Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 17(5): 505-513, 2016 05.
Article in English | MEDLINE | ID: mdl-26998764

ABSTRACT

The effect of alterations in intestinal microbiota on microbial metabolites and on disease processes such as graft-versus-host disease (GVHD) is not known. Here we carried out an unbiased analysis to identify previously unidentified alterations in gastrointestinal microbiota-derived short-chain fatty acids (SCFAs) after allogeneic bone marrow transplant (allo-BMT). Alterations in the amount of only one SCFA, butyrate, were observed only in the intestinal tissue. The reduced butyrate in CD326(+) intestinal epithelial cells (IECs) after allo-BMT resulted in decreased histone acetylation, which was restored after local administration of exogenous butyrate. Butyrate restoration improved IEC junctional integrity, decreased apoptosis and mitigated GVHD. Furthermore, alteration of the indigenous microbiota with 17 rationally selected strains of high butyrate-producing Clostridia also decreased GVHD. These data demonstrate a heretofore unrecognized role of microbial metabolites and suggest that local and specific alteration of microbial metabolites has direct salutary effects on GVHD target tissues and can mitigate disease severity.


Subject(s)
Epithelial Cells/immunology , Gastrointestinal Microbiome/immunology , Graft vs Host Disease/immunology , Intestines/immunology , Metabolome/immunology , Acetylation/drug effects , Animals , Bone Marrow Transplantation/adverse effects , Bone Marrow Transplantation/methods , Butyrates/immunology , Butyrates/metabolism , Butyrates/pharmacology , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Fatty Acids, Volatile/immunology , Fatty Acids, Volatile/metabolism , Female , Gas Chromatography-Mass Spectrometry , Gastrointestinal Microbiome/physiology , Gene Expression/immunology , Graft vs Host Disease/etiology , Graft vs Host Disease/microbiology , Histone Acetyltransferases/genetics , Histone Acetyltransferases/immunology , Histone Acetyltransferases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/immunology , Histone Deacetylases/metabolism , Histones/immunology , Histones/metabolism , Immunoblotting , Intestines/cytology , Intestines/microbiology , Mice, Inbred BALB C , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Homologous
2.
Immunity ; 48(5): 992-1005.e8, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768180

ABSTRACT

Dietary fiber protects against chronic inflammatory diseases by dampening immune responses through short-chain fatty acids (SCFAs). Here we examined the effect of dietary fiber in viral infection, where the anti-inflammatory properties of SCFAs in principle could prevent protective immunity. Instead, we found that fermentable dietary fiber increased survival of influenza-infected mice through two complementary mechanisms. High-fiber diet (HFD)-fed mice exhibited altered bone marrow hematopoiesis, characterized by enhanced generation of Ly6c- patrolling monocytes, which led to increased numbers of alternatively activated macrophages with a limited capacity to produce the chemokine CXCL1 in the airways. Blunted CXCL1 production reduced neutrophil recruitment to the airways, thus limiting tissue immunopathology during infection. In parallel, diet-derived SCFAs boosted CD8+ T cell effector function by enhancing cellular metabolism. Hence, dietary fermentable fiber and SCFAs set an immune equilibrium, balancing innate and adaptive immunity so as to promote the resolution of influenza infection while preventing immune-associated pathology.


Subject(s)
Antigens, Ly/immunology , CD8-Positive T-Lymphocytes/immunology , Dietary Fiber/pharmacology , Hematopoiesis/immunology , Monocytes/immunology , Orthomyxoviridae Infections/immunology , Adaptive Immunity/drug effects , Adaptive Immunity/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Dietary Fiber/administration & dosage , Fatty Acids, Volatile/immunology , Fatty Acids, Volatile/metabolism , Hematopoiesis/drug effects , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Monocytes/metabolism , Protective Agents/administration & dosage , Protective Agents/pharmacology
3.
Immunity ; 43(2): 304-17, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26253786

ABSTRACT

Antimicrobial peptides (AMPs) expressed by epithelial and immune cells are largely described for the defense against invading microorganisms. Recently, their immunomodulatory functions have been highlighted in various contexts. However how AMPs expressed by non-immune cells might influence autoimmune responses in peripheral tissues, such as the pancreas, is unknown. Here, we found that insulin-secreting ß-cells produced the cathelicidin related antimicrobial peptide (CRAMP) and that this production was defective in non-obese diabetic (NOD) mice. CRAMP administrated to prediabetic NOD mice induced regulatory immune cells in the pancreatic islets, dampening the incidence of autoimmune diabetes. Additional investigation revealed that the production of CRAMP by ß-cells was controlled by short-chain fatty acids produced by the gut microbiota. Accordingly, gut microbiota manipulations in NOD mice modulated CRAMP production and inflammation in the pancreatic islets, revealing that the gut microbiota directly shape the pancreatic immune environment and autoimmune diabetes development.


Subject(s)
Cathelicidins/metabolism , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , Intestines/immunology , Microbiota/physiology , Pancreas/immunology , Animals , Antimicrobial Cationic Peptides , Cathelicidins/genetics , Diabetes Mellitus, Type 1/microbiology , Fatty Acids, Volatile/immunology , Female , Intestines/microbiology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Pancreas/microbiology
4.
J Infect Dis ; 223(12 Suppl 2): S194-S200, 2021 06 16.
Article in English | MEDLINE | ID: mdl-33326565

ABSTRACT

Clostridioides difficile is an urgent antimicrobial-resistant bacterium, causing mild to moderate and sometimes life-threatening disease. Commensal gut microbes are critical for providing colonization resistance against C difficile and can be leveraged as non-antibiotic alternative therapeutics for the prevention and treatment of C difficile infection.


Subject(s)
Clostridioides difficile/growth & development , Clostridium Infections/immunology , Animals , Anti-Bacterial Agents/immunology , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/therapeutic use , Antibiosis , Bile Acids and Salts/immunology , Bile Acids and Salts/metabolism , Clostridioides difficile/drug effects , Clostridioides difficile/metabolism , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Fatty Acids, Volatile/immunology , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Humans , Nutrients/immunology , Nutrients/metabolism
5.
Infect Immun ; 89(9): e0018821, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34097474

ABSTRACT

Short-chain fatty acids (SCFAs) are the main metabolites produced by the gut microbiota via the fermentation of complex carbohydrates and fibers. Evidence suggests that SCFAs play a role in the control of infections through direct action both on microorganisms and on host signaling. This review summarizes the main microbicidal effects of SCFAs and discusses studies highlighting the effect of SCFAs in the virulence and viability of microorganisms. We also describe the diverse and complex modes of action of the SCFAs on the immune system in the face of infections with a specific focus on bacterial and viral respiratory infections. A growing body of evidence suggests that SCFAs protect against lung infections. Finally, we present potential strategies that may be leveraged to exploit the biological properties of SCFAs for increasing effectiveness and optimizing patient benefits.


Subject(s)
Anti-Infective Agents/therapeutic use , Fatty Acids, Volatile/therapeutic use , Infections/drug therapy , Lung/drug effects , Animals , Anti-Infective Agents/immunology , Anti-Infective Agents/metabolism , Fatty Acids, Volatile/immunology , Fatty Acids, Volatile/metabolism , Humans , Infections/immunology , Infections/microbiology , Lung/immunology , Lung/microbiology , Lung/virology , Microbial Viability , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/virology , Signal Transduction/immunology , Virulence
6.
Mol Cell Neurosci ; 105: 103493, 2020 06.
Article in English | MEDLINE | ID: mdl-32333962

ABSTRACT

Neuroinflammation contributes to neurodegenerative disorders, including Alzheimer's disease (AD). Gut microbes are involved in regulating systemic inflammation. Short-chain fatty acids (SCFAs), which are among the many metabolites released by gut microbes, can cross the blood-brain barrier (BBB) and interact with microglia. High concentrations of individual SCFAs decrease the inflammatory responses of peripheral monocytes; therefore, we hypothesized that SCFAs act on their own or in combinations to reduce the inflammatory response of microglia. Cultured human THP-1 monocytic cells and differentiated human HL-60 myelomonocytic cells were used to model select immune functions of human microglia. Acetate, propionate, butyrate, formate, and valerate were added to cells alone or as a mixture containing SCFAs at an approximate physiological concentration ratio. The SCFA mixture, as well as several individual SCFAs at the highest concentrations used in the mixture (15-236 µM), decreased the secretion of interleukin (IL)-1ß, monocyte chemoattractant protein (MCP)-1, tumor necrosis factor (TNF)-α, and cytotoxins by immune-stimulated THP-1 cells. GLPG 0974, a free fatty acid receptor (FFAR) 2/3 antagonist, did not block the inhibitory effect of the SCFA mixture on IL-1ß secretion by THP-1 cells while blocking the inhibitory effect of formate alone. We demonstrated that formate and valerate alone reduced the phagocytic activity of immune-stimulated THP-1 cells. Formate, but not valerate, alone also inhibited the N-formylmethionine-leucyl-phenylalanine (fMLP)-induced respiratory burst of HL-60 cells, reducing the production of reactive oxygen species (ROS). Our data indicate that SCFAs could regulate select microglial functions that are disrupted in AD.


Subject(s)
Butyrates/pharmacology , Fatty Acids, Volatile/metabolism , Microglia/metabolism , Monocytes/metabolism , Thiophenes/pharmacology , Chemokine CCL2/metabolism , Fatty Acids, Volatile/immunology , Humans , Inflammation/drug therapy , Inflammation/metabolism , Microglia/drug effects , Microglia/immunology , Monocytes/drug effects , Tumor Necrosis Factor-alpha/metabolism
7.
J Am Soc Nephrol ; 31(7): 1445-1461, 2020 07.
Article in English | MEDLINE | ID: mdl-32482686

ABSTRACT

BACKGROUND: Short-chain fatty acids derived from gut microbial fermentation of dietary fiber have been shown to suppress autoimmunity through mechanisms that include enhanced regulation by T regulatory cells (Tregs). METHODS: Using a murine kidney transplantation model, we examined the effects on alloimmunity of a high-fiber diet or supplementation with the short-chain fatty acid acetate. Kidney transplants were performed from BALB/c(H2d) to B6(H2b) mice as allografts in wild-type and recipient mice lacking the G protein-coupled receptor GPR43 (the metabolite-sensing receptor of acetate). Allograft mice received normal chow, a high-fiber diet, or normal chow supplemented with sodium acetate. We assessed rejection at days 14 (acute) and 100 (chronic), and used 16S rRNA sequencing to determine gut microbiota composition pretransplantation and post-transplantation. RESULTS: Wild-type mice fed normal chow exhibited dysbiosis after receiving a kidney allograft but not an isograft, despite the avoidance of antibiotics and immunosuppression for the latter. A high-fiber diet prevented dysbiosis in allograft recipients, who demonstrated prolonged survival and reduced evidence of rejection compared with mice fed normal chow. Allograft mice receiving supplemental sodium acetate exhibited similar protection from rejection, and subsequently demonstrated donor-specific tolerance. Depletion of CD25+ Tregs or absence of the short-chain fatty acid receptor GPR43 abolished this survival advantage. CONCLUSIONS: Manipulation of the microbiome by a high-fiber diet or supplementation with sodium acetate modified alloimmunity in a kidney transplant model, generating tolerance dependent on Tregs and GPR43. Diet-based therapy to induce changes in the gut microbiome can alter systemic alloimmunity in mice, in part through the production of short-chain fatty acids leading to Treg cell development, and merits study as a potential clinical strategy to facilitate transplant acceptance.


Subject(s)
Dietary Fiber/administration & dosage , Fatty Acids, Volatile/immunology , Gastrointestinal Microbiome/immunology , Graft Rejection/prevention & control , Immune Tolerance/drug effects , T-Lymphocytes, Regulatory , Acute Disease , Allografts/immunology , Animals , Butyric Acid/pharmacology , Chronic Disease , Dietary Supplements , Dysbiosis/etiology , Dysbiosis/microbiology , Dysbiosis/prevention & control , Gastrointestinal Microbiome/drug effects , Graft Rejection/pathology , Graft Rejection/physiopathology , Graft Survival/drug effects , Graft Survival/immunology , Kidney Transplantation/adverse effects , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Sodium Acetate/pharmacology
8.
Int J Mol Sci ; 22(16)2021 Aug 06.
Article in English | MEDLINE | ID: mdl-34445184

ABSTRACT

The microbiome, i.e., the communities of microbes that inhabit the surfaces exposed to the external environment, participates in the regulation of host physiology, including the immune response against pathogens. At the same time, the immune response shapes the microbiome to regulate its composition and function. How the crosstalk between the immune system and the microbiome regulates the response to fungal infection has remained relatively unexplored. We have previously shown that strict anaerobes protect from infection with the opportunistic fungus Aspergillus fumigatus by counteracting the expansion of pathogenic Proteobacteria. By resorting to immunodeficient mouse strains, we found that the lung microbiota could compensate for the lack of B and T lymphocytes in Rag1-/- mice by skewing the composition towards an increased abundance of protective anaerobes such as Clostridia and Bacteroidota. Conversely, NSG mice, with major defects in both the innate and adaptive immune response, showed an increased susceptibility to infection associated with a low abundance of strict anaerobes and the expansion of Proteobacteria. Further exploration in a murine model of chronic granulomatous disease, a primary form of immunodeficiency characterized by defective phagocyte NADPH oxidase, confirms the association of lung unbalance between anaerobes and Proteobacteria and the susceptibility to aspergillosis. Consistent changes in the lung levels of short-chain fatty acids between the different strains support the conclusion that the immune system and the microbiota are functionally intertwined during Aspergillus infection and determine the outcome of the infection.


Subject(s)
Aspergillosis/immunology , Aspergillus fumigatus/immunology , Lung/microbiology , Adaptive Immunity , Animals , Aspergillosis/microbiology , Aspergillus fumigatus/physiology , Fatty Acids, Volatile/immunology , Host-Pathogen Interactions , Immunity, Innate , Lung/immunology , Mice , Mice, Inbred C57BL , Microbiota
9.
Int J Mol Sci ; 21(14)2020 Jul 11.
Article in English | MEDLINE | ID: mdl-32664466

ABSTRACT

Short-chain fatty acids (SCFA), bacterial metabolites released from dental biofilm, are supposed to target the oral epithelium. There is, however, no consensus on how SCFA affect the oral epithelial cells. The objective of the present study was to systematically review the available in vitro evidence of the impact of SCFA on human oral epithelial cells in the context of periodontal disease. A comprehensive electronic search using five databases along with a grey literature search was performed. In vitro studies that evaluated the effects of SCFA on human oral epithelial cells were eligible for inclusion. Risk of bias was assessed by the University of Bristol's tool for assessing risk of bias in cell culture studies. Certainty in cumulative evidence was evaluated using GRADE criteria (grading of recommendations assessment, development, and evaluation). Of 3591 records identified, 10 were eligible for inclusion. A meta-analysis was not possible due to the heterogeneity between the studies. The risk of bias across the studies was considered "serious" due to the presence of methodological biases. Despite these limitations, this review showed that SCFA negatively affect the viability of oral epithelial cells by activating a series of cellular events that includes apoptosis, autophagy, and pyroptosis. SCFA impair the integrity and presumably the transmigration of leucocytes through the epithelial layer by changing junctional and adhesion protein expression, respectively. SCFA also affect the expression of chemokines and cytokines in oral epithelial cells. Future research needs to identify the underlying signaling cascades and to translate the in vitro findings into preclinical models.


Subject(s)
Dysbiosis/complications , Epithelial Cells/drug effects , Fatty Acids, Volatile/adverse effects , Mouth Mucosa/microbiology , Periodontitis/etiology , Apoptosis/drug effects , Biofilms , Butyrates/pharmacology , Cell Adhesion/drug effects , Cell Shape , Dysbiosis/microbiology , Fatty Acids, Volatile/immunology , Humans , Microbiota , Mouth Mucosa/cytology , Periodontal Pocket/microbiology , Periodontitis/drug therapy , Sodium Bicarbonate/pharmacology , Sodium Bicarbonate/therapeutic use
10.
Int J Mol Sci ; 21(22)2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33227973

ABSTRACT

Inflammation is the key for the initiation and progression of atherosclerosis. Accumulating evidence has revealed that an altered gut microbiome (dysbiosis) triggers both local and systemic inflammation to cause chronic inflammatory diseases, including atherosclerosis. There have been some microbiome-relevant pro-inflammatory mechanisms proposed to link the relationships between dysbiosis and atherosclerosis such as gut permeability disruption, trigger of innate immunity from lipopolysaccharide (LPS), and generation of proatherogenic metabolites, such as trimethylamine N-oxide (TMAO). Meanwhile, immune responses, such as inflammasome activation and cytokine production, could reshape both composition and function of the microbiota. In fact, the immune system delicately modulates the interplay between microbiota and atherogenesis. Recent clinical trials have suggested the potential of immunomodulation as a treatment strategy of atherosclerosis. Here in this review, we present current knowledge regarding to the roles of microbiota in contributing atherosclerotic pathogenesis and highlight translational perspectives by discussing the mutual interplay between microbiota and immune system on atherogenesis.


Subject(s)
Atherosclerosis/immunology , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Immunity, Innate , Immunomodulation , Animals , Atherosclerosis/drug therapy , Atherosclerosis/microbiology , Atherosclerosis/pathology , Clinical Trials as Topic , Cytokines/immunology , Cytokines/metabolism , Disease Progression , Dysbiosis/drug therapy , Dysbiosis/microbiology , Dysbiosis/pathology , Fatty Acids, Volatile/immunology , Fatty Acids, Volatile/metabolism , Humans , Immunologic Factors/therapeutic use , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Methylamines/immunology , Methylamines/metabolism
11.
Curr Opin Lipidol ; 30(2): 125-133, 2019 04.
Article in English | MEDLINE | ID: mdl-30664014

ABSTRACT

PURPOSE OF REVIEW: Not all of the risk of cardiovascular disease can be explained by diet and genetics, and the human microbiome, which lies at the interface of these two factors, may help explain some of the unaccounted risk. This review examines some of the well established links between the microbiome and cardiovascular health, and proposes relatively unexplored associations. RECENT FINDINGS: Byproducts of microbial metabolism are associated with health and disease: Trimethylamine N oxide is associated with atherosclerosis; whereas short-chain fatty acids are associated with decreased inflammation and increased energy expenditure. More broadly, a large number of association studies have been conducted to explore the connections between bacterial taxa and metabolic syndrome. In contrast, the relationship between the microbiome and triglycerides levels remains poorly understood. SUMMARY: We suggest that deeper understanding of the molecular mechanisms that drive linkages between the microbiome and disease can be determined by replacing 16S rRNA gene sequencing with shotgun metagenomic sequencing or other functional approaches. Furthermore, to ensure translatability and reproducibility of research findings, a combination of multiple different complementary '-omic' approaches should be employed.


Subject(s)
Atherosclerosis/microbiology , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/immunology , Lipid Metabolism/immunology , Metabolic Syndrome/microbiology , Methylamines/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Bile Acids and Salts/immunology , Bile Acids and Salts/metabolism , Carnitine/immunology , Carnitine/metabolism , Choline/immunology , Choline/metabolism , Energy Metabolism/genetics , Energy Metabolism/immunology , Fatty Acids, Volatile/immunology , Gastrointestinal Microbiome/genetics , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Humans , Lipid Metabolism/genetics , Metabolic Syndrome/genetics , Metabolic Syndrome/immunology , Metabolic Syndrome/pathology , Methylamines/immunology , Methylamines/pharmacology , Phosphatidylcholines/immunology , Phosphatidylcholines/metabolism , RNA, Ribosomal, 16S/genetics , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Triglycerides/immunology , Triglycerides/metabolism
12.
Pharmacol Res ; 141: 521-529, 2019 03.
Article in English | MEDLINE | ID: mdl-30660825

ABSTRACT

There is increasing evidence that the intestinal microbiota plays a mechanistic role in the etiology of non-alcoholic fatty liver disease (NAFLD). Animal and human studies have linked small molecule metabolites produced by commensal bacteria in the gut contribute to not only intestinal inflammation, but also to hepatic inflammation. These immunomodulatory metabolites are capable of engaging host cellular receptors, and may mediate the observed association between gut dysbiosis and NAFLD. This review focuses on the effects and potential mechanisms of three specific classes of metabolites that synthesized or modified by gut bacteria: short chain fatty acids, amino acid catabolites, and bile acids. In particular, we discuss their role as ligands for cell surface and nuclear receptors regulating metabolic and inflammatory pathways in the intestine and liver. Studies reveal that the metabolites can both agonize and antagonize their cognate receptors to reduce or exacerbate liver steatosis and inflammation, and that the effects are metabolite- and context-specific. Further studies are warranted to more comprehensively understand bacterial metabolite-mediated gut-liver in NAFLD. This understanding could help identify novel therapeutics and therapeutic targets to intervene in the disease through the gut microbiota.


Subject(s)
Dysbiosis/complications , Dysbiosis/immunology , Gastrointestinal Microbiome , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/immunology , Amino Acids/immunology , Amino Acids/metabolism , Animals , Bacteria/immunology , Bacteria/metabolism , Bile Acids and Salts/immunology , Bile Acids and Salts/metabolism , Dysbiosis/metabolism , Fatty Acids, Volatile/immunology , Fatty Acids, Volatile/metabolism , Humans , Inflammation/etiology , Inflammation/immunology , Inflammation/metabolism , Non-alcoholic Fatty Liver Disease/metabolism
13.
Eur J Immunol ; 47(7): 1188-1199, 2017 07.
Article in English | MEDLINE | ID: mdl-28543188

ABSTRACT

Dysbiosis in gut microbiome has been shown to be associated with inflammatory and autoimmune diseases. Previous studies from our laboratory demonstrated the pivotal role played by CD44 in the regulation of EAE, a murine model of multiple sclerosis. In the current study, we determined whether these effects resulted from an alteration in gut microbiota and the short-chain fatty acid (SCFA) production in CD44 knockout (CD44KO) mice. Fecal transfer from naïve CD44KO but not C57BL/6 wild type (CD44WT) mice, into EAE-induced CD44WT mice, led to significant amelioration of EAE. High-throughput bacterial 16S rRNA gene sequencing, followed by clustering sequences into operational taxonomic units (OTUs) and biochemical analysis, revealed that EAE-induced CD44KO mice showed significant diversity, richness, and evenness when compared to EAE-induced CD44WT mice at the phylum level, with dominant Bacteroidetes (68.5%) and low Firmicutes (26.8%). Further, data showed a significant change in the abundance of SCFAs, propionic acid, and i-butyric acid in EAE-CD44KO compared to EAE-CD44WT mice. In conclusion, our results demonstrate that the attenuation of EAE seen following CD44 gene deletion in mice may result from alterations in the gut microbiota and SCFAs. Furthermore, our studies also demonstrate that the phenotype of gene knock-out animals may be shaped by gut microbiota.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Gastrointestinal Microbiome/immunology , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , Animals , Bacteroidetes/genetics , Bacteroidetes/immunology , Bacteroidetes/isolation & purification , Disease Models, Animal , Dysbiosis , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Fatty Acids, Volatile/immunology , Fecal Microbiota Transplantation , Feces/microbiology , Firmicutes/genetics , Firmicutes/immunology , Firmicutes/isolation & purification , Gastrointestinal Microbiome/genetics , Gene Deletion , Metagenomics , Mice , Mice, Inbred C57BL , Mice, Knockout , Propionates/metabolism , RNA, Ribosomal, 16S
14.
Int J Mol Sci ; 18(7)2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28654020

ABSTRACT

The incidence of inflammatory bowel disease (IBD) has increased considerably over the past few decades. In the present review, we discuss several disadvantages existing in the treatment of IBD and current understandings of the structures, sources, and natures of various kinds of non-starch polysaccharides (NSPs). Available evidences for the use of different sources of NSPs in IBD treatment both in vitro and in vivo are analyzed, including glucan from oat bran, mushroom, seaweed, pectin, gum, prebiotics, etc. Their potential mechanisms, especially their related molecular mechanism of protective action in the treatment and prevention of IBD, are also summarized, covering the anti-inflammation, immune-stimulating, and gut microbiota-modulating activities, as well as short-chain fatty acids (SCFAs) production, anti-oxidative stress accompanied with inflammation, the promotion of gastric epithelial cell proliferation and tissue healing, and the reduction of the absorption of toxins of NSPs, thus ameliorating the symptoms and reducing the reoccurrence rate of IBD. In summary, NSPs exhibit the potential to be promising agents for an adjuvant therapy and for the prevention of IBD. Further investigating of the crosstalk between immune cells, epithelial cells, and gut microorganisms in addition to evaluating the effects of different kinds and different molecular weights of NSPs will lead to well-designed clinical intervention trials and eventually improve the treatment and prevention of IBD.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Dietary Fiber/therapeutic use , Inflammatory Bowel Diseases/therapy , Polysaccharides/therapeutic use , Animals , Anti-Inflammatory Agents/chemistry , Antioxidants/chemistry , Antioxidants/therapeutic use , Dietary Fiber/analysis , Fatty Acids, Volatile/immunology , Gastrointestinal Microbiome , Humans , Inflammation/immunology , Inflammation/therapy , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Polysaccharides/chemistry
15.
Gastroenterology ; 145(2): 396-406.e1-10, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23665276

ABSTRACT

BACKGROUND & AIMS: Short-chain fatty acids (SCFAs), the most abundant microbial metabolites in the intestine, activate cells via G-protein-coupled receptors (GPRs), such as GPR41 and GPR43. We studied regulation of the immune response by SCFAs and their receptors in the intestines of mice. METHODS: Inflammatory responses were induced in GPR41(-/-), GPR43(-/-), and C57BL6 (control) mice by administration of ethanol; 2, 4, 6-trinitrobenzene sulfonic-acid (TNBS); or infection with Citrobacter rodentium. We examined the effects of C rodentium infection on control mice fed SCFAs and/or given injections of antibodies that delay the immune response. We also studied the kinetics of cytokine and chemokine production, leukocyte recruitment, intestinal permeability, and T-cell responses. Primary colon epithelial cells were isolated from GPR41(-/-), GPR43(-/-), and control mice; signaling pathways regulated by SCFAs were identified using immunohistochemical, enzyme-linked immunosorbent assay, and flow cytometry analyses. RESULTS: GPR41(-/-) and GPR43(-/-) mice had reduced inflammatory responses after administration of ethanol or TNBS compared with control mice, and had a slower immune response against C rodentium infection, clearing the bacteria more slowly. SCFAs activated intestinal epithelial cells to produce chemokines and cytokines in culture and mice after administration of ethanol, TNBS, or C rodentium. These processes required GPR41 and GPR43 and were required to recruit leukocytes and activate effector T cells in the intestine. GPR41 and GPR43 activated extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase signaling pathways in epithelial cells to induce production of chemokines and cytokines during immune responses. CONCLUSIONS: SCFAs activate GPR41 and GPR43 on intestinal epithelial cells, leading to mitogen-activated protein kinase signaling and rapid production of chemokines and cytokines. These pathways mediate protective immunity and tissue inflammation in mice.


Subject(s)
Colitis/immunology , Epithelial Cells/immunology , Fatty Acids, Volatile/immunology , Intestinal Mucosa/cytology , MAP Kinase Signaling System/immunology , Receptors, G-Protein-Coupled/immunology , Animals , Citrobacter rodentium , Disease Models, Animal , Enterobacteriaceae Infections/immunology , Ethanol/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Trinitrobenzenesulfonic Acid/immunology
16.
Microb Cell Fact ; 12: 71, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23876056

ABSTRACT

The human gut is one of the most complex ecosystems, composed of 1013-1014 microorganisms which play an important role in human health. In addition, some food products contain live bacteria which transit through our gastrointestinal tract and could exert beneficial effects on our health (known as probiotic effect). Among the numerous proposed health benefits attributed to commensal and probiotic bacteria, their capacity to interact with the host immune system is now well demonstrated. Currently, the use of recombinant lactic acid bacteria to deliver compounds of health interest is gaining importance as an extension of the probiotic concept. This review summarizes some of the recent findings and perspectives in the study of the crosstalk of both commensal and probiotic bacteria with the human host as well as the latest studies in recombinant commensal and probiotic bacteria. Our aim is to highlight the potential roles of recombinant bacteria in this ecosystem.


Subject(s)
Inflammatory Bowel Diseases/microbiology , Animals , Bacteria/immunology , Bacteria/metabolism , Dysbiosis , Fatty Acids, Volatile/immunology , Gastrointestinal Tract/microbiology , Humans , Inflammatory Bowel Diseases/immunology , Organisms, Genetically Modified/immunology , Organisms, Genetically Modified/metabolism , Probiotics
17.
Inflammation ; 45(1): 372-386, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34792688

ABSTRACT

Intestinal inflammation is a common disease which can further lead to inflammatory bowel disease and even intestinal cancer. The increasing focus has come to the role of short-chain fatty acid (SCFA) in various bowel diseases. Hence, this study was designed to explore the specific role of SCFA in intestinal inflammation. In vivo and in vitro models of intestinal inflammation were constructed by lipopolysaccharide (LPS) injection in mice and LPS treatment on intestinal epithelial cells. A possible regulatory mechanism involving SCFA, CCAAT enhancer-binding protein beta (CEBPB), microRNA-145 (miR-145), and dual-specificity phosphatase 6 (DUSP6) in intestinal inflammation was verified by ChIP assay and dual-luciferase reporter gene assay. To evaluate the effects of SCFA on LPS-treated intestinal epithelial cells, the expression of relevant genes and inflammatory factors (IL-6, TNF-α, and IL-1ß) were determined. Last, the role of SCFA in vivo was explored through the scoring of disease activity index (DAI) and observation of colonic histology of LPS-treated mice. SCFA decreased the CEBPB expression in mouse colon tissues and small intestine epithelial cells induced by LPS. Furthermore, CEBPB could bind to the miR-145 promoter to inhibit its expression, thereby promoting the expression of DUSP6. In addition, SCFA improved the DAI, colonic histology, and the expression of serum inflammatory factors in LPS-treated mice and cells, noting that SCFA alleviated intestinal inflammation in vitro and in vivo. To sum up, SCFA inhibited DUSP6 by upregulating miR-145 through CEBPB repression and thus prevented the development of intestinal inflammation.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Colitis/metabolism , Colon/metabolism , Dual Specificity Phosphatase 6/metabolism , Fatty Acids, Volatile/metabolism , Intestinal Mucosa/metabolism , MicroRNAs/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/immunology , Colitis/immunology , Colitis/pathology , Colon/immunology , Colon/pathology , Dual Specificity Phosphatase 6/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fatty Acids, Volatile/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/immunology
18.
Int Arch Allergy Immunol ; 156(3): 325-32, 2011.
Article in English | MEDLINE | ID: mdl-21720179

ABSTRACT

BACKGROUND: The present study was designed to compare the faecal microbiota and concentrations of faecal short-chain fatty acid and ammonia between healthy and cow's milk protein allergic (CMPA) infants. METHODS: The population comprised 92 infants aged 2-12 months who were nonallergic (n = 46) or diagnosed as having CMPA (n = 46). Faecal samples were analyzed by fluorescent in situ hybridization and flow cytometry, using a panel of 10 rRNA targeted group- and species-specific oligonucleotide probes. Acetic, propionic, butyric, isocaproic and branched-chain short fatty acids (BCSFA) were measured by gas-liquid chromatography, lactate by enzymatic reaction, and pH and ammonia levels were determined. RESULTS: CMPA infant faeces had significantly higher proportions of the Clostridium coccoides group and Atopobium cluster and a higher sum of the proportions of the different bacterial groups in comparison to healthy infant faeces. Faecal pH and ammonia did not significantly differ between CMPA and healthy infants. Faeces concentrations and percentages of butyric acid and BCSFA were higher in CMPA infants than in healthy infants. CONCLUSIONS: The findings clearly set a link between a dysbiosis in gut microbiota composition and the pathogenesis of CMPA. No single species or genus appeared to play an essential role, but dysbiosis led to biomarkers of CMPA among bacterial fermentation products.


Subject(s)
Fatty Acids, Volatile/analysis , Feces/chemistry , Feces/microbiology , Milk Hypersensitivity/immunology , Actinobacteria/isolation & purification , Ammonia/analysis , Butyric Acid/analysis , Clostridium/isolation & purification , Fatty Acids, Volatile/immunology , Female , Humans , Infant , Intestines/microbiology , Male , Metagenome , Milk Proteins/immunology
19.
Biomed Pharmacother ; 141: 111817, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34126349

ABSTRACT

In recent years, an overwhelming amount of evidence has positively recommended a significant role of microbiota in human health and disease. Microbiota also plays a crucial role in the initiation, preparation, and function of the host immune response. Recently, it has been shown that short-chain fatty acids (SCFAs) are the primary metabolites of the intestinal microbiota produced by anaerobic fermentation, which contributes to the host-pathogen interaction. SCFAs, such as propionate, acetate, and butyrate, are bacterial metabolites with immunomodulatory activity, and they are indispensable for the maintenance of homeostasis. Some evidence indicates that they are involved in the development of infections. In the present study, we provide the latest findings on the role of SCFAs in response to bacterial infections.


Subject(s)
Bacterial Infections/immunology , Fatty Acids, Volatile/pharmacology , Immunologic Factors/pharmacology , Microbiota , Animals , Fatty Acids, Volatile/immunology , Gastrointestinal Microbiome , Humans
20.
Cells ; 10(1)2021 01 10.
Article in English | MEDLINE | ID: mdl-33435197

ABSTRACT

Although the etiology of multiple sclerosis (MS) is still unknown, it is commonly accepted that environmental factors could contribute to the disease. The objective of this study was to analyze the humoral response to Epstein-Barr virus, human herpesvirus 6A/B and cytomegalovirus, and the levels of 25-hydroxyvitamin D (25(OH)D) and the three main short-chain fatty acids (SCFA), propionate (PA), butyrate (BA) and acetate (AA), in MS patients and healthy controls (HC) to understand how they could contribute to the pathogenesis of the disease. With this purpose, we analyzed the correlations among them and with different clinical variables and a wide panel of cell subsets. We found statistically significant differences for most of the environmental factors analyzed when we compared MS patients and HC, supporting their possible involvement in the disease. The strongest correlations with the clinical variables and the cell subsets analyzed were found for 25(OH)D and SCFAs levels. A correlation was also found between 25(OH)D and PA/AA ratio, and the interaction between these factors negatively correlated with interleukin 17 (IL-17)-producing CD4+ and CD8+ T cells in untreated MS patients. Therapies that simultaneously increase vitamin D levels and modify the proportion of SCFA could be evaluated in the future.


Subject(s)
Antibodies, Viral/immunology , Fatty Acids, Volatile/immunology , Herpesvirus 4, Human/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/virology , Vitamin D/metabolism , Adult , Case-Control Studies , Environment , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Male , Middle Aged , Vitamin D/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL