Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 739
Filtrer
1.
PeerJ ; 12: e17477, 2024.
Article de Anglais | MEDLINE | ID: mdl-39006008

RÉSUMÉ

Objective: The primary objective is to study the impact of gut microbiota and their interactions with diverse immunological markers on the development of rheumatoid arthritis. Methods: This study was performed in Astana, Kazakhstan, and included 77 Kazakh female patients older than 18 years, who met the American College of Rheumatology 2010 classification criteria for rheumatoid arthritis (RA), and 113 healthy controls. The DNA was extracted from fecal samples obtained from all study participants for subsequent sequencing at the 16S rRNA gene V1-V3 locus, facilitating the analysis of the gut microbiome. The Multiplex immunoassay was employed to measure the concentrations of inflammatory cytokines, chemokines, and immunoglobulins in both fecal and plasma samples. Results: Our taxonomic analysis revealed significant differences in the composition of the gut microbiota between the healthy control cohort and the cohort with rheumatoid arthritis RA. Alpha diversity was significantly lower in the RA group. Lachnospiraceae were the most abundant taxon and found to be crucial, showing correlations with immunological markers such as IL5. Additionally, Lachnospiraceae and Oscillospiraceae exhibited the most predictable power and distinguished the composition of both study groups. Conclusion: Our study identifies key differences in the gut microbiome of RA patients, revealing distinct microbial patterns and specific taxa abundance. We highlight potential biomarkers in immunological and bacterial pathways, offering insights into RA development and indicating possibilities for personalized treatment.


Sujet(s)
Polyarthrite rhumatoïde , Fèces , Microbiome gastro-intestinal , ARN ribosomique 16S , Humains , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/microbiologie , Microbiome gastro-intestinal/immunologie , Femelle , Adulte d'âge moyen , Adulte , Fèces/microbiologie , ARN ribosomique 16S/génétique , Études cas-témoins , Kazakhstan , Marqueurs biologiques/sang , Cytokines/métabolisme , Cytokines/génétique , Cytokines/immunologie , Cytokines/sang
2.
J Med Virol ; 96(7): e29781, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38961767

RÉSUMÉ

Rheumatoid arthritis-associated interstitial lung disease (RA-ILD) is a serious and common extra-articular disease manifestation. Patients with RA-ILD experience reduced bacterial diversity and gut bacteriome alterations. However, the gut mycobiome and virome in these patients have been largely neglected. In this study, we performed whole-metagenome shotgun sequencing on fecal samples from 30 patients with RA-ILD, and 30 with RA-non-ILD, and 40 matched healthy controls. The gut bacteriome and mycobiome were explored using a reference-based approach, while the gut virome was profiled based on a nonredundant viral operational taxonomic unit (vOTU) catalog. The results revealed significant alterations in the gut microbiomes of both RA-ILD and RA-non-ILD groups compared with healthy controls. These alterations encompassed changes in the relative abundances of 351 bacterial species, 65 fungal species, and 4,367 vOTUs. Bacteria such as Bifidobacterium longum, Dorea formicigenerans, and Collinsella aerofaciens were enriched in both patient groups. Ruminococcus gnavus (RA-ILD), Gemmiger formicilis, and Ruminococcus bromii (RA-non-ILD) were uniquely enriched. Conversely, Faecalibacterium prausnitzii, Bacteroides spp., and Roseburia inulinivorans showed depletion in both patient groups. Mycobiome analysis revealed depletion of certain fungi, including Saccharomyces cerevisiae and Candida albicans, in patients with RA compared with healthy subjects. Notably, gut virome alterations were characterized by an increase in Siphoviridae and a decrease in Myoviridae, Microviridae, and Autographiviridae in both patient groups. Hence, multikingdom gut microbial signatures showed promise as diagnostic indicators for both RA-ILD and RA-non-ILD. Overall, this study provides comprehensive insights into the fecal virome, bacteriome, and mycobiome landscapes of RA-ILD and RA-non-ILD gut microbiota, thereby offering potential biomarkers for further mechanistic and clinical research.


Sujet(s)
Polyarthrite rhumatoïde , Bactéries , Fèces , Microbiome gastro-intestinal , Pneumopathies interstitielles , Humains , Pneumopathies interstitielles/microbiologie , Pneumopathies interstitielles/virologie , Polyarthrite rhumatoïde/complications , Polyarthrite rhumatoïde/microbiologie , Fèces/microbiologie , Fèces/virologie , Femelle , Mâle , Adulte d'âge moyen , Bactéries/classification , Bactéries/isolement et purification , Bactéries/génétique , Sujet âgé , Virome , Mycobiome , Adulte , Virus/classification , Virus/isolement et purification , Virus/génétique , Champignons/isolement et purification , Champignons/classification
3.
Clin Exp Rheumatol ; 42(7): 1343-1349, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38829018

RÉSUMÉ

The present narrative review explores the multifactorial aetiology of rheumatoid arthritis (RA) and other immunemediated inflammatory disorders (IMIDs), emphasising the significant role of various environmental factors in disease development and exacerbation. Key modifiable environmental factors such as cigarette smoking and air pollution are identified as major contributors to RA. We will also focus on the influence of weather, seasonality, and particularly vitamin D levels, on RA activity, suggesting potential for seasonal management and supplementation to mitigate disease severity. The emerging role of diet and the gut microbiome in RA pathogenesis and progression is discussed as well, with dietary interventions and specific nutrients like omega-3 fatty acids offering protective benefits against inflammation. Despite the mounting evidence around these factors, further research is needed, to better understand the clinical impacts on RA, including well-designed randomised clinical trials.


Sujet(s)
Polyarthrite rhumatoïde , Microbiome gastro-intestinal , Humains , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/microbiologie , Facteurs de risque , Saisons , Temps (météorologie) , Exposition environnementale/effets indésirables , Régime alimentaire/effets indésirables , Pollution de l'air/effets indésirables , Vitamine D/usage thérapeutique , Vitamine D/sang , Compléments alimentaires
4.
Int Immunopharmacol ; 137: 112501, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38885604

RÉSUMÉ

Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by inflammation and joint damage. Existing treatment options primarily focus on managing symptoms and slowing disease progression, often with side effects and limitations. The gut microbiome, a vast community of microorganisms present in the gastrointestinal tract, plays a crucial role in health and disease. Recent research suggests a bidirectional relationship between the gut microbiome and RA, highlighting its potential as a therapeutic option. This review focuses on the interaction between the gut microbiome and RA development, by discussing how dysbiosis, an imbalance in gut bacteria, can contribute to RA through multiple mechanisms such as molecular mimicry, leaky gut, and metabolic dysregulation. Probiotics, live microorganisms with health benefits, are emerging as promising tools for managing RA. They can prevent the negative effects of dysbiosis by displacing harmful bacteria, producing anti-inflammatory metabolites like short-chain fatty acids (SCFA), Directly influencing immune cells, and modifying host metabolism. animal and clinical studies demonstrate the potential of probiotics in improving RA symptoms and disease outcomes. However, further research is needed to optimize probiotic strains, dosages, and treatment protocols for personalized and effective management of RA. This review summarizes the current understanding of the gut microbiome and its role in RA and discusses future research directions. In addition to the established role of gut dysbiosis in RA, emerging strategies like fecal microbiota transplantation, prebiotics, and postbiotics offer exciting possibilities. However, individual variations in gut composition necessitate personalized treatment plans. Long-term effects and clear regulations need to be established. Future research focusing on metagenomic analysis, combination therapies, and mechanistic understanding will unlock the full potential of gut microbiome modulation for effective RA management.


Sujet(s)
Polyarthrite rhumatoïde , Dysbiose , Microbiome gastro-intestinal , Homéostasie , Probiotiques , Humains , Polyarthrite rhumatoïde/thérapie , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/microbiologie , Polyarthrite rhumatoïde/traitement médicamenteux , Probiotiques/usage thérapeutique , Animaux , Dysbiose/thérapie , Transplantation de microbiote fécal
5.
Bone Res ; 12(1): 31, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38782893

RÉSUMÉ

Rheumatoid arthritis (RA) is an autoimmune disease. Early studies hold an opinion that gut microbiota is environmentally acquired and associated with RA susceptibility. However, accumulating evidence demonstrates that genetics also shape the gut microbiota. It is known that some strains of inbred laboratory mice are highly susceptible to collagen-induced arthritis (CIA), while the others are resistant to CIA. Here, we show that transplantation of fecal microbiota of CIA-resistant C57BL/6J mice to CIA-susceptible DBA/1J mice confer CIA resistance in DBA/1J mice. C57BL/6J mice and healthy human individuals have enriched B. fragilis than DBA/1J mice and RA patients. Transplantation of B. fragilis prevents CIA in DBA/1J mice. We identify that B. fragilis mainly produces propionate and C57BL/6J mice and healthy human individuals have higher level of propionate. Fibroblast-like synoviocytes (FLSs) in RA are activated to undergo tumor-like transformation. Propionate disrupts HDAC3-FOXK1 interaction to increase acetylation of FOXK1, resulting in reduced FOXK1 stability, blocked interferon signaling and deactivation of RA-FLSs. We treat CIA mice with propionate and show that propionate attenuates CIA. Moreover, a combination of propionate with anti-TNF etanercept synergistically relieves CIA. These results suggest that B. fragilis or propionate could be an alternative or complementary approach to the current therapies.


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Microbiome gastro-intestinal , Histone deacetylases , Souris de lignée C57BL , Cellules synoviales , Animaux , Humains , Mâle , Souris , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/métabolisme , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/microbiologie , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Facteurs de transcription Forkhead/métabolisme , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Histone deacetylases/métabolisme , Souris de lignée DBA , Transduction du signal/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/anatomopathologie
6.
Int J Mol Sci ; 25(10)2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38791230

RÉSUMÉ

The human microbiome exists throughout the body, and it is essential for maintaining various physiological processes, including immunity, and dysbiotic events, which are associated with autoimmunity. Peptidylarginine deiminase (PAD) enzymes can citrullinate self-proteins related to rheumatoid arthritis (RA) that induce the production of anti-citrullinated protein antibodies (ACPAs) and lead to inflammation and joint damage. The present investigation was carried out to demonstrate the expression of homologs of PADs or arginine deiminases (ADs) and citrullinated proteins in members of the human microbiota. To achieve the objective, we used 17 microbial strains and specific polyclonal antibodies (pAbs) of the synthetic peptide derived from residues 100-200 of human PAD2 (anti-PAD2 pAb), and the recombinant fragment of amino acids 326 and 611 of human PAD4 (anti-PAD4 pAb), a human anti-citrulline pAb, and affinity ACPAs of an RA patient. Western blot (WB), enzyme-linked immunosorbent assay (ELISA), elution, and a test with Griess reagent were used. This is a cross-sectional case-control study on patients diagnosed with RA and control subjects. Inferential statistics were applied using the non-parametric Kruskal-Wallis test and Mann-Whitney U test generated in the SPSS program. Some members of phyla Firmicutes and Proteobacteria harbor homologs of PADs/ADs and citrullinated antigens that are reactive to the ACPAs of RA patients. Microbial citrullinome and homolog enzymes of PADs/ADs are extensive in the human microbiome and are involved in the production of ACPAs. Our findings suggest a molecular link between microorganisms of a dysbiotic microbiota and RA pathogenesis.


Sujet(s)
Anticorps anti-protéines citrullinées , Polyarthrite rhumatoïde , Citrullination , Microbiote , Protein-arginine deiminases , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Anticorps anti-protéines citrullinées/immunologie , Anticorps anti-protéines citrullinées/métabolisme , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/microbiologie , Études cas-témoins , Citrulline/métabolisme , Études transversales , Hydrolases/métabolisme , Protein-arginine deiminase Type 2/métabolisme , Protein-arginine deiminase Type 4/métabolisme , Protein-arginine deiminases/métabolisme , Protein-arginine deiminases/génétique
7.
Biosci Trends ; 18(2): 187-194, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38599880

RÉSUMÉ

Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic and symmetric in-flammation. Our previous research revealed an imbalance in the gut flora of RA patients and showed that certain gut microbiota can accelerate RA progression by enhancing vitamin C degradation. However, it is unclear whether vitamin C supplementation could improve the gut microbiota to prevent the development of arthritis by interfering with the gut-joint axis. In this work, we aimed to evaluate the effects of vitamin C in regulating the gut microbiota and to elucidate its potential role in the onset and progression of RA in a mouse model, thus providing a basis for the development of new intervention strategies and treatments for RA. In this study, collagen-induced arthritis (CIA) mouse models, biochemical, histological and 16S rRNA microbiological methods were used to investigate the role and possible mechanism of vitamin C in rheumatoid arthritis. The results showed that treatment of CIA mice with vitamin C effectively rescued the gut mi-crobiota imbalance and suppressed the inflammatory response associated with RA, and effectively alleviated arthritis symptoms in mice in which levels of the pro-inflammatory cytokines IL-6 and TNF-α were specifi-cally reduced. In conclusion, our results demonstrate the potential of vitamin C as a potential therapeutic choice for RA.


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Acide ascorbique , Microbiome gastro-intestinal , Animaux , Acide ascorbique/usage thérapeutique , Acide ascorbique/administration et posologie , Acide ascorbique/pharmacologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/microbiologie , Souris , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/microbiologie , Arthrite expérimentale/immunologie , Mâle , Souris de lignée DBA , Facteur de nécrose tumorale alpha/métabolisme , Interleukine-6/métabolisme , Modèles animaux de maladie humaine , ARN ribosomique 16S/génétique
8.
Comput Biol Chem ; 110: 108076, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38678728

RÉSUMÉ

Clinical studies have already illustrated the associations between gut microbes and diseases, yet fundamental questions remain unclear that how we can universalize this knowledge. Considering the important role of human gut microbial composition in maintaining overall health, it is important to understand the microbial diversity and altered disease conditions of the human gut. Metagenomics provides a way to analyze and understand the microbes and their role in a community manner. It provides qualitative as well as quantitative measurements, in terms of relative abundance. Various studies are already going on to find out the association between microbes and diseases; still, the mined knowledge is limited. Considering the current scenario, using the targeted metagenomics approach, we analyzed the gut microbiome of 99 samples from healthy and diseased individuals. Our metagenomic analysis mainly targeted five diseased microbiomes (i.e., Age-related macular degeneration, Autism spectrum disorder, Rheumatoid arthritis, Type 2 diabetes and Vogt-Koyanagi harada), with compare to healthy microbiome, and reported disease-associated microbiome shift in different conditions.


Sujet(s)
Polyarthrite rhumatoïde , Trouble du spectre autistique , Diabète de type 2 , Microbiome gastro-intestinal , Dégénérescence maculaire , Humains , Microbiome gastro-intestinal/génétique , Polyarthrite rhumatoïde/microbiologie , Trouble du spectre autistique/microbiologie , Diabète de type 2/microbiologie , Dégénérescence maculaire/microbiologie , Dégénérescence maculaire/génétique , Métagénome , Métagénomique
9.
Microb Pathog ; 191: 106644, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38616001

RÉSUMÉ

OBJECTIVE: To assess and compare the composition of tongue coating microbiota among patients at different stages of rheumatoid arthritis (RA). METHODS: A total of 47 patients diagnosed with RA, as per the American College of Rheumatology criteria, and 10 healthy individuals were enrolled in this study. The RA patients were stratified considering their Disease Activity Score 28 (DAS28), a composite measure based on the 28 tender and swollen joint count and erythrocyte sedimentation rate (ESR). The study population was further categorized into active phase group (LMH group) and inactive phase group (RE group) according to their DAS28 values. DNA extraction was extracted from tongue coating samples. Subsequently, the V3-V4 16S rDNA region was selectively amplified and sequenced through high-throughput 16S rDNA analysis. The resulting data were then utilized to ascertain the microbial contents. RESULTS: Significant variations were observed in the tongue coating microbiota of patients with RA during active and inactive phases, in comparison to healthy individuals (p < 0.05). At the genus level, the presence of Prevotellan, Veillonella, Rothia, and Neisseria in RA patients was notably more evident than in the healthy control (HC) group. These disparities find support in existing research on gut and oral microbiota. During the active phase of RA, the relative abundance of Veillonella, Rothia, and Neisseria in the tongue coating microbiota of patients was significantly higher than in those with inactive RA. These findings underscore the need for further and in-depth research on the potential impact of these microorganisms on the progression of RA disease. CONCLUSION: The results substantiate the hypothesis that tongue coating microbes actively contribute to the progression of RA.


Sujet(s)
Polyarthrite rhumatoïde , Bactéries , Évolution de la maladie , Microbiote , ARN ribosomique 16S , Langue , Humains , Polyarthrite rhumatoïde/microbiologie , Langue/microbiologie , Langue/anatomopathologie , Femelle , Mâle , Adulte d'âge moyen , ARN ribosomique 16S/génétique , Adulte , Microbiote/génétique , Bactéries/classification , Bactéries/génétique , Bactéries/isolement et purification , ADN bactérien/génétique , Sujet âgé , Indice de gravité de la maladie
10.
Biomed Pharmacother ; 174: 116515, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38569276

RÉSUMÉ

Mesenchymal stem cell exosome (MSCs-exo) is a class of products secreted by mesenchymal stem cells (MSCs) that contain various biologically active substances. MSCs-exo is a promising alternative to MSCs due to their lower immunogenicity and lack of ethical constraints. Ginsenoside Rh2 (Rh2) is a hydrolyzed component of the primary active substance of ginsenosides. Rh2 has a variety of pharmacological functions, including anti-inflammatory, anti-tumor, and antioxidant. Studies have demonstrated that gut microbiota and metabolites are critical in developing rheumatoid arthritis (RA). In this study, we constructed a collagen-induced arthritis (CIA) model in rats. We used MSCs-exo combined with Rh2 to treat CIA rats. To observe the effect of MSCs-exo combined with Rh2 on joint inflammation, rat feces were collected for 16 rRNA amplicon sequencing and untargeted metabolomics analysis. The results showed that the arthritis index score and joint swelling of CIA rats treated with MSCs-exo in combination with Rh2 were significantly lower than those of the model and MSCs-exo alone groups. MSCs-exo and Rh2 significantly ameliorated the disturbed gut microbiota in CIA rats. The regulation of Candidatus_Saccharibacteria and Clostridium_XlVb regulation may be the most critical. Rh2 enhanced the therapeutic effect of MSCs-exo compared with the MSCs-exo -alone group. Furthermore, significant changes in gut metabolites were observed in the CIA rat group, and these differentially altered metabolites may act as messengers for host-microbiota interactions. These differential metabolites were enriched into relevant critical metabolic pathways, revealing possible pathways for host-microbiota interactions.


Sujet(s)
Arthrite expérimentale , Microbiome gastro-intestinal , Ginsénosides , Cellules souches mésenchymateuses , Animaux , Humains , Mâle , Rats , Arthrite expérimentale/induit chimiquement , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/microbiologie , Arthrite expérimentale/thérapie , Polyarthrite rhumatoïde/induit chimiquement , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/microbiologie , Polyarthrite rhumatoïde/thérapie , Exosomes/métabolisme , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Ginsénosides/pharmacologie , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cordon ombilical , Collagène/métabolisme , Collagène/pharmacologie
11.
APMIS ; 132(6): 382-415, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38469726

RÉSUMÉ

Rheumatoid arthritis (RA) is a multifaceted autoimmune disorder characterized by chronic inflammation and joint destruction. Recent research has elucidated the intricate interplay between gut microbiota and RA pathogenesis, underscoring the role of microbiota-derived metabolites as pivotal contributors to disease development and progression. The human gut microbiota, comprising a vast array of microorganisms and their metabolic byproducts, plays a crucial role in maintaining immune homeostasis. Dysbiosis of this microbial community has been linked to numerous autoimmune disorders, including RA. Microbiota-derived metabolites, such as short-chain fatty acids (SCFAs), tryptophan derivatives, Trimethylamine-N-oxide (TMAO), bile acids, peptidoglycan, and lipopolysaccharide (LPS), exhibit immunomodulatory properties that can either exacerbate or ameliorate inflammation in RA. Mechanistically, these metabolites influence immune cell differentiation, cytokine production, and gut barrier integrity, collectively shaping the autoimmune milieu. This review highlights recent advances in understanding the intricate crosstalk between microbiota metabolites and RA pathogenesis and also discusses the potential of specific metabolites to trigger or suppress autoimmunity, shedding light on their molecular interactions with immune cells and signaling pathways. Additionally, this review explores the translational aspects of microbiota metabolites as diagnostic and prognostic tools in RA. Furthermore, the challenges and prospects of translating these findings into clinical practice are critically examined.


Sujet(s)
Polyarthrite rhumatoïde , Marqueurs biologiques , Dysbiose , Microbiome gastro-intestinal , Humains , Polyarthrite rhumatoïde/microbiologie , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/métabolisme , Marqueurs biologiques/métabolisme , Dysbiose/microbiologie , Animaux , Acides gras volatils/métabolisme
12.
Int J Mol Sci ; 25(6)2024 Mar 13.
Article de Anglais | MEDLINE | ID: mdl-38542216

RÉSUMÉ

Dysregulation of the gut microbiota and their metabolites is involved in the pathogenic process of intestinal diseases, and several pieces of evidence within the current literature have also highlighted a possible connection between the gut microbiota and the unfolding of inflammatory pathologies of the joints. This dysregulation is defined as the "gut-joint axis" and is based on the joint-gut interaction. It is widely recognized that the microbiota of the gut produce a variety of compounds, including enzymes, short-chain fatty acids, and metabolites. As a consequence, these proinflammatory compounds that bacteria produce, such as that of lipopolysaccharide, move from the "leaky gut" to the bloodstream, thereby leading to systemic inflammation which then reaches the joints, with consequences such as osteoarthritis, rheumatoid arthritis, and spondylarthritis. In this state-of-the-art research, the authors describe the connections between gut dysbiosis and osteoarthritis, rheumatoid arthritis, and spondylarthritis. Moreover, the diagnostic tools, outcome measures, and treatment options are elucidated. There is accumulating proof suggesting that the microbiota of the gut play an important part not only in immune-mediated, metabolic, and neurological illnesses but also in inflammatory joints. According to the authors, future studies should concentrate on developing innovative microbiota-targeted treatments and their effects on joint pathology as well as on organizing screening protocols to predict the onset of inflammatory joint disease based on gut dysbiosis.


Sujet(s)
Polyarthrite rhumatoïde , Microbiome gastro-intestinal , Arthrose , Spondylarthrite , Humains , Microbiome gastro-intestinal/physiologie , Dysbiose/microbiologie , Polyarthrite rhumatoïde/microbiologie
13.
Intern Emerg Med ; 19(4): 891-900, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38141117

RÉSUMÉ

A growing amount of evidence suggests that gut microbiota plays an important role in human health, including a possible role in the pathogenesis of rheumatic and musculoskeletal diseases (RMD). We analysed the current evidence about the role of microbiota in rheumatoid arthritis (RA), spondyloarthritis (SpA), systemic lupus erythematosus (SLE) and systemic sclerosis (SSc). In RA, we found a general consensus regarding a reduction of diversity and a specific bacterial signature, with consistent changes according to the different ethnic and geographical areas. The major pathogenetic role in RA is recognised for P. copri, L. salivarius and Collinsella, even if findings become more heterogeneous when considering established disease. In SpA, we found a relative gut abundance of Akkermansia, Coprococcus, Ruminoccocus and a relative reduction in Bacterioides and Firmicutes spp. Human and preclinical data suggest loss of mucosal barrier, increased permeability and Th1- and Th17-mediated inflammation. Additionally, HLA-B27 seems to play a role in shaping the intestinal microbiota and the consequent inflammation. In SLE, the typical gut microbiota signature was characterised by a reduction in the Firmicutes/Bacteroidetes ratio and by enrichment of Rhodococcus, Eggerthella, Klebsiella, Prevotella, Eubacterium and Flavonifractor, even if their real pathogenic impact remains unclear. In SSc, gastrointestinal dysbiosis is well documented with an increase of pro-inflammatory species (Fusobacterium, Prevotella, Ruminococcus, Akkermansia, γ-Proteobacteria, Erwinia, Trabsulsiella, Bifidobacterium, Lactobacillus, Firmicutes and Actinobacteria) and a reduction of species as Faecalibacterium, Clostridium, Bacteroidetes and Rikenella. In conclusion, seems possible to recognise a distinct gut microbiota profile for each RMD, even if significant differences in bacterial species do exist between different studies and there is a high risk of bias due to the cross-sectional nature of such studies. Therefore longitudinal studies are needed, especially on patients with preclinical and early disease, to investigate the real role of gut microbiota in the pathogenesis of RMD.


Sujet(s)
Microbiome gastro-intestinal , Rhumatismes , Humains , Microbiome gastro-intestinal/physiologie , Rhumatismes/microbiologie , Polyarthrite rhumatoïde/microbiologie
15.
Front Cell Infect Microbiol ; 12: 956417, 2022.
Article de Anglais | MEDLINE | ID: mdl-35923803

RÉSUMÉ

Rheumatoid arthritis (RA) is a systematical autoimmune disease, characterized by chronic synovial joint inflammation and hurt. Porphyromonas gingivalis(P. gingivalis) can cause life-threatening inflammatory immune responses in humans when the host pathogenic clearance machinery is disordered. Some epidemiological studies have reported that P. gingivalis exposure would increase the prevalence of RA. However, the results remain inconsistent. Therefore, a meta-analysis was done to systematically analyze the relationship between P. gingivalis exposure and the prevalence of rheumatoid arthritis. Database including Cochrane Library, Web of Science, PubMed, and EMBASE were searched for published epidemiological articles assessed the relationship between P. gingivalis and RA. Obtained studies were screened based on the predefined inclusion and exclusion criteria. The overall Odds Ratios (ORs) of incorporated articles were pooled by random-effect model with STATA 15.1 software. The literature search returned a total of 2057 studies. After exclusion, 28 articles were included and analyzed. The pooled ORs showed a significant increase in the risk of RA in individuals with P. gingivalis exposure (OR = 1.86; 95% CI: 1.43-2.43). Subgroup analysis revealed that pooled ORs from populations located in Europe (OR = 2.17; 95% CI: 1.46-3.22) and North America (OR = 2.50; 95% CI: 1.23-5.08) were significantly higher than that from population in Asia (OR = 1.11; 95% CI: 1.03-1.20). Substantial heterogeneity was observed but did not significantly influence the overall outcome. In conclusion, our results indicated P. gingivalis exposure was a risk factor in RA. Prompt diagnosis and management decisions on P. gingivalis antimicrobial therapy would prevent rheumatoid arthritis development and progression.


Sujet(s)
Polyarthrite rhumatoïde , Porphyromonas gingivalis , Polyarthrite rhumatoïde/épidémiologie , Polyarthrite rhumatoïde/microbiologie , Humains , Prévalence
16.
J Immunol Res ; 2022: 6839356, 2022.
Article de Anglais | MEDLINE | ID: mdl-35224112

RÉSUMÉ

Intestinal bacterial compositions of rheumatoid arthritis (RA) patients have been reported to be different from those of healthy people. Dysbiosis, imbalance of the microbiota, is widely known to cause gut barrier damage, resulting in an influx of bacteria and their substances into host bloodstreams in animal studies. However, few studies have investigated the effect of bacterial substances on the pathophysiology of RA. In this study, eighty-seven active RA patients who had inadequate responses to conventional synthetic disease-modifying antirheumatic drugs or severe comorbidities were analyzed for correlations between many factors such as disease activities, disease biomarkers, intestinal bacterial counts, fecal and serum lipopolysaccharide (LPS), LPS-binding protein (LBP), endotoxin neutralizing capacity (ENC), and serum antibacterial substance IgG and IgA antibody levels by multiple regression analysis with consideration for demographic factors such as age, sex, smoking, and methotrexate treatment. Serum LBP levels, fecal LPS levels, total bacteria counts, serum anti-LPS from Porphyromonas gingivalis (Pg-LPS) IgG antibody levels, and serum anti-Pg-LPS IgA antibody levels were selected for multiple regression analysis using Spearman's correlation analysis. Serum LBP levels were correlated with disease biomarker levels, such as erythrocyte sedimentation rate (p < 0.001), C-reactive protein (p < 0.001), matrix metalloproteinase-3 (p < 0.001), and IL-6 (p = 0.001), and were inversely correlated with hemoglobin (p = 0.005). Anti-Pg-LPS IgG antibody levels were inversely correlated with activity indices such as patient global assessments using visual analogue scale (VAS) (p = 0.002) and painVAS (p < 0.001). Total bacteria counts were correlated with ENC (p < 0.001), and inversely correlated with serum LPS (p < 0.001) and anti-Pg-LPS IgA antibody levels (p < 0.001). These results suggest that substances from oral and gut microbiota may influence disease activity in RA patients.


Sujet(s)
Polyarthrite rhumatoïde/microbiologie , Infections à Bacteroidaceae/microbiologie , Dysbiose/microbiologie , Bouche/microbiologie , Porphyromonas gingivalis/physiologie , Protéine de la phase aigüe/métabolisme , Sujet âgé , Polyarthrite rhumatoïde/immunologie , Autoanticorps/sang , Charge bactérienne , Infections à Bacteroidaceae/immunologie , Marqueurs biologiques/métabolisme , Protéines de transport/métabolisme , Études transversales , Dysbiose/immunologie , Femelle , Microbiome gastro-intestinal , Humains , Immunoglobuline A/métabolisme , Lipopolysaccharides/métabolisme , Mâle , Glycoprotéines membranaires/métabolisme , Adulte d'âge moyen
17.
Toxins (Basel) ; 14(1)2022 01 11.
Article de Anglais | MEDLINE | ID: mdl-35051027

RÉSUMÉ

Leukotoxin A (LtxA) is the major virulence factor of an oral bacterium known as Aggregatibacter actinomycetemcomitans (Aa). LtxA is associated with elevated levels of anti-citrullinated protein antibodies (ACPA) in rheumatoid arthritis (RA) patients. LtxA targets leukocytes and triggers an influx of extracellular calcium into cytosol. The current proposed model of LtxA-mediated hypercitrullination involves the dysregulated activation of peptidylarginine deiminase (PAD) enzymes to citrullinate proteins, the release of hypercitrullinated proteins through cell death, and the production of autoantigens recognized by ACPA. Although model-based evidence is yet to be established, its interaction with the host's immune system sparked interest in the role of LtxA in RA. The first part of this review summarizes the current knowledge of Aa and LtxA. The next part highlights the findings of previous studies on the association of Aa or LtxA with RA aetiology. Finally, we discuss the unresolved aspects of the proposed link between LtxA of Aa and RA.


Sujet(s)
Aggregatibacter actinomycetemcomitans/physiologie , Polyarthrite rhumatoïde/microbiologie , Infections à Pasteurellaceae/microbiologie , Anticorps anti-protéines citrullinées/immunologie , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/anatomopathologie , Exotoxines/immunologie , Humains , Infections à Pasteurellaceae/immunologie , Infections à Pasteurellaceae/anatomopathologie
18.
Nutrients ; 14(2)2022 Jan 14.
Article de Anglais | MEDLINE | ID: mdl-35057535

RÉSUMÉ

BACKGROUND: We aimed to provide a systematic review and meta-analysis of randomized controlled trials assessing the effect of probiotics supplementation on symptoms and disease activity in patients with chronic inflammatory rheumatic diseases (rheumatoid arthritis (RA), spondylarthritis (SpA), or psoriatic arthritis). METHODS: A systematic literature review and meta-analysis from RA and SpA randomized controlled trials were conducted searching for articles in MEDLINE/PubMed and abstracts from recent international rheumatology meetings. The control group was a placebo or another dietary intervention. The risk of bias of the selected studies was evaluated using the Cochrane Collaboration tool and the Jadad scale. RESULTS: The initial search yielded 173 articles. Of these, 13 studies were included in the qualitative synthesis, 8 concerning a total of 344 RA patients and 2 concerning a total of 197 SpA patients. Three meta-analyses were also analyzed. Probiotic strains and quantities used were different among trials (5 studies using Lactobacillus sp., 1 trial Bacillus coagulans and the others a mix of different probiotic strains). Time to assess response ranged from 8 weeks to one year. Two studies associated probiotic supplementation with a dietary intervention. Meta-analysis showed a statistically significant decrease of C-reactive protein (CRP) concentration (mean difference (MD)) -3.04 (95% CI -4.47, -1.62) mg/L, p < 0.001; I2 = 20%, n patients = 209) with probiotics in RA. However, after excluding high-risk-of-bias trials of meta-analysis, there was no difference between probiotics and placebo on DAS28 (standard MD -0.54; 95% CI -1.94 to 0.85, p = 0.45, I2 93%, n patients = 143). The two studies on SpA patients showed no efficacy of probiotics. CONCLUSIONS: Probiotic supplementation might decrease RA activity with a moderate decrease effect on CRP, but lack of evidence and studies' heterogeneity do not allow us to propose them to patients with inflammatory arthritis to control their disease. Further RCTs are required in the future to determinate the efficacy of probiotics and the optimal administration design.


Sujet(s)
Polyarthrite rhumatoïde/microbiologie , Polyarthrite rhumatoïde/thérapie , Probiotiques/usage thérapeutique , Spondylarthrite/microbiologie , Spondylarthrite/thérapie , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Essais contrôlés randomisés comme sujet , Résultat thérapeutique
19.
Article de Anglais | MEDLINE | ID: mdl-34998201

RÉSUMÉ

Yaobitong capsule (YBTC), a Chinese medicine compound preparation, has been demonstrated to affect multiple pathways associated with inflammation and exhibit potential anti-arthritis effect. In this study, an integrated omic approach based on UHPLC-Q-TOF MS and 16S rRNA sequencing analyses was proposed to reveal the anti-arthritis effect and possible mechanism of YBTC. The AIA rat model showed that YBTC significantly alleviated the typical symptoms of AIA rats such as weight, spleen index and pro-inflammatory cytokines. Fecal metabolomics results identified 41 differential metabolites, which mainly referred to tryptophan, bile acid and fatty acid metabolism. The gut microbiota played a crucially important role in anti-inflammatory immunity, 16S rRNA results indicated that YBTC changed the community structure and alleviated the microecological imbalance caused by rheumatoid arthritis (RA). Further ROC curve analysis demonstrated that it was reliable to identify RA by using 5 metabolites and 3 microorganisms (AUC > 0.83). In summary, it was the first time that the preventive effect of YBTC in RA was confirmed. The secretion of the microbiota-mediated metabolites was significantly improved by YBTC, through its callback effect on the disturbed gut microbiota. Thus, we have indicated a potential novel strategy for the prevention of RA via evaluation of intervention effects of YBTC on AIA rat model.


Sujet(s)
Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/microbiologie , Médicaments issus de plantes chinoises/administration et posologie , Fèces/composition chimique , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Animaux , Polyarthrite rhumatoïde/métabolisme , Bactéries/classification , Bactéries/génétique , Bactéries/isolement et purification , Bactéries/métabolisme , Modèles animaux de maladie humaine , Fèces/microbiologie , Humains , Mâle , Métabolomique , Microbiote/effets des médicaments et des substances chimiques , Rats , Rat Sprague-Dawley
20.
Probiotics Antimicrob Proteins ; 14(1): 99-113, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34036479

RÉSUMÉ

Microbiota is a balanced ecosystem that has important functions to the host health including development, defense, digestion, and absorption of dietary fibers and minerals, vitamin synthesizes, protection, and training the host immune system. On the other hand, its dysbiosis is linked to many human diseases such as rheumatoid arthritis (RA). The RA is an inflammatory autoimmune disorder caused by genetic and environmental factors; microbiota may be considered as a risk environmental factor for it. Citrullination is a post-translation modification (PMT) that converts the amino acid arginine to amino acid citrulline in certain proteins. These citrullinated proteins are recognized as a foreign antigen by the immune system resulting in the upregulation of inflammatory action such as in RA. The current work highlights the effect of both gut and oral microbiota dysbiosis on the development of RA, as well as discusses how the alteration in microbiota composition leads to the overgrowth of some bacterial species that entangled in RA pathogenicity. The evidence suggested that some oral and gut microbial species such as Porphyromonas gingivalis and Prevotella copri, respectively, contribute to RA pathogenesis. During dysbiosis, these bacteria can mediate the citrullination of either human or bacteria proteins to trigger an immune response that leads to the generation of autoantibodies.


Sujet(s)
Polyarthrite rhumatoïde , Microbiote , Parodontite , Polyarthrite rhumatoïde/microbiologie , Citrullination , Humains , Parodontite/microbiologie , Porphyromonas gingivalis/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE