Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 455
Filter
1.
Microbiome ; 12(1): 157, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39182099

ABSTRACT

BACKGROUND: Diabetic cardiomyopathy (DCM) is one of leading causes of diabetes-associated mortality. The gut microbiota-derived branched-chain amino acids (BCAA) have been reported to play a central role in the onset and progression of DCM, but the potential mechanisms remain elusive. RESULTS: We found the type 1 diabetes (T1D) mice had higher circulating BCAA levels due to a reduced BCAA degradation ability of the gut microbiota. Excess BCAA decreased hepatic FGF21 production by inhibiting PPARα signaling pathway and thereby resulted in a higher expression level of cardiac LAT1 via transcription factor Zbtb7c. High cardiac LAT1 increased the levels of BCAA in the heart and then caused mitochondrial damage and myocardial apoptosis through mTOR signaling pathway, leading to cardiac fibrosis and dysfunction in T1D mice. Additionally, transplant of faecal microbiota from healthy mice alleviated cardiac dysfunction in T1D mice, but this effect was abolished by FGF21 knockdown. CONCLUSIONS: Our study sheds light on BCAA-mediated crosstalk among the gut microbiota, liver and heart to promote DCM and FGF21 serves as a key mediator. Video Abstract.


Subject(s)
Amino Acids, Branched-Chain , Diabetic Cardiomyopathies , Fibroblast Growth Factors , Gastrointestinal Microbiome , Liver , Animals , Fibroblast Growth Factors/metabolism , Mice , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/microbiology , Liver/metabolism , Amino Acids, Branched-Chain/metabolism , Signal Transduction , Diabetes Mellitus, Type 1/microbiology , Diabetes Mellitus, Type 1/metabolism , Male , Myocardium/metabolism , Myocardium/pathology , PPAR alpha/metabolism , Mice, Inbred C57BL , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/microbiology
2.
Int J Mol Sci ; 25(13)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-39000406

ABSTRACT

Diabetes mellitus (DM) poses a significant challenge to global health, with its prevalence projected to rise dramatically by 2045. This narrative review explores the bidirectional relationship between periodontitis (PD) and type 1 diabetes mellitus (T1DM), focusing on cellular and molecular mechanisms derived from the interplay between oral microbiota and the host immune response. A comprehensive search of studies published between 2008 and 2023 was conducted to elucidate the association between these two diseases. Preclinical and clinical evidence suggests a bidirectional relationship, with individuals with T1DM exhibiting heightened susceptibility to periodontitis, and vice versa. The review includes recent findings from human clinical studies, revealing variations in oral microbiota composition in T1DM patients, including increases in certain pathogenic species such as Porphyromonas gingivalis, Prevotella intermedia, and Aggregatibacter actinomycetemcomitans, along with shifts in microbial diversity and abundance. Molecular mechanisms underlying this association involve oxidative stress and dysregulated host immune responses, mediated by inflammatory cytokines such as IL-6, IL-8, and MMPs. Furthermore, disruptions in bone turnover markers, such as RANKL and OPG, contribute to periodontal complications in T1DM patients. While preventive measures to manage periodontal complications in T1DM patients may improve overall health outcomes, further research is needed to understand the intricate interactions between oral microbiota, host response, periodontal disease, and systemic health in this population.


Subject(s)
Diabetes Mellitus, Type 1 , Microbiota , Periodontal Diseases , Humans , Diabetes Mellitus, Type 1/microbiology , Diabetes Mellitus, Type 1/complications , Periodontal Diseases/microbiology , Periodontitis/microbiology , Periodontitis/complications , Periodontitis/immunology
3.
Front Immunol ; 15: 1413177, 2024.
Article in English | MEDLINE | ID: mdl-38903498

ABSTRACT

Introduction: Type 1 diabetes (T1D) is an autoimmune disease characterized by the destruction of insulin-producing ß cells. Toll-like receptor 9 (TLR9) plays a role in autoimmune diseases, and B cell-specific TLR9 deficiency delays T1D development. Gut microbiota are implicated in T1D, although the relationship is complex. However, the impact of B cell-specific deficiency of TLR9 on intestinal microbiota and the impact of altered intestinal microbiota on the development of T1D are unclear. Objectives: This study investigated how gut microbiota and the intestinal barrier contribute to T1D development in B cell-specific TLR9-deficient NOD mice. Additionally, this study explored the role of microbiota in immune regulation and T1D onset. Methods: The study assessed gut permeability, gene expression related to gut barrier integrity, and gut microbiota composition. Antibiotics depleted gut microbiota, and fecal samples were transferred to germ-free mice. The study also examined IL-10 production, Breg cell differentiation, and their impact on T1D development. Results: B cell-specific TLR9-deficient NOD mice exhibited increased gut permeability and downregulated gut barrier-related gene expression. Antibiotics restored gut permeability, suggesting microbiota influence. Altered microbiota were enriched in Lachnospiraceae, known for mucin degradation. Transferring this microbiota to germ-free mice increased gut permeability and promoted IL-10-expressing Breg cells. Rag-/- mice transplanted with fecal samples from Tlr9 fl/fl Cd19-Cre+ mice showed delayed diabetes onset, indicating microbiota's impact. Conclusion: B cell-specific TLR9 deficiency alters gut microbiota, increasing gut permeability and promoting IL-10-expressing Breg cells, which delay T1D. This study uncovers a link between TLR9, gut microbiota, and immune regulation in T1D, with implications for microbiota-targeted T1D therapies.


Subject(s)
Diabetes Mellitus, Type 1 , Gastrointestinal Microbiome , Interleukin-10 , Mice, Inbred NOD , Toll-Like Receptor 9 , Animals , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism , Gastrointestinal Microbiome/immunology , Interleukin-10/metabolism , Mice , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/microbiology , Mice, Knockout , B-Lymphocytes, Regulatory/immunology , Female , B-Lymphocytes/immunology , B-Lymphocytes/metabolism
4.
Molecules ; 29(12)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38930856

ABSTRACT

The extract of Dendrobium huoshanense, a traditional Chinese medicinal and food homologous plant belonging to the family Orchidaceae, was previously reported to have hypoglycemic and antioxidant effects. In this study, the direct effects of polysaccharide (DHP) and non-polysaccharide (NDHP) components of D. huoshanense, as well as its water extract (DHWE) were compared with that of metformin (an antidiabetic drug) on the gut microbiota (collected from fecal flora) of rats with streptozotocin-induced type 1 diabetes (T1D) using an in vitro fermentation method. The results showed that DHWE, DHP, and NDHP reduced pH and increased bacterial proliferation and short-chain fatty acid (SCFA) content in fermentation broth. DHWE, DHP, NDHP and metformin promoted the production of acetic and propionic acid, acetic acid, propionic acid and butyric acid, and propionic acid, respectively. DHWE, DHP, and NDHP reduced the abundance of Proteobacteria (subdominant pathogenic bacteria) and increased the abundance of Firmicutes (dominant beneficial gut bacteria). NDHP also reduced the abundance of Bacteroidetes (beneficial and conditional pathogenic). Metformin increased the abundance of Proteobacteria and reduced the abundance of Firmicutes and Bacteroidetes. At the genus level, NDHP promoted the proliferation of Megamonas and Megasphaera and decreased harmful bacteria (e.g., Klebsiella), and DHP increased the abundance of Prevotellaceae (opportunistic and usually harmless). By contrast, metformin increased the abundance of harmful bacteria (e.g., Citrobacter) and reduced the abundance of beneficial bacteria (e.g., Oscillospira). Our study indicates that DHWE, DHP, and NDHP are potentially more beneficial than metformin on the gut microbiota of T1D rats in vitro.


Subject(s)
Dendrobium , Diabetes Mellitus, Type 1 , Fatty Acids, Volatile , Gastrointestinal Microbiome , Metformin , Polysaccharides , Animals , Gastrointestinal Microbiome/drug effects , Metformin/pharmacology , Dendrobium/chemistry , Polysaccharides/pharmacology , Polysaccharides/chemistry , Rats , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/microbiology , Fatty Acids, Volatile/metabolism , Hypoglycemic Agents/pharmacology , Plant Extracts/pharmacology , Plant Extracts/chemistry , Male , Diabetes Mellitus, Experimental/drug therapy
5.
Nutrients ; 16(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38892608

ABSTRACT

Gut microbiome-modulating agents (MMAs), including probiotics, prebiotics, postbiotics, and synbiotics, are shown to ameliorate type 1 diabetes (T1D) by restoring the microbiome from dysbiosis. The objective of this systematic review and meta-analysis was to assess the impact of MMAs on hemoglobin A1c (HbA1c) and biomarkers associated with (T1D). A comprehensive search was conducted in PubMed, Web of Science, Embase, Cochrane Library, National Knowledge Infrastructure, WeiPu, and WanFang Data up to 30 November 2023. Ten randomized controlled trials (n = 630) were included, with study quality evaluated using the Cochrane risk-of-bias tool. Random-effect models with standardized mean differences (SMDs) were utilized. MMA supplementation was associated with improvements in HbA1c (SMD = -0.52, 95% CI [-0.83, -0.20]), daily insulin usage (SMD = -0.41, 95% confidence interval (CI) [-0.76, -0.07]), and fasting C-peptide (SMD = 0.99, 95% CI [0.17, 1.81]) but had no effects on FBG, CRP, TNF-α, IL-10, LDL, HDL, and the Shannon index. Subgroup analysis of HbA1c indicated that a long-term intervention (>3 months) might exert a more substantial effect. These findings suggest an association between MMAs and glycemic control in T1D. Further large-scale clinical trials are necessary to confirm these findings with investigations on inflammation and gut microbiota composition while adjusting confounding factors such as diet, physical activity, and the dose and form of MMA intervention.


Subject(s)
Diabetes Mellitus, Type 1 , Gastrointestinal Microbiome , Glycated Hemoglobin , Probiotics , Randomized Controlled Trials as Topic , Diabetes Mellitus, Type 1/microbiology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/blood , Humans , Gastrointestinal Microbiome/drug effects , Glycated Hemoglobin/metabolism , Probiotics/therapeutic use , Prebiotics/administration & dosage , Biomarkers/blood , Synbiotics/administration & dosage , Dietary Supplements , Female , Dysbiosis , Adult , Male
6.
Immunol Rev ; 325(1): 46-63, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38752578

ABSTRACT

Type 1 diabetes (T1D) is a chronic autoimmune disease targeting insulin-producing pancreatic beta cells. T1D is a multifactorial disease incorporating genetic and environmental factors. In recent years, the advances in high-throughput sequencing have allowed researchers to elucidate the changes in the gut microbiota taxonomy and functional capacity that accompany T1D development. An increasing number of studies have shown a role of the gut microbiota in mediating immune responses in health and disease, including autoimmunity. Fecal microbiota transplantations (FMT) have been largely used in murine models to prove a causal role of the gut microbiome in disease progression and have been shown to be a safe and effective treatment in inflammatory human diseases. In this review, we summarize and discuss recent research regarding the gut microbiota-host interactions in T1D, the current advancement in therapies for T1D, and the usefulness of FMT studies to explore microbiota-host immunity encounters in murine models and to shape the course of human type 1 diabetes.


Subject(s)
Autoimmunity , Diabetes Mellitus, Type 1 , Disease Models, Animal , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/microbiology , Diabetes Mellitus, Type 1/therapy , Humans , Animals , Gastrointestinal Microbiome/immunology , Mice
7.
Diabetes Obes Metab ; 26(8): 3306-3317, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38751358

ABSTRACT

AIM: To assess and verify the effect of the gut microbiome on the susceptibility and complications of type 1 diabetes (T1D). MATERIALS AND METHODS: To achieve this aim, a two-sample and reverse Mendelian randomization (MR) analysis was conducted. In addition, an external validation study was performed using individual microbiome data of patients with T1D from the gutMEGA datasets and the National Clinical Research Center for Metabolic Diseases. The circulating metabolites facilitated two-sample MR analysis, mediation and multivariable MR analysis to evaluate the direct relationship between the gut microbiome and T1D complications. RESULTS: The MR analysis results from the discovery and validation phases confirmed that Veillonellaceae can potentially reduce the susceptibility of T1D. In the gutMEGA dataset, the average relative abundance of Veillonellaceae in patients with T1D was 0.66%, compared with 1.09% in the controls. Furthermore, the external validation, which included 60 patients with T1D and 30 matched healthy controls, found that the median relative abundance of Veillonellaceae was also lower than controls at 1.10% (95% CI 0.50%-1.80%). Specifically, the Eubacterium coprostanoligenes group, known for its ability to regulate cholesterol, was significantly associated with a lower risk of developing renal, neurological and ophthalmic complications in T1D. Moreover, high cholesterol in small high-density lipoprotein and cholesteryl esters in high-density lipoprotein were associated with a reduced risk of T1D renal and ophthalmic complications. The mediation and multivariable MR analysis combining cholesterol indicated that the E. coprostanoligenes group is the most dominant factor influencing T1D complications. CONCLUSIONS: Our findings supported the potential causal effect of gut microbiota on the susceptibility and complications of T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Gastrointestinal Microbiome , Mendelian Randomization Analysis , Humans , Diabetes Mellitus, Type 1/microbiology , Diabetes Mellitus, Type 1/complications , Gastrointestinal Microbiome/physiology , Male , Female , Adult , Disease Susceptibility , Diabetes Complications/microbiology
8.
PLoS One ; 19(5): e0303863, 2024.
Article in English | MEDLINE | ID: mdl-38781241

ABSTRACT

Type 1 diabetes (T1D)-associated hyperglycemia develops, in part, from loss of insulin-secreting beta cells. The degree of glycemic dysregulation and the age at onset of disease can serve as indicators of the aggressiveness of the disease. Tracking blood glucose levels in prediabetic mice may demonstrate the onset of diabetes and, along with animal age, also presage disease severity. In this study, an analysis of blood glucose levels obtained from female NOD mice starting at 4 weeks until diabetes onset was undertaken. New onset diabetic mice were orally vaccinated with a Salmonella-based vaccine towards T1D-associated preproinsulin combined with TGFß and IL10 along with anti-CD3 antibody. Blood glucose levels were obtained before and after development of disease and vaccination. Animals were classified as acute disease if hyperglycemia was confirmed at a young age, while other animals were classified as progressive disease. The effectiveness of the oral T1D vaccine was greater in mice with progressive disease that had less glucose excursion compared to acute disease mice. Overall, the Salmonella-based vaccine reversed disease in 60% of the diabetic mice due, in part, to lessening of islet inflammation, improving residual beta cell health, and promoting tolerance. In summary, the age of disease onset and severity of glucose dysregulation in NOD mice predicted response to vaccine therapy. This suggests a similar disease categorization in the clinic may predict therapeutic response.


Subject(s)
Blood Glucose , Diabetes Mellitus, Type 1 , Mice, Inbred NOD , Animals , Female , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/microbiology , Mice , Administration, Oral , Blood Glucose/metabolism , Salmonella Vaccines/immunology , Salmonella Vaccines/administration & dosage , Salmonella/immunology , Insulin/immunology , Disease Progression , Acute Disease , Protein Precursors
9.
Diabetes Obes Metab ; 26(6): 2158-2166, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38433703

ABSTRACT

AIM: Type 1 diabetes results from autoimmune events influenced by environmental variables, including changes in diet. This study investigated how feeding refined versus unrefined (aka 'chow') diets affects the onset and progression of hyperglycaemia in non-obese diabetic (NOD) mice. METHODS: Female NOD mice were fed either unrefined diets or matched refined low- and high-fat diets. The onset of hyperglycaemia, glucose tolerance, food intake, energy expenditure, circulating insulin, liver gene expression and microbiome changes were measured for each dietary group. RESULTS: NOD mice consuming unrefined (chow) diets developed hyperglycaemia at similar frequencies. By contrast, mice consuming the defined high-fat diet had an accelerated onset of hyperglycaemia compared to the matched low-fat diet. There was no change in food intake, energy expenditure, or physical activity within each respective dietary group. Microbiome changes were driven by diet type, with chow diets clustering similarly, while refined low- and high-fat bacterial diversity also grouped closely. In the defined dietary cohort, liver gene expression changes in high-fat-fed mice were consistent with a greater frequency of hyperglycaemia and impaired glucose tolerance. CONCLUSION: Glucose intolerance is associated with an enhanced frequency of hyperglycaemia in female NOD mice fed a defined high-fat diet. Using an appropriate matched control diet is an essential experimental variable when studying changes in microbiome composition and diet as a modifier of disease risk.


Subject(s)
Diabetes Mellitus, Type 1 , Diet, High-Fat , Hyperglycemia , Mice, Inbred NOD , Animals , Diet, High-Fat/adverse effects , Female , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/microbiology , Mice , Hyperglycemia/etiology , Glucose Intolerance/etiology , Energy Metabolism , Liver/metabolism , Diet, Fat-Restricted , Insulin/metabolism , Insulin/blood , Blood Glucose/metabolism
10.
Brief Funct Genomics ; 23(4): 464-474, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38376798

ABSTRACT

Gut microbes is a crucial factor in the pathogenesis of type 1 diabetes (T1D). However, it is still unclear which gut microbiota are the key factors affecting T1D and their influence on the development and progression of the disease. To fill these knowledge gaps, we constructed a model to find biomarker from gut microbiota in patients with T1D. We first identified microbial markers using Linear discriminant analysis Effect Size (LEfSe) and random forest (RF) methods. Furthermore, by constructing co-occurrence networks for gut microbes in T1D, we aimed to reveal all gut microbial interactions as well as major beneficial and pathogenic bacteria in healthy populations and type 1 diabetic patients. Finally, PICRUST2 was used to predict Kyoto Encyclopedia of Genes and Genomes (KEGG) functional pathways and KO gene levels of microbial markers to investigate the biological role. Our study revealed that 21 identified microbial genera are important biomarker for T1D. Their AUC values are 0.962 and 0.745 on discovery set and validation set. Functional analysis showed that 10 microbial genera were significantly positively associated with D-arginine and D-ornithine metabolism, spliceosome in transcription, steroid hormone biosynthesis and glycosaminoglycan degradation. These genera were significantly negatively correlated with steroid biosynthesis, cyanoamino acid metabolism and drug metabolism. The other 11 genera displayed an inverse correlation. In summary, our research identified a comprehensive set of T1D gut biomarkers with universal applicability and have revealed the biological consequences of alterations in gut microbiota and their interplay. These findings offer significant prospects for individualized management and treatment of T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Gastrointestinal Microbiome , Machine Learning , Humans , Gastrointestinal Microbiome/genetics , Diabetes Mellitus, Type 1/microbiology , Biomarkers/metabolism , Bacteria/genetics , Bacteria/metabolism , Male
11.
Int. microbiol ; 26(3): 675-690, Ene-Agos, 2023. ilus, tab, graf
Article in English | IBECS | ID: ibc-223992

ABSTRACT

Type 1 diabetes mellitus (T1DM) has been increasing in prevalence in the last decades and has become a global burden. Autoantibodies against human glutamate decarboxylase (GAD65) are among the first to be detected at the onset of T1DM. Diverse viruses have been proposed to be involved in the triggering of T1DM because of molecular mimicry, i.e., similarity between parts of some viral proteins and one or more epitopes of GAD65. However, the possibility that bacterial proteins might also be responsible for GAD65 mimicry has been seldom investigated. To date, many genomes of Streptococcus pneumoniae (the pneumococcus), a prominent human pathogen particularly prevalent among children and the elderly, have been sequenced. A dataset of more than 9000 pneumococcal genomes was mined and two different (albeit related) genes (gadA and gadB), presumably encoding two glutamate decarboxylases similar to GAD65, were found. The various gadASpn alleles were present only in serotype 3 pneumococci belonging to the global lineage GPSC83, although some homologs have also been discovered in two subspecies of Streptococcus constellatus (pharyngis and viborgensis), an isolate of the group B streptococci, and several strains of Lactobacillus delbrueckii. Besides, gadBSpn alleles are present in > 10% of the isolates in our dataset and represent 16 GPSCs with 123 sequence types and 20 different serotypes. Sequence analyses indicated that gadA- and gadB-like genes have been mobilized among different bacteria either by prophage(s) or by integrative and conjugative element(s), respectively. Substantial similarities appear to exist between the putative pneumococcal glutamate decarboxylases and well-known epitopes of GAD65. In this sense, the use of broader pneumococcal conjugate vaccines such as PCV20 would prevent the majority of serotypes expressing those genes that might potentially contribute to T1DM.(AU)


Subject(s)
Humans , Streptococcus pneumoniae/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/microbiology , Glutamate Decarboxylase/genetics , Molecular Mimicry , Pneumococcal Vaccines , Microbiology , Microbiological Techniques , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/prevention & control
12.
Exp Cell Res ; 422(1): 113429, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36402426

ABSTRACT

This study aimed to explore the effect of PF in regulating the progression of T1D through regulating gut microbiota and inhibiting TLR4-myD88/TRIF pathway. T1D mouse models were established and received PF treatment through intraperitoneal injection. The glucose, sugar tolerance, the incidence of T1D and H&E staining were detected to verify the effect of PF on T1D. Meanwhile, the changes of gut microbiota and the permeability of intestines in mice were also measured. On parallel, the number and function of immune cells were detected by Flow Cytometry. The expressions of ZO-1, ZO-2 and TLR4-myD88/TRIF pathway related proteins were detected by western blotting. Mice received PF treatment had decreased incidence of T1D and inflammatory infiltration in islet tissues compared with those received PBS treatment. In addition to that, PF treated mice had increased Sutterella species and decreased intestinal permeability, in which the decreased ratio of Th1/Th17 and increased Treg cells were also identified. The expression of TLR4-myD88/TRIF pathway was also suppressed in response to PF treatment. Moreover, further treatment with TLR4 agonist, LPS, could reverse the effect of PF on T1D mice. PF can suppress the TLR4 mediated myD88/TRIF pathway to change the distribution of gut microbiota, so as to protect NOD mice from T1D.


Subject(s)
Diabetes Mellitus, Experimental , Gastrointestinal Microbiome , Animals , Mice , Adaptor Proteins, Vesicular Transport/genetics , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/microbiology , Gastrointestinal Microbiome/drug effects , Mice, Inbred C57BL , Mice, Inbred NOD , Myeloid Differentiation Factor 88/metabolism , Signal Transduction , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/microbiology
13.
Microbiome ; 10(1): 243, 2022 12 28.
Article in English | MEDLINE | ID: mdl-36578059

ABSTRACT

BACKGROUND: Alterations to the gut microbiome have been linked to multiple chronic diseases. However, the drivers of such changes remain largely unknown. The oral cavity acts as a major route of exposure to exogenous factors including pathogens, and processes therein may affect the communities in the subsequent compartments of the gastrointestinal tract. Here, we perform strain-resolved, integrated meta-genomic, transcriptomic, and proteomic analyses of paired saliva and stool samples collected from 35 individuals from eight families with multiple cases of type 1 diabetes mellitus (T1DM). RESULTS: We identified distinct oral microbiota mostly reflecting competition between streptococcal species. More specifically, we found a decreased abundance of the commensal Streptococcus salivarius in the oral cavity of T1DM individuals, which is linked to its apparent competition with the pathobiont Streptococcus mutans. The decrease in S. salivarius in the oral cavity was also associated with its decrease in the gut as well as higher abundances in facultative anaerobes including Enterobacteria. In addition, we found evidence of gut inflammation in T1DM as reflected in the expression profiles of the Enterobacteria as well as in the human gut proteome. Finally, we were able to follow transmitted strain-variants from the oral cavity to the gut at the individual omic levels, highlighting not only the transfer, but also the activity of the transmitted taxa along the gastrointestinal tract. CONCLUSIONS: Alterations of the oral microbiome in the context of T1DM impact the microbial communities in the lower gut, in particular through the reduction of "mouth-to-gut" transfer of Streptococcus salivarius. Our results indicate that the observed oral-cavity-driven gut microbiome changes may contribute towards the inflammatory processes involved in T1DM. Through the integration of multi-omic analyses, we resolve strain-variant "mouth-to-gut" transfer in a disease context. Video Abstract.


Subject(s)
Diabetes Mellitus, Type 1 , Gastrointestinal Microbiome , Microbiota , Humans , Gastrointestinal Microbiome/genetics , Diabetes Mellitus, Type 1/microbiology , Proteomics , Multiomics , Microbiota/genetics , Mouth/microbiology , Enterobacteriaceae
14.
PLoS Biol ; 20(3): e3001556, 2022 03.
Article in English | MEDLINE | ID: mdl-35235560

ABSTRACT

Evaluating the relationship between the human gut microbiome and disease requires computing reliable statistical associations. Here, using millions of different association modeling strategies, we evaluated the consistency-or robustness-of microbiome-based disease indicators for 6 prevalent and well-studied phenotypes (across 15 public cohorts and 2,343 individuals). We were able to discriminate between analytically robust versus nonrobust results. In many cases, different models yielded contradictory associations for the same taxon-disease pairing, some showing positive correlations and others negative. When querying a subset of 581 microbe-disease associations that have been previously reported in the literature, 1 out of 3 taxa demonstrated substantial inconsistency in association sign. Notably, >90% of published findings for type 1 diabetes (T1D) and type 2 diabetes (T2D) were particularly nonrobust in this regard. We additionally quantified how potential confounders-sequencing depth, glucose levels, cholesterol, and body mass index, for example-influenced associations, analyzing how these variables affect the ostensible correlation between Faecalibacterium prausnitzii abundance and a healthy gut. Overall, we propose our approach as a method to maximize confidence when prioritizing findings that emerge from microbiome association studies.


Subject(s)
Bacteria/genetics , Biomedical Research/methods , Gastrointestinal Microbiome/genetics , Metagenome/genetics , Metagenomics/methods , Algorithms , Bacteria/classification , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/microbiology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/microbiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/microbiology , Feces/microbiology , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/microbiology , Models, Theoretical , RNA, Ribosomal, 16S/genetics
15.
PLoS One ; 17(2): e0261103, 2022.
Article in English | MEDLINE | ID: mdl-35196314

ABSTRACT

A variety of islet autoantibodies (AAbs) can predict and possibly dictate eventual type 1 diabetes (T1D) diagnosis. Upwards of 75% of those with T1D are positive for AAbs against glutamic acid decarboxylase (GAD65 or GAD), a producer of gamma-aminobutyric acid (GABA) in human pancreatic beta cells. Interestingly, bacterial populations within the human gut also express GAD and produce GABA. Evidence suggests that dysbiosis of the microbiome may correlate with T1D pathogenesis and physiology. Therefore, autoimmune linkages between the gut microbiome and islets susceptible to autoimmune attack need to be further elucidated. Utilizing in silico analyses, we show that 25 GAD sequences from human gut bacterial sources show sequence and motif similarities to human beta cell GAD65. Our motif analyses determined that most gut GAD sequences contain the pyroxical dependent decarboxylase (PDD) domain of human GAD65, which is important for its enzymatic activity. Additionally, we showed overlap with known human GAD65 T cell receptor epitopes, which may implicate the immune destruction of beta cells. Thus, we propose a physiological hypothesis in which changes in the gut microbiome in those with T1D result in a release of bacterial GAD, thus causing miseducation of the host immune system. Due to the notable similarities we found between human and bacterial GAD, these deputized immune cells may then target human beta cells leading to the development of T1D.


Subject(s)
Autoantibodies/immunology , Bacteria/enzymology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/microbiology , Gastrointestinal Microbiome/immunology , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/immunology , Animals , Antigen-Presenting Cells/immunology , Computer Simulation , Diabetes Mellitus, Type 1/enzymology , Epitopes, T-Lymphocyte/immunology , Genes, Bacterial , Humans , Islets of Langerhans/enzymology , Islets of Langerhans/immunology , Mice , Pan troglodytes/microbiology , Phylogeny , Protein Domains , Sequence Alignment/methods , gamma-Aminobutyric Acid/metabolism
16.
Int J Mol Sci ; 23(1)2022 Jan 01.
Article in English | MEDLINE | ID: mdl-35008906

ABSTRACT

Diabetes mellitus is a significant clinical and therapeutic problem because it can lead to serious long-term complications. Its pathogenesis is not fully understood, but there are indications that dysbiosis can play a role in the development of diabetes, or that it appears during the course of the disease. Changes in microbiota composition are observed in both type 1 diabetes (T1D) and type 2 diabetes (T2D) patients. These modifications are associated with pro-inflammation, increased intestinal permeability, endotoxemia, impaired ß-cell function and development of insulin resistance. This review summarizes the role of the gut microbiota in healthy individuals and the changes in bacterial composition that can be associated with T1D or T2D. It also presents new developments in diabetes therapy based on influencing the gut microbiota as a promising method to alter the course of diabetes. Moreover, it highlights the lacking data and suggests future directions needed to prove the causal relationship between dysbiosis and diabetes, both T1D and T2D.


Subject(s)
Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/microbiology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/microbiology , Gastrointestinal Microbiome , Animals , Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/prevention & control , Diabetes Mellitus, Type 2/therapy , Homeostasis , Humans , Models, Biological
17.
Invest Ophthalmol Vis Sci ; 63(1): 5, 2022 01 03.
Article in English | MEDLINE | ID: mdl-34985498

ABSTRACT

Purpose: The gut microbiome has been linked to disease pathogenesis through their interaction in metabolic, endocrine, and immune functions. The goal of this study was to determine whether the gut and plasma microbiota could transfer microbes to the retina in type 1 diabetic mice with retinopathy. Methods: We analyzed the fecal, plasma, whole globe, and retina microbiome in Akita mice and compared with age-matched wild-type (WT) mice using 16S rRNA sequencing and metatranscriptomic analysis. To eliminate the contribution of the ocular surface and plasma microbiome, mice were perfused with sterile saline solution, the whole globes were extracted, and the neural retina was removed under sterile conditions for retinal microbiome. Results: Our microbiome analysis revealed that Akita mice demonstrated a distinct pattern of microbes within each source: feces, plasma, whole globes, and retina. WT mice and Akita mice experienced transient bacteremia in the plasma and retina. Bacteria were identified in the retina of the Akita mice, specifically Corynebacterium, Pseudomonas, Lactobacillus, Staphylococcus, Enterococcus, and Bacillus. Significantly increased levels of peptidoglycan (0.036 ± 0.001 vs. 0.023 ± 0.002; P < 0.002) and TLR2 (3.47 ± 0.15 vs. 1.99 ± 0.07; P < 0.0001) were observed in the retina of Akita mice compared to WT. Increased IBA+ cells in the retina, reduced a- and b-waves on electroretinography, and increased acellular capillary formation demonstrated the presence of retinopathy in the Akita cohort compared to WT mice. Conclusions: Together, our findings suggest that transient bacteremia exists in the plasma and retina of both cohorts. The bacteria found in Akita mice are distinct from WT mice and may contribute to development of retinal inflammation and barrier dysfunction in retinopathy.


Subject(s)
Bacteremia/microbiology , Bacteria/isolation & purification , Diabetic Retinopathy/microbiology , Feces/microbiology , Retina/microbiology , Animals , Bacteria/genetics , Diabetes Mellitus, Type 1/microbiology , Disease Models, Animal , Electroretinography , Enzyme-Linked Immunosorbent Assay , Eye/microbiology , Gastrointestinal Microbiome/physiology , Male , Mice , Mice, Inbred C57BL , Microbiota/physiology , RNA, Ribosomal, 16S/genetics
18.
BMC Microbiol ; 22(1): 8, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34983374

ABSTRACT

BACKGROUND: Type 1 diabetes (T1D) is an autoimmune disease that is increasing in prevalence worldwide. One of the contributing factors to the pathogenesis of T1D is the composition of the intestinal microbiota, as has been demonstrated. in T1D patients, with some studies demonstrating a deficiency in their levels of Prevotella. We have isolated a strain of Prevotella histicola from a duodenal biopsy that has anti-inflammatory properties, and in addition, alters the development of autoimmune diseases in mouse models. Therefore, our hypothesis is that the oral administration of P. histicola might delay the development of T1D in the non-obese diabetic (NOD) mice. To assess this, we used the following materials and methods. Female NOD mice (ages 5-8 weeks) were administered every other day P. histicola that was cultured in-house. Blood glucose levels were measured every other week. Mice were sacrificed at various time points for histopathological analysis of the pancreas. Modulation of immune response by the commensal was tested by analyzing regulatory T-cells and NKp46+ cells using flow cytometry and intestinal cytokine mRNA transcript levels using quantitative RT-PCR. For microbial composition, 16 s rRNA gene analysis was conducted on stool samples collected at various time points. RESULTS: Administration of P. histicola in NOD mice delayed the onset of T1D. Beta diversity in the fecal microbiomes demonstrated that the microbial composition of the mice administered P. histicola was different from those that were not treated. Treatment with P. histicola led to a significant increase in regulatory T cells with a concomitant decrease in NKp46+ cells in the pancreatic lymph nodes as compared to the untreated group after 5 weeks of treatment. CONCLUSIONS: These observations suggest that P. histicola treatment delayed onset of diabetes by increasing the levels of regulatory T cells in the pancreatic lymph nodes. This preliminary work supports the rationale that enteral exposure to a non pathogenic commensal P. histicola be tested as a future therapy for T1D.


Subject(s)
Diabetes Mellitus, Type 1/diet therapy , Gastrointestinal Microbiome/physiology , Prevotella/physiology , Probiotics/administration & dosage , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Cytokines/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/microbiology , Duodenum/immunology , Duodenum/microbiology , Feces/microbiology , Female , Humans , Mice , Mice, Inbred NOD , Pancreas/immunology , Pancreas/pathology
19.
Diabetes Care ; 45(3): 555-563, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35045174

ABSTRACT

OBJECTIVE: Previous studies have demonstrated an association between gut microbiota composition and type 1 diabetes (T1D) pathogenesis. However, little is known about the composition and function of the gut microbiome in adults with longstanding T1D or its association with host glycemic control. RESEARCH DESIGN AND METHODS: We performed a metagenomic analysis of the gut microbiome obtained from fecal samples of 74 adults with T1D, 14.6 ± 9.6 years following diagnosis, and compared their microbial composition and function to 296 age-matched healthy control subjects (1:4 ratio). We further analyzed the association between microbial taxa and indices of glycemic control derived from continuous glucose monitoring measurements and blood tests and constructed a prediction model that solely takes microbiome features as input to evaluate the discriminative power of microbial composition for distinguishing individuals with T1D from control subjects. RESULTS: Adults with T1D had a distinct microbial signature that separated them from control subjects when using prediction algorithms on held-out subjects (area under the receiver operating characteristic curve = 0.89 ± 0.03). Linear discriminant analysis showed several bacterial species with significantly higher scores in T1D, including Prevotella copri and Eubacterium siraeum, and species with higher scores in control subjects, including Firmicutes bacterium and Faecalibacterium prausnitzii (P < 0.05, false discovery rate corrected for all). On the functional level, several metabolic pathways were significantly lower in adults with T1D. Several bacterial taxa and metabolic pathways were associated with the host's glycemic control. CONCLUSIONS: We identified a distinct gut microbial signature in adults with longstanding T1D and associations between microbial taxa, metabolic pathways, and glycemic control indices. Additional mechanistic studies are needed to identify the role of these bacteria for potential therapeutic strategies.


Subject(s)
Diabetes Mellitus, Type 1 , Gastrointestinal Microbiome , Adult , Blood Glucose , Blood Glucose Self-Monitoring , Diabetes Mellitus, Type 1/microbiology , Feces/microbiology , Gastrointestinal Microbiome/genetics , Glycemic Control , Humans
20.
Gut ; 71(2): 296-308, 2022 02.
Article in English | MEDLINE | ID: mdl-33593807

ABSTRACT

OBJECTIVE: Type 1 diabetes (T1D) is an autoimmune disease caused by the destruction of pancreatic ß-cells producing insulin. Both T1D patients and animal models exhibit gut microbiota and mucosa alterations, although the exact cause for these remains poorly understood. We investigated the production of key cytokines controlling gut integrity, the abundance of segmented filamentous bacteria (SFB) involved in the production of these cytokines, and the respective role of autoimmune inflammation and hyperglycaemia. DESIGN: We used several mouse models of autoimmune T1D as well as mice rendered hyperglycaemic without inflammation to study gut mucosa and microbiota dysbiosis. We analysed cytokine expression in immune cells, epithelial cell function, SFB abundance and microbiota composition by 16S sequencing. We assessed the role of anti-tumour necrosis factor α on gut mucosa inflammation and T1D onset. RESULTS: We show in models of autoimmune T1D a conserved loss of interleukin (IL)-17A, IL-22 and IL-23A in gut mucosa. Intestinal epithelial cell function was altered and gut integrity was impaired. These defects were associated with dysbiosis including progressive loss of SFB. Transfer of diabetogenic T-cells recapitulated these gut alterations, whereas induction of hyperglycaemia with no inflammation failed to do so. Moreover, anti-inflammatory treatment restored gut mucosa and immune cell function and dampened diabetes incidence. CONCLUSION: Our results demonstrate that gut mucosa alterations and dysbiosis in T1D are primarily linked to inflammation rather than hyperglycaemia. Anti-inflammatory treatment preserves gut homeostasis and protective commensal flora reducing T1D incidence.


Subject(s)
Bacteria/isolation & purification , Diabetes Mellitus, Type 1/complications , Dysbiosis/etiology , Gastrointestinal Microbiome , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Animals , Cytokines/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/microbiology , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Hyperglycemia/etiology , Inflammation/etiology , Intestinal Mucosa/metabolism , Mice
SELECTION OF CITATIONS
SEARCH DETAIL