
[23] Lai Y, Gallo RL. AMPed up immunity: how antimicrobial peptides have multiple
Declaration of competing interest roles in immune defense. Trends Immunol 2009;30:131–41. https://doi.org/
10.1016/j.it.2008.12.003.
[24] Basler K, Bergmann S, Heisig M, Naegel A, Zorn-Kruppa M, Brandner JM. The role
The authors declare no competing financial interest. of tight junctions in skin barrier function and dermal absorption. J Contr Release :
official journal of the Controlled Release Society 2016;242:105–18. https://
doi.org/10.1016/j.jconrel.2016.08.007.
Acknowledgements
[25] Proksch E, Brandner JM, Jensen JM. The skin: an indispensable barrier. Exp
Dermatol 2008;17:1063–72. https://doi.org/10.1111/j.1600-0625.2008.00786.x.
We thank Dr. Fu-Tong Liu for proofreading the article and providing [26] Bigliardi PL. Role of skin pH in psoriasis. Curr Probl Dermatol 2017;54:108–14.
https://doi.org/10.1159/000489524.
insights to the pathogenesis of psoriasis.
[27] Dainichi T, Kitoh A, Otsuka A, Nakajima S, Nomura T, Kaplan DH, Kabashima K.
This work was supported by the Next-generation Pathway of Taiwan The epithelial immune microenvironment (EIME) in atopic dermatitis and psoriasis.
Cancer Precision Medicine Program (Grant No. ASKPQ-107-TCPMP), Nat Immunol 2018;19:1286–98. https://doi.org/10.1038/s41590-018-0256-2.
Academia Sinica, Taiwan. [28] Grice EA, Kong HH, Conlan S, Deming CB, Davis J, Young AC, Bouffard GG,
Blakesley RW, Murray PR, Green ED, Turner ML, Segre JA. Topographical and
temporal diversity of the human skin microbiome. Science (New York, N.Y.) 2009;
References 324:1190–2. https://doi.org/10.1126/science.1171700.
[29] Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knight R. Bacterial
community variation in human body habitats across space and time. Science (New
[1] World Health O. Global report on psoriasis. Geneva: World Health Organization;
York, N.Y.) 2009;326:1694–7. https://doi.org/10.1126/science.1177486.
2016.
[30] Assarsson M, Duvetorp A, Dienus O, S€ oderman J, Seifert O. Significant changes in
[2] Sondermann W, Djeudeu Deudjui DA, Korber A, Slomiany U, Brinker TJ, Erbel R,
the skin microbiome in patients with chronic plaque psoriasis after treatment with
Moebus S. Psoriasis, cardiovascular risk factors and metabolic disorders: sex-
narrowband ultraviolet B. Acta Derm Venereol 2018;98:428–36. https://doi.org/
specific findings of a population-based study. J Eur Acad Dermatol Venereol 2019.
10.1007/s00005-018-0528-4.
https://doi.org/10.1111/jdv.16029.
[31] Alekseyenko AV, Perez-Perez GI, De Souza A, Strober B, Gao Z, Bihan M, Li K,
[3] Rachakonda TD, Schupp CW, Armstrong AW. Psoriasis prevalence among adults in
Methe BA, Blaser MJ. Community differentiation of the cutaneous microbiota in
the United States. J Am Acad Dermatol 2014;70:512–6. https://doi.org/10.1016/
psoriasis. Microbiome 2013;1:31. https://doi.org/10.1186/2049-2618-1-31.
j.jaad.2013.11.013.
[32] Grice EA. The skin microbiome: potential for novel diagnostic and therapeutic
[4] Parisi R, Symmons DP, Griffiths CE, Ashcroft DM. Global epidemiology of psoriasis:
approaches to cutaneous disease. Semin Cutan Med Surg 2014;33:98–103. https://
a systematic review of incidence and prevalence. J Invest Dermatol 2013;133:
doi.org/10.12788/j.sder.0087.
377–85. https://doi.org/10.1038/jid.2012.339.
[33] Prast-Nielsen S, Tobin A-M, Adamzik K, Powles A, Hugerth LW, Sweeney C, Kirby B,
[5] Gisondi P, Fostini AC, Fossa I, Girolomoni G, Targher G. Psoriasis and the metabolic
Engstrand L, Fry L. Investigation of the skin microbiome: swabs vs. biopsies. Br J
syndrome. Clin Dermatol 2018;36:21–8. https://doi.org/10.1016/
Dermatol 2019;181:572–9. https://doi.org/10.1111/bjd.17691.
j.clindermatol.2017.09.005.

6
D.K. Hsu et al. Medicine in Microecology 4 (2020) 100016

[34] Visser MJE, Kell DB, Pretorius E. Bacterial dysbiosis and translocation in psoriasis [57] Dainichi T, Kitoh A, Otsuka A, Nakajima S, Nomura T, Kaplan DH, Kabashima K.
vulgaris. Front Cell Infect Microbiol 2019;9:7. https://doi.org/10.3389/ The epithelial immune microenvironment (EIME) in atopic dermatitis and psoriasis.
fcimb.2019.00007. Nat Immunol 2018;19:1286–98. https://doi.org/10.1038/s41590-018-0256-2.
[35] Luger TA, Loser K. Novel insights into the pathogenesis of psoriasis. Clin Immunol [58] Chimenti MS, Perricone C, Novelli L, Caso F, Costa L, Bogdanos D, Conigliaro P,
2018;186:43–5. https://doi.org/10.1016/j.clim.2017.07.014. Triggianese P, Ciccacci C, Borgiani P, Perricone R. Interaction between microbiome
[36] Fyhrquist N, Muirhead G, Prast-Nielsen S, Jeanmougin M, Olah P, Skoog T, Jules- and host genetics in psoriatic arthritis. Autoimmun Rev 2018;17:276–83. https://
Clement G, Feld M, Barrientos-Somarribas M, Sinkko H, van den Bogaard EH, doi.org/10.1016/j.autrev.2018.01.002.
Zeeuwen PLJM, Rikken G, Schalkwijk J, Niehues H, D€aubener W, Eller SK, [59] Wolf P, Weger W, Patra V, Gruber-Wackernagel A, Byrne SN. Desired response to
Alexander H, Pennino D, Suomela S, Tessas I, Lybeck E, Baran AM, Darban H, phototherapy vs photoaggravation in psoriasis: what makes the difference? Exp
Gangwar RS, Gerstel U, Jahn K, Karisola P, Yan L, Hansmann B, Katayama S, Dermatol 2016;25:937–44. https://doi.org/10.1111/exd.13137.
Meller S, Bylesj€o M, Hup e P, Levi-Schaffer F, Greco D, Ranki A, Schr€
oder JM, [60] Flutter B, Nestle FO. TLRs to cytokines: mechanistic insights from the imiquimod
Barker J, Kere J, Tsoka S, Lauerma A, Soumelis V, Nestle FO, Homey B, mouse model of psoriasis. Eur J Immunol 2013;43:3138–46. https://doi.org/
Andersson B, Alenius H. Microbe-host interplay in atopic dermatitis and psoriasis. 10.1002/eji.201343801.
Nat Commun 2019;10:4703. https://doi.org/10.1038/s41467-019-12253-y. [61] Dalkilic E, Bulbul Baskan E, Alkis N, Gullulu M, Yavuz M, Dilek K, Ersoy A,
[37] Hidalgo-Cantabrana C, G omez J, Delgado S, Requena-L opez S, Queiro-Silva R, Yurtkuran M. Tumor necrosis factor-alpha antagonist therapy-induced psoriasis in
Margolles A, Coto E, S anchez B, Coto-Segura P. Gut microbiota dysbiosis in a cohort Turkey: analysis of 514 patients. Mod Rheumatol 2012;22:738–42. https://doi.org/
of patients with psoriasis. Br J Dermatol 2019. https://doi.org/10.1111/bjd.17931. 10.1007/s10165-011-0590-9.
[38] Chen Y-J, Ho HJ, Tseng C-H, Lai Z-L, Shieh J-J, Wu C-Y. Intestinal microbiota [62] Naldi L, Chatenoud L, Linder D, Belloni Fortina A, Peserico A, Virgili AR, Bruni PL,
profiling and predicted metabolic dysregulation in psoriasis patients. Exp Dermatol Ingordo V, Lo Scocco G, Solaroli C, Schena D, Barba A, Di Landro A, Pezzarossa E,
2018;27:1336–43. https://doi.org/10.1111/exd.13786. Arcangeli F, Gianni C, Betti R, Carli P, Farris A, Barabino GF, La Vecchia C. Cigarette
[39] McKenzie C, Tan J, Macia L, Mackay CR. The nutrition-gut microbiome-physiology smoking, body mass index, and stressful life events as risk factors for psoriasis:
axis and allergic diseases. Immunol Rev 2017;278:277–95. https://doi.org/ results from an Italian case-control study. J Invest Dermatol 2005;125:61–7.
10.1111/imr.12556. https://doi.org/10.1111/j.0022-202X.2005.23681.x.
[40] Mazidi M, Rezaie P, Kengne AP, Mobarhan MG, Ferns GA. Gut microbiome and [63] Bardazzi F, Balestri R, Baldi E, Antonucci A, De Tommaso S, Patrizi A. Correlation
metabolic syndrome. Diabetes & metabolic syndrome 2016;10:S150–7. https:// between BMI and PASI in patients affected by moderate to severe psoriasis
doi.org/10.1016/j.dsx.2016.01.024. undergoing biological therapy. Dermatol Ther 2010;23(Suppl 1):S14–9. https://
[41] Santoru ML, Piras C, Murgia A, Palmas V, Camboni T, Liggi S, Ibba I, Lai MA, doi.org/10.1111/j.1529-8019.2009.01281.x.
Orru S, Blois S, Loizedda AL, Griffin JL, Usai P, Caboni P, Atzori L, Manzin A. Cross [64] Farkas A, Kemeny L, Szell M, Dobozy A, Bata-Csorgo Z. Ethanol and acetone
sectional evaluation of the gut-microbiome metabolome axis in an Italian cohort of stimulate the proliferation of HaCaT keratinocytes: the possible role of alcohol in
IBD patients. Sci Rep 2017;7:9523. https://doi.org/10.1038/s41598-017-10034-5. exacerbating psoriasis. Arch Dermatol Res 2003;295:56–62. https://doi.org/
[42] Pascal V, Pozuelo M, Borruel N, Casellas F, Campos D, Santiago A, Martinez X, 10.1007/s00403-003-0399-2.
Varela E, Sarrabayrouse G, Machiels K, Vermeire S, Sokol H, Guarner F, [65] Hajishengallis G, Darveau RP, Curtis MA. The keystone-pathogen hypothesis. Nat
Manichanh C. A microbial signature for Crohn's disease. Gut 2017;66:813–22. Rev Microbiol 2012;10:717–25. https://doi.org/10.1038/nrmicro2873.
https://doi.org/10.1136/gutjnl-2016-313235. [66] Hashiguchi Y, Yabe R, Chung SH, Murayama MA, Yoshida K, Matsuo K, Kubo S,
[43] Fu Y, Lee CH, Chi CC. Association of psoriasis with inflammatory bowel disease: a Saijo S, Nakamura Y, Matsue H, Iwakura Y. IL-36alpha from skin-resident cells plays
systematic review and meta-analysis. JAMA Dermatol 2018;154:1417–23. https:// an important role in the pathogenesis of imiquimod-induced psoriasiform
doi.org/10.1001/jamadermatol.2018.3631. dermatitis by forming a local autoamplification loop. J Immunol 2018;201:167–82.
[44] Verstockt B, Ferrante M, Vermeire S, Van Assche G. New treatment options for https://doi.org/10.4049/jimmunol.1701157.
inflammatory bowel diseases. J Gastroenterol 2018;53:585–90. https://doi.org/ [67] Zeng B, Shi S, Ashworth G, Dong C, Liu J, Xing F. ILC3 function as a double-edged
10.1007/s00535-018-1449-z. sword in inflammatory bowel diseases. Cell Death Dis 2019;10:315. https://
[45] Ely PH. Is psoriasis a bowel disease? Successful treatment with bile acids and doi.org/10.1038/s41419-019-1540-2.
bioflavonoids suggests it is. Clin Dermatol 2018;36:376–89. https://doi.org/ [68] Zakostelska Z, Malkova J, Klimesova K, Rossmann P, Hornova M, Novosadova I,
10.1016/j.clindermatol.2018.03.011.  ankova R, Jůzlova K, Hercogova J,
Stehlíkova Z, Kostovcík M, Hudcovic T, Step
[46] Eppinga H, Sperna Weiland CJ, Thio HB, van der Woude CJ, Nijsten TEC, Tlaskalova-Hogenova H, Kverka M. Intestinal microbiota promotes psoriasis-like
Peppelenbosch MP, Konstantinov SR. Similar depletion of protective skin inflammation by enhancing Th17 response. PloS One 2015;11:e0159539.
Faecalibacterium prausnitzii in psoriasis and inflammatory bowel disease, but not https://doi.org/10.1371/journal.pone.0159539.
in hidradenitis suppurativa. J Crohns Colitis 2016;10:1067–75. https://doi.org/ [69] Zhang H, Carnevale G, Polese B, Simard M, Thurairajah B, Khan N, Gentile ME,
10.1093/ecco-jcc/jjw070. Fontes G, Vinh DC, Pouliot R, King IL. CD109 restrains activation of cutaneous IL-
[47] Oliveira Mde F, Rocha Bde O, Duarte GV. Psoriasis: classical and emerging 17-producing γδ T cells by commensal microbiota. Cell Rep 2019;29:391–405.
comorbidities. An Bras Dermatol 2015;90:9–20. https://doi.org/10.1590/abd1806- https://doi.org/10.1016/j.celrep.2019.09.003. e395.
4841.20153038. [70] Quan C, Chen X-Y, Li X, Xue F, Chen L-H, Liu N, Wang B, Wang L-Q, Wang X-P,
[48] Bedoya SK, Lam B, Lau K, Larkin J. Th17 cells in immunity and autoimmunity. Clin Yang H, Zheng J. Psoriatic lesions are characterized by higher bacterial load and
Dev Immunol 2012;2013:986789. https://doi.org/10.1155/2013/986789. imbalance between Cutibacterium and Corynebacterium. J Am Acad Dermatol
[49] Tan L, Zhao S, Zhu W, Wu L, Li J, Shen M, Lei L, Chen X, Peng C. The Akkermansia 2019. https://doi.org/10.1016/j.jaad.2019.06.024.
muciniphila is a gut microbiota signature in psoriasis. Exp Dermatol 2018;27: [71] Thio HB. The microbiome in psoriasis and psoriatic arthritis: the skin perspective.
144–9. https://doi.org/10.1111/exd.13463. J Rheumatol Suppl 2018;94:30–1. https://doi.org/10.3899/jrheum.180133.
[50] Sitkin S, Pokrotnieks J. Clinical potential of anti-inflammatory effects of [72] Langan EA, Griffiths CEM, Solbach W, Knobloch JK, Zillikens D, Thaçi D. The role of
Faecalibacterium prausnitzii and butyrate in inflammatory bowel disease. Inflamm the microbiome in psoriasis: moving from disease description to treatment
Bowel Dis 2019;25:e40–1. https://doi.org/10.1093/ibd/izy258. selection? Br J Dermatol 2018;178:1020–7. https://doi.org/10.1111/bjd.16081.
[51] Bian X, Wu W, Yang L, Lv L, Wang Q, Li Y, Ye J, Fang D, Wu J, Jiang X, Shi D, Li L. [73] Yan D, Issa N, Afifi L, Jeon C, Chang HW, Liao W. The role of the skin and gut
Administration of Akkermansia muciniphila ameliorates dextran sulfate sodium- microbiome in psoriatic disease. Curr Dermatol Rep 2017;6:94–103. https://
induced ulcerative colitis in mice. Front Microbiol 2019;10:2259. https://doi.org/ doi.org/10.1007/s13671-017-0178-5.
10.3389/fmicb.2019.02259. [74] Drago L, De Grandi R, Altomare G, Pigatto P, Rossi O, Toscano M. Skin microbiota
[52] Trembath RC, Clough RL, Rosbotham JL, Jones AB, Camp RD, Frodsham A, of first cousins affected by psoriasis and atopic dermatitis. Clin Mol Allergy 2015;
Browne J, Barber R, Terwilliger J, Lathrop GM, Barker JN. Identification of a major 14:2. https://doi.org/10.1186/s12948-016-0038-z.
susceptibility locus on chromosome 6p and evidence for further disease loci [75] Statnikov A, Alekseyenko AV, Li Z, Henaff M, Perez-Perez GI, Blaser MJ, Aliferis CF.
revealed by a two stage genome-wide search in psoriasis. Hum Mol Genet 1997;6: Microbiomic signatures of psoriasis: feasibility and methodology comparison. Sci
813–20. https://doi.org/10.1093/hmg/6.5.813. Rep 2012;3:2620. https://doi.org/10.1038/srep02620.
[53] Burden AD, Javed S, Bailey M, Hodgins M, Connor M, Tillman D. Genetics of [76] Fahlen A, Engstrand L, Baker BS, Powles A, Fry L. Comparison of bacterial
psoriasis: paternal inheritance and a locus on chromosome 6p. J Invest Dermatol microbiota in skin biopsies from normal and psoriatic skin. Arch Dermatol Res
1998;110:958–60. https://doi.org/10.1046/j.1523-1747.1998.00213.x. 2011;304:15–22. https://doi.org/10.1007/s00403-011-1189-x.
[54] Mallon E, Bunce M, Wojnarowska F, Welsh K. HLA-CW*0602 is a susceptibility [77] Gao Z, Tseng C-h, Strober BE, Pei Z, Blaser MJ. Substantial alterations of the
factor in type I psoriasis, and evidence Ala-73 is increased in male type I psoriatics. cutaneous bacterial biota in psoriatic lesions. PloS One 2008;3:e2719. https://
J Invest Dermatol 1997;109:183–6. https://doi.org/10.1111/1523- doi.org/10.1371/journal.pone.0002719.
1747.ep12319304. [78] Valdimarsson H, Baker BS, Jonsdottir I, Powles A, Fry L. Psoriasis: a T-cell-mediated
[55] Asumalahti K, Veal C, Laitinen T, Suomela S, Allen M, Elomaa O, Moser M, de Cid R, autoimmune disease induced by streptococcal superantigens? Immunol today 1995;
Ripatti S, Vorechovsky I, Marcusson JA, Nakagawa H, Lazaro C, Estivill X, Capon F, 16:145–9. https://doi.org/10.1016/0167-5699(95)80132-4.
Novelli G, Saarialho-Kere U, Barker J, Trembath R, Kere J. Coding haplotype [79] Leung DY, Travers JB, Giorno R, Norris DA, Skinner R, Aelion J, Kazemi LV,
analysis supports HCR as the putative susceptibility gene for psoriasis at the MHC Kim MH, Trumble AE, Kotb M, et al. Evidence for a streptococcal superantigen-
PSORS1 locus. Hum Mol Genet 2002;11:589–97. https://doi.org/10.1093/hmg/ driven process in acute guttate psoriasis. J Clin Invest 1995;96:2106–12. https://
11.5.589. doi.org/10.1172/jci118263.
[56] Allen MH, Veal C, Faassen A, Powis SH, Vaughan RW, Trembath RC, Barker JN. [80] Codo~ ner FM, Ramírez-Bosca A, Climent E, Carri on-Gutierrez M, Guerrero M, Perez-
A non-HLA gene within the MHC in psoriasis. Lancet (London, England) 1999;353: Orquín JM, Horga de la Parte J, Genoves S, Ram on D, Navarro-L
opez V, Chenoll E.
1589–90. https://doi.org/10.1016/s0140-6736(99)01618-9. Gut microbial composition in patients with psoriasis. Sci Rep 2018;8:3812. https://
doi.org/10.1038/s41598-018-22125-y.

You might also like