Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Annu Rev Immunol ; 35: 371-402, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28446062

ABSTRACT

Nutrition and the gut microbiome regulate many systems, including the immune, metabolic, and nervous systems. We propose that the host responds to deficiency (or sufficiency) of dietary and bacterial metabolites in a dynamic way, to optimize responses and survival. A family of G protein-coupled receptors (GPCRs) termed the metabolite-sensing GPCRs bind to various metabolites and transmit signals that are important for proper immune and metabolic functions. Members of this family include GPR43, GPR41, GPR109A, GPR120, GPR40, GPR84, GPR35, and GPR91. In addition, bile acid receptors such as GPR131 (TGR5) and proton-sensing receptors such as GPR65 show similar features. A consistent feature of this family of GPCRs is that they provide anti-inflammatory signals; many also regulate metabolism and gut homeostasis. These receptors represent one of the main mechanisms whereby the gut microbiome affects vertebrate physiology, and they also provide a link between the immune and metabolic systems. Insufficient signaling through one or more of these metabolite-sensing GPCRs likely contributes to human diseases such as asthma, food allergies, type 1 and type 2 diabetes, hepatic steatosis, cardiovascular disease, and inflammatory bowel diseases.


Subject(s)
Cardiovascular Diseases/immunology , Diabetes Mellitus, Type 1/immunology , Gastrointestinal Microbiome/immunology , Hypersensitivity/immunology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Diet , Homeostasis , Humans , Immunity , Receptors, G-Protein-Coupled/immunology
2.
Nat Immunol ; 18(5): 552-562, 2017 05.
Article in English | MEDLINE | ID: mdl-28346408

ABSTRACT

Gut dysbiosis might underlie the pathogenesis of type 1 diabetes. In mice of the non-obese diabetic (NOD) strain, we found that key features of disease correlated inversely with blood and fecal concentrations of the microbial metabolites acetate and butyrate. We therefore fed NOD mice specialized diets designed to release large amounts of acetate or butyrate after bacterial fermentation in the colon. Each diet provided a high degree of protection from diabetes, even when administered after breakdown of immunotolerance. Feeding mice a combined acetate- and butyrate-yielding diet provided complete protection, which suggested that acetate and butyrate might operate through distinct mechanisms. Acetate markedly decreased the frequency of autoreactive T cells in lymphoid tissues, through effects on B cells and their ability to expand populations of autoreactive T cells. A diet containing butyrate boosted the number and function of regulatory T cells, whereas acetate- and butyrate-yielding diets enhanced gut integrity and decreased serum concentration of diabetogenic cytokines such as IL-21. Medicinal foods or metabolites might represent an effective and natural approach for countering the numerous immunological defects that contribute to T cell-dependent autoimmune diseases.


Subject(s)
Acetates/metabolism , B-Lymphocytes/immunology , Butyrates/metabolism , Colon/metabolism , Diabetes Mellitus, Type 1/diet therapy , Dysbiosis/diet therapy , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity , B-Lymphocytes/microbiology , Cells, Cultured , Colon/pathology , Diet Therapy , Gastrointestinal Microbiome , Interleukins/blood , Mice , Mice, Inbred NOD , T-Lymphocytes, Regulatory/microbiology
4.
J Neurosci ; 43(37): 6460-6475, 2023 09 13.
Article in English | MEDLINE | ID: mdl-37596052

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder with poorly understood etiology. AD has several similarities with other "Western lifestyle" inflammatory diseases, where the gut microbiome and immune pathways have been associated. Previously, we and others have noted the involvement of metabolite-sensing GPCRs and their ligands, short-chain fatty acids (SCFAs), in protection of numerous Western diseases in mouse models, such as Type I diabetes and hypertension. Depletion of GPR43, GPR41, or GPR109A accelerates disease, whereas high SCFA yielding diets protect in mouse models. Here, we extended the concept that metabolite-sensing receptors and SCFAs may be a more common protective mechanism against Western diseases by studying their role in AD pathogenesis in the 5xFAD mouse model. Both male and female mice were included. Depletion of GPR41 and GPR43 accelerated cognitive decline and impaired adult hippocampal neurogenesis in 5xFAD and WT mice. Lack of fiber/SCFAs accelerated a memory deficit, whereas diets supplemented with high acetate and butyrate (HAMSAB) delayed cognitive decline in 5xFAD mice. Fiber intake impacted on microglial morphology in WT mice and microglial clustering phenotype in 5xFAD mice. Lack of fiber impaired adult hippocampal neurogenesis in both W and AD mice. Finally, maternal dietary fiber intake significantly affects offspring's cognitive functions in 5xFAD mice and microglial transcriptome in both WT and 5xFAD mice, suggesting that SCFAs may exert their effect during pregnancy and lactation. Together, metabolite-sensing GPCRs and SCFAs are essential for protection against AD, and reveal a new strategy for disease prevention.Significance Statement Alzheimer's disease (AD) is one of the most common neurodegenerative diseases; currently, there is no cure for AD. In our study, short-chain fatty acids and metabolite receptors play an important role in cognitive function and pathology in AD mouse model as well as in WT mice. SCFAs also impact on microglia transcriptome, and immune cell recruitment. Out study indicates the potential of specialized diets (supplemented with high acetate and butyrate) releasing high amounts of SCFAs to protect against disease.


Subject(s)
Alzheimer Disease , Microbiota , Female , Male , Pregnancy , Animals , Mice , Cognition , Dietary Fiber , Butyrates , Disease Models, Animal
6.
J Immunol ; 206(10): 2441-2452, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33941658

ABSTRACT

Intestinal barrier is essential for dietary products and microbiota compartmentalization and therefore gut homeostasis. When this barrier is broken, cecal content overflows into the peritoneal cavity, leading to local and systemic robust inflammatory response, characterizing peritonitis and sepsis. It has been shown that IL-1ß contributes with inflammatory storm during peritonitis and sepsis and its inhibition has beneficial effects to the host. Therefore, we investigated the mechanisms underlying IL-1ß secretion using a widely adopted murine model of experimental peritonitis. The combined injection of sterile cecal content (SCC) and the gut commensal bacteria Bacteroides fragilis leads to IL-1ß-dependent peritonitis, which was mitigated in mice deficient in NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome components. Typically acting as a damage signal, SCC, but not B. fragilis, activates canonical pathway of NLRP3 promoting IL-1ß secretion in vitro and in vivo. Strikingly, absence of fiber in the SCC drastically reduces IL-1ß production, whereas high-fiber SCC conversely increases this response in an NLRP3-dependent manner. In addition, NLRP3 was also required for IL-1ß production induced by purified dietary fiber in primed macrophages. Extending to the in vivo context, IL-1ß-dependent peritonitis was worsened in mice injected with B. fragilis and high-fiber SCC, whereas zero-fiber SCC ameliorates the pathology. Corroborating with the proinflammatory role of dietary fiber, IL-1R-deficient mice were protected from peritonitis induced by B. fragilis and particulate bran. Overall, our study highlights a function, previously unknown, for dietary fibers in fueling peritonitis through NLRP3 activation and IL-1ß secretion outside the gut.


Subject(s)
Bacteroides Infections/immunology , Bacteroides fragilis/immunology , Dietary Fiber/adverse effects , Inflammasomes/metabolism , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Peritonitis/immunology , Animals , Bacteroides Infections/microbiology , Diet , Dietary Fiber/administration & dosage , Disease Models, Animal , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Peritonitis/microbiology , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Signal Transduction/drug effects , Signal Transduction/immunology
7.
Int J Mol Sci ; 24(5)2023 Feb 25.
Article in English | MEDLINE | ID: mdl-36901964

ABSTRACT

Butyrate produced by the gut microbiota has beneficial effects on metabolism and inflammation. Butyrate-producing bacteria are supported by diets with a high fiber content, such as high-amylose maize starch (HAMS). We investigated the effects of HAMS- and butyrylated HAMS (HAMSB)-supplemented diets on glucose metabolism and inflammation in diabetic db/db mice. Mice fed HAMSB had 8-fold higher fecal butyrate concentration compared to control diet-fed mice. Weekly analysis of fasting blood glucose showed a significant reduction in HAMSB-fed mice when the area under the curve for all five weeks was analyzed. Following treatment, fasting glucose and insulin analysis showed increased homeostatic model assessment (HOMA) insulin sensitivity in the HAMSB-fed mice. Glucose-stimulated insulin release from isolated islets did not differ between the groups, while insulin content was increased by 36% in islets of the HAMSB-fed mice. Expression of insulin 2 was also significantly increased in islets of the HAMSB-fed mice, while no difference in expression of insulin 1, pancreatic and duodenal homeobox 1, MAF bZIP transcription factor A and urocortin 3 between the groups was observed. Hepatic triglycerides in the livers of the HAMSB-fed mice were significantly reduced. Finally, mRNA markers of inflammation in liver and adipose tissue were reduced in mice fed HAMSB. These findings suggest that HAMSB-supplemented diet improves glucose metabolism in the db/db mice, and reduces inflammation in insulin-sensitive tissues.


Subject(s)
Butyrates , Starch , Rats , Mice , Animals , Rats, Sprague-Dawley , Amylose/metabolism , Inflammation , Liver/metabolism , Mice, Inbred Strains , Insulin , Homeostasis , Glucose , Mice, Inbred C57BL , Blood Glucose/metabolism
8.
Exp Dermatol ; 31(12): 1872-1880, 2022 12.
Article in English | MEDLINE | ID: mdl-36054650

ABSTRACT

Altered gut microbiota composition has been observed in individuals with hidradenitis suppurutiva (HS) and many other inflammatory diseases, including obesity, type 1 and type 2 diabetes. Here, we addressed whether adalimumab, a systemic anti-inflammatory therapy, may impact the microbiota biochemical profile, particularly on beneficial metabolites such as short-chain fatty acids (SCFAs). We conducted an observational single-arm pilot trial to assess gut microbiota composition by 16S rRNA gene sequence analysis and to detect metabolite signatures by gas chromatography in stool samples from participants with HS prior to and 12 weeks after commencing adalimumab therapy. HS individuals that better responded to adalimumab treatment showed a shift in the composition and function of the gut microbiota with significantly increased SCFA acetate and propionate compared to age, gender and BMI-matched healthy controls. A positive correlation was observed between propionate with Prevotella sp and Faecalibacterium prausnitsii. Increased SCFAs, changes in gut microbiota composition, function and metabolic profile following 12 weeks of adalimumab suggest that targeting SCFAs may be considered a potential biomarker to be evaluated as a complementary protective factor or as a diagnostically relevant signal in HS.


Subject(s)
Diabetes Mellitus, Type 2 , Hidradenitis Suppurativa , Humans , Hidradenitis Suppurativa/drug therapy , Adalimumab/therapeutic use , RNA, Ribosomal, 16S/genetics , Propionates/therapeutic use , Fatty Acids, Volatile/metabolism
9.
Adv Exp Med Biol ; 1307: 499-519, 2021.
Article in English | MEDLINE | ID: mdl-32193865

ABSTRACT

Diet-microbiota related inflammatory conditions such as obesity, autoimmune type 1 diabetes (T1D), type 2 diabetes (T2D), cardiovascular disease (CVD) and gut infections have become a stigma in Western societies and developing nations. This book chapter examines the most relevant pre-clinical and clinical studies about diet-gut microbiota approaches as an alternative therapy for diabetes. We also discuss what we and others have extensively investigated- the power of dietary short-chain fatty acids (SCFAs) technology that naturally targets the gut microbiota as an alternative method to prevent and treat diabetes and its related complications.


Subject(s)
Diabetes Mellitus, Type 2/diet therapy , Fatty Acids, Volatile/administration & dosage , Gastrointestinal Microbiome , Immunotherapy , Diet , Humans
10.
FASEB J ; 33(11): 11894-11908, 2019 11.
Article in English | MEDLINE | ID: mdl-31366236

ABSTRACT

Butyrate is a short-chain fatty acid derived from the metabolism of indigestible carbohydrates by the gut microbiota. Butyrate contributes to gut homeostasis, but it may also control inflammatory responses and host physiology in other tissues. Butyrate inhibits histone deacetylases, thereby affecting gene transcription, and also signals through the metabolite-sensing G protein receptor (GPR)109a. We produced an mAb to mouse GPR109a and found high expression on podocytes in the kidney. Wild-type and Gpr109a-/- mice were induced to develop nephropathy by a single injection of Adriamycin and treated with sodium butyrate or high butyrate-releasing high-amylose maize starch diet. Butyrate improved proteinuria by preserving podocyte at glomerular basement membrane and attenuated glomerulosclerosis and tissue inflammation. This protective phenotype was associated with increased podocyte-related proteins and a normalized pattern of acetylation and methylation at promoter sites of genes essential for podocyte function. We found that GPR109a is expressed by podocytes, and the use of Gpr109a-/- mice showed that the protective effects of butyrate depended on GPR109a expression. A prebiotic diet that releases high amounts of butyrate also proved highly effective for protection against kidney disease. Butyrate and GPR109a play a role in the pathogenesis of kidney disease and provide one of the important molecular connections between diet, the gut microbiota, and kidney disease.-Felizardo, R. J. F., de Almeida, D. C., Pereira, R. L., Watanabe, I. K. M., Doimo, N. T. S., Ribeiro, W. R., Cenedeze, M. A., Hiyane, M. I., Amano, M. T., Braga, T. T., Ferreira, C. M., Parmigiani, R. B., Andrade-Oliveira, V., Volpini, R. A., Vinolo, M. A. R., Mariño, E., Robert, R., Mackay, C. R., Camara, N. O. S. Gut microbial metabolite butyrate protects against proteinuric kidney disease through epigenetic- and GPR109a-mediated mechanisms.


Subject(s)
Butyrates/pharmacology , Epigenesis, Genetic , Gastrointestinal Microbiome/physiology , Kidney Diseases/prevention & control , Proteinuria/prevention & control , Receptors, G-Protein-Coupled/genetics , Animals , Bacteria/metabolism , Butyrates/metabolism , Cells, Cultured , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Podocytes/drug effects , Podocytes/metabolism , Protective Agents/metabolism , Protective Agents/pharmacology , Receptors, G-Protein-Coupled/metabolism
11.
Immunol Rev ; 245(1): 164-76, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22168419

ABSTRACT

Certain autoimmune diseases as well as asthma have increased in recent decades, particularly in developed countries. The hygiene hypothesis has been the prevailing model to account for this increase; however, epidemiology studies also support the contribution of diet and obesity to inflammatory diseases. Diet affects the composition of the gut microbiota, and recent studies have identified various molecules and mechanisms that connect diet, the gut microbiota, and immune responses. Herein, we discuss the effects of microbial metabolites, such as short chain fatty acids, on epithelial integrity as well as immune cell function. We propose that dysbiosis contributes to compromised epithelial integrity and disrupted immune tolerance. In addition, dietary molecules affect the function of immune cells directly, particularly through lipid G-protein coupled receptors such as GPR43.


Subject(s)
Bacteria/immunology , Bacterial Infections/immunology , Fatty Acids/immunology , Immune System Diseases/immunology , Intestinal Mucosa/immunology , Animals , Bacteria/metabolism , Bacterial Infections/complications , Bacterial Infections/microbiology , Diet , Dietary Supplements , Fatty Acids/metabolism , Humans , Immune System Diseases/etiology , Immune System Diseases/microbiology , Immune Tolerance , Inflammation/immunology , Inflammation/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Receptors, G-Protein-Coupled/metabolism
12.
Eur J Immunol ; 44(4): 983-93, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24435807

ABSTRACT

Targeting the BAFF/APRIL system has shown to be effective in preventing T-cell dependent autoimmune disease in the NOD mouse, a spontaneous model of type 1 diabetes. In this study we generated BAFF-deficient NOD mice to examine how BAFF availability would influence T-cell responses in vivo and the development of spontaneous diabetes. BAFF-deficient NOD mice which lack mature B cells, were protected from diabetes and showed delayed rejection of an allogeneic islet graft. Diabetes protection correlated with a failure to expand pathogenic IGRP-reactive CD8(+) T cells, which were maintained in the periphery at correspondingly low levels. Adoptive transfer of IGRP-reactive CD8(+) T cells with B cells into BAFF-deficient NOD mice enhanced IGRP-reactive CD8(+) T-cell expansion. Furthermore, when provoked with cyclophosphamide, or transferred to a secondary lymphopenic host, the latent pool of self-reactive T cells resident in BAFF-deficient NOD mice could elicit beta cell destruction. We conclude that lack of BAFF prevents the procurement of B-cell-dependent help necessary for the emergence of destructive diabetes. Indeed, treatment of NOD mice with the BAFF-blocking compound, BR3-Fc, resulted in a delayed onset and reduced incidence of diabetes.


Subject(s)
Autoimmunity/immunology , B-Cell Activating Factor/immunology , B-Lymphocytes/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Autoimmunity/genetics , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Female , Flow Cytometry , Glucose-6-Phosphatase/immunology , Glucose-6-Phosphatase/metabolism , Graft Rejection/genetics , Graft Rejection/immunology , Graft Survival/genetics , Graft Survival/immunology , Immunophenotyping , Islets of Langerhans Transplantation/methods , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , T-Lymphocytes/metabolism , Time Factors
13.
J Immunol ; 191(1): 97-109, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23740954

ABSTRACT

Marginal zone (MZ) B cells are an innate-like population that oscillates between MZ and follicular areas of the splenic white pulp. Differentiation of B cells into the MZ subset is governed by BCR signal strength and specificity, NF-κB activation through the B cell-activating factor belonging to the TNF family (BAFF) receptor, Notch2 signaling, and migration signals mediated by chemokine, integrin, and sphingosine-1-phosphate receptors. An imbalance in splenic B cell development resulting in expansion of the MZ subset has been associated with autoimmune pathogenesis in various murine models. One example is the NOD inbred mouse strain, in which MZ B cell expansion has been linked to development of type 1 diabetes and Sjögren's syndrome. However, the cause of MZ B cell expansion in this strain remains poorly understood. We have determined that increased MZ B cell development in NOD mice is independent of T cell autoimmunity, BCR specificity, BCR signal strength, and increased exposure to BAFF. Rather, mixed bone marrow chimeras showed that the factor(s) responsible for expansion of the NOD MZ subset is B cell intrinsic. Analysis of microarray expression data indicated that NOD MZ and precursor transitional 2-MZ subsets were particularly dysregulated for genes controlling cellular trafficking, including Apoe, Ccbp2, Cxcr7, Lgals1, Pla2g7, Rgs13, S1pr3, Spn, Bid, Cd55, Prf1, and Tlr3. Furthermore, these B cell subsets exhibited an increased steady state dwell time within splenic MZ areas. Our data therefore reveal that precursors of mature B cells in NOD mice exhibit an altered migration set point, allowing increased occupation of the MZ, a niche favoring MZ B cell differentiation.


Subject(s)
B-Cell Activating Factor/physiology , B-Lymphocyte Subsets/immunology , Cell Differentiation/immunology , Receptor, Notch2/physiology , Receptors, Antigen, B-Cell/physiology , Receptors, Lysosphingolipid/physiology , Spleen/immunology , Animals , B-Cell Activating Factor/deficiency , B-Cell Activation Factor Receptor/physiology , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/pathology , Cell Differentiation/genetics , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred NOD , Mice, Transgenic , Proprotein Convertases/physiology , Serine Endopeptidases/physiology , Spleen/metabolism , Spleen/pathology
14.
Trends Immunol ; 32(6): 287-94, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21531625

ABSTRACT

B cells play a pathogenic role as antigen-presenting cells and autoantibody secretors in the lead up to T cell-mediated autoimmune destruction of insulin-producing ß cells in type 1 diabetes (T1D). This has led to significant interest in the use of B cell depletion therapies as a treatment for T1D. In this review, we compare results from five recent studies that used distinct B cell-depleting agents and protocols to successfully prevent and even reverse T1D in the non-obese diabetic (NOD) mouse model. We discuss how information gained from animal studies could be used to improve on the positive outcomes of a completed phase II clinical trial of the B cell-depleting drug rituximab in humans with recent-onset T1D.


Subject(s)
B-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Clinical Trials, Phase II as Topic , Humans , Kinetics , Rituximab
15.
iScience ; 27(1): 108694, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38213620

ABSTRACT

An altered gut microbiota is associated with type 1 diabetes (T1D), affecting the production of short-chain fatty acids (SCFA) and glucose homeostasis. We previously demonstrated that enhancing serum acetate and butyrate using a dietary supplement (HAMSAB) improved glycemia in non-obese diabetic (NOD) mice and patients with established T1D. The effects of SCFA on immune-infiltrated islet cells remain to be clarified. Here, we performed single-cell RNA sequencing on islet cells from NOD mice fed an HAMSAB or control diet. HAMSAB induced a regulatory gene expression profile in pancreas-infiltrated immune cells. Moreover, HAMSAB maintained the expression of ß-cell functional genes and decreased cellular stress. HAMSAB-fed mice showed preserved pancreatic endocrine cell identity, evaluated by decreased numbers of poly-hormonal cells. Finally, SCFA increased insulin levels in human ß-like cells and improved transplantation outcome in NOD/SCID mice. Our findings support the use of metabolite-based diet as attractive approach to improve glucose control in T1D.

16.
Trends Mol Med ; 29(11): 912-925, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37730461

ABSTRACT

Recent research has confirmed the strong connection between imbalances in the oral and gut microbiome (oral-gut dysbiosis), periodontitis, and inflammatory conditions such as diabetes, Alzheimer's disease, and cardiovascular diseases. Microbiome modulation is crucial for preventing and treating several autoimmune and inflammatory diseases, including periodontitis. However, the causal relationships between the microbiome and its derived metabolites that mediate periodontitis and chronic inflammation constitute a notable knowledge gap. Here we review the mechanisms involved in the microbiome-host crosstalk, and describe novel precision medicine for the control of systemic inflammation. As microbiome-targeted therapies begin to enter clinical trials, the success of these approaches relies upon understanding these reciprocal microbiome-host interactions, and it may provide new therapeutic avenues to reduce the risk of periodontitis-associated diseases.

17.
Antioxidants (Basel) ; 11(4)2022 Mar 25.
Article in English | MEDLINE | ID: mdl-35453317

ABSTRACT

Geraniin, an ellagitannin, has ameliorative properties against high-fat diet (HFD)-induced metabolic syndrome. Since geraniin has poor bioavailability, we hypothesised the interaction of this compound with gut microbiota as the main mechanism for improving metabolic aberrations. Male Sprague Dawley rats were divided into normal diet (ND)- and HFD-fed animals and treated with geraniin and an enriched extract of geraniin (GEE). We observed that 5 mg geraniin and 115 mg GEE supplementation significantly attenuated glucose intolerance, lipopolysaccharide-binding protein, total cholesterol, triacylglyceride, and low-density lipoprotein; improved insulin sensitivity; and significantly increased adiponectin and hepatic PPARα expression. Although geraniin and GEE did not significantly alter the gut microbial composition, we found an increment in the relative abundance of a few butyrate producers such as Alloprevotella, Blautia, Lachnospiraceae NK4A136 group, and Clostridium sensu stricto 1. Geraniin and its enriched extract's ability to ameliorate metabolic syndrome parameters while positively affecting the growth of butyrate-producing bacteria suggests its potential prebiotic role.

18.
Microbiome ; 10(1): 9, 2022 01 19.
Article in English | MEDLINE | ID: mdl-35045871

ABSTRACT

BACKGROUND: Short-chain fatty acids (SCFAs) produced by the gut microbiota have beneficial anti-inflammatory and gut homeostasis effects and prevent type 1 diabetes (T1D) in mice. Reduced SCFA production indicates a loss of beneficial bacteria, commonly associated with chronic autoimmune and inflammatory diseases, including T1D and type 2 diabetes. Here, we addressed whether a metabolite-based dietary supplement has an impact on humans with T1D. We conducted a single-arm pilot-and-feasibility trial with high-amylose maize-resistant starch modified with acetate and butyrate (HAMSAB) to assess safety, while monitoring changes in the gut microbiota in alignment with modulation of the immune system status. RESULTS: HAMSAB supplement was administered for 6 weeks with follow-up at 12 weeks in adults with long-standing T1D. Increased concentrations of SCFA acetate, propionate, and butyrate in stools and plasma were in concert with a shift in the composition and function of the gut microbiota. While glucose control and insulin requirements did not change, subjects with the highest SCFA concentrations exhibited the best glycemic control. Bifidobacterium longum, Bifidobacterium adolescentis, and vitamin B7 production correlated with lower HbA1c and basal insulin requirements. Circulating B and T cells developed a more regulatory phenotype post-intervention. CONCLUSION: Changes in gut microbiota composition, function, and immune profile following 6 weeks of HAMSAB supplementation were associated with increased SCFAs in stools and plasma. The persistence of these effects suggests that targeting dietary SCFAs may be a mechanism to alter immune profiles, promote immune tolerance, and improve glycemic control for the treatment of T1D. TRIAL REGISTRATION: ACTRN12618001391268. Registered 20 August 2018, https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=375792 Video Abstract.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Microbiota , Animals , Diabetes Mellitus, Type 2/microbiology , Dietary Supplements , Fatty Acids, Volatile , Humans , Mice
19.
J Immunol ; 182(2): 793-801, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19124722

ABSTRACT

The cytokine B cell activation factor of the TNF family (BAFF) is considered to perform a proinflammatory function. This paradigm is particularly true for B cell-dependent immune responses; however the exact role for BAFF in regulating T cell immunity is ill-defined. To directly assess the effect of BAFF upon T cells, we analyzed T cell-dependent immune responses in BAFF-transgenic (Tg) mice. We found that T cell responses in BAFF-Tg mice are profoundly compromised, as indicated by their acceptance of islet allografts and delayed skin graft rejection. However, purified BAFF-Tg effector T cells could reject islet allografts with a normal kinetic, suggesting that the altered response did not relate to a defect in T cell function per se. Rather, we found that BAFF-Tg mice harbored an increased number of peripheral CD4+Foxp3+ T cells. A large proportion of the BAFF-expanded CD4+CD25+Foxp3+ regulatory T cells (Tregs) were CD62LlowCD103high and ICAM-1high, a phenotype consistent with an ability to home to inflammatory sites and prevent T cell effector responses. Indeed, depletion of the endogenous BAFF-Tg Tregs allowed allograft rejection to proceed, demonstrating that the increased Tregs were responsible for preventing alloimmunity. The ability of BAFF to promote Treg expansion was not T cell intrinsic, as Tregs did not express high levels of BAFF receptor 3, nor did excessive BAFF trigger NF-kappaB2 processing in Tregs. In contrast, we found that BAFF engendered Treg expansion through an indirect, B cell-dependent mechanism. Thus, under certain conditions, BAFF can play a surprising anti-inflammatory role in T cell biology by promoting the expansion of Treg cells.


Subject(s)
B-Cell Activating Factor/genetics , Cell Proliferation , Forkhead Transcription Factors/biosynthesis , Graft Rejection/prevention & control , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Cells, Cultured , Coculture Techniques , Graft Rejection/genetics , Graft Rejection/immunology , Graft Survival/genetics , Graft Survival/immunology , Islets of Langerhans Transplantation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Skin Transplantation/immunology , Transplantation Tolerance/genetics
20.
J Neurotrauma ; 38(18): 2610-2621, 2021 09 15.
Article in English | MEDLINE | ID: mdl-33957773

ABSTRACT

Traumatic brain injury (TBI) alters microbial populations present in the gut, which may impact healing and tissue recovery. However, the duration and impact of these changes on outcome from TBI are unknown. Short-chain fatty acids (SCFAs), produced by bacterial fermentation of dietary fiber, are important signaling molecules in the microbiota gut-brain axis. We hypothesized that TBI would lead to a sustained reduction in SCFA producing bacteria, fecal SCFAs concentration, and administration of soluble SCFAs would improve functional outcome after TBI. Adult mice (n = 10) had the controlled cortical impact (CCI) model of TBI performed (6 m/sec, 2-mm depth, 50-msec dwell). Stool samples were collected serially until 28 days after CCI and analyzed for SCFA concentration by high-performance liquid chromatography-mass spectrometry/mass spectrometry and microbiome analyzed by 16S gene sequencing. In a separate experiment, mice (n = 10/group) were randomized 2 weeks before CCI to standard drinking water or water supplemented with the SCFAs acetate (67.5 mM), propionate (25.9 mM), and butyrate (40 mM). Morris water maze performance was assessed on post-injury Days 14-19. Alpha diversity remained stable until 72 h, at which point a decline in diversity was observed without recovery out to 28 days. The taxonomic composition of post-TBI fecal samples demonstrated depletion of bacteria from Lachnospiraceae, Ruminococcaceae, and Bacteroidaceae families, and enrichment of bacteria from the Verrucomicrobiaceae family. Analysis from paired fecal samples revealed a reduction in total SCFAs at 24 h and 28 days after TBI. Acetate, the most abundant SCFA detected in the fecal samples, was reduced at 7 days and 28 days after TBI. SCFA administration improved spatial learning after TBI versus standard drinking water. In conclusion, TBI is associated with reduced richness and diversity of commensal microbiota in the gut and a reduction in SCFAs detected in stool. Supplementation of soluble SCFAs improves spatial learning after TBI.


Subject(s)
Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/psychology , Dysbiosis/etiology , Fatty Acids, Volatile/metabolism , Feces/chemistry , Nervous System Diseases/etiology , Nervous System Diseases/psychology , Animals , Brain Injuries, Traumatic/metabolism , Brain-Gut Axis , Dietary Supplements , Fatty Acids, Volatile/chemistry , Fatty Acids, Volatile/pharmacology , Feces/microbiology , Gastrointestinal Microbiome , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Nervous System Diseases/metabolism , Psychomotor Performance/drug effects , RNA, Ribosomal, 16S/genetics , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL