Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Ann Rheum Dis ; 80(5): 573-581, 2021 05.
Article in English | MEDLINE | ID: mdl-33397732

ABSTRACT

OBJECTIVES: Analysis of oral dysbiosis in individuals sharing genetic and environmental risk factors with rheumatoid arthritis (RA) patients may illuminate how microbiota contribute to disease susceptibility. We studied the oral microbiota in a prospective cohort of patients with RA, first-degree relatives (FDR) and healthy controls (HC), then genomically and functionally characterised streptococcal species from each group to understand their potential contribution to RA development. METHODS: After DNA extraction from tongue swabs, targeted 16S rRNA gene sequencing and statistical analysis, we defined a microbial dysbiosis score based on an operational taxonomic unit signature of disease. After selective culture from swabs, we identified streptococci by sequencing. We examined the ability of streptococcal cell walls (SCW) from isolates to induce cytokines from splenocytes and arthritis in ZAP-70-mutant SKG mice. RESULTS: RA and FDR were more likely to have periodontitis symptoms. An oral microbial dysbiosis score discriminated RA and HC subjects and predicted similarity of FDR to RA. Streptococcaceae were major contributors to the score. We identified 10 out of 15 streptococcal isolates as S. parasalivarius sp. nov., a distinct sister species to S. salivarius. Tumour necrosis factor and interleukin 6 production in vitro differed in response to individual S. parasalivarius isolates, suggesting strain specific effects on innate immunity. Cytokine secretion was associated with the presence of proteins potentially involved in S. parasalivarius SCW synthesis. Systemic administration of SCW from RA and HC-associated S. parasalivarius strains induced similar chronic arthritis. CONCLUSIONS: Dysbiosis-associated periodontal inflammation and barrier dysfunction may permit arthritogenic insoluble pro-inflammatory pathogen-associated molecules, like SCW, to reach synovial tissue.


Subject(s)
Arthritis, Rheumatoid/microbiology , Biopolymers/isolation & purification , Dysbiosis/microbiology , Peptidoglycan/isolation & purification , Periodontitis/microbiology , Streptococcus/isolation & purification , Adult , Animals , Disease Susceptibility/microbiology , Female , Humans , Male , Mice , Microbiota , Middle Aged , Mouth/microbiology , Pedigree , RNA, Ribosomal, 16S
2.
Immunol Cell Biol ; 97(1): 97-103, 2019 01.
Article in English | MEDLINE | ID: mdl-30191611

ABSTRACT

The microbial community making up the gut microbiota can profoundly influence intestinal homeostasis and immune system development, and is believed to influence the development of complex diseases including type 1 diabetes (T1D). T1D susceptible nonobese diabetic (NOD) mice have been shown to harbor a distinct microbiota to disease-protected mice. We hypothesized that the T1D susceptible genetic background of NOD mice would be resistant to the introduction of a C57BL/6-derived microbiota. NOD and C57BL/6 mice were cohoused either continually from birth, from birth until weaning or from weaning onwards, allowing transfer of microbiota between the mice. Cohousing NOD with C57BL/6 mice from before birth, resulted in moderate changes to the gut microbiota, whereas initiating cohousing at weaning only led to minimal changes. Terminating cohousing at weaning reduced the changes in the microbiota composition. However, diabetes onset was not significantly delayed and there was no reduction in intestinal inflammation or the proportion of regulatory T cells in the cohoused NOD mice. However, insulin but not islet-specific glucose-6-phosphatase catalytic subunit-related protein-specific CD8+ T cells were reduced by cohousing suggesting an epitope-specific modulation of the autoreactive response by the gut microbiota. These results suggest that the T1D susceptible genetic background of the NOD mouse was resistant to the introduction of a C57BL/6-derived microbiota.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Gastrointestinal Microbiome/immunology , Age Factors , Animals , Animals, Newborn , Diabetes Mellitus, Type 1/microbiology , Diabetes Mellitus, Type 1/prevention & control , Fecal Microbiota Transplantation , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , T-Lymphocytes, Regulatory/immunology
3.
Microbiome ; 6(1): 51, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29558994

ABSTRACT

Following publication of the original article [1] it came to the attention of the Production Editor that Figs. 1 and 2 had not been replaced with the newly revised figures supplied by the authors (the originals being unusable due to poor quality image and text).

4.
Microbiome ; 6(1): 35, 2018 02 17.
Article in English | MEDLINE | ID: mdl-29454391

ABSTRACT

BACKGROUND: Dysbiosis of the gut microbiota has been implicated in the pathogenesis of many autoimmune conditions including type 1 diabetes (T1D). It is unknown whether changes in the gut microbiota observed in T1D are due to environmental drivers, genetic risk factors, or both. Here, we have performed an analysis of associations between the gut microbiota and T1D genetic risk using the non-obese diabetic (NOD) mouse model of T1D and the TwinsUK cohort. RESULTS: Through the analysis of five separate colonies of T1D susceptible NOD mice, we identified similarities in NOD microbiome that were independent of animal facility. Introduction of disease protective alleles at the Idd3 and Idd5 loci (IL2, Ctla4, Slc11a1, and Acadl) resulted in significant alterations in the NOD microbiome. Disease-protected strains exhibited a restoration of immune regulatory pathways within the gut which could also be reestablished using IL-2 therapy. Increased T1D disease risk from IL-2 pathway loci in the TwinsUK cohort of human subjects resulted in some similar microbiota changes to those observed in the NOD mouse. CONCLUSIONS: These findings demonstrate for the first time that type 1 diabetes-associated genetic variants that restore immune tolerance to islet antigens also result in functional changes in the gut immune system and resultant changes in the microbiota.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Dysbiosis/pathology , Gastrointestinal Microbiome , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Interleukin-2/metabolism , Animals , CTLA-4 Antigen/genetics , Cation Transport Proteins/genetics , Clostridiales/isolation & purification , Diabetes Mellitus, Type 1/immunology , Dysbiosis/microbiology , Female , Genetic Predisposition to Disease/genetics , Humans , Interleukin-2/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Ruminococcus/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...