Next Article in Journal
Curcumin Epigenetically Represses Histone Acetylation of Echinocandin B Producing Emericella rugulosa
Next Article in Special Issue
The Effects of a Multi-Ingredient Supplement Containing Wasabia Japonica Extract, Theacrine, and Copper (I) Niacin Chelate on Peripheral Blood Mononuclear Cell DNA Methylation, Transcriptomics, and Sirtuin Activity
Previous Article in Journal
Anticancer Potential and Molecular Mechanisms of Cinnamaldehyde and Its Congeners Present in the Cinnamon Plant
Previous Article in Special Issue
Heart Rate Variability Parameters to Evaluate Autonomic Functions in Healthy Young Subjects during Short-Term “Dry” Immersion
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Gut Microbiome in Psoriatic Arthritis—A Literature Review

by
Cristina Alexandru
1,
Carmen Catalina Iorgus
1,
Ionut Melesteu
2,
Elena Daniela Șerban
3,4,
Florin Bobircă
5,*,
Maria Magdalena Constantin
3,6,
Razvan Simu
7,
Ioan Ancuța
1,8,
Mihai Bojincă
1,8 and
Anca Bobircă
1,8
1
Department of Internal Medicine and Rheumatology, Dr I. Cantacuzino Clinical Hospital, 011437 Bucharest, Romania
2
Department of Gastroenterology, Colentina Clinical Hospital, 020125 Bucharest, Romania
3
2nd Department of Dermatology, Colentina Clinical Hospital, 020125 Bucharest, Romania
4
Department of Dermatology, Assistance Publique des Hôpitaux de Paris (AP-HP), Henri Mondor Hospital, 94000 Créteil, France
5
Department of General Surgery, Carol Davila University of Medicine and Pharmacy, Dr I. Cantacuzino Clinical Hospital, 011437 Bucharest, Romania
6
Department of Dermatology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
7
Department of Internal Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
8
Department of Internal Medicine and Rheumatology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
*
Author to whom correspondence should be addressed.
Physiologia 2023, 3(2), 208-220; https://doi.org/10.3390/physiologia3020014
Submission received: 30 January 2023 / Revised: 4 March 2023 / Accepted: 30 March 2023 / Published: 4 April 2023
(This article belongs to the Special Issue Feature Papers in Human Physiology–2nd Edition)

Abstract

:
Psoriatic arthritis is a heterogeneous chronic autoimmune disorder characterized principally by skin lesions, arthritis, dactylitis and enthesitis. The exact etiology of the disease is yet to be discovered, with genetic predisposition alongside environmental factors being a well-known theory. In recent years, new discoveries have emphasized the role of gut microbiome in perpetuating inflammation in spondylarthritis. The exact mechanism through which dysbiosis underlies the pathophysiology of psoriatic arthritis is not defined. One of the current areas of focus in rheumatic research with new studies emerging annually is the link between microbiome and psoriatic arthritis. In this review, we synthesized the recent knowledge on intestinal microbiome and psoriatic arthritis. We screened two databases for articles, PubMed and Medline, using the following keywords: “microbiome”, “microbiota” and “psoriatic arthritis”. We described the current expertise on diversity and composition of gut microbiome in psoriatic arthritis, comparing the results with other inflammatory diseases. In the future, preventing the dysbiosis process that leads to the development of psoriatic arthritis could open the door to new therapeutic modalities. Moreover, fecal microbiota transplantation and probiotics’ benefits in modulating the gut microbiome are being intensively researched at the moment.

1. Introduction

Psoriatic arthritis (PsA) is a heterogeneous, chronic inflammatory disorder characterized principally by axial and/or peripheral arthritis, dactylitis, enthesitis and psoriasis, with clinical symptoms in line with other spondyloarthritis (SpA). In the last few decades, intestinal involvement with microscopic gut inflammation in over 50% of SpA patients without gastrointestinal conditions has been verified [1,2]. Additionally, approximately 20–30% of people with psoriasis will develop psoriatic arthritis [3,4]. Although the nature of the pathology is still undetermined, hypotheses claim that the main cause is a combination of genetic predispositions and environmental influences [5]. Moreover, investigations on animals have shown that HLA-B27 rats grown in a microbe-free environment do not acquire SpA; rather, they show symptoms when exposed to a certain intestinal population [6].
Recently, a new theory has emerged, focusing on the gastrointestinal barrier, which can contribute to the production of inflammatory cytokines such as interleukin (IL)-23 and IL-17, with the microbiota playing an important role in this process [7,8,9]. The increased risk of Crohn’s disease in PsA and psoriasis patients provides additional evidence that the gut microbiota is involved in these conditions [10]. The human microbiome encompasses the microorganisms existing in, on, and within a human body (such as bacteria, archaea, viruses and eukaryotes) together with their genetic components and their habitat [5,11]. Every anatomical region is made up of distinct microbial communities with an important contribution to the host physiology [10,12]. The taxonomic characterization of these microbial communities has been recently identified owing to important progress in sequencing techniques [13]. Moreover, as a result of the research advances, it seems that the human microbiome is a dynamic community influenced by numerous factors such as genetics, age, dietary customs, antibiotic use and geography [14]. It is well-known that interactions between a host and a microbe are important in the development of a healthy organism [15]. Disturbances of the intestinal flora, dysbiosis, can be associated with different forms of inflammation in the organism and therefore with various autoimmune diseases [16]. Dysbiosis may be caused by a decrease in commensal bacteria such as lactobacilli, bifidobacterial and Faecalibacterium prausnitzii and/or an increase in pathogens such as Escherichia coli, Salmonella, and Helicobacter [17,18]. In patients diagnosed with psoriatic arthritis, a lower consistency of multi-gut bacteria was observed; however, the exact characteristics of the microbial components and their influence on immune diseases activity is yet to be discovered [5,16].
Interleukin-22, tumor necrosis factor (TNF), interferon gamma and IL-23/IL-17 axis are the key cytokines involved in the pathophysiology of PsA, with the latter playing the primary role in this condition [19,20]. The central role of IL-23/IL-17 and increased levels of lymphocyte T helper, mainly Th17, have been proven in patients with inflammatory bowel disease [21,22,23]. Activated macrophages and dendritic cells regulate the synthesis of IL-23, as a result of environmental variables such as various infections, but also the existence of self-molecules identified as autoantigens by CD4+ and CD8+ T cells [4].
Recent studies have highlighted the importance of innate lymphoid cells (ILCs), which are a subtype derived from natural killers’ cells, but are not yet fully described [24]. ILCs have the ability to secrete cytokine, resulting in the initiation and perpetuation of inflammation [25,26]. Furthermore, the relationship between the microbiome and spondyloarthritis is backed up by research, showing that individuals with ankylosing spondylitis (AS) had higher blood and synovial fluid concentrations of gut-derived ILCs that secrete IL-17 and IL-22 [23,27]. Moreover, the strong relation between psoriatic arthritis and inflammatory bowel diseases supports the role of gut dysbiosis in the pathophysiology of PsA, even though the main structures affected in this disorder are the enthesis and joints [28].
There is a commensal and mutualistic link between specific intestinal flora and immune system. Since the gut flora contains the greatest variety of bacteria, the involvement of the intestinal microbiota in autoimmune disorders has attracted increasing study interest [14]. Genetic, epigenetic factors, but also factors related to the external environment, especially diet and the probiotic intake recently analyzed, determine the profile of the intestinal microbiota [15,29]. Recently, studies have been focusing on the existence of a bidirectional signaling pathway between the gut microbiome and the osteoarticular system [30]. The concept of the skin–joint–gut axis describes the relationship between the intestinal microbiota and the function of the skin, joints, relevant especially in spondyloarthritis, such as PsA [11,16]. Unrelated to non-steroidal anti-inflammatory medicines, increased intestinal permeability caused by epithelial barrier dysfunction has been linked to SpA. This increased permeability is also linked to bacterial translocation, which starts and maintains inflammation [31].
Moreover, in dermatology, it has also been demonstrated that inflammatory skin diseases are associated with disruption of the skin and gut microbiota; the main diseases with this implication are psoriasis, rosacea and atopic dermatitis [32]. The function and differentiation of T cells with the imbalance of regulatory T cells, mostly Th17, are involved in the mechanism of the gut–skin axis in psoriasis. The interaction between pattern recognition receptors expressed by the host cell and the bacterial antigen are important factors in this mechanism [33]. Commensal bacteria play a crucial role in the balance of effector and regulatory T cells, the induction of immunoglobulin A that activates B cells, and the subsequent generation of particular immunoglobulin A antibodies. These processes all have an impact on adaptive immunity [16].
Gut dysbiosis is involved in Th17-mediated skin inflammation and in metabolite production, which results in an anti-microbial signaling changing immune cell activation through the IL-23/IL-17 signaling pathway through IL-22 and interferon gamma production [23]. The result of this mechanism is the hyperproliferation of keratinocytes, as illustrated in Figure 1 [34].
The effects of probiotics in rheumatic disorders, particularly PsA, have drawn considerable scientific attention. Probiotics might improve the quality of life of patients with psoriasis disease by promoting specific bacteria and reshaping the composition of the intestinal microbiome; however, further research is needed to establish the beneficial effects of this therapeutic option for autoimmune conditions [35,36].
Studies examining the role of the microbiome in inflammatory diseases are proliferating, with an emphasis on AS, inflammatory bowel disease (IBD) and psoriasis, with PsA being a recently considered disease in this topic. The goal of this narrative review is to compile pertinent data available on gut microbiome heterogeneity and its potential link to the inflammatory pathway in psoriatic arthritis.

2. Results

In this review, we have included a total of five studies relevant to gut microbiota in PsA published until December 2022. One study is a double-blind trial on fecal microbiome transplantation and its impact on PsA; one study focuses on the probiotic intake, analyzing the effect on the disease activity; and three studies are on gut microbiota components, using 16S rRNA sequencing from fecal samples. In the description of the results section, we included one study realized on mice, due to the valuable information provided by the authors on the chosen theme. The main characteristics of the results are illustrated in Table 1.
In a recent study, Chun-Yu Lin et al. analyzed the fecal samples from a small cohort of 9 PsA patients, compering the results with a control group [16]. They showed that PsA patients had a specific microbiome species (p = 0.048) and a different microbial density (p < 0.05). Bacteroides, Firmicutes, Proteobacteria and Actinobacteria were the bacterial communities more frequent in PsA, with significantly increased levels for Actinobacteria compared to those in No-PsA. Moreover, as regards symptomatology, dactylitis and enthesitis are especially associated with a specific microbiome, and this theory is supported by a higher frequency of Megasphaera elsdenii among patients with PsA with dactylitis and/or enthesitis compared with the No-PsA group. Another finding was the negative association of Magasphaera elsdenii and Bifidobacterium longum with eosinophil count. Additionally, they carried out a metagenome sequencing and showed bacterial phylotypes differences between the two groups. Across the groups, they reported various microbial compositions for multiple species, including Actinobacteria Phylum, Fusobacteriales Order, Acidaminococcaceae Family, Erysipelotrichaceae Family, Clostridium innocuum Genus, with Family: XIII_AD3011 being significantly higher for No-PsA controls, p = 0.039.
In a small study, the authors examined the impact of probiotic intake for 12 weeks on PsA activity, analyzing the enteric permeability, gut inflammatory marker, calprotectin and possible correlations with peripheral Th17 cells frequency [35]. The results showed that probiotic intake, by influencing the gut microbiome, can interfere with the immune process and significantly ameliorate the disease activity. Moreover, they brought to light the association of Th17 cells levels, suggesting that patients with a positive effect of the probiotic usage are those with lower levels of Th17 peripheral cells. Regarding the gut permeability, analyzing fecal zonulin and α1-antitrypain, the results showed higher levels of those markers. Gut inflammation was investigated using fecal calprotectin, and 60% of patients in this study had high levels of calprotectin. After probiotic intake, the markers analyzed significantly decreased. Thus, this paper stated that PsA has an increase degree of gut permeability and inflammation, and that probiotic usage has a beneficial effect [35]. However, the effects of probiotic treatment are not long-lasting; however, further investigations are needed to establish a correct correlation.
J. Manasson and colleagues characterized the gut microbiome in PsA/AS patients before and after biological therapy with TN necrosis factor inhibitors (TNFi) and with anti-interleukin-17A agents [37]. In this study, all 14 patients treated with TNFi were diagnosed with PsA, while only 64.3% (N = 9) of patients treated with anti-interleukin-17A had their PsA faecal samples analyzed before treatment with a biological agent and compared with controls. They reported an increase in Clostridiales and Erysuoelotrichales, and a reduction in Bacteroidales compared to controls. Comparing the groups with pre- and post-exposure to biologic therapy, the results showed a reduction in all subjects from TNFi group and in one-third of anti-IL17 group for Bacteroidales. For the anti-IL17 patients, there was expansion and reduction in Clostridiales levels throughout the therapy; these changes not identified in the TNFi treatment. Regarding fungal perturbation after biological treatment, the authors observed increased levels of Saccharomycetales after therapy. Moreover, 29% of the cohort had an expansion of Candida, while a reduction in Candida was registered in 14% of cohort treated with anti-IL17 [37]. The shift in Candida albicans in patients treated with anti-IL17 was significantly higher than TNFi treatment, p < 0.005. 40% of TNFi cohort showed an enlargement in Saccharomyces cerevisiae, which was significantly higher than that in anti-IL17.
The first study that analyzed gut microbiome in PsA patients was carried out by J. Scher et al. in 2015 [28]. They compared the gut microbiome in recent-onset PsA patients, naïve to treatment and psoriasis patients, further comparing the results to controls. PsA subjects had an important decrease in Coprococcus sp., Akkermansia, Ruminococcus and Pseudobutyrivibrio. The results also showed a reduction in Clostridia in PsA patients. In contrast, reduced levels of Parabacteroides and Coprobacillus were registered in psoriatic patients. Furthermore, they analyzed the levels of proinflammatory markers in fecal samples and serum IgA and the results showed an increased fecal IgA level (p = 0.06) and a significant decreased in RANKL level (p < 0.005) with no difference in OPG levels between PsA and controls, but significantly lower osteoprotegerin levels in psoriasis (p < 0.005). Analyzing the fatty acids in those patients, these evidenced that medium chain fatty acids have lower quantities in samples from PsA group. Comparing the fecal and serum levels of inflammatory cytokines, no differences were registered in the study.
In 2020, Miguens Blanco et al. published a study protocol for a multicentric, prospective, observational study (Mi-PART study) with the aim of identifying changes in the gut microbiome using 16S rRNA gene sequencing and manopore sequencing from fecal sample [39]. In order to complete this research, they enrolled 65 PsA patients, 30 ankylosis spondylitis patients and 30 healthy controls. The principal aims of this paper are to link disease activity to microbiota, to identify new biomarkers from gut microbiome and a potential mechanism in order to better understand how microbiome drives the immune disease. These findings could enrich not only the knowledge of physiological role of gut microbiota in PsA, but also some new therapy options [39].
In our review, we included one trial on fecal microbiota transplantation (FMT) in psoriatic arthritis patients, conducted by MS Kragsnaes et al. and published in 2020 [38]. They realized a randomized double-blind trial on 31 patients with PsA which underwent FMT or sham transplantation. Their results showed no significant side effects of the procedure. Furthermore, they investigated the disease activity using ACR20 score and HAD-DI score. In contrast to the sham group, FMT patients registered more frequent treatment failure (p = 0.018) and slightly less improvement of the HAQ-DI score (by 0.23 points, p = 0.031). For a certain conclusion of the FMT effect regarding the disease activity, a bigger cohort is needed.
P. Lin et al., in a study from 2014, analyzed the gut microbiota in HLA-B27 rats [40]. This paper revealed that HLA-B27 gene expression is associated with gut microbiome disfunction. Clostridia and Helicobacter had similar levels in HLA-B27 rats and wild rats. There was a reduction in the Rikenellaceae family for HLA-B27 group, while Paraprevotella registered an increase. A more abundant Bacterioides vulgatus and a reduction in Akkermansia muciniohila are the principal changes observed in HLA-B27 rats [40].

3. Discussion

The intestine constitutes an internal barrier, acting similarly to a semipermeable structure, which allows the uptake of beneficial nutrients while restricting the passage to toxic substances and pathogens [41]. The skin is the largest organ of the human body and constitutes an external barrier between the body and the environment. There are many factors contributing to the integrity of this barrier, one of them being the human microbiome, both cutaneous and intestinal.
The interaction between the host and the intestinal microorganisms is essential for maintaining human health since intestinal microorganisms are involved in various processes such as digestion, synthesis of vitamins or adaptive immune homeostasis [42]. Perturbation of this interaction can lead to intestinal dysbiosis, further contributing to the pathogenesis of various diseases such as psoriatic disease [43], but also other well-established comorbidities of psoriatic disease such as spondylarthritis [44], diabetes mellitus [45], cardiovascular disease [46] and inflammatory bowel disease [47].
Psoriasis can affect an individual in various ways, and this is why psoriatic disease would be a more appropriate term that encompasses its manifestations. Therefore, psoriatic arthritis and psoriasis (PsO) should be considered part of the spectrum of psoriatic disease [48], developing from an intricate interaction between genetic predisposition and environmental factors. It is now well-known that the intestinal microbiome is a major regulator of this complex interaction as an important component of the skin–joint–gut axis, a concept that perfectly describes the relationship between the intestinal microbiome, PsO and PsA, but also the development of other comorbidities [18].
The interaction between human microbiome, both cutaneous and intestinal, and the development of PsO and PsA has been a popular research topic recently, as proven by the high number of papers regarding this subject. The findings of these studies can be confounding and the purpose of the present review is to focus on the major findings regarding PsA patients.
It is well-established that the major driver of PsO and PsA is the IL-23/IL-17 signaling pathway [49]. It seems that the intestinal dysbiosis is involved in the Th17-mediated skin inflammation and in metabolite production, resulting in an anti-microbial signaling changing immune cell activation through the IL-23/IL-17 signaling pathway through IL-22 and interferon gamma production [34]. Increased intestinal permeability was reported in spondyloarthritis and Crohn’s disease patients, as demonstrated by the elevated serum zonulin levels [50,51]. Nevertheless, there was no correlation found between disease activity and intestinal permeability in AP patients [52,53]. Gut microbiome in autoinflammatory disease can open up new therapeutical developments in the future. Probiotics are live organisms that help to restore the balance of the intestinal microbiome, thus increasing the function of the intestinal barrier [54,55]. The influence of this off-label treatment is yet undefined in immune diseases such as SpA. Another therapeutic option increasingly researched is fecal microbiota transplantation, which had a beneficial effect in IBD and some metabolic syndrome patients [56,57].

3.1. The Intestinal Microbiome in Psoriatic Arthritis

At the phylum level, the intestinal microbiome of PsA patients is composed predominantly of Bacteroides, Firmicutes, Proteobacteria and Actinobacteria, the latter being more represented compared to patients with undifferentiated arthritis. Furthermore, Megasphaera elsdenii was more frequently found among patients with dactylitis and/or enthesitis [16]. These findings complement the previous information stating that PsA patients have an important decrease in Coprococcus, Akkermansia, Ruminococcus and Pseudobutyrivibrio, with a global decrease in Clostridia class. Moreover, this study was among the first to demonstrate the lower concentration of medium chain fatty acids in PsA patients’ stools [28].
Furthermore, questions have arisen regarding the intestinal microbiome dynamics in relation to biological therapies. Fecal samples of the PsA and AS patients treated with tumor necrosis factor inhibitors and with anti-interleukin-17 inhibitors were analyzed before and after treatment [37]. In the pre-TNFi phase, there were two clusters dominated by Bacteroides and Ruminococcaceae that became less prominent after treatment. In the pre-IL-17i phase, there was a strong positive correlation between Bacteroides and Ruminococcaceae, with nodes arranged in a linear structure that separated into two negatively correlated clusters after treatment. Quite uniquely, the mycobiome was analyzed as well and an increase in Saccharomycetales was found in both groups, in addition to the expansion of Candida and C. albicans in a subgroup of patients of varying clinical phenotypes, with a few subjects demonstrating a reduction in the IL-17i group.
Regarding the impact of probiotic intake on PsA activity, the authors concluded that PsA patients suffer from enhanced enteric permeability and inflammation (by measuring fecal zonulin, α1-antitrypsin and calprotectin, as well as peripheral immune phenotyping) and that probiotics may ameliorate disease activity in PsA by targeting these alterations [35].
Fecal transplantation appeared to be inferior to sham in treating active peripheral PsA since treatment failure occurred more frequently in the FMT group than in the sham group and the improvement in HAQ-DI was in favor of sham, with no difference in the proportion of ACR20 responders between the groups [38].

3.2. The Intestinal Microbiome in Psoriasis

Regarding the microbiome composition, the most frequently reported findings are the reduction in Bacteroides and Akkermansia, with the increase in Firmicutes and Actinobacteria [29,58]. Altered intestine leads to bacterial translocation triggering inflammation, which is able to influence extra-intestinal sites [59]. It is well-known that psoriasis can be associated with inflammatory bowel diseases, as patients with Crohn’s disease have a five-fold risk of developing psoriasis [60]. Moreover, in these patients, Faecalibacterium praunitziiis is reduced, limiting its anti-inflammatory effect through the inhibition of the NFkB pathway [33].
Comparison between the PsA and PsO patients revealed that Akkermansia and Ruminococcus (including Firmicutes/Clostridiales and Verrucomicrobiales, respectively) were significantly less abundant in PsA patients, whereas the Bacteroidetes phylum and Coprobacillus genus were less abundant in PsO patients [28].
Antibiotics used to treat intestinal dysbiosis can lead to improvements in PsO activity; however, it cannot be a long-term solution since it eradicates beneficial microbial communities [33,61].
Analogously to PsA patients, probiotics can decrease PsO activity, but can also provide a better response to treatment, lower the need for steroids and lower the risk of relapse [29]. Prebiotics and symbiotics can be used as adjunctive therapies in psoriasis due to their role in immunomodulation [12].

3.3. The Cutaneous Microbiome in Psoriasis

It seems that Firmicutes, Actinobacteria and Proteobacteria are the predominant phyla identified in the cutaneous plaques. Furthermore, there is a decrease in Actinobacteria and Bacteroides and an increase in Coprobacillus, Ruminococcus and Streptococcus [33,62].
There is also an important increase in the number of Staphylococcus in both non-lesional (more precisely, S. sciuri and S. aureus) and lesional skin (S. aureus and S. ptettenkoferi) when compared to the skin of healthy individuals [63,64]. S. aureus can be identified in the cutaneous lesions of 60% of PsO patients and its colonization is linked to an inflammatory Th17 response [63]. Moreover, in up to 60% of cases, S. aureus can secrete enterotoxins and toxic shock syndrome toxin-1. S. pyogenes is also frequently identified as a trigger for both the development and the exacerbations of PsO [60].

3.4. The Gut Microbiome in Ankylosing Spondylitis

The strong link between ankylosing spondylitis and dysbiosis is supported by recent research, which verified composition changes of microbial components. B Liu et al., in analyzing the microbiome in AS patients, have showed increased levels of Cyanobacteria, Deinococcota, Patescibacteria and Actinobacteria and decreased levels of Acidobacteria, Proteobacteria, Campyobacteria when compared to the those of healthy controls [65]. The diversity of AS microbiota is demonstrated in another study investigating the stool of 127 AS patients in comparison with 123 healthy controls [66]. The authors showed that microbiome in AS patients consisted of increased levels of Clostridiales bacterium, Clostridium bolteae and Clostridium hatheway and decreased levels of Bifidobacterium, Coprococcus, Lachnospiraceae and Roseburia. Additionally, they examined the impact of TNFi on microbial alterations, and their findings demonstrated that patients who had never had TNFi therapy were significantly different from healthy controls; however, there was no distinction between patients treated with TNFi and controls, indicating that the medication can restore the balance of the intestinal microbiota. [66]. Similar to IBD, Fecalibacterium prausnitzii, a species with a crucial role due to the anti-inflammatory properties, is deficient in AS patients. It is interesting to note that following TNFi therapy, Fecalibacterium prausnitzii levels reverted to normal [66]. Dialister sp. are more frequent among AS, being significant correlated with this disease, compared not only with healthy individuals, but with non-inflammatory intestinal biopsy from AS patients as well [67].
The link between gut dysbiosis and disease activity was analyzed, and a recent study showed a strong correlation of dysbiosis with an active disease and physical dysfunction in axial-SpA [68]. Regarding therapeutic improvements, although there is some evidence that probiotic intake in rats reduced arthritis and the levels of pro-inflammatory cytokines, the usefulness of probiotics in ankylosing spondylitis has not been yet established [69,70].

3.5. The Gut Microbiome in Inflammatory Bowel Disease

More research has been conducted on the influence of gut microbiota in IBD than on PsA; currently being investigated is not only the presence of dysbiosis, but also the potential increased intestinal bacterial translocation [18]. The results from animal and human studies have revealed the indisputable and critical role of the gut microbiome in the pathophysiology of inflammatory bowel disease. Dysbiosis in IBD is supported by the disturbance of microbial distribution with decreased beneficial pathogens and increased harmful ones. Aberrant microbial composition was observed in IBD patients, mainly represented by reduced levels of Firmicutes and Faecalibacterium, and increased amounts of Bacteroidetes, Enterobacteriaceae and Escherchia, which has been connected to NOD2 mutations [18,71,72]. Fecalibacterium prausnitzii, from the Clostridium leptum subgroup, has increased levels in normal microbiota, having a beneficial role by producing a protein with anti-inflammatory properties that is able to inhibit the nuclear factor-κB pathway. Quevrain et al. showed that Faecalibacterium prausnitzii has decreased levels in the microbiome from Crohn’s disease patients [73]. Similar to AS and PsA patients, Bifidobacterium adolescentis and Coprococcus levels are depleted in IBD patients [66,74].
Even though probiotics can enhance the diversity and richness of the microbiome and promote intestinal barrier function, a certain probiotic effect on IBD has not been determined, and more research is required to determine the therapeutic benefits of probiotics for use in day-to-day care [55]. On the other hand, FMT can improve the diversity of gut microbial components in IBD patients; however, at present, there are mixed results regarding this therapeutic option in Crohn’s disease or ulcerative colitis [75,76,77].
This narrative review highlights the new data about the microbiota in psoriatic arthritis, a rheumatic pathology that has been less researched on this topic until now. The principal limitation of this literature research is the small number of studies conducted on this subject, especially the small number of enrolled patients in each study. Second, it is more difficult to compare and validate the findings across studies due to the variety of the gut microbiota and various component analysis.

4. Materials and Methods

This review was conducted by searching two databases, PubMed and Embase, using the combinations of words “gut microbiota” or “microbiome” and “psoriatic arthritis”, until December 2022. A total of 175 articles were generated. After excluding the duplicates, we analyzed the article content, applying the inclusion and exclusion criteria. The inclusion criteria were as follows: original studies, written in English, that focused on the importance of gut microbiota in PsA. Exclusion criteria were: reviews, studies with a focus only on psoriasis, not PsA, studies without significant information on gut microbiome in PsA, and studies conducted on animals (although we described a relevant study in the Results section, we did not include it in the Results table). The research strategy is illustrated in Figure 2.

5. Conclusions

According to recent research, the microbiome has a role in the development and progression of psoriatic arthritis. The composition and diversity of the gut microbiome are different in psoriatic arthritis patients compared to healthy people. Studies have revealed that alterations in the gut microbiota may be related to a specific inflammatory disease, with differences between psoriatic arthritis and psoriasis, ankylosis spondylitis and inflammatory bowel disease. Furthermore, there is evidence that various microbial components are associated with specific symptoms such as enthesitis and dactylitis. As this is still subject under investigation, more research is required to completely understand the gut microbiota’s role in the pathophysiology of psoriatic arthritis. The effects of biologic therapy on the microbiome and the association of HLA-B27 and dysbiosis are arguments to support the importance of gut microbiota in Spondylarthritis. Moreover, the possibility of fecal transplantation and the scarce evidence of beneficial probiotic usage in treating chronic inflammatory diseases such as psoriatic arthritis encourage the development of novel therapeutic options targeting microbiome.

Author Contributions

Conceptualization, A.B. and F.B.; methodology, I.A.; software, M.M.C.; validation, A.B., I.A. and M.B.; formal analysis, C.A.; investigation, C.C.I.; resources, E.D.Ș.; data curation, R.S.; writing—original draft preparation, C.C.I. and I.M.; writing—review and editing, C.A. and E.D.Ș.; visualization, F.B.; supervision, I.A.; project administration, M.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gilis, E.; Mortier, C.; Venken, K.; Debusschere, K.; Vereecke, L.; Elewaut, D. The Role of the Microbiome in Gut and Joint Inflammation in Psoriatic Arthritis and Spondyloarthritis. J. Rheumatol. 2018, 94, 36–39. [Google Scholar] [CrossRef] [PubMed]
  2. Haque, N.; Lories, R.J.; de Vlam, K. Comorbidities Associated with Psoriatic Arthritis Compared with Non-psoriatic Spondyloarthritis: A Cross-sectional Study. J. Rheumatol. 2016, 43, 376–382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Karmacharya, P.; Chakradhar, R.; Ogdie, A. The epidemiology of psoriatic arthritis: A literature review. Best Pract. Res. Clin. Rheumatol. 2021, 35, 101692. [Google Scholar] [CrossRef] [PubMed]
  4. Chimenti, M.S.; Perricone, C.; Novelli, L.; Caso, F.; Costa, L.; Bogdanos, D.; Conigliaro, P.; Triggianese, P.; Ciccacci, C.; Borgiani, P.; et al. Interaction between microbiome and host genetics in psoriatic arthritis. Autoimmun. Rev. 2018, 17, 276–283. [Google Scholar] [CrossRef]
  5. Veale, D. Psoriatic arthritis: Recent progress in pathophysiology and drug development. Arthritis Res. Ther. 2013, 15, 224. [Google Scholar] [CrossRef] [Green Version]
  6. Rath, H.C.; Herfarth, H.H.; Ikeda, J.S.; Grenther, W.B.; Hamm, T.E.; Balish, E.; Taurog, J.D.; Hammer, R.E.; Wilson, K.H.; Sartor, R.B. Normal luminal bacteria, especially Bacteroides species, mediate chronic colitis, gastritis, and arthritis in HLA-B27/human beta2 microglobulin transgenic rats. J. Clin. Investig. 1996, 98, 945–953. [Google Scholar] [CrossRef] [Green Version]
  7. Tiwari, V.; Brent, L.H. Psoriatic Arthritis. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK547710/ (accessed on 20 December 2022).
  8. Zhao, R.; Zhou, H.; Su, S.B. A critical role for interleukin-1β in the progression of autoimmune diseases. Int. Immunopharmacol. 2013, 17, 658–669. [Google Scholar] [CrossRef]
  9. Raychaudhuri, S.P.; Raychaudhuri, S.K. Mechanistic rationales for targeting interleukin-17A in spondyloarthritis. Arthritis Res. Ther. 2017, 19, 51. [Google Scholar] [CrossRef] [Green Version]
  10. Hsu, D.K.; Fung, M.A.; Chen, H.-L. Role of skin and gut microbiota in the pathogenesis of psoriasis, an inflammatory skin disease. Med. Microecol. 2020, 4, 100016. [Google Scholar] [CrossRef]
  11. Szychlinska, M.A.; Di Rosa, M.; Castorina, A.; Mobasheri, A.; Musumeci, G. A correlation between intestinal microbiota dysbiosis and osteoarthritis. Heliyon 2019, 5, e01134. [Google Scholar] [CrossRef] [Green Version]
  12. Chen, L.; Li, J.; Zhu, W.; Kuang, Y.; Liu, T.; Zhang, W.; Chen, X.; Peng, C. Skin and Gut Microbiome in Psoriasis: Gaining Insight into the Pathophysiology of It and Finding Novel Therapeutic Strategies. Front. Microbiol. 2020, 11, 589726. [Google Scholar] [CrossRef] [PubMed]
  13. Ursell, L.K.; Metcalf, J.L.; Parfrey, L.W.; Knight, R. Defining the human microbiome. Nutr. Rev. 2012, 70, S38–S44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Belvoncikova, P.; Maronek, M.; Gardlik, R. Gut Dysbiosis and Fecal Microbiota Transplantation in Autoimmune Diseases. Int. J. Mol. Sci. 2022, 23, 10729. [Google Scholar] [CrossRef]
  15. Myers, B.; Brownstone, N.; Reddy, V.; Chan, S.; Thibodeaux, Q.; Truong, A.; Bhutani, T.; Chang, H.-W.; Liao, W. The gut microbiome in psoriasis and psoriatic arthritis. Best Pract. Res. Clin. Rheumatol. 2019, 33, 101494. [Google Scholar] [CrossRef] [PubMed]
  16. Lin, C.-Y.; Hsu, C.-Y.; He, H.-R.; Chiang, W.-Y.; Lin, S.-H.M.; Huang, Y.-L.M.; Kuo, Y.-H.B.; Su, Y.-J.M. Gut microbiota differences between psoriatic arthritis and other undifferentiated arthritis: A pilot study. Medicine 2022, 101, e29870. [Google Scholar] [CrossRef] [PubMed]
  17. Favazzo, L.J.; Hendesi, H.; Villani, D.A.; Soniwala, S.; Dar, Q.A.; Schott, E.M.; Gill, S.R.; Zuscik, M.J. The gut microbiome-joint connection: Implications in osteoarthritis. Curr. Opin. Rheumatol. 2020, 32, 92–101. [Google Scholar] [CrossRef]
  18. Eppinga, H.; Konstantinov, S.R.; Peppelenbosch, M.P.; Thio, H.B. The Microbiome and Psoriatic Arthritis. Curr. Rheumatol. Rep. 2014, 16, 407. [Google Scholar] [CrossRef]
  19. Chimenti, M.S.; Ballanti, E.; Perricone, C.; Cipriani, P.; Giacomelli, R.; Perricone, R. Immunomodulation in psoriatic arthritis: Focus on cellular and molecular pathways. Autoimmun. Rev. 2013, 12, 599–606. [Google Scholar] [CrossRef]
  20. Doss, G.P.; Agoramoorthy, G.; Chakraborty, C. TNF/TNFR: Drug target for autoimmune diseases and immune-mediated inflammatory diseases. Front. Biosci. 2014, 19, 1028. [Google Scholar] [CrossRef] [Green Version]
  21. Benham, H.; Rehaume, L.M.; Hasnain, S.Z.; Velasco, J.; Baillet, A.C.; Ruutu, M.; Kikly, K.; Wang, R.; Tseng, H.-W.; Thomas, G.P.; et al. Interleukin-23 Mediates the Intestinal Response to Microbial β-1,3-Glucan and the Development of Spondyloarthritis Pathology in SKG Mice. Arthritis Rheumatol. 2014, 66, 1755–1767. [Google Scholar] [CrossRef] [Green Version]
  22. Appel, H.; Maier, R.; Wu, P.; Scheer, R.; Hempfing, A.; Kayser, R.; Thiel, A.; Radbruch, A.; Loddenkemper, C.; Sieper, J. Analysis of IL-17+ cells in facet joints of patients with spondyloarthritis suggests that the innate immune pathway might be of greater relevance than the Th17-mediated adaptive immune response. Arthritis Res. Ther. 2011, 13, R95. [Google Scholar] [CrossRef] [Green Version]
  23. Durham, L.E.; Kirkham, B.W.; Taams, L.S. Contribution of the IL-17 Pathway to Psoriasis and Psoriatic Arthritis. Curr. Rheumatol. Rep. 2015, 17, 55. [Google Scholar] [CrossRef] [PubMed]
  24. Vély, F.; Barlogis, V.; Vallentin, B.; Neven, B.; Piperoglou, C.; Ebbo, M.; Perchet, T.; Petit, M.; Yessaad, N.; Touzot, F.; et al. Evidence of innate lymphoid cell redundancy in humans. Nat. Immunol. 2016, 17, 1291–1299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Leijten, E.F.; van Kempen, T.S.; Boes, M.; Michels-van Amelsfort, J.M.; Hijnen, D.; Hartgring, S.A.; van Roon, J.A.; Wenink, M.H.; Radstake, T.R. Brief Report: En-richment of Activated Group 3 Innate Lymphoid Cells in Psoriatic Arthritis Synovial Fluid. Arthritis Rheumatol. 2015, 67, 2673–2678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Gasteiger, G.; Fan, X.; Dikiy, S.; Lee, S.Y.; Rudensky, A.Y. Tissue residency of innate lymphoid cells in lymphoid and non-lymphoid organs. Science 2015, 350, 981–985. [Google Scholar] [CrossRef] [Green Version]
  27. Sonnenberg, G.F.; Artis, D. Innate lymphoid cells in the initiation, regulation and resolution of inflammation. Nat. Med. 2015, 21, 698–708. [Google Scholar] [CrossRef] [Green Version]
  28. Scher, J.U.; Ubeda, C.; Artacho, A.; Attur, M.; Isaac, S.; Reddy, S.M.; Marmon, S.; Neimann, A.; Brusca, S.; Patel, T.; et al. Decreased Bacterial Diversity Characterizes the Altered Gut Microbiota in Patients with Psoriatic Arthritis, Resembling Dysbiosis in Inflammatory Bowel Disease. Arthritis Rheumatol. 2015, 67, 128–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Navarro-López, V.; Martínez-Andrés, A.; Ramírez-Boscá, A.; Ruzafa-Costas, B.; Núñez-Delegido, E.; Carrión-Gutiérrez, M.; Prieto-Merino, D.; Codoñer-Cortés, F.; Ramón-Vidal, D.; Genovés-Martínez, S.; et al. Efficacy and Safety of Oral Administration of a Mixture of Probiotic Strains in Patients with Psoriasis: A Randomized Clinical Trial. Acta Derm. -Venereol. 2019, 99, 1078–1084. [Google Scholar] [CrossRef] [Green Version]
  30. Jacques, P.; Mielants, H.; Coppieters, K.; de Vos, M.; Elewaut, D. The intimate relationship between gut and joint in spondyloarthropathies. Curr. Opin. Rheumatol. 2007, 19, 353–357. [Google Scholar] [CrossRef]
  31. Hecquet, S.; Totoson, P.; Martin, H.; Prati, C.; Wendling, D.; Demougeot, C.; Verhoeven, F. AB0073 Intestinal Permeability in Spondyloarthritis: A Systematic Review of the Literature. Ann. Rheum. Dis. 2021, 80 (Suppl. S1), 1067. [Google Scholar] [CrossRef]
  32. de Pessemier, B.; Grine, L.; Debaere, M.; Maes, A.; Paetzold, B.; Callewaert, C. Gut–Skin Axis: Current Knowledge of the Interrelationship between Microbial Dysbiosis and Skin Conditions. Microorganisms 2021, 9, 353. [Google Scholar] [CrossRef]
  33. Visser, M.J.E.; Kell, D.; Pretorius, E. Bacterial Dysbiosis and Translocation in Psoriasis Vulgaris. Front. Cell. Infect. Microbiol. 2019, 9, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Thye, A.Y.-K.; Bah, Y.-R.; Law, J.W.-F.; Tan, L.T.-H.; He, Y.-W.; Wong, S.-H.; Thurairajasingam, S.; Chan, K.-G.; Lee, L.-H.; Letchumanan, V. Gut–Skin Axis: Unravelling the Connection between the Gut Microbiome and Psoriasis. Biomedicines 2022, 10, 1037. [Google Scholar] [CrossRef]
  35. Haidmayer, A.; Bosch, P.; Lackner, A.; D’Orazio, M.; Fessler, J.; Stradner, M.H. Effects of Probiotic Strains on Disease Activity and Enteric Permeability in Psoriatic Arthritis—A Pilot Open-Label Study. Nutrients 2020, 12, 2337. [Google Scholar] [CrossRef]
  36. Sanchez, P.; Letarouilly, J.-G.; Nguyen, Y.; Sigaux, J.; Barnetche, T.; Czernichow, S.; Flipo, R.-M.; Sellam, J.; Daïen, C. Efficacy of Probiotics in Rheumatoid Arthritis and Spondyloarthritis: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutrients 2022, 14, 354. [Google Scholar] [CrossRef]
  37. Manasson, J.; Wallach, D.S.; Guggino, G.; Stapylton, M.; Badri, M.H.; Solomon, G.; Reddy, S.M.; Coras, R.; Aksenov, A.A.; Jones, D.R.; et al. Interleukin-17 Inhibition in Spondyloarthritis Is Associated with Subclinical Gut Microbiome Perturbations and a Distinctive Interleukin-25–Driven Intestinal Inflammation. Arthritis Rheumatol. 2020, 72, 645–657. [Google Scholar] [CrossRef]
  38. Kragsnaes, M.S.; Kjeldsen, J.; Horn, H.C.; Munk, H.L.; Pedersen, J.K.; Just, S.A.; Ahlquist, P.; Pedersen, F.M.; de Wit, M.; Möller, S.; et al. Safety and efficacy of faecal microbiota transplantation for active peripheral psoriatic arthritis: An exploratory randomised placebo-controlled trial. Ann. Rheum. Dis. 2021, 80, 1158–1167. [Google Scholar] [CrossRef] [PubMed]
  39. Miguens Blanco, J.; Borghese, F.; McHugh, N.; Kelleher, P.; Sengupta, R.; Marchesi, J.R.; Abraham, S. Longitudinal profiling of the gut microbiome in patients with psoriatic arthritis and ankylosing spondylitis: A multicentre, prospective, observational study. BMC Rheumatol. 2020, 4, 60. [Google Scholar] [CrossRef]
  40. Lin, P.; Bach, M.; Asquith, M.; Lee, A.Y.; Akileswaran, L.; Stauffer, P.; Davin, S.; Pan, Y.; Cambronne, E.D.; Dorris, M.; et al. HLA-B27 and Human beta2-Microglobulin Affect the Gut Microbiota of Transgenic Rats. PLoS ONE 2014, 9, e105684. [Google Scholar] [CrossRef]
  41. Vancamelbeke, M.; Vermeire, S. The intestinal barrier: A fundamental role in health and disease. Expert Rev. Gastroenterol. Hepatol. 2017, 11, 821–834. [Google Scholar] [CrossRef] [PubMed]
  42. Honda, K.; Littman, D.R. The microbiota in adaptive immune homeostasis and disease. Nature 2016, 535, 75–84. [Google Scholar] [CrossRef] [PubMed]
  43. Yan, D.; Issa, N.; Afifi, L.; Jeon, C.; Chang, H.-W.; Liao, W. The Role of the Skin and Gut Microbiome in Psoriatic Disease. Curr. Dermatol. Rep. 2017, 6, 94–103. [Google Scholar] [CrossRef]
  44. Costello, M.-E.; Robinson, P.C.; Benham, H.; Brown, M.A. The intestinal microbiome in human disease and how it relates to arthritis and spondyloarthritis. Best Pract. Res. Clin. Rheumatol. 2015, 29, 202–212. [Google Scholar] [CrossRef] [PubMed]
  45. Dao, M.C.; Everard, A.; Aron-Wisnewsky, J.; Sokolovska, N.; Prifti, E.; Verger, E.O.; Kayser, B.D.; Levenez, F.; Chilloux, J.; Hoyles, L.; et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: Relationship with gut microbiome richness and ecology. Gut 2016, 65, 426–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Jin, M.; Qian, Z.; Yin, J.; Xu, W.; Zhou, X. The role of intestinal microbiota in cardiovascular disease. J. Cell. Mol. Med. 2019, 23, 2343–2350. [Google Scholar] [CrossRef] [Green Version]
  47. Sartor, R.B.; Wu, G.D. Roles for Intestinal Bacteria, Viruses, and Fungi in Pathogenesis of Inflammatory Bowel Diseases and Therapeutic Approaches. Gastroenterology 2017, 152, 327–339.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Raychaudhuri, S.P. A Cutting Edge Overview: Psoriatic Disease. Clin. Rev. Allergy Immunol. 2012, 44, 109–113. [Google Scholar] [CrossRef]
  49. Hawkes, J.E.; Yan, B.Y.; Chan, T.C.; Krueger, J.G. Discovery of the IL-23/IL-17 Signaling Pathway and the Treatment of Psoriasis. J. Immunol. 2018, 201, 1605–1613. [Google Scholar] [CrossRef] [Green Version]
  50. Hecquet, S.; Totoson, P.; Martin, H.; Prati, C.; Wendling, D.; Demougeot, C.; Verhoeven, F. Intestinal permeability in spondyloarthritis and rheumatoid arthritis: A systematic review of the literature. Semin. Arthritis Rheum. 2021, 51, 712–718. [Google Scholar] [CrossRef]
  51. Tajik, N.; Frech, M.; Schulz, O.; Schälter, F.; Lucas, S.; Azizov, V.; Dürholz, K.; Steffen, F.; Omata, Y.; Rings, A.; et al. Targeting zonulin and intestinal epithelial barrier function to prevent onset of arthritis. Nat. Commun. 2020, 11, 1995. [Google Scholar] [CrossRef] [Green Version]
  52. Morris, A.J.; Howden, C.W.; Robertson, C.; Duncan, A.; Torley, H.; Sturrock, R.D.; Russell, R.I. Increased intestinal permeability in ankylosing spondylitis—Primary lesion or drug effect? Gut 1991, 32, 1470–1472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Ciccia, F.; Guggino, G.; Rizzo, A.; Alessandro, R.; Luchetti, M.M.; Milling, S.; Saieva, L.; Cypers, H.; Stampone, T.; Di Benedetto, P.; et al. Dysbiosis and zonulin upregulation alter gut epithelial and vascular barriers in patients with ankylosing spondylitis. Ann. Rheum. Dis. 2017, 76, 1123–1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Singh, N.; Yadav, H.; Marotta, F.; Singh, V. Probiotics—A Probable Therapeutic Agent for Spondyloarthropathy. Int. J. Probiotics Prebiotics 2017, 12, 57–68. [Google Scholar]
  55. Abraham, B.P.; Quigley, E.M.M. Probiotics in Inflammatory Bowel Disease. Gastroenterol. Clin. N. Am. 2017, 46, 769–782. [Google Scholar] [CrossRef] [PubMed]
  56. Vrieze, A.; Van Nood, E.; Holleman, F.; Salojärvi, J.; Kootte, R.S.; Bartelsman, J.F.; Dallinga-Thie, G.M.; Ackermans, M.T.; Serlie, M.J.; Oozeer, R.; et al. Transfer of Intestinal Microbiota from Lean Donors Increases Insulin Sensitivity in Individuals with Metabolic Syndrome. Gastroenterology 2012, 143, 913–916.e7. [Google Scholar] [CrossRef]
  57. Damman, C.J.; Miller, S.I.; Surawicz, C.M.; Zisman, T.L. The Microbiome and Inflammatory Bowel Disease: Is There a Therapeutic Role for Fecal Microbiota Transplantation? Am. J. Gastroenterol. 2012, 107, 1452–1459. [Google Scholar] [CrossRef]
  58. Chen, P.; He, G.; Qian, J.; Zhan, Y.; Xiao, R. Potential role of the skin microbiota in Inflammatory skin diseases. J. Cosmet. Dermatol. 2020, 20, 400–409. [Google Scholar] [CrossRef]
  59. Ramírez-Boscá, A.; Navarro-López, V.; Martínez-Andrés, A.; Such, J.; Francés, R.; de la Parte, J.H.; Asín-Llorca, M. Identification of Bacterial DNA in the Peripheral Blood of Patients with Active Psoriasis. JAMA Dermatol. 2015, 151, 670–671. [Google Scholar] [CrossRef] [Green Version]
  60. Lewis, D.J.; Chan, W.H.; Hinojosa, T.; Hsu, S.; Feldman, S.R. Mechanisms of microbial pathogenesis and the role of the skin microbiome in psoriasis: A review. Clin. Dermatol. 2019, 37, 160–166. [Google Scholar] [CrossRef]
  61. Saxena, V.N.; Dogra, J. Long-term use of penicillin for the treatment of chronic plaque psoriasis. Eur. J. Dermatol. 2005, 15, 359–362. [Google Scholar]
  62. Thio, H.B. The Microbiome in Psoriasis and Psoriatic Arthritis: The Skin Perspective. J. Rheumatol. 2018, 94, 30–31. [Google Scholar] [CrossRef] [PubMed]
  63. Chang, H.W.; Yan, D.; Singh, R.; Liu, J.; Lu, X.; Ucmak, D.; Lee, K.; Afifi, L.; Fadrosh, D.; Leech, J.; et al. Alteration of the cutaneous microbiome in psoriasis and potential role in Th17 polarization. Microbiome 2018, 6, 154. [Google Scholar] [CrossRef]
  64. Tett, A.; Pasolli, E.; Farina, S.; Truong, D.T.; Asnicar, F.; Zolfo, M.; Beghini, F.; Armanini, F.; Jousson, O.; De Sanctis, V.; et al. Unexplored diversity and strain-level structure of the skin microbiome associated with psoriasis. NPJ Biofilms Microbiomes 2017, 3, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Liu, B.; Ding, Z.; Xiong, J.; Heng, X.; Wang, H.; Chu, W. Gut Microbiota and Inflammatory Cytokine Changes in Patients with Ankylosing Spondylitis. BioMed Res. Int. 2022, 2022, 1005111. [Google Scholar] [CrossRef] [PubMed]
  66. Yin, J.; Sternes, P.R.; Wang, M.; Song, J.; Morrison, M.; Li, T.; Zhou, L.; Wu, X.; He, F.; Zhu, J.; et al. Shotgun metagenomics reveals an enrichment of potentially cross-reactive bacterial epitopes in ankylosing spondylitis patients, as well as the effects of TNFi therapy upon microbiome composition. Ann. Rheum. Dis. 2020, 79, 132–140. [Google Scholar] [CrossRef]
  67. Tito, R.Y.; Cypers, H.; Joossens, M.; Varkas, G.; Van Praet, L.; Glorieus, E.; Bosch, F.V.D.; De Vos, M.; Raes, J.; Elewaut, D. Brief Report: Dialister as a Microbial Marker of Disease Activity in Spondyloarthritis. Arthritis Rheumatol. 2017, 69, 114–121. [Google Scholar] [CrossRef] [PubMed]
  68. Sagard, J.; Olofsson, T.; Mogard, E.; Marsal, J.; Andréasson, K.; Geijer, M.; Kristensen, L.E.; Lindqvist, E.; Wallman, J.K. Gut dysbiosis associated with worse disease activity and physical function in axial spondyloarthritis. Thromb. Haemost. 2022, 24, 42. [Google Scholar] [CrossRef]
  69. Regel, A.; Sepriano, A.; Baraliakos, X.; Van Der Heijde, D.; Braun, J.D.; Landewé, R.; Bosch, F.V.D.; Falzon, L.; Ramiro, S. Efficacy and safety of non-pharmacological and non-biological pharmacological treatment: A systematic literature review informing the 2016 update of the ASAS/EULAR recommendations for the management of axial spondyloarthritis. RMD Open 2017, 3, e000397. [Google Scholar] [CrossRef] [Green Version]
  70. Bedaiwi, M.K.; Inman, R. Microbiome and probiotics. Curr. Opin. Rheumatol. 2014, 26, 410–415. [Google Scholar] [CrossRef]
  71. Al Nabhani, Z.; Dietrich, G.; Hugot, J.P.; Barreau, F. Nod2: The intestinal gate keeper. PLoS Pathog. 2017, 13, e1006177. [Google Scholar] [CrossRef] [Green Version]
  72. Li, E.; Hamm, C.M.; Gulati, A.S.; Sartor, R.B.; Chen, H.; Wu, X.; Zhang, T.; Rohlf, F.J.; Zhu, W.; Gu, C.; et al. Inflammatory Bowel Diseases Phenotype, C. difficile and NOD2 Genotype Are Associated with Shifts in Human Ileum Associated Microbial Composition. PLoS ONE 2012, 7, e26284. [Google Scholar] [CrossRef] [Green Version]
  73. Quévrain, E.; Maubert, M.A.; Michon, C.; Chain, F.; Marquant, R.; Tailhades, J.; Miquel, S.; Carlier, L.; Bermúdez-Humarán, L.G.; Pigneur, B.; et al. Identification of an anti-inflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn’s disease. Gut 2016, 65, 415–425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Gevers, D.; Kugathasan, S.; Denson, L.A.; Vázquez-Baeza, Y.; Van Treuren, W.; Ren, B.; Schwager, E.; Knights, D.; Song, S.J.; Yassour, M.; et al. The Treatment-Naive Microbiome in New-Onset Crohn’s Disease. Cell Host Microbe 2014, 15, 382–392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Weingarden, A.R.; Vaughn, B.P. Intestinal microbiota, fecal microbiota transplantation, and inflammatory bowel disease. Gut Microbes 2017, 8, 238–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Zhang, F.-M. Fecal microbiota transplantation for severe enterocolonic fistulizing Crohn’s disease. World J. Gastroenterol. 2013, 19, 7213–7216. [Google Scholar] [CrossRef] [PubMed]
  77. Moayyedi, P.; Surette, M.G.; Kim, P.T.; Libertucci, J.; Wolfe, M.; Onischi, C.; Armstrong, D.; Marshall, J.K.; Kassam, Z.; Reinisch, W.; et al. Fecal Microbiota Transplantation Induces Remission in Patients with Active Ulcerative Colitis in a Randomized Controlled Trial. Gastroenterology 2015, 149, 102–109.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Gut microbiota relationship with psoriasis disease. Dietary, genetic and environmental factors are key regulators of the microbiome. If there is no balance of these factors, it can lead to dysbiosis; this, by increasing permeability and destabilizing the integrity of the intestinal barrier, leads to inflammation and an aberrant cytokine response. The result is inflammation of the skin with keratocyte proliferation and the appearance of psoriatic arthropathy.
Figure 1. Gut microbiota relationship with psoriasis disease. Dietary, genetic and environmental factors are key regulators of the microbiome. If there is no balance of these factors, it can lead to dysbiosis; this, by increasing permeability and destabilizing the integrity of the intestinal barrier, leads to inflammation and an aberrant cytokine response. The result is inflammation of the skin with keratocyte proliferation and the appearance of psoriatic arthropathy.
Physiologia 03 00014 g001
Figure 2. Database search for gut microbiota in psoriatic arthritis.
Figure 2. Database search for gut microbiota in psoriatic arthritis.
Physiologia 03 00014 g002
Table 1. Results on gut microbiome in PsA.
Table 1. Results on gut microbiome in PsA.
Studies on Microbiota
StudyYearCohortMethodResults
CY Lin et al. [16]20229 PsA patients
10 No-PsA patients
16S rRNA amplicon sequencing from fecal sample
    • Increased Family: XIII_AD3011in No-PsA > PsA.
    • Increased Megasphaera elsdenii, Bacteroides, Firmicutes, Proteobacteria and Actinobacteria in PsA > No-PsA.
A Haidmayer et al. [35]202010 PsA patients who received probioticsFecal zonulin,
α1-antitrypsin and calprotectin
Peripheral immune phenotyping
    • Increased enteric permeability marker (zonulin) correlated with Th17 cells frequency.
    • Elevated Calprotectin.
    • Reduction in disease activity.
J. Manasson et al. [37]202015 patients PsA/AS treated with TNFi
vs.
14 patients treated with anti-interleukin-17A
vs.
controls.
16S rRNA gene sequencing from fecal sample
  • Comparison to controls before treatment:
    • Increased in Clostridiales and Erysuoelotrichales.
    • Reduction in Bacteroidales.
  • Comparison after treatment:
    • Reduction in Bacteroidales in all TNFi group, and in thirt of anti-IL-17A group. Reduction in Ruminococcaceae in TNFi.
    • Increased Candida albicans and Clostridiales after treatment with anti-IL-17A.
    • Increased Saccharomycetales after both therapies.
    • Increased Saccharomyces cerevisiae in TNFi.
J. Scher et al. [28]201516 PsA patients
15 PsO patients
17 controls.
16S ribosomal RNA pyrosequencing from fecal samples
    • Microbiota less diverse in PsA and PsO than controls.
    • Decreased Coprococcus sp. in PsA and PsO.
    • Decreased Akkermansia, Ruminococcus, Clostridia and Pseudobutyrivibrio in PsA.
Studies on Fecal Microbiota Transplantation
StudyYearCohort SampleScore UsedResults
MS Kragsnaes et al.
(Double-blind study) [38]
202131 patients
15 FMT group vs. 16 sham group
Side effects
ACR20 score
HAQ-DI
    • No adverse effects.
    • Sham group had better improvement of disease activity than FMT.
Abbreviations: PsA = psoriatic arthritis; No-PsA = controls without psoriatic arthritis; TNFi = tumor necrosis factor inhibitors; AS = Ankylosis spondylitis; PsO = psoriasis; IL = interleukin; sp. = species; FMT = fecal microbiota transplantation; ACR20 = American College of Rheumatology 20% score; HAQ-DI = The Health Assessment Questionnaire Disability Index.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alexandru, C.; Iorgus, C.C.; Melesteu, I.; Șerban, E.D.; Bobircă, F.; Constantin, M.M.; Simu, R.; Ancuța, I.; Bojincă, M.; Bobircă, A. The Role of Gut Microbiome in Psoriatic Arthritis—A Literature Review. Physiologia 2023, 3, 208-220. https://doi.org/10.3390/physiologia3020014

AMA Style

Alexandru C, Iorgus CC, Melesteu I, Șerban ED, Bobircă F, Constantin MM, Simu R, Ancuța I, Bojincă M, Bobircă A. The Role of Gut Microbiome in Psoriatic Arthritis—A Literature Review. Physiologia. 2023; 3(2):208-220. https://doi.org/10.3390/physiologia3020014

Chicago/Turabian Style

Alexandru, Cristina, Carmen Catalina Iorgus, Ionut Melesteu, Elena Daniela Șerban, Florin Bobircă, Maria Magdalena Constantin, Razvan Simu, Ioan Ancuța, Mihai Bojincă, and Anca Bobircă. 2023. "The Role of Gut Microbiome in Psoriatic Arthritis—A Literature Review" Physiologia 3, no. 2: 208-220. https://doi.org/10.3390/physiologia3020014

Article Metrics

Back to TopTop