Open access peer-reviewed chapter

Skin and Gut Microbiota in Psoriasis: A Systematic Review

Written By

Atiya Rungjang, Jitlada Meephansan and Hok Bing Thio

Submitted: 17 March 2020 Reviewed: 28 April 2020 Published: 02 June 2020

DOI: 10.5772/intechopen.92686

From the Edited Volume

Human Microbiome

Edited by Natalia V. Beloborodova and Andrey V. Grechko

Chapter metrics overview

960 Chapter Downloads

View Full Metrics

Abstract

Paying attention to a microbial approach may lead to improvements in diagnosis, treatment, prevention, and prognosis of psoriasis. A systematic review was performed according to the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines searching strategy to identify the pattern of the microbiome and the association of skin and gut microbiota with psoriasis, including the factors that may affect the results of the microbial study. In total, 16 studies were included in this systematic review. Ten studies investigated the skin microbiome, of which six studies were cross-sectional and four studies were prospective studies. Six studies investigated the gut microbiome, including five cross-sectional studies and one prospective study. The understanding of the relationship between microbiota and psoriasis may lead to diagnostics and treatment improvements. Currently, there is a slight consensus on some specific features that define psoriasis. However, no specific taxa have been identified as biomarkers of the disease, even from large-scale cohort studies. Thus, future cohort studies with standardized methodologies and proof-of-concept investigations in animal models may uncover the role of microbiota and the microbial pathways in psoriasis.

Keywords

  • psoriasis
  • microbiome
  • alpha diversity
  • beta diversity
  • dysbiosis

1. Introduction

Psoriasis is one of the most common immune-mediated inflammatory skin diseases. The prevalence of the disease has been reported, with ranges from 0.09 to 11.43% by the WHO Global Report 2016 [1]. Psoriatic skin lesions are characterized by hyperproliferation of keratinocytes, infiltration of immune cells, including neutrophils, T cells, dendritic cells, and macrophages. To date the etiology of this disease is not fully understood; genetic and environmental interaction plays a crucial role in the disease development [2, 3]. Recently, the immunological approach has helped to significantly clarify the pathophysiology of the disease. Dysregulation of both the bacteria, including Staphylococcus aureus [4], Streptococcus pyogenes [5], and fungi such as Malassezia [6] through innate and adaptive immune systems in genetically susceptible individuals, such as immune cells in the skin, Tumor necrosis factor α, dendritic cells—particularly pathogenic T cells that produce high levels of IL-17 in response to IL-23, all contribute substantially to the pathogenic process [7].

Previous studies have indicated an association between psoriasis and numerous comorbidities that share the chronic inflammatory state. Moreover, increasing evidence indicates that the gut microbe contributes to the onset of the low-grade inflammation, which is a pathological phenotype of these metabolic disorders [8].

Additionally, it has been known that several microorganisms contribute to psoriasis exacerbation alterations in the innate and adaptive immune processes [9]. The increasing evidence here suggests that the microbiota may play a critical role in psoriasis pathogenesis. This systematic review aims to elucidate the correlation between the microbiome and psoriasis pathogenesis, and the microbiota modulation that may lead to possible therapeutic interventions.

Advertisement

2. Psoriasis and microbiota

The initial search revealed a total of 629 studies of which 501 studies were excluded based on their title and abstract. The full texts were reviewed, and a further 116 studies were excluded. An additional four studies from the reference lists of already included studies were included in the systematic review. In total, 16 studies were included in this systematic review; 10 studies investigated the skin microbiome, of which 6 studies were cross-sectional and 4 studies were prospective study. Six studies investigated the gut microbiome, including five cross-sectional studies and one prospective study. The most commonly used method was 16S r RNA (skin swab, biopsies, curette); Langan et al. [10] used traditional culture combined with mass spectrometry (MALDI-TOF) (Table 1).

2.1 Skin microbiota in psoriasis

Several studies reported the characteristic features of microbiota in psoriatic skin (Table 2). Significant differences were observed between psoriatic lesion and control skin, but the changes were different in each study. Gao et al. [19] and Chang et al. [14] reported an increase in lesional skin diversity compared to non-lesional and control. In contrast, subsequent studies by Fahlen et al. [18] found wider range of Shannon index values in the control suggesting that the trend of decrease in lesional psoriasis microbiome diversity is consistent with the findings by Alexseyenko et al. [17] who observed a decrease in the diversity and significantly lower Shannon index in lesional skin. Consistent with previous studies, Tett et al. [16] found that psoriatic plaques at the ear are characterized by a significant decrease in microbial diversity. When beta diversity was analyzed to describe heterogeneity of microbial community, Fahlen et al. [18] reported a lower beta diversity in psoriasis compared to control, while Alexseyenko et al. [17] found that beta diversity was the highest in lesional skin, followed by unaffected skin, and the lowest in healthy skin. In line with the study by Tett et al. [16], which reported that ear lesions revealed higher beta diversity, Loeshe et al. [12] and Chang et al. [14] also reported a higher beta diversity at dry skin sites in psoriasis. At the phylum level, most skin bacterial composition fall into four major phyla: Actinobacteria, Firmicutes, Bacteroidetes, and Proteobacteria. Within these phyla, the three most abundant genera are: Propionibacterium, Corynebacterium, and Staphylococcus. From the studies of Gao et al. [19], Fahlen et al. [18], and Langan et al. [10], it has been revealed that at the phylum level, compared to healthy skin, psoriatic skin was associated with an increase in the relative abundance of Firmicutes but a decrease for Actinobacteria, which is partially consistent with Alexseyenko et al. [17] who identified psoriatic lesion as cutaneo type 2, which was dominated by Firmicutes and Actinobacteria. In contrast, Firmicutes were lower in the studies by Loeshe et al. [12], Assarsson et al. [13], and Drago et al. [15]. Proteobacteria showed inconsistent abundance, lower in lesional skin as observed by Gao et al. [19] whereas Fahlen et al. [18] observed an increase, and Drago et al. [15] reported that Proteobacteria and Bacteroidetes were the dominant microbiota in psoriasis lesion. At the genus level, Streptococcus were higher in lesional skin by Gao et al. [19], Fahlen et al. [18], Alexseyenko et al. [17], Stehlikova et al. [11], and Drago et al. [15] while Loeshe et al. [12] found no correlation between psoriasis lesional and unaffected skin. Staphylococcus were detected more frequently in the lesion by Gao et al. [19] and Tett et al. [16] opposite to Fahlen et al. [18] who found that Staphylococcus were increased in abundance in healthy controls. Lower abundance of Propionibacterium in lesional skin was reported by Gao et al. [19], Fahlen et al. [18], Drago et al. [15], Stehlikova et al. [11], and Loeshe et al. [12], which is in contrast to Alexseyenko et al. [17] who reported an increase in the relative abundance of combined Gram positives such as Corynebacterium, Propionibacterium, Staphylococcus, and Streptococcus. In the subsequent study by Langan et al. [10], the presence of Corynebacterium and Staphylococcus was found to be significantly correlated with PASI scores while Anaerococcus and Propionibacterium were associated with non-lesional skin. These are consistent with the reports by Gao et al. [19] and Chang et al. [14] that at species level lesional skin psoriasis had an increased level of S. aureus but a decreased level of P. acne. On the other hand, study on the importance of site-specific microbiota without related disease reported that at the species level the most abundant bacteria were S. epidermidis and P. acne irrespective of disease status and hence suggested that an underlying subject-specific microbial signature better defines the microbiome.

StudyStudy design/ratings of the qualityPopulationResult
Langan et al. [10] (2019)Cross-sectional/423 Pso, 20 CPso L
At the phylum level:
Firmicutes, Actinobacteria
At the genus level:
Prevotella, Staphylococcus, Anaerococcus and Propionibacterium
Prevotella and Staphylococcus significantly associated with Pso L
Pso NL
Anaerococcus, Propionibacterium
Prospective systemic treatment/2Actinobacteria-to-Firmicutes ratio, partially reversible during treatment
Biological therapies demonstrated the largest impact on the ratio of Actinobacteria to Firmicutes
Corynebacterium, Staphylococcus, significantly correlated with PASI scores
Stehlikova et al. [11] (2019)Cross-sectional/434 Pso, 25 CBeta diversity: no significant differences between Pso L and Pso NL
Pso L
Streptococcus regardless of the sampling site
Brevibacterium richness and evenness in in the elbow lesions, compared to back lesions
Propionibacterium PsoL, PsoNL compared C in elbow lesions
Remark
Alpha diversity and bacterial taxa from skin swab, scraping, and biopsy are comparable
Loesche et al. [12] (2018)
Remark: study did not include healthy control
Cross-sectional/4114 PsoBeta diversity: Pso L > Pso NL
Pso L
At the phylum level: Actinobacteria in leg, scalp, and trunk lesions Firmicutes in scalp and trunk lesions
At the genus level: Caulobacteraceae, Corynebacterium leg lesions
At the species level: BacilliPropionibacterium acnes in scalp lesions
Streptococcus colonization of skin does not correlate with severity in lesional and non-lesional skin
Longitudinal RCT/189 PsoPso L and Pso NL respond similarly to ustekinumab
Significant change in abundance from baseline in all body sites
No difference diversity in Pso L vs. Pso NL except in trunk
Pso L microbiota was not converging with Pso NL as treatment progressed
Microbiota diverged further between Pso L and Pso NL across body sites
Assarsson et al. [13] (2018)
Remark: study did not include healthy controls
Cross-sectional/426 PsoPso L
Firmicutes Staphylococcus
Longitudinal Narrowband UVB/2Pso L
Firmicutes, Staphylococcus, Finegoldia, Anaerococcus, Peptoniphilus, Gardnerella, Prevotella, Clostridium
Pso NL
Firmicutes
Pseudomonas in treatment responders
Chang et al. [14] (2018)Cross-sectional/428 Pso, 26 CAlpha diversity: Pso L > Pso NL > C
Beta diversity in all dry skin sites
Psol L
Alpha diversity at dry skin sites, with a trend at the sebaceous (scalp) site, and no increase at moist sites
S. aureus and S. pettenkoferi
Pso NL
S. sciuri
C
P. acnes, P. granulosum
Drago et al. [15] (2016)Cross-sectional/43 adult first cousins—1 AD, 1 Pso, 1 C (same lifestyle and environmental factors)Pso L
At the phylum level
Firmicutes, Proteobacteria in Pso L compare to AD and C
At the family level
Streptococcaceae, Rhodobacteraceae, Campylobacteraceae, Moraxellaceae in Pso L compare to AD and C
Staphylococcaceae, Propionibacteriaceae in Pso L compare to AD, C.
At the species level: Propionibacterium acnes in Pso compare to AD and C.
S. aureus in Pso L < C < AD, no difference in Psp NL
Tett et al. [16] (2017)
Remark: study did not include healthy control
Cross-sectional/428 PsoAlpha diversity: Pso L < Pso NL in ear lesions (richness did not correlate with PASI score)
Beta diversity: Pso L > Pso NL in ear lesions
Pso L
At the phylum level:
Actinobacteria and Firmicutes.
At the genus level:
Staphylococcus
At the species level:
S. epidermidis, P. acnes, S. caprae/capitis, and M. luteus
Alekseyenko et al. [17] (2013)Cross-sectional/475 Pso, 124 CAlpha diversity: Pso L < Pso NL and C
Beta diversity: Pso L > Pso NL > C.
Pso L
At the phylum level:
Cutaneotype 2 (dominated by Actinobacteria, Firmicutes)
At the genus level:
combined relative abundance of Corynebacterium, Propionibacterium, Staphylococcus, Streptococcus
Cupriavidus, Flavisolibacter, Methylobacterium, Schlegelella.
At the species level:
Acidobacteria, Schlegelella
Acidobacteria positively correlated with PASI
C:Cutaneotype 1 (dominated by Proteobacteria)
Cupriavidus, Flavisolibacter
Longitudinal 12 weeks, 36 weeks after systemic treatment/117 Pso, 15 cNo statistically significant difference was observed between the lesion and unaffected groups, or longitudinally within groups
Pso L↑Relative abundance of Corynebacterium, Propionibacterium, Staphylococcus and Streptococcus
Fahlen et al. [18] (2012)Cross-sectional/410 Pso, 12 CAlpha diversity: no difference observed when using the Shannon index
Beta diversity: Pso L < C
Pso LAt the phylum level: ↓ Actinobacteria, ↑ Proteobacteria in trunk lesions
At the phylum level:
Propionibacterium in all sites, ↓Staphylococcus
Streptococcus/Propionibacterium ratio
Gao et al. [19] (2008)Cross-sectional/46 Pso, 6 CAlpha diversity: Pso L > Pso NL, C
Beta diversity: Pso L > C
At the phylum level: ↑ Firmicutes, ↓ Actinobacteria, Proteobacteria
At genus level: ↓ Propionibacterium, ↑ Streptococcus
At the species level:
P. acne, ↓ Anaerobic species

Table 1.

Skin microbiota in patients with psoriasis.

Pso—Psoriasis, C—Control, Pso L—Psoriasis lesional skin, Pso NL—Psoriasis non-lesional skin.

FindingBy
Alpha diversityIncreasedGao et al. [19], Chang et al. [14]
DecreasedFahlen et al. [18], Alexseyenko et al. [17], Tett et al. [16]
Beta diversityLowerFahlen et al. [18]
HigheAlexseyenko et al. [17], Tett et al. [16], Loeshe et al. [12], Chang et al. [14]
Phylum level
FirmicutesIncreased FirmicutesGao et al. [19], Fahlen et al. [18], Langan et al. [10]
Decreased FirmicutesLoeshe et al. [12], Assarsson et al. [13], Drago et al. [15]
ActinobacteriaDecreased ActinobacteriaGao et al. [19], Fahlen et al. [18], Langan et al. [10]
Increased Firmicutes and ActinobacteriaAlexseyenko et al. [17]
ProteobacteriaDecreased ProteobacteriaGao et al. [19]
Increased ProteobacteriaFahlen et al. [18], Drago et al. [15]
Genus level
StreptococcusIncreased StreptococcusGao et al. [19], Fahlen et al. [18], Alexseyenko et al. [17], Stehlikova et al. [11], Drago et al. [15]
No correlationLoeshe et al. [12]
StaphylococcusIncreased StaphylococcusGao et al. [19], Tett et al. [16]
Decreased StaphylococcusFahlen et al. [18]
PropionibacteriumLower PropionibacteriumGao et al. [19], Fahlen et al. [18], Drago et al. [15], Stehlikova et al. [11], Loeshe et al. [12]
Gram positivesIncreased relative abundance of combined Gram positives: Corynebacterium, Propionibacterium, Staphylococcus, and StreptococcusAlexseyenko et al. [17]
Corynebacterium and Staphylococcus were significantly correlated with PASI scoresLangan et al. [10]
Increased S. aureus, decreased P. acneGao et al. [19]
Site-specific microbiota without related diseaseTett et al. [16]

Table 2.

Summary of skin microbiota findings in psoriasis.

There is a challenge to identify the explicit features of healthy or psoriasis microbiomes. Investigations of such a complex system of bacteria, fungi, and viruses are difficult and there is also high variation between samples. The composition of these communities of microorganisms depends on skin characteristics, such as sebaceous gland concentration, moisture content, topography, and temperature, as well as on host genetics and exogenous environmental factors [20]. Thus, the skin microbiome is biogeographically specific for each body site [21]. Demographic differences, such as gender, age, place of residence, living with animals, hygiene habits, occupation, and ethnicity also influence the composition of the skin microbiome [22]. The underlying disease and/or disease severity may also have an effect on the microbiome diversity or alterations in microbial communities due to disease states.

Moreover, microbiome diversities differ between studies; however, the more recent studies demonstrate decreased alpha diversity with increased beta diversity in psoriasis. Also, there are data that demonstrate a trend toward a changing microbial composition in psoriasis-affected skin. Propionibacterium is known as a protective commensal bacterium that is related with SCFA and propionate production, which regulates immune function. The decrease in the relative abundance of this microorganism in psoriasis may be related to the course of disease. In most studies, Staphylococcus are dominant in psoriatic skin, as species such as S. aureus proposed pathogenic Th17 activation while S. epidermidis appear to modulate immune and barrier functions. Interestingly, a study by Tett et al. [16] reported S. epidermidis strains contain known virulence-related genes that are predominate in psoriasis-affected skin. Therefore, a future study at the species and the strain level may provide more information.

2.2 Gut microbiota in psoriasis

The fecal sample study by Scher et al. [23] revealed that gut microbiome in skin psoriasis and psoriatic arthritis had a decrease in alpha diversity compared to control, and Actinobacteria had a decrease in relative abundance at the phylum level. This is in line with Masallat et al. [24] who found that the relative abundance of Actinobacteria was reduced in psoriasis versus healthy controls with a negative correlation of PASI score whereas the ratio of Firmicutes to Bacteroidetes was positively correlated with PASI score. This is consistent with Codoner et al. [25] who found a lower abundance of Bacteroides at the genus level and characterized core microbiome of psoriasis by an increase in Feacalibacterium but a decrease in Bacteroides spp. The abundance of Akkermansia, Ruminococcus, and Pseudobutyrivibrio was found to be lower in psoriatic arthritis compared to controls by Scher et al. [23]. Eppinga et al. [26] found that the abundance of Faecalibacterium prausnitzii was reduced in psoriasis with a significant increase in the relative abundance of Escherichia coli (Table 3).

StudyStudy design/ratings of the qualityPopulationResult
Shapiro et al. [27] (2019)Cross-sectional/424 Pso 22 CAlpha diversity, beta diversity: no significant differences
At the phylum level: ↑Firmicutes, Actinobacteria
Bacteroidetes, Proteobacteria
At the species level:
Ruminococcus gnavus, Doreaformici generans and Collinsella aerofaciens, Prevotella copri and Parabacteroides distasonis
Tan et al. [28] (2018)Cross-sectional/414 Pso 14CPso L
At the phylum level: Verrucomicrobia, Tenericutes
At the class level: Mollicutes, Verrucomicrobiae
At the order level: Verrucomicrobiales, RF39
At the family level: Verrucomicrobiaceae, S24–7
At the genus level: ↑Bacteroidaceae, Enterococcaceae, Akkermansia
At the species level: Akkermansia muciniphila, ↑Clostridium citroniae
Codoner et al. [25] (2018)
Remark: study did not include healthy control
Cross-sectional/452Pso compared with a cohort of over 300 healthy individuals extracted from the human microbiome projectPso:
↑Beta diversity
Enterotype 2 (predominance of Prevotella) tended to experience more frequent bacterial translocation and higher inflammatory status
Bacteroides, ↑Akkermansia, Faecalibacterium
Chen et al. [29] (2018)Cross-sectional/432Pso 64 CDiversity: no significant difference between Pso and C
At the phylum level: ↑Firmicutes, Bacteroidetes
At the genus level: ↑ Ruminococcus, Megasphaera
At the family level: Bacteroidaceae, PrevotellaceaeRuminococcaceae, Lachnospiraceae
Other covariates: sex, disease activity assessed by PASI score, phototherapy, arthritis, as well as diet, alcohol, smoking, coffee, tea, and habit of exercise, did not significantly affect the abundance profile of intestinal microbiota among Pso and C
Patients receiving systemic treatment (DMARDs or biologics drugs BioSysDrug) subgroup analyses /220 Pso↓the species Prevotella stercorea, belonging to Prevotellaceae, of the phylum Bacteroidetes, in patients receiving BioSysDrug
Scher et al. [23] (2015)Cross-sectional/415Pso, 16PsA 17CDiversity: Pso, PsA < C
At the phylum level:
Firmicutes, Clostridiales, Verrucomicrobiales
Bacteroidetes in PsA vs. Pso
Actinobacteria in Pso vs. C
At the genus level:
Coprococcus spp. in Pso and PsA vs. C
Akkermansia, Ruminococcus, Pseudobutyrivibrio in PsA vs. C
Parabacteroides, Coprobacillus in Pso vs. C
Masallat et al. [24] (2016)Case control/445Pso 45CFirmicutes/Bacteroidetes ratio correlated with PASI
Actinobacteria

Table 3.

Gut microbiota in patients with psoriasis.

Pso—Psoriasis, C—Control, Pso L—Psoriasis lesional skin, Pso NL—Psoriasis non-lesional skin.

The gut is considered as a major immune organ, with gut-associated lymphoid tissue (GALT) being the most complex immune compartment [30]. It is well known that change in the microbe composition may promote both health and disease [31]. Intestinal dysbiosis has been implicated in the etiology of various diseases [32], such as Crohn’s disease and obesity [33, 34]. Moreover, there is strong evidence that indicates intestinal dysbiosis is clinically relevant to psoriasis [35, 36]. The importance of the gut-skin axis in the pathogenesis of psoriasis has recently been documented in humans, as well as in animal models of psoriasis [9, 37]. A study by Tan et al. identified that the signature of gut microbiota and its function are significantly altered in the gut of patients with psoriasis [28]. Intestinal and skin microbiota directly regulate imiquimod-induced skin inflammation (IISI), and emphasizes the importance of microbiota in the pathogenesis of psoriasis [38]. A study by Zákostelská et al. has shown that exposure of mice to antibiotics inhibited the induction of psoriasis [37].

To identify bacterial pathways, which may be involved in the pathogenesis of psoriasis, it should be highlighted that SCFAs potentially regulate the generation and function of Th17 cells [39]. Moreover, in psoriasis the loss or depletion of Faecalibacterium prausnitzii, a major source of the protective SCFAs in the gut, may be associated with disease development [26]. In psoriatic arthritis, decreased Akkermansia and Ruminococcus, which are protective bacteria that regulate the intestinal barrier that produces SCFA, may be related with disease severity. Gut dysbiosis markedly reduced butyrate production, which inhibits NF-ĸβ, an inflammation pathway that impacts gut epithelial integrity and consequential cross-talk between gut proteins, bacteria, and the innate and humoral immune systems [27]. Alterations in the pathways involved in LPS function were also observed in psoriasis patients. Additionally, LPS is also thought to be involved in gut inflammation and has been linked to the pathogenesis of insulin resistance and diabetes mellitus [40], which has been epidemiologically associated with psoriasis. A decrease in Bacteroides, which are known to play an immunomodulatory role in the gut through the production of polysaccharide A that induces regulatory T cells, may result in an altered immune response [41]. Whereas a decrease in Actinobacteria, a phylum that includes the Bifidobacterium species that have been shown to reduce intestinal inflammation, suppresses autoimmunity, and induces regulatory T cell expression. There are also several studies that have shown how bacterial translocation from the gut and skin into the bloodstream may take place in psoriasis, and be responsible for driving the chronic, systemic inflammatory nature of the disease [42].

2.3 Skin mycobiota

An investigation by Findley et al. suggests that fungal diversity is increased in psoriatic lesions, compared to healthy skin sites. Furthermore the skin of psoriatic patients, at the genus level, is dominant with Malassezia [43]. Whereas the study by Takemoto et al. found that psoriatic skin revealed higher diversity and decreased relative abundance of Malassezia, which is still the most abundant phylum compared to controls. Moreover, the ratio of M. globosa to M. restricta is lower in psoriatic lesions [44]. Stehlikova et al. [11] found no significant difference in alpha and beta diversity and a significant increase in abundance of M. restricta in back lesions and M. sympodialis in the elbow lesions. Conversely, however, Paulino et al. showed that psoriatic lesions on the back, in decreasing order of abundance, are predominated by M. restricta, followed by M. globosa and M. sympodialis, respectively. Paulino et al. concluded that there was no significant difference between the fungal compositions of psoriatic and healthy skin [45]. Furthermore, Paulino et al. also showed there was no consistent variation between psoriasis and healthy controls [46] as M. furfur was found only in the skin of psoriasis participants in the study by Jagielski et al. [47] compared to healthy controls and atopic dermatitis.

2.4 Factors affecting microbiota study

So far, no specific patterns of microbiota in psoriatic patients have been identified (Tables 1 and 3).

The difficulty to establish such precise features, although a plethora of published studies have attempted to do so, is due to the lack of standardized protocols. Differences in sample collection and processing, sequencing methods, and analysis procedures between studies may impact the study results [48], and can confound comparisons and results in incompatible outcomes (Table 4).

FindingsBy
Alpha diversityDecreasedScher et al. [23]
ActinobacteriaDecreasedScher et al. [23], Masallat et al. [24]
BacteroidesLowerCodoner et al. [25], Masallat et al. [24]
FeacalibacteriumIncreasedCodoner et al. [25]

Table 4.

Summary of gut microbiota findings in psoriasis.

Different factors that may affect microbiome study

  • Host factors: Not many studies accounted the interpersonal and intrapersonal factors that affect the microbial community.

  • Samples collected from different body sites cannot be compared due to site-specific niches, as described previously [21].

  • Sampling method: Several studies showed that different skin layers contain different bacterial communities [48]. Most of the published research on cutaneous microbiota has been based on skin swabs, which represent the surface of the skin. Prast-Nielsen et al. [49] found differences in both diversity and taxonomic composition of the microbiome obtained from swabs and biopsies of the same individual, while an investigation by Stehlikova et al. showed that various sampling approaches (swab, scraping, and biopsy) in affected and unaffected skin of psoriatic patients and in healthy control skin results in similar bacterial diversity despite the different genera abundance that is observed [11]. Grice et al. used three different sampling strategies in the antecubital fossa of five patients: swabs, skin scrapes, and punch biopsies, and concluded that similar microbial populations were captured by each technique and that the dominant species was present in the noninvasive swabs [50]. Recent studies have also reported that the tape stripping method may capture more viable bacteria than the swabbing method [51].

Sequencing methods, analysis procedures, and techniques for studying the microbiome:

  • Langan et al. demonstrated that the changes in the microbiome during treatment that were detected by 16S rRNA were not detected by culture data. This suggested that changes in bacterial populations may have been too subtle to be detected by culture, or that changes are predominantly in nonculturable species [10].

  • Studies using the most often used 16S rRNA have shown that the accuracy of molecular signatures depends on DNA sequencing and downstream analysis protocols. Therefore numerous combinations of primer pairs have been previously tested to select the most appropriate one for skin microbiome surveys; however, standardized methodology is still lacking [52].

    • Several studies suggested that primers for V1V3 and V3V4 hypervariable regions were described to sufficiently cover the skin bacterial diversity [20].

    • Statnikov et al. concluded that using 16S rRNA data from the V3–V5 locus leads to accurate and statistically significant molecular signatures, whereas data from the V1–V3 locus carry a limited diagnostic signal [53].

    • The latter study by Stehlikova et al. observed that variable regions of the V3V4 region capture a wider microbial diversity than the V1V2 region, where observed and estimated richness was significantly higher when using the V3V4 region compared to the V1V2 region [11].

  • Whole-genome shotgun metagenomics offers the most comprehensive and robust data; however, as a result of its high cost, only a few shotgun metagenomic studies have been conducted on the microbiota associated with the skin, such as the Human Microbiome Project [54]. We found very few studies on psoriasis microbiome.

2.5 Therapeutic implements

Several studies reveal that psoriasis treatment changes the gut and skin microbiome, such as the correlation between psoriasis systemic treatment and the Actinobacteria-to-Firmicutes ratio. Biological therapies demonstrated the largest impact [10] during ustekinumab treatment; the composition of microbiota diverged further between lesional and non-lesional skin, across body sites, which could be due to the regression of lesions that returns the skin to more normal environments and increases the body site-specific niches [12]. Secukinumab (anti-IL17) therapy is associated with distinct and more profound gut microbiome shifts than ustekinumab therapy (anti-IL 12/23), in patients with psoriasis by increasing the relative abundance of Proteobacteria and decreases in Bacteroidetes and Firmicutes [55]. Burns et al. demonstrates that UVR has profound qualitative and quantitative influences on the composition of the skin microbiome by an increase in the phylum Cyanobacteria and a decrease in the family Lactobacillaceae and Pseudomonadaceae [56]. This suggests that skin microbiome alterations after UVB treatment could be related to treatment and treatment responses [13]. Thus, it may be implied that the modulation of the gut and skin microbiota can improve disease condition.

Therapeutic modalities that target the shifting microbiota:

The use of orally administered antibiotics, prebiotics, probiotics, and most recently, fecal transplantation [57] also appears to improve the disease condition and may be a practical prospect as a therapeutic avenue.

  • Antibiotic treatment of psoriasis can alter the bowel flora toward normality, and therapy might include the use of appropriate antibiotics to reduce susceptible microbes while permitting others to flourish [58]. Saxena and Dogra reported that administration of benzathine penicillin in psoriasis vulgaris patients showed a significant improvement [59] and administration of azithromycin revealed a significant improvement at 12 weeks in the patient with psoriasis [60].

  • Pro- and prebiotics are commonly used to modulate the microbiome by promoting the growth of specific species. Three studies using three distinct probiotic species affecting distinct pathways of the pathomechanism of psoriasis [61, 62] have all shown improvement in the course of the disease. The probiotics resulted in the improvement of epithelial barrier function, increased production of TNF-alpha by epithelial cells, and regulated activation of the NF-ĸβ pathway [63]. An issue with probiotic supplementation is that the colonization of probiotic bacteria in the gut is mostly transient as they are only detectable for less than 2 weeks after cessation of intake [64]. However, a study by Maldonado-Gómez et al. demonstrated that a certain Bifidobacterium longum (B. longum) strain was able to persist for over 6 months in a subset of subjects where it was originally absent [65]. A recent, randomized, double-blind, placebo-controlled trial evaluated the effect of a probiotic mixture as co-adjutant treatment together with topical steroids in 90 patients with plaque psoriasis. The results showed a large reduction in the score of severity indexes in the probiotic group, compared with the placebo group. Gut microbiota analysis demonstrated the efficacy of the probiotic in modulation of the composition of the microbiota. After the end of the probiotic or placebo intake, patients were followed up for 6 months. The results showed a lower risk of relapse in patients in the probiotic group [66].

  • Topical probiotics show that after sequential applications of a donor microbiome, the recipient microbiome becomes more similar to the donor [67]. The use of topical probiotics may have special subclinical significance, for example, to improve skin defense with probiotic-containing cosmeceuticals. It has been reported that B. longum strains exert pro-differentiating, as well as and pro-regenerating, effects on primary human epidermal keratinocytes [68]. Thus, using the most suitable oral probiotic strain in combination with topical probiotics and/or prebiotics might help in the personalized treatment of skin disorders.

  • Fecal microbiota transplantation (FMT) is currently being used to restore the balance of the intestinal microbiota [69, 70]. Particularly, this procedure has demonstrated >90% clinical resolution of recurrent, or refractory, Clostridium difficile infections [71]. Also, multiple FMTs seem to be able to induce remission in patients with IBD [72]. Due to these results, FMT is now being tested as a potential novel treatment for other gastrointestinal and extraintestinal diseases [73] as it greatly improves outcomes compared with those before treatment.

Advertisement

3. Conclusion

The function of microbiota may be more important in psoriasis. The metabolic activity of microbiota may become an upcoming research area in near future for identifying crucial biomarkers and new therapeutic approaches for psoriasis. Future cohort studies with standardized methodologies and proof-of-concept investigations in animal models may uncover the role of microbiota and the microbial pathway in psoriasis. This, then, may lead to the development of diagnostics and therapeutic opportunities.

Advertisement

Conflict of interest

The authors have no conflict of interest to declare.

Advertisement

Notes/Thanks/Other declarations

The authors gratefully acknowledge the Bualuang ASEAN Chair Professorship grant award provided by Thammasat University. The funders had no role in any parts of this study.

References

  1. 1. WHO. Global Report on Psoriasis. World Health Organization; 2016. Available on the WHO website (www.who.\r\nint) or can be purchased from WHO Press. ISBN: 9789241565189. Available from: https://apps.who.int/iris/handle/10665/204417
  2. 2. Vandenbroeck K. Cytokine Gene Polymorphisms in Multifactorial Conditions. Boca Raton/FL: CRC Press, Taylor & Francis Group; 2006. p. 459
  3. 3. Mattozzi C, Richetta AG, Cantisani C, MacAluso L, Calvieri S. Psoriasis: New insight about pathogenesis, role of barrier organ integrity, NLR/CATERPILLER family genes and microbial flora. The Journal of Dermatology. 2012;39(9):752-760. DOI: 10.1111/j.1346-8138.2012.01606.x
  4. 4. Tomi NS, Kränke B, Aberer E. Staphylococcal toxins in patients with psoriasis, atopic dermatitis, and erythroderma, and in healthy control subjects. Journal of the American Academy of Dermatology. 2005;53(1):67-72. DOI: 10.1016/j.jaad.2005.02.034
  5. 5. Munz OH, Sela S, Baker BS, Griffiths CEM, Powles AV, Fry L. Evidence for the presence of bacteria in the blood of psoriasis patients. Archives of Dermatological Research. 2010;302:495-498. DOI: 10.1007/s00403-010-1065-0
  6. 6. Rudramurthy SM, Honnavar P, Chakrabarti A, Dogra S, Singh P, Handa S. Association of Malassezia species with psoriatic lesions. Mycoses. 2014;57(8):483-488. DOI: 10.1111/myc.12186
  7. 7. Griffiths CE, Barker JN. Pathogenesis and clinical features of psoriasis. Lancet. 2007;370(9583):263-271. DOI: 10.1016/S0140-6736(07)61128-3
  8. 8. Correia B, Torres T. Obesity: A key component of psoriasis. Acta Biomedica. 2015;86(2):121-129
  9. 9. Fry L, Baker BS, Powles AV, Fahlen A, Engstrand L. Is chronic plaque psoriasis triggered by microbiota in the skin? The British Journal of Dermatology. 2013;169(1):47-52. DOI: 10.1111/bjd.12322
  10. 10. Langan EA, Künstner A, Miodovnik M, Zillikens D, Thaçi D, Baines JF, et al. Combined culture and metagenomic analyses reveal significant shifts in the composition of the cutaneous microbiome in psoriasis. British Journal of Dermatology. 2019. DOI: 10.1111/bjd.17989
  11. 11. Stehlikova Z, Kostovcik M, Kostovcikova K, Kverka M, Juzlova K, Rob F, et al. Dysbiosis of skin microbiota in psoriatic patients: Co-occurrence of fungal and bacterial communities. Frontiers in Microbiology. 2019;10:438. DOI: 10.3389/fmicb.2019.00438
  12. 12. Loesche MA, Farahi K, Capone K, Fakharzadeh S, Blauvelt A, Duffin KC, et al. Longitudinal study of the psoriasis-associated skin microbiome during therapy with ustekinumab in a randomized phase 3b clinical trial. Journal of Investigative Dermatology. 2018;138(9):1973-1981. DOI: 10.1016/j.jid.2018.03.1501
  13. 13. Assarsson M, Duvetorp A, Dienus O, Söderman J, Seifert O. Significant changes in the skin microbiome in patients with chronic plaque psoriasis after treatment with narrowband ultraviolet B. Acta Dermato-Venereologica. 2018;98(4):428-436. DOI: 10.2340/00015555-2859
  14. 14. Chang H-W, Yan D, Singh R, Liu J, Lu X, Ucmak D, et al. Alteration of the cutaneous microbiome in psoriasis and potential role in Th17 polarization. Microbiome. 2018;6(1):154. DOI: 10.1186/s40168-018-0533-1
  15. 15. Drago L, De Grandi R, Altomare G, Pigatto P, Rossi O, Toscano M. Skin microbiota of first cousins affected by psoriasis and atopic dermatitis. Clinical and Molecular Allergy. 2016;14(1):2. DOI: 10.1186/s12948-016-0038-z
  16. 16. Tett A, Pasolli E, Farina S, et al. Unexplored diversity and strain-level structure of the skin microbiome associated with psoriasis. NPJ Biofilms Microbiomes. 2017;3(1):14. DOI: 10.1038/s41522-017-0022-5
  17. 17. Alekseyenko AV, Perez-Perez GI, De Souza A, et al. Community differentiation of the cutaneous microbiota in psoriasis. Microbiome. 2013;1(1):31. DOI: 10.1186/2049-2618-1-31
  18. 18. Fahlén A, Engstrand L, Baker BS, Powles A, Fry L. Comparison of bacterial microbiota in skin biopsies from normal and psoriatic skin. Archives of Dermatological Research. 2012;304(1):15-22. DOI: 10.1007/s00403-011-1189-x
  19. 19. Gao Z, Tseng C, Strober BE, Pei Z, Blaser MJ. Substantial alterations of the cutaneous bacterial biota in psoriatic lesions. PLoS One. 2008;3(7):e2719. DOI: 10.1371/journal.pone.0002719
  20. 20. Grice EA, Segre JA. The skin microbiome. Nature Reviews. Microbiology. 2013;9(4):244-253. DOI: 10.1038/nrmicro2537
  21. 21. Oh J, Byrd AL, Deming C, et al. Biogeography and individuality shape function in the human skin metagenome. Nature. 2014;514(7520):59-64. DOI: 10.1038/nature13786
  22. 22. Fierer N, Hamady M, Lauber CL, Knight R. The influence of sex, handedness, and washing on the diversity of hand surface bacteria. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(46):17994-17999. DOI: 10.1073/pnas.0807920105
  23. 23. Scher JU, Ubeda C, Artacho A, et al. Decreased bacterial diversity characterizes the altered gut microbiota in patients with psoriatic arthritis, resembling dysbiosis in inflammatory bowel disease. Arthritis & Rhematology. 2015;67(1):128-139. DOI: 10.1002/art.38892
  24. 24. Doaa M, Dalia M, Ahmed FS. Gut bacterial microbiota in psoriasis: A case control study. African Journal of. Microbiology Research. 2016;10(33):1337-1343. DOI: 10.5897/ajmr2016.8046
  25. 25. Codoñer FM, Ramírez-Bosca A, Climent E, et al. Gut microbial composition in patients with psoriasis. Scientific Reports. 2018;8(1):3812. DOI: 10.1038/s41598-018-22125-y
  26. 26. Eppinga H, Sperna Weiland CJ, Thio HB, et al. Similar depletion of protective Faecalibacterium prausnitzii in psoriasis and inflammatory bowel disease, but not in Hidradenitis suppurativa. Journal of Crohn’s and Colitis. 2016;10(9):1067-1075. DOI: 10.1093/ecco-jcc/jjw070
  27. 27. Shapiro J, Cohen NA, Shalev V, Uzan A, Koren O, Maharshak N. Psoriatic patients have a distinct structural and functional fecal microbiota compared with controls. The Journal of Dermatology. 2019;46(7):595-603. DOI: 10.1111/1346-8138.14933
  28. 28. Tan L, Zhao S, Zhu W, et al. The Akkermansia muciniphila is a gut microbiota signature in psoriasis. Experimental Dermatology. 2018;27(2):144-149. DOI: 10.1111/exd.13463
  29. 29. Chen Y, Ho HJ, Tseng C, Lai Z, Shieh J, Wu C. Intestinal microbiota profiling and predicted metabolic dysregulation in psoriasis patients. Experimental Dermatology. 2018;27(12):1336-1343. DOI: 10.1111/exd.13786
  30. 30. Ahluwalia B, Magnusson MK, Öhman L. Mucosal immune system of the gastrointestinal tract: Maintaining balance between the good and the bad. Scandinavian Journal of Gastroenterology. 2017;52(11):1185-1193. DOI: 10.1080/00365521.2017.1349173
  31. 31. Blum HE. The human microbiome. Advances in Medical Sciences. 2017;62(2):414-420. DOI: 10.1016/j.advms.2017.04.005
  32. 32. de Faria GF, Oliveira DG, de Oliveira JM, de Castro Ferreira LEVV, Cesar DE, Moreira APB. Influence of gut microbiota on the development and progression of nonalcoholic steatohepatitis. European Journal of Nutrition. 2018;57(3):861-876. DOI: 10.1007/s00394-017-1524-x
  33. 33. Miele L, Giorgio V, Alberelli MA, De Candia E, Gasbarrini A, Grieco A. Impact of gut microbiota on obesity, diabetes, and cardiovascular disease risk. Current Cardiology Reports. 2015;17(12):120. DOI: 10.1007/s11886-015-0671-z
  34. 34. Manichanh C, Rigottier-Gois L, Bonnaud E, et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut. 2006;55(2):205-211. DOI: 10.1136/gut.2005.073817
  35. 35. Park HS, Koh SJ, Park GY, et al. Psoriasis concurrent with inflammatory bowel disease. Journal of the European Academy of Dermatology and Venereology. 2014;28(11):1436-1441. DOI: 10.1111/jdv.12305
  36. 36. Egeberg A, Mallbris L, Warren RB, et al. Association between psoriasis and inflammatory bowel disease: A Danish nationwide cohort study. The British Journal of Dermatology. 2016;175(3):487-492. DOI: 10.1111/bjd.14528
  37. 37. Zákostelská Z, Málková J, Klimešová K, et al. Intestinal microbiota promotes psoriasis-like skin inflammation by enhancing Th17 response. PLoS One. 2016;11(7):e0159539. DOI: 10.1371/journal.pone.0159539
  38. 38. Stehlikova Z, Kostovcikova K, Kverka M, et al. Crucial role of microbiota in experimental psoriasis revealed by a gnotobiotic mouse model. Frontiers in Microbiology. 2019;10(feb):236. DOI: 10.3389/fmicb.2019.00236
  39. 39. Ohnmacht C, Park J-H, Cording S, et al. Mucosal Immunology. The microbiota regulates type 2 immunity through RORγt+ T cells. Science. 2015;349(6251):989-993. DOI: 10.1126/science.aac4263
  40. 40. Cani PD, Amar J, Iglesias MA, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56(7):1761-1772. DOI: 10.2337/db06-1491
  41. 41. Mosca A, Leclerc M, Hugot JP. Gut microbiota diversity and human diseases: Should we reintroduce key predators in our ecosystem? Frontiers in Microbiology. 2016;7:455. DOI: 10.3389/fmicb.2016.00455
  42. 42. Visser MJE, Kell DB, Pretorius E. Bacterial dysbiosis and translocation in psoriasis vulgaris. Frontiers in Cellular and Infection Microbiology. 2019;9:7. DOI: 10.3389/fcimb.2019.00007
  43. 43. Findley K, Oh J, Yang J, et al. Human skin fungal diversity. Nature. 2013;498(7454):367-370. DOI: 10.1038/nature12171.Human
  44. 44. Takemoto A, Cho O, Morohoshi Y, Sugita T, Muto M. Molecular characterization of the skin fungal microbiome in patients with psoriasis. The Journal of Dermatology. 2015;42(2):166-170. DOI: 10.1111/1346-8138.12739
  45. 45. Paulino LC, Tseng CH, Strober BE, Blaser MJ. Molecular analysis of fungal microbiota in samples from healthy human skin and psoriatic lesions. Journal of Clinical Microbiology. 2006;44(8):2933-2941. DOI: 10.1128/JCM.00785-06
  46. 46. Paulino LC, Tseng CH, Blaser MJ. Analysis of Malassezia microbiota in healthy superficial human skin and in psoriatic lesions by multiplex real-time PCR. FEMS Yeast Research. 2008;8(3):460-471. DOI: 10.1111/j.1567-1364.2008.00359.x
  47. 47. Jagielski T, Rup E, Ziółkowska A, Roeske K, Macura AB, Bielecki J. Distribution of Malassezia species on the skin of patients with atopic dermatitis, psoriasis, and healthy volunteers assessed by conventional and molecular identification methods. BMC Dermatology. 2014;14(1):3. doi: 10.1186/1471-5945-14-3
  48. 48. Kong HH, Andersson B, Clavel T, et al. Performing skin microbiome research: A method to the madness. The Journal of Investigative Dermatology. 2017;137(3):561-568. DOI: 10.1016/j.jid.2016.10.033
  49. 49. Prast-Nielsen S, Tobin A-M, Adamzik K, et al. Investigation of the skin microbiome: Swabs vs. biopsies. British Journal of Dermatology. 2019;181(3):572-579. doi: 10.1111/bjd.17691
  50. 50. Grice EA, Kong HH, Renaud G, et al. A diversity profile of the human skin microbiota. Genome Research. 2008;18(7):1043-1050. DOI: 10.1101/gr.075549.107
  51. 51. Ogai K, Nagase S, Mukai K, et al. A comparison of techniques for collecting skin microbiome samples: Swabbing versus tape-stripping. Frontiers in Microbiology. 2018;9:2362. DOI: 10.3389/fmicb.2018.02362
  52. 52. Meisel JS, Hannigan GD, Tyldsley AS, et al. Skin microbiome surveys are strongly influenced by experimental design. The Journal of Investigative Dermatology. May 2016;136(5):947-956. DOI: 10.1016/j.jid.2016.01.016
  53. 53. Statnikov A, Alekseyenko AV, Li Z, et al. Microbiomic signatures of psoriasis: Feasibility and methodology comparison. Scientific Reports. 2013;3:1-7. DOI: 10.1038/srep02620
  54. 54. Huttenhower C, Gevers D, Knight R, et al. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207-214. DOI: 10.1038/nature11234
  55. 55. Yeh N-L, Hsu C-Y, Tsai T-F, Chiu H-Y. Gut microbiome in psoriasis is perturbed differently during secukinumab and ustekinumab therapy and associated with response to treatment. Clinical Drug Investigation. 2019;39(12):1195-1203. DOI: 10.1007/s40261-019-00849-7
  56. 56. Burns EM, Ahmed H, Isedeh PN, et al. Ultraviolet radiation, both UVA and UVB, influences the composition of the skin microbiome. Experimental Dermatology. 2019;28(2):136-141. DOI: 10.1111/exd.13854
  57. 57. Mikó E, Vida A, Bai P. Translational aspects of the microbiome—To be exploited. Cell Biology and Toxicology. 2016;32:153-156. DOI: 10.1007/s10565-016-9320-6
  58. 58. Wang GL, Li XY, Wang MY, et al. Cell-wall-deficient bacteria: A major etiological factor for psoriasis? Chinese Medical Journal. 2009;122(24):3011-3016. DOI: 10.3760/cma.j.issn.0366-6999.2009.24.019
  59. 59. Saxena VN, Dogra J. Long-term use of penicillin for the treatment of chronic plaque psoriasis. European Journal of Dermatology. 2005;15(5):359-362. DOI: 10.1016/s0093-3619(08)70381-2
  60. 60. Saxena VN, Dogra J. Long-term oral azithromycin in chronic plaque psoriasis: A controlled trial. European Journal of Dermatology. 2010;20(3):329-333. DOI: 10.1684/ejd.2010.0930
  61. 61. Chen YH, Wu CS, Chao YH, et al. Lactobacillus pentosus GMNL-77 inhibits skin lesions in imiquimod-induced psoriasis-like mice. Journal of Food and Drug Analysis. 2017;25(3):559-566. DOI: 10.1016/j.jfda.2016.06.003
  62. 62. Vijayashankar M, Raghunath N. Pustular psoriasis responding to probiotics—A new insight. Our Dermatology Online. 2012;3(4):326-328. DOI: 10.7241/ourd.20124.71
  63. 63. Parisi R, Symmons DPM, Griffiths CEM, Ashcroft DM. Global epidemiology of psoriasis: A systematic review of incidence and prevalence. The Journal of Investigative Dermatology. 2013;133(2):377-385. DOI: 10.1038/jid.2012.339
  64. 64. Frese S, Hutkins RW, Walter J. Comparison of the colonization ability of autochthonous and allochthonous strains of Lactobacilli in the human gastrointestinal tract. Advances in Microbiology. 2012;2(3):399-409. DOI: 10.4236/aim.2012.23051
  65. 65. Maldonado-Gómez MX, Martínez I, Bottacini F, et al. Stable engraftment of Bifidobacterium longum AH1206 in the human gut depends on individualized features of the resident microbiome. Cell Host & Microbe. 2016;20(4):515-526. DOI: 10.1016/j.chom.2016.09.001
  66. 66. Navarro-López V, Martínez-Andrés A, Ramírez-Boscá A, et al. Efficacy and safety of oral administration of a mixture of probiotic strains in patients with psoriasis: A randomized clinical trial. Acta Dermato-Venereologica. 2019;9:0. DOI: 10.2340/00015555-3305
  67. 67. Paetzold B, Willis JR, Pereira de Lima J, et al. Skin microbiome modulation induced by probiotic solutions. Microbiome. 2019;7(1):95. DOI: 10.1186/s40168-019-0709-3
  68. 68. Szöllősi AG, Gueniche A, Jammayrac O, et al. Bifidobacterium longum extract exerts pro-differentiating effects on human epidermal keratinocytes, in vitro. Experimental Dermatology. 2017;26(1):92-94. DOI: 10.1111/exd.13130
  69. 69. Kragsnaes MS, Kjeldsen J, Horn HC, et al. Efficacy and safety of faecal microbiota transplantation in patients with psoriatic arthritis: Protocol for a 6-month, double-blind, randomised, placebo-controlled trial. BMJ Open. 2018;8(4):e019231. DOI: 10.1136/bmjopen-2017-019231
  70. 70. Cammarota G, Pecere S, Ianiro G, Masucci L, Currò D. Principles of DNA-based gut microbiota assessment and therapeutic efficacy of fecal microbiota transplantation in gastrointestinal diseases. Digestive Diseases. 2016;34(3):279-285. DOI: 10.1159/000443362
  71. 71. Van Nood E, Vrieze A, Nieuwdorp M, et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. The New England Journal of Medicine. 2013;368(5):407-415. DOI: 10.1056/NEJMoa1205037
  72. 72. Paramsothy S, Kamm MA, Kaakoush NO, et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: A randomised placebo-controlled trial. Lancet. 2017;389(10075):1218-1228. DOI: 10.1016/S0140-6736(17)30182-4
  73. 73. Cui B, Feng Q , Wang H, et al. Fecal microbiota transplantation through mid-gut for refractory Crohn’s disease: Safety, feasibility, and efficacy trial results. Journal of Gastroenterology and Hepatology. 2015;30(1):51-58. DOI: 10.1111/jgh.12727

Written By

Atiya Rungjang, Jitlada Meephansan and Hok Bing Thio

Submitted: 17 March 2020 Reviewed: 28 April 2020 Published: 02 June 2020