Microbiome and Rheumatoidarthritis
Microbiome and Rheumatoidarthritis
Microbiome and Rheumatoidarthritis
specific bacterial lineages and alterations in gut microbiota shaping their function and phenotypes. The gut microbiota
metabolism led to changes in the host immune profile that mediates constant bidirectional communication with the host
contribute to RA (8). It has been proposed that the mechanism immune system in a delicate balance of inducing pathogenic
by which gut microbiota imbalance leads to RA may be related to infection or residing in the human body in a commensal state
regulation of immune function by metabolites produced by gut (27). The innate immune cells in the gut-associated lymphoid
microbes (9, 10). Immune T and B cells have position-specific tissue comprise the first-line of defense against xenobiotics from
phenotypes and functions in the mucosa, influenced by the the gastrointestinal tract. Disturbed gut microbiota can trigger the
microbiota (11). In turn, bacterial peptidoglycan components aberrant activation of innate immune cells, which leads to the
are found in the synovial tissue of RA patients, which may upregulation of proinflammatory cytokine including interleukin-
contribute to inflammation within the microenvironment of the 12 (IL-12), IL-23, and type I interferons, etc., as well as reduction
joint (12, 13). Substantial data published in the past few years of anti-inflammatory cytokines including transforming growth
demonstrate that an altered composition of the gut microbiota in factor b and IL-10, etc. (28). Moreover, the adaptive lymphocytes
RA patients is one of the major factors triggering aberrant are critical players in autoimmunity, and aberrant activation of T
systemic immunity (14–16). Notably, different strains of gut and B cells instigates RA. Gut pathogens with proinflammatory
bacteria can have profoundly different regulatory effects on capacities can reshape the immune milieu through innate immune
immune system function. Some strains can stimulate an overactivation, followed by aberrant activation of the adaptive
immune response, benefiting immunocompromised patients, immune system. Microbial antigens can be presented to CD4+ T
while others can suppress the immune response, affecting cells by dendritic cells and macrophages, leading to differentiation
immune regulation in RA patients (17–21). For example, of inflammatory T cell subtypes. Th17 cells are a subset of
segmented filamentous bacteria (SFB) have a unique ability to proinflammatory CD4+ T cells characterized by production of
drive T helper 17 (Th17) cell accumulation in the small interleukin-17 (IL-17) (23). Tregs are also derived from CD4+ T
intestine’s lamina propria through SFB-derived antigens cells, show instead immunosuppressive actions, and may inhibit
presented by dendritic cells (22–24). In contrast, the Th17 responses (29, 30). Studies have demonstrated that an
colonization of Bacteroides fragilis is associated with enhanced increased Th17/Treg ratio is closely related to RA, and that the
activity of regulatory T cells (Tregs), which may alleviate Th17/Treg balance is strongly regulated by gut microbiota and
autoimmune disease (25, 26). Therefore, the relative their metabolites (31, 32). Microbial antigens can also induce
abundance of different bacterial lineages may lead to changes overactivation of B lymphocytes with the help of T follicular
in the host immune profile and drive inflammatory responses helper cells, differentiating into plasma cells and producing
contributing to RA. pathogenic autoantibodies. This may influence the pathogenesis
This mini-review discusses the role of the gut microbiota in of RA (28). Therefore, the dysbiosis gut microbiota, inflammatory
the pathogenesis of RA, summarizes the diagnostic value of gut factors, and immune responses are interrelated and jointly affect
microbe-based biomarkers, and outlines mutual influences the development of RA (33) (Figure 1).
between the gut microbiota and some drugs used to treat RA.
Some suggestions and outlooks are also raised to guide future
research efforts. Intestinal barrier dysfunction
FIGURE 1
Gut microbiota in the pathogenesis of rheumatoid arthritis (RA) and effects of RA therapeutic drugs on the gut microbiota. (A) Changes in the composition of
gut microbiota at different stages of RA. Levels of Prevotella copri and Lactobacillus are increased, while those of Bacteroidetes, Bifidobacteria and Eubacterium
rectale are decreased, at an early stage; Abundance of Lactobacillus salivarius, Collinsella, and Akkermansia is increased, while that of Haemophilus spp. is
decreased, in the active RA phase. (B) RA treatment drugs can improve gut microbiota imbalance and relieve disease symptoms, mainly including methotrexate,
sulfasalazine, hydroxychloroquine, etanercept, and traditional Chinese medicine. (C) Gut microbiota can lead to damage of the epithelium and to the opening
of the paracellular pathway and can cross the epithelium and get in contact with the immune cells beneath the epithelial layer, which leads to inflammation.
Furthermore, bacterial antigens promote activation of autoreactive immune cells (B cell and T cell) in the lymphoid tissues, resulting in an imbalance between
regulatory T cells (Tregs) and T helper 17 (Th17) cells, leading to expansion of inflammatory response. Activated B cells produce autoantibodies (anti-citrullinated
protein antibody and rheumatoid factor). (D) Gut microbiota imbalance can trigger the migration of autoreactive cells to the joints, causing cartilage and bone
damage. ① Bacterial antigens trigger promotes inflammation in synovial membrane, attracting leukocytes into the tissue. ② Autoreactive cells activate
macrophages, resulting in inflammatory cytokine production. ③ Cytokines induce fibroblasts to produce MMPs (matrix metalloproteinases) and RANKL
(receptor activator of nuclear factor kB ligand), which mediate destruction of bone and cartilage tissue, leading to the development of RA.
has shown that Collinsella aerofaciens, a commensal gut transgenic mice subjected to collagen-induced arthritis (CIA) by
bacterium found to be overrepresented in RA patients, reduces disrupting the integrity of the intestinal mucosa (36). In contrast,
the expression of tight junction proteins in human intestinal Faecalibacterium prausnitzii, a prominent member of the
epithelial cells and increases disease incidence in HLA-DQ8 human commensal gut microbiota whose abundance is
reduced in patients with RA, was shown to sustain intestinal major advances, the mechanisms linking dysbiosis of the gut
barrier function, maintain Th17/Treg balance, and exert microbiota and RA remain still incompletely characterized.
significant anti-inflammatory effects (37). These findings
indicate that changes in gut microbiota diversity may impair
intestinal mucosal permeability, facilitating the onset of RA (38, Gut microbiota biomarkers for
39). can trigger the migration of autoreactive cells to the joints, diagnosing RA
causing cartilage and bone damage.
Early diagnosis of RA is critical to provide prompt treatment
to slow down joint damage. Scher et al. (2013) reported that the
Molecular mimicry abundance of Prevotella copri was increased, while that of
Bacteroidetes was decreased, in patients with early RA; the
Molecular mimicry is a mechanism by which pathogen- presence of P. copri was correlated with a reduction in the
derived antigens that share sequence homology with self- abundance of other bacterial groups, including many beneficial
peptides may lead to cross-activation of autoreactive T or B microbes (46). Transplanting gut microbiota (with a high
cells, triggering autoimmunity. Many identical peptides between abundance of Prevotella) from patients with early RA into
human tissues and gut microbes bind to HLA-II alleles. The germ-free SKG mice can induce severe arthritis (47). In
autoimmune candidates have been shown to be enriched in addition, increased number and species of intestinal
bacterial species belonging to the Firmicutes and Proteobacteria, Lactobacillus were observed in early RA patients than in
which may have a higher disease impact in genetically susceptible healthy individuals (48). Studies have shown that the
individuals (40). N-acetylglucosamine-6-sulfatase and filamin A monocontamination of germ-free IL-1 receptor antagonist–
were identified as T- and B-cell-targeted autoantigens in more deficient (IL1rn-/-) mice, which develop spontaneous arthritis
than 50% of RA patients (41). The HLA-DR-presented N- due to excessive IL-1 signaling, with indigenous Lactobacillus
acetylglucosamine-6-sulfatase peptide has marked sequence bifidus resulted in rapid onset of arthritis that reached incidence
homology with epitopes from sulfatase proteins of the Prevotella rate and severity scores comparable to those recorded in non–
sp. and Parabacteroides sp., whereas the HLA-DR-presented germ-free mice (49). It was in turn reported that in early RA
filamin A peptide has homology with epitopes from proteins of patients the fecal microbiota contained significantly less
the Prevotella sp. and Butyricimonas sp., another gut commensal Bifidobacteria, B. fragilis, and Eubacterium rectale than in
(41). In turn, the presence of shared sequences between Collinsella patients with fibromyalgia (50).
and DRB1*0401 suggested that Collinsella may induce RA In the active period of RA, depletion of Haemophilus spp. in
through molecular mimicry (36). These findings thus identify the patients’ gut, teeth, and saliva correlated negatively with RA
molecular mimicry as a plausible link between disrupted mucosal severity and serum antibody levels. In contrast, Lactobacillus
immune tolerance and systemic immunity in RA patients. salivarius was present in increased amounts in cases of very
active RA (16). Meanwhile, gut microbiota analysis revealed
higher relative abundances of the genera Collinsella and
Other factors Akkermansia in patients with active, compared to inactive, RA
disease status (51). A study indicated that treatment with
Gut microbiota composition can alter sex hormone levels to Collinsella exacerbated CIA in HLA-DQ8 transgenic mice (36).
affect the occurrence of RA. Sex hormone deficiency increases These observations reaffirm the impact of alterations in the gut
intestinal permeability, thereby increasing the number of Th17 microbiota on RA severity and suggest that changes in gut
cells in peripheral blood and the levels of osteoclastogenic microbiota composition may serve as markers for the
cytokines RANKL, IL-17 and TNF-a, promoting bone diagnosis of RA. However, further research is needed to
resorption (42). Clostridium can produce enzymes that conclusively identify reliable gut microbiota biomarkers for
catalyze the conversion of glucocorticoids into androgens, diagnosing RA (Figure 1).
exerting immunomodulatory effects (43). Implying a strong
connection between periodontitis and RA, Porphyromonas
gingivalis, a major pathogen in periodontitis, promotes Therapeutic modulation of the gut
citrullination of proteins and production of anti-citrullinated
protein antibodies, a hallmark of RA. This is mediated by
microbiota and impact of gut
production of peptidyl arginine deiminase (PAD) and microbes on the efficacy of
leukotoxin A, which triggers in turn endogenous PAD RA drugs
activation in neutrophils (44). Besides, several metabolites
including short-chain fatty acids, serotonin, as well as plant The gut microbiota was found to predict drug response in
enzymes in the gut can also influence RA (45). However, despite RA (52). On the other hand, in recent years oral probiotics and
fecal flora transplantation have shown promising results when system and appears to normalize lymphocyte activity by
used as adjuvant therapy for treating RA, by directly and regulating gut microbiota (68). A study in active RA patients
indirectly modulating the gut microbiota. Indirect regulation showed that sulfasalazine therapy led to a substantial fall in fecal
of gut microbiota in RA patients and animal models is also counts of Clostridium perfringens and Escherichia coli (69).
exerted by disease-modifying anti-rheumatic drugs (DMARDs), Furthermore, sulfasalazine treatment significantly altered fecal
traditional Chinese medicine (TCM) herbs and prescriptions, microflora of RA patients by reducing total aerobic bacteria,
and by adjusting diet structure. Bacteroides, and Escherichia coli, and increasing the numbers of
Bacillus (70). However, thorough characterization of the effects
of sulfasalazine therapy on the gut microbiota in RA is
Direct regulation of gut microbiota still lacking.
Treatment with etanercept, a TNF-a antagonist, was shown
Probiotics, defined as “live microorganisms that, when to beneficially impact gut microbiota composition. In RA
administered in adequate amounts, confer a health benefit on patients, etanercept treatment was associated with enrichment
the host” (53), can reduce the abundance of pathogenic bacteria of Cyanobacteria, including members of the Nostocophycideae
by competing for nutrition and colonization sites. At the same class and the Nostocales order (which were not represented
time, probiotics can alleviate RA symptoms by producing among naïve patients), as well as with decreased abundance of
antibiotics and strengthening the intestinal barrier, with Clostridiaceae and Deltaproteobacteria (71). In CIA mice,
beneficial modulation of the immune function (54). Studies etanercept treatment led to decreased microbial community
conducted in rats with adjuvant-induced arthritis (AIA) richness and diversity, increasing the abundance of Escherichia
showed that oral administration of L. casei or L. acidophilus and Shigella and decreasing the abundance of Clostridium XIVa,
reduced arthritic inflammation, pannus formation, and cartilage Tannerella, and Lactobacillus (72). In contrast,
destruction (55–57). More recently, it was reported that hydroxychloroquine treatment in RA patients was associated
administration of L. casei to AIA rats significantly suppressed with increased intestinal bacterial richness and diversity,
arthritis and protected against bone loss by reducing dysbiosis of suggestive of restoration of normal microbiota. In addition,
the gut microbiota (58). However, supplementation with L. the abundance of Faecalibacterium, found to be decreased
reuteri and L. rhamnosus GG did not significantly reduce RA prior to treatment, was positively correlated with the use of
disease activity (59, 60), suggesting that different Lactobacillus hydroxychloroquine (36, 71).
species may act differently on RA. Numerous studies indicated that TCM-based therapies
Fecal microbiota transplantation (FMT) refers to the provide significant curative effects and elicit minor adverse
introduction of gut microbiota obtained from the feces of a reactions in RA (73). Notably, part of the beneficial effects of
healthy donor into a patient’s gastrointestinal tract (61). TCM on RA may be ascribed to regulation of the intestinal flora.
Normalizing the gut microbiota through FMT may potentially Mei et al. (2021) showed that depletion of Clostridium celatum in
improve RA symptoms. A case of a patient with refractory RA RA patients could be reversed by treatment with the Huayu-
successfully treated with FMT indicated that FMT may have an Qiangshen-Tongbi formula (74). Administration of total
excellent therapeutic effect on RA (62). However, clinical studies glucosides of paeony to CIA rats corrected 78% of the
examining the efficacy of FMT in RA patients are so far scarce. taxonomic differences in microbial structure, while also
increasing the relative abundance of certain forms of beneficial
commensal bacteria (75). Similarly, most of the 19 types of
Indirect regulation of gut microbiota bacteria found to be altered at the family level in CIA rats could
be regulated by the Zushima tablet (76). It was also reported that
DMARDs may indirectly affect and remodel the structure Qingluo Tongbi decoction can effectively ameliorate arthritis in
and function of gut microbiota to regulate systemic immunity. AIA rats at least partly by decreasing inflammatory responses
Studies have demonstrated that microbial differences in the regulated by the gut microbiota (77).
gastrointestinal tract of RA patients may partially determine In addition to therapeutic drugs, dietary nutrients also affect
the bioavailability and subsequent clinical outcome of the composition and function of the gut microbiota and may
methotrexate (63, 64). In turn, methotrexate treatment was thus have an important impact on the prevention and treatment
shown to partially restore normal gut microbiota composition of RA. Dietary fiber, abundant in vegetarian diets, can improve
in RA patients (16, 65). Thus, the gut microbiota may be a gut microbiota composition in RA patients and reduce joint pain
predictor of clinical response to methotrexate, influencing the (78). Reducing the intake of carbohydrates can help improve the
treatment response rate. balance of intestinal flora and immune function (79, 80). The
Another DMARD, i.e. sulfasalazine, is cleaved by the action omega-3 polyunsaturated fatty acids can help to maintain
of bacterial azoreductases in the large intestine into sulfapyridine the intestinal barrier integrity and interact with host immune
and mesalazine (66, 67). Sulfapyridine affects the immune cells (81). A low ratio of omega-3/omega-6 fatty acids has been
References
1. Weyand CM, Goronzy JJ. The immunology of rheumatoid arthritis. Nat 2. Firestein GS. Evolving concepts of rheumatoid arthritis. Nature (2003) 423
Immunol (2021) 22(1):10–8. doi: 10.1038/s41590-020-00816-x (6937):356–61. doi: 10.1038/nature01661
3. McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med human CD39(+)Foxp3(+) T cells and treg function. Gut Microbes (2015) 6(4):234–
(2011) 365(23):2205–19. doi: 10.1056/NEJMra1004965 42. doi: 10.1080/19490976.2015.1056973
4. Edilova MI, Akram A, Abdul-Sater AA. Innate immunity drives pathogenesis 27. Shin C, Kim YK. Autoimmunity in microbiome-mediated diseases and
of rheumatoid arthritis. Biomed J (2021) 44(2):172–82. doi: 10.1016/ novel therapeutic approaches. Curr Opin Pharmacol (2019) 49:34–42. doi: 10.1016/
j.bj.2020.06.010 j.coph.2019.04.018
5. Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune 28. Zhang X, Chen BD, Zhao LD, Li H. The gut microbiota: Emerging evidence
responses during health and disease. Nat Rev Immunol (2009) 9(5):313–23. in autoimmune diseases. Trends Mol Med (2020) 26(9):862–73. doi: 10.1016/
doi: 10.1038/nri2515 j.molmed.2020.04.001
6. Scher JU, Abramson SB. The microbiome and rheumatoid arthritis. Nat Rev 29. Lina C, Conghua W, Nan L, Ping Z. Combined treatment of etanercept and
Rheumatol (2011) 7(10):569–78. doi: 10.1038/nrrheum.2011.121 MTX reverses Th1/Th2, Th17/Treg imbalance in patients with rheumatoid
7. Gomez A, Luckey D, Yeoman CJ, Marietta EV, Berg Miller ME, Murray JA, arthritis. J Clin Immunol (2011) 31(4):596–605. doi: 10.1007/s10875-011-9542-6
et al. Loss of sex and age driven differences in the gut microbiome characterize 30. Chen X, Oppenheim JJ. Th17 cells and tregs: Unlikely allies. J Leukoc Biol
arthritis-susceptible 0401 mice but not arthritis-resistant 0402 mice. PLoS One (2014) 95(5):723–31. doi: 10.1189/jlb.1213633
(2012) 7(4):e36095. doi: 10.1371/journal.pone.0036095 31. Kelly D, Mulder IE. Microbiome and immunological interactions. Nutr Rev
8. Wang Q, Zhang SX, Chang MJ, Qiao J, Wang CH, Li XF, et al. Characteristics (2012) 70 Suppl 1:S18–30. doi: 10.1111/j.1753-4887.2012.00498.x
of the gut microbiome and its relationship with peripheral CD4(+) T cell 32. Cheng H, Guan X, Chen D, Ma W. The Th17/Treg cell balance: A gut
subpopulations and cytokines in rheumatoid arthritis. Front Microbiol (2022) microbiota-modulated story. Microorganisms (2019) 7(12):583. doi: 10.3390/
13:799602. doi: 10.3389/fmicb.2022.799602 microorganisms7120583
9. He J, Chu Y, Li J, Meng Q, Liu Y, Jin J, et al. Intestinal butyrate-metabolizing 33. Li Y, Zhang SX, Yin XF, Zhang MX, Qiao J, Xin XH, et al. The gut
species contribute to autoantibody production and bone erosion in rheumatoid microbiota and its relevance to peripheral lymphocyte subpopulations and
arthritis. Sci Adv (2022) 8(6):eabm1511. doi: 10.1126/sciadv.abm1511 cytokines in patients with rheumatoid arthritis. J Immunol Res (2021)
10. Yu D, Du J, Pu X, Zheng L, Chen S, Wang N, et al. The gut microbiome and 2021:6665563. doi: 10.1155/2021/6665563
metabolites are altered and interrelated in patients with rheumatoid arthritis. Front 34. Brandl C, Bucci L, Schett G, Zaiss MM. Crossing the barriers: Revisiting the
Cell Infect Microbiol (2021) 11:763507. doi: 10.3389/fcimb.2021.763507 gut feeling in rheumatoid arthritis. Eur J Immunol (2021) 51(4):798–810.
11. Honda K, Littman DR. The microbiota in adaptive immune homeostasis doi: 10.1002/eji.202048876
and disease. Nature (2016) 535(7610):75–84. doi: 10.1038/nature18848 35. Pacifici R. T Cells, osteoblasts, and osteocytes: Interacting lineages key for
12. Schrijver IA, Melief MJ, Tak PP, Hazenberg MP, Laman JD. Antigen- the bone anabolic and catabolic activities of parathyroid hormone. Ann N Y Acad
presenting cells containing bacterial peptidoglycan in synovial tissues of Sci (2016) 1364(1):11–24. doi: 10.1111/nyas.12969
rheumatoid arthritis patients coexpress costimulatory molecules and cytokines. 36. Chen J, Wright K, Davis JM, Jeraldo P, Marietta EV, Murray J, et al. An
Arthritis Rheum (2000) 43(10):2160–8. doi: 10.1002/1529-0131(200010) expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis.
43:10<2160::Aid-anr3>3.0.Co;2-t Genome Med (2016) 8(1):43. doi: 10.1186/s13073-016-0299-7
13. Pianta A, Arvikar S, Strle K, Drouin EE, Wang Q, Costello CE, et al. 37. Zhou L, Zhang M, Wang Y, Dorfman RG, Liu H, Yu T, et al.
Evidence of the immune relevance of prevotella copri, a gut microbe, in patients Faecalibacterium prausnitzii produces butyrate to maintain Th17/Treg balance
with rheumatoid arthritis. Arthritis Rheumatol (2017) 69(5):964–75. doi: 10.1002/ and to ameliorate colorectal colitis by inhibiting histone deacetylase 1.
art.40003 Inflammation Bowel Dis (2018) 24(9):1926–40. doi: 10.1093/ibd/izy182
14. Kinashi Y. Hase k. partners in leaky gut syndrome: Intestinal dysbiosis and 38. Tajik N, Frech M, Schulz O, Schälter F, Lucas S, Azizov V, et al. Targeting
autoimmunity. Front Immunol (2021) 12:673708. doi: 10.3389/fimmu.2021.673708 zonulin and intestinal epithelial barrier function to prevent onset of arthritis. Nat
15. Chen B, Sun L, Zhang X. Integration of microbiome and epigenome to Commun (2020) 11(1):1995. doi: 10.1038/s41467-020-15831-7
decipher the pathogenesis of autoimmune diseases. J Autoimmun (2017) 83:31–42. 39. Matei DE, Menon M, Alber DG, Smith AM, Nedjat-Shokouhi B, Fasano A,
doi: 10.1016/j.jaut.2017.03.009 et al. Intestinal barrier dysfunction plays an integral role in arthritis pathology and
16. Zhang X, Zhang D, Jia H, Feng Q, Wang D, Liang D, et al. The oral and gut can be targeted to ameliorate disease. Med (N Y) (2021) 2(7):864–83.e9.
microbiomes are perturbed in rheumatoid arthritis and partly normalized after doi: 10.1016/j.medj.2021.04.013
treatment. Nat Med (2015) 21(8):895–905. doi: 10.1038/nm.3914 40. Negi S, Singh H, Mukhopadhyay A. Gut bacterial peptides with
17. Jung H, Jung SM, Rim YA, Park N, Nam Y, Lee J, et al. Arthritic role of autoimmunity potential as environmental trigger for late onset complex
porphyromonas gingivalis in collagen-induced arthritis mice. PLoS One (2017) 12 diseases: In-silico study. PloS One (2017) 12(7):e0180518. doi: 10.1371/journal.
(11):e0188698. doi: 10.1371/journal.pone.0188698 pone.0180518
18. Ferreira-Halder CV, Faria AVS, Andrade SS. Action and function of 41. Pianta A, Arvikar SL, Strle K, Drouin EE, Wang Q, Costello CE, et al. Two
faecalibacterium prausnitzii in health and disease. Best Pract Res Clin rheumatoid arthritis-specific autoantigens correlate microbial immunity with
Gastroenterol (2017) 31(6):643–8. doi: 10.1016/j.bpg.2017.09.011 autoimmune responses in joints. J Clin Invest (2017) 127(8):2946–56.
19. Iljazovic A, Roy U, Gá lvez EJC, Lesker TR, Zhao B, Gronow A, et al. doi: 10.1172/jci93450
Perturbation of the gut microbiome by prevotella spp. enhances host susceptibility 42. Li JY, Chassaing B, Tyagi AM, Vaccaro C, Luo T, Adams J, et al. Sex steroid
to mucosal inflammation. Mucosal Immunol (2021) 14(1):113–24. doi: 10.1038/ deficiency-associated bone loss is microbiota dependent and prevented by
s41385-020-0296-4 probiotics. J Clin Invest (2016) 126(6):2049–63. doi: 10.1172/jci86062
20. Amdekar S, Singh V, Kumar A, Sharma P, Singh R. Lactobacillus casei and 43. Ridlon JM, Ikegawa S, Alves JM, Zhou B, Kobayashi A, Iida T, et al.
lactobacillus acidophilus regulate inflammatory pathway and improve antioxidant Clostridium scindens: A human gut microbe with a high potential to convert
status in collagen-induced arthritic rats. J Interferon Cytokine Res (2013) 33(1):1–8. glucocorticoids into androgens. J Lipid Res (2013) 54(9):2437–49. doi: 10.1194/
doi: 10.1089/jir.2012.0034 jlr.M038869
21. Alpizar-Rodriguez D, Lesker TR, Gronow A, Gilbert B, Raemy E, Lamacchia 44. Farquharson D, Butcher JP, Culshaw S. Periodontitis, porphyromonas, and
C, et al. Prevotella copri in individuals at risk for rheumatoid arthritis. Ann Rheum the pathogenesis of rheumatoid arthritis. Mucosal Immunol (2012) 5(2):112–20.
Dis (2019) 78(5):590–3. doi: 10.1136/annrheumdis-2018-214514 doi: 10.1038/mi.2011.66
22. Flannigan KL, Denning TL. Segmented filamentous bacteria-induced 45. Rosser EC, Piper CJM, Matei DE, Blair PA, Rendeiro AF, Orford M, et al.
immune responses: A balancing act between host protection and autoimmunity. Microbiota-derived metabolites suppress arthritis by amplifying aryl-hydrocarbon
Immunology (2018) 154(4):537–46. doi: 10.1111/imm.12950 receptor activation in regulatory b cells. Cell Metab (2020) 31(4):837–51.e10.
23. Wang Y, Yin Y, Chen X, Zhao Y, Wu Y, Li Y, et al. Induction of intestinal doi: 10.1016/j.cmet.2020.03.003
Th17 cells by flagellins from segmented filamentous bacteria. Front Immunol 46. Scher JU, Sczesnak A, Longman RS, Segata N, Ubeda C, Bielski C, et al.
(2019) 10:2750. doi: 10.3389/fimmu.2019.02750 Expansion of intestinal prevotella copri correlates with enhanced susceptibility to
24. Atarashi K, Tanoue T, Ando M, Kamada N, Nagano Y, Narushima S, et al. arthritis. Elife (2013) 2:e01202. doi: 10.7554/eLife.01202
Th17 cell induction by adhesion of microbes to intestinal epithelial cells. Cell (2015) 47. Maeda Y, Kurakawa T, Umemoto E, Motooka D, Ito Y, Gotoh K, et al.
163(2):367–80. doi: 10.1016/j.cell.2015.08.058 Dysbiosis contributes to arthritis development via activation of autoreactive T
25. Round JL, Lee SM, Li J, Tran G, Jabri B, Chatila TA, et al. The toll-like cells in the intestine. Arthritis Rheumatol (2016) 68(11):2646–61. doi: 10.1002/
receptor 2 pathway establishes colonization by a commensal of the human art.39783
microbiota. Science (2011) 332(6032):974–7. doi: 10.1126/science.1206095 48. Liu X, Zou Q, Zeng B, Fang Y, Wei H. Analysis of fecal lactobacillus
26. Telesford KM, Yan W, Ochoa-Reparaz J, Pant A, Kircher C, Christy MA, community structure in patients with early rheumatoid arthritis. Curr Microbiol
et al. A commensal symbiotic factor derived from bacteroides fragilis promotes (2013) 67(2):170–6. doi: 10.1007/s00284-013-0338-1
49. Abdollahi-Roodsaz S, Joosten LA, Koenders MI, Devesa I, Roelofs MF, 69. Neumann VC, Shinebaum R, Cooke EM, Wright V. Effects of sulphasalazine on
Radstake TR, et al. Stimulation of TLR2 and TLR4 differentially skews the balance faecal flora in patients with rheumatoid arthritis: A comparison with penicillamine. Br J
of T cells in a mouse model of arthritis. J Clin Invest (2008) 118(1):205–16. Rheumatol (1987) 26(5):334–7. doi: 10.1093/rheumatology/26.5.334
doi: 10.1172/jci32639 70. Kanerud L, Scheynius A, Nord CE, Hafström I. Effect of sulphasalazine on
50. Vaahtovuo J, Munukka E, Korkeamäki M, Luukkainen R, Toivanen P. gastrointestinal microflora and on mucosal heat shock protein expression in
Fecal microbiota in early rheumatoid arthritis. J Rheumatol (2008) 35(8):1500–5. patients with rheumatoid arthritis. Br J Rheumatol (1994) 33(11):1039–48.
51. Chiang HI, Li JR, Liu CC, Liu PY, Chen HH, Chen YM, et al. An association doi: 10.1093/rheumatology/33.11.1039
of gut microbiota with different phenotypes in Chinese patients with rheumatoid 71. Picchianti-Diamanti A, Panebianco C, Salemi S, Sorgi ML, Di Rosa R,
arthritis. J Clin Med (2019) 8(11):1770. doi: 10.3390/jcm8111770 Tropea A, et al. Analysis of gut microbiota in rheumatoid arthritis patients:
52. Onuora S. Gut microbiome could predict drug response in RA. Nat Rev Disease-related dysbiosis and modifications induced by etanercept. Int J Mol Sci
Rheumatol (2021) 17(3):129. doi: 10.1038/s41584-021-00582-6 (2018) 19(10):2938. doi: 10.3390/ijms19102938
53. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. Expert 72. Wang B, He Y, Tang J, Ou Q, Lin J. Alteration of the gut microbiota in
consensus document. the international scientific association for probiotics and tumor necrosis factor-a antagonist-treated collagen-induced arthritis mice. Int J
prebiotics consensus statement on the scope and appropriate use of the term Rheum Dis (2020) 23(4):472–9. doi: 10.1111/1756-185x.13802
probiotic. Nat Rev Gastroenterol Hepatol (2014) 11(8):506–14. doi: 10.1038/ 73. Wang Y, Chen S, Du K, Liang C, Wang S, Owusu Boadi E, et al. Traditional
nrgastro.2014.66 herbal medicine: Therapeutic potential in rheumatoid arthritis. J Ethnopharmacol
54. Ferro M, Charneca S, Dourado E, Guerreiro CS, Fonseca JE. Probiotic (2021) 279:114368. doi: 10.1016/j.jep.2021.114368
supplementation for rheumatoid arthritis: A promising adjuvant therapy in the gut 74. Mei L, Yang Z, Zhang X, Liu Z, Wang M, Wu X, et al. Sustained drug
microbiome era. Front Pharmacol (2021) 12:711788. doi: 10.3389/ treatment alters the gut microbiota in rheumatoid arthritis. Front Immunol (2021)
fphar.2021.711788 12:704089. doi: 10.3389/fimmu.2021.704089
55. So JS, Kwon HK, Lee CG, Yi HJ, Park JA, Lim SY, et al. Lactobacillus casei 75. Peng J, Lu X, Xie K, Xu Y, He R, Guo L, et al. Dynamic alterations in the gut
suppresses experimental arthritis by down-regulating T helper 1 effector functions. microbiota of collagen-induced arthritis rats following the prolonged
Mol Immunol (2008) 45(9):2690–9. doi: 10.1016/j.molimm.2007.12.010 administration of total glucosides of paeony. Front Cell Infect Microbiol (2019)
56. Bungau SG, Behl T, Singh A, Sehgal A, Singh S, Chigurupati S, et al. 9:204. doi: 10.3389/fcimb.2019.00204
Targeting probiotics in rheumatoid arthritis. Nutrients (2021) 13(10):3376. 76. Shan J, Peng L, Qian W, Xie T, Kang A, Gao B, et al. Integrated serum and
doi: 10.3390/nu13103376 fecal metabolomics study of collagen-induced arthritis rats and the therapeutic
57. Paul AK, Paul A, Jahan R, Jannat K, Bondhon TA, Hasan A, et al. Probiotics effects of the zushima tablet. Front Pharmacol (2018) 9:891. doi: 10.3389/
and amelioration of rheumatoid arthritis: Significant roles of lactobacillus casei and fphar.2018.00891
lactobacillus acidophilus. Microorganisms (2021) 9(5):1070. doi: 10.3390/ 77. Huang Y, Li M, Zhou L, Xu D, Qian F, Zhang J, et al. Effects of qingluo
microorganisms9051070 tongbi decoction on gut flora of rats with adjuvant-induced arthritis and the
58. Pan H, Guo R, Ju Y, Wang Q, Zhu J, Xie Y, et al. A single bacterium underlying mechanism. Evid Based Complement Alternat Med (2019)
restores the microbiome dysbiosis to protect bones from destruction in a rat model 2019:6308021. doi: 10.1155/2019/6308021
of rheumatoid arthritis. Microbiome (2019) 7(1):107. doi: 10.1186/s40168-019- 78. Alwarith J, Kahleova H, Rembert E, Yonas W, Dort S, Calcagno M, et al.
0719-1 Nutrition interventions in rheumatoid arthritis: The potential use of plant-based
59. Hatakka K, Martio J, Korpela M, Herranen M, Poussa T, Laasanen T, et al. diets. A Review Front Nutr (2019) 6:141. doi: 10.3389/fnut.2019.00141
Effects of probiotic therapy on the activity and activation of mild rheumatoid 79. DeChristopher LR, Uribarri J, Tucker KL. Intake of high-fructose corn syrup
arthritis–a pilot study. Scand J Rheumatol (2003) 32(4):211–5. doi: 10.1080/ sweetened soft drinks, fruit drinks and apple juice is associated with prevalent
03009740310003695 arthritis in US adults, aged 20-30 years. Nutr Diabetes (2016) 6(3):e199.
60. Pineda Mde L, Thompson SF, Summers K, de Leon F, Pope J, Reid G. A doi: 10.1038/nutd.2016.7
randomized, double-blinded, placebo-controlled pilot study of probiotics in active 80. Wagnerberger S, Spruss A, Kanuri G, Volynets V, Stahl C, Bischoff SC, et al.
rheumatoid arthritis. Med Sci Monit (2011) 17(6):Cr347–54. doi: 10.12659/ Toll-like receptors 1-9 are elevated in livers with fructose-induced hepatic steatosis.
msm.881808 Br J Nutr (2012) 107(12):1727–38. doi: 10.1017/s0007114511004983
61. Antushevich H. Fecal microbiota transplantation in disease therapy. Clin 81. Costantini L, Molinari R, Farinon B, Merendino N. Impact of omega-3 fatty
Chim Acta (2020) 503:90–8. doi: 10.1016/j.cca.2019.12.010 acids on the gut microbiota. Int J Mol Sci (2017) 18(12):2645. doi: 10.3390/
62. Zeng J, Peng L, Zheng W, Huang F, Zhang N, Wu D, et al. Fecal microbiota ijms18122645
transplantation for rheumatoid arthritis: A case report. Clin Case Rep (2021) 9 82. Gioxari A, Kaliora AC, Marantidou F, Panagiotakos DP. Intake of w-3
(2):906–9. doi: 10.1002/ccr3.3677 polyunsaturated fatty acids in patients with rheumatoid arthritis: A systematic
63. Yan H, Su R, Xue H, Gao C, Li X, Wang C. Pharmacomicrobiology of review and meta-analysis. Nutrition (2018) 45:114–24.e4. doi: 10.1016/
methotrexate in rheumatoid arthritis: Gut microbiome as predictor of j.nut.2017.06.023
therapeutic response. Front Immunol (2021) 12:789334. doi: 10.3389/ 83. Di Giuseppe D, Wallin A, Bottai M, Askling J, Wolk A. Long-term intake of
fimmu.2021.789334 dietary long-chain n-3 polyunsaturated fatty acids and risk of rheumatoid arthritis:
64. Zaharko DS, Bruckner H, Oliverio VT. Antibiotics alter methotrexate A prospective cohort study of women. Ann Rheum Dis (2014) 73(11):1949–53.
metabolism and excretion. Science (1969) 166(3907):887–8. doi: 10.1126/ doi: 10.1136/annrheumdis-2013-203338
science.166.3907.887 84. Minamino H, Katsushima M, Hashimoto M, Fujita Y, Yoshida T, Ikeda K,
65. Rodrigues GSP, Cayres LCF, Gonçalves FP, Takaoka NNC, Lengert AH, et al. Urinary sodium-to-potassium ratio associates with hypertension and current
Tansini A, et al. Detection of increased relative expression units of bacteroides and disease activity in patients with rheumatoid arthritis: A cross-sectional study.
prevotella, and decreased clostridium leptum in stool samples from Brazilian Arthritis Res Ther (2021) 23(1):96. doi: 10.1186/s13075-021-02479-x
rheumatoid arthritis patients: A pilot study. Microorganisms (2019) 7(10):413. 85. Wang C, Huang Z, Yu K, Ding R, Ye K, Dai C, et al. High-salt diet has a
doi: 10.3390/microorganisms7100413 certain impact on protein digestion and gut microbiota: A sequencing and
66. Krook A. Effect of metronidazole and sulfasalazine on the normal human proteome combined study. Front Microbiol (2017) 8:1838. doi: 10.3389/
faecal flora. Scand J Gastroenterol (1981) 16(5):587–92. doi: 10.3109/ fmicb.2017.01838
00365528109182016 86. Yang YH, Istomine R, Alvarez F, Al-Aubodah TA, Shi XQ, Takano T, et al.
67. Peppercorn MA, Goldman P. The role of intestinal bacteria in the Salt sensing by Serum/Glucocorticoid-regulated kinase 1 promotes Th17-like
metabolism of salicylazosulfapyridine. J Pharmacol Exp Ther (1972) 181(3):555–62. inflammatory adaptation of Foxp3(+) regulatory T cells. Cell Rep (2020) 30
(5):1515–29.e4. doi: 10.1016/j.celrep.2020.01.002
68. Pullar T, Hunter JA, Capell HA. Which component of sulphasalazine is
active in rheumatoid arthritis? Br Med J (Clin Res Ed) (1985) 290(6481):1535–8. 87. Khanna S, Jaiswal KS, Gupta B. Managing rheumatoid arthritis with dietary
doi: 10.1136/bmj.290.6481.1535 interventions. Front Nutr (2017) 4:52. doi: 10.3389/fnut.2017.00052