Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell ; 187(12): 2952-2968.e13, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38795705

ABSTRACT

Recent studies suggest that human-associated bacteria interact with host-produced steroids, but the mechanisms and physiological impact of such interactions remain unclear. Here, we show that the human gut bacteria Gordonibacter pamelaeae and Eggerthella lenta convert abundant biliary corticoids into progestins through 21-dehydroxylation, thereby transforming a class of immuno- and metabo-regulatory steroids into a class of sex hormones and neurosteroids. Using comparative genomics, homologous expression, and heterologous expression, we identify a bacterial gene cluster that performs 21-dehydroxylation. We also uncover an unexpected role for hydrogen gas production by gut commensals in promoting 21-dehydroxylation, suggesting that hydrogen modulates secondary metabolism in the gut. Levels of certain bacterial progestins, including allopregnanolone, better known as brexanolone, an FDA-approved drug for postpartum depression, are substantially increased in feces from pregnant humans. Thus, bacterial conversion of corticoids into progestins may affect host physiology, particularly in the context of pregnancy and women's health.


Subject(s)
Gastrointestinal Microbiome , Glucocorticoids , Hydrogen , Progestins , Humans , Progestins/metabolism , Hydrogen/metabolism , Female , Glucocorticoids/metabolism , Pregnancy , Animals , Multigene Family , Feces/microbiology , Pregnanolone/metabolism , Mice
2.
Nat Chem Biol ; 20(4): 410-421, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38347214

ABSTRACT

Recent studies have demonstrated that metabolites produced by commensal bacteria causally influence health and disease. The sulfated metabolome is one class of molecules that has recently come to the forefront due to efforts to understand the role of these metabolites in host-microbiome interactions. Sulfated compounds have canonically been classified as waste products; however, studies have revealed a variety of physiological roles for these metabolites, including effects on host metabolism, immune response and neurological function. Moreover, recent research has revealed that commensal bacteria either chemically modify or synthesize a variety of sulfated compounds. In this Review, we explore how host-microbiome collaborative metabolism transforms the sulfated metabolome. We describe bacterial and mammalian enzymes that sulfonate and desulfate biologically relevant carbohydrates, amino acid derivatives and cholesterol-derived metabolites. We then discuss outstanding questions and future directions in the field, including potential roles of sulfated metabolites in disease detection, prevention and treatment. We hope that this Review inspires future research into sulfated compounds and their effects on physiology.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Mammals , Metabolome , Sulfates
3.
Nat Microbiol ; 7(9): 1404-1418, 2022 09.
Article in English | MEDLINE | ID: mdl-35982310

ABSTRACT

Members of the human gut microbiome enzymatically process many bioactive molecules in the gastrointestinal tract. Most gut bacterial modifications characterized so far are hydrolytic or reductive in nature. Here we report that abundant human gut bacteria from the phylum Bacteroidetes perform conjugative modifications by selectively sulfonating steroidal metabolites. While sulfonation is a ubiquitous biochemical modification, this activity has not yet been characterized in gut microbes. Using genetic and biochemical approaches, we identify a widespread biosynthetic gene cluster that encodes both a sulfotransferase (BtSULT, BT0416) and enzymes that synthesize the sulfonate donor adenosine 3'-phosphate-5'-phosphosulfate (PAPS), including an APS kinase (CysC, BT0413) and an ATP sulfurylase (CysD and CysN, BT0414-BT0415). BtSULT selectively sulfonates steroidal metabolites with a flat A/B ring fusion, including cholesterol. Germ-free mice monocolonized with Bacteroides thetaiotaomicron ΔBT0416 exhibited reduced gastrointestinal levels of cholesterol sulfate (Ch-S) compared with wild-type B. thetaiotaomicron-colonized mice. The presence of BtSULT and BtSULT homologues in bacteria inhibited leucocyte migration in vitro and in vivo, and abundances of cluster genes were significantly reduced in patients with inflammatory bowel disease. Together, these data provide a mechanism by which gut bacteria sulfonate steroidal metabolites and suggest that these compounds can modulate immune cell trafficking in the host.


Subject(s)
Bacteroides thetaiotaomicron , Biosynthetic Pathways , Animals , Bacteria , Gastrointestinal Tract , Humans , Mice , Sulfate Adenylyltransferase
4.
Sci Adv ; 8(34): eabo2794, 2022 08 26.
Article in English | MEDLINE | ID: mdl-36026454

ABSTRACT

Altered host-microbe interactions and increased intestinal permeability have been implicated in disease pathogenesis. However, the mechanisms by which intestinal microbes affect epithelial barrier integrity remain unclear. Here, we investigate the impact of bacterial metabolism of host-produced bile acid (BA) metabolites on epithelial barrier integrity. We observe that rats fed a choline-deficient, l-amino acid-defined, high-fat diet (CDAHFD) exhibit reduced intestinal abundance of host-produced conjugated BAs at early time points, coinciding with increased gut permeability. We show that in vitro, conjugated BAs protect gut epithelial monolayers from damage caused by bacterially produced unconjugated BAs through micelle formation. We then demonstrate that inhibition of bacterial BA deconjugation with a small-molecule inhibitor prevents the development of pathologic intestinal permeability and hepatic inflammation in CDAHFD-fed rats. Our study identifies a signaling-independent, physicochemical mechanism for conjugated BA-mediated protection of epithelial barrier function and suggests that rational manipulation of microbial BA metabolism could be leveraged to regulate gut barrier integrity.


Subject(s)
Bile Acids and Salts , Gastrointestinal Microbiome , Animals , Liver , Micelles , Permeability , Rats
5.
Cell Host Microbe ; 30(4): 435-438, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35421338

ABSTRACT

Here, I reflect on my trajectory from a graduate student in organic chemistry to an early-career scientist in the microbiome field. I discuss strategies for discovering microbiome-derived molecules and their activities, and I contemplate how we will uncover which of the molecules we identify are responsible for driving host phenotypes.


Subject(s)
Microbiota , Humans
6.
Nature ; 603(7903): 907-912, 2022 03.
Article in English | MEDLINE | ID: mdl-35296854

ABSTRACT

The microbiota modulates gut immune homeostasis. Bacteria influence the development and function of host immune cells, including T helper cells expressing interleukin-17A (TH17 cells). We previously reported that the bile acid metabolite 3-oxolithocholic acid (3-oxoLCA) inhibits TH17 cell differentiation1. Although it was suggested that gut-residing bacteria produce 3-oxoLCA, the identity of such bacteria was unknown, and it was unclear whether 3-oxoLCA and other immunomodulatory bile acids are associated with inflammatory pathologies in humans. Here we identify human gut bacteria and corresponding enzymes that convert the secondary bile acid lithocholic acid into 3-oxoLCA as well as the abundant gut metabolite isolithocholic acid (isoLCA). Similar to 3-oxoLCA, isoLCA suppressed TH17 cell differentiation by inhibiting retinoic acid receptor-related orphan nuclear receptor-γt, a key TH17-cell-promoting transcription factor. The levels of both 3-oxoLCA and isoLCA and the 3α-hydroxysteroid dehydrogenase genes that are required for their biosynthesis were significantly reduced in patients with inflammatory bowel disease. Moreover, the levels of these bile acids were inversely correlated with the expression of TH17-cell-associated genes. Overall, our data suggest that bacterially produced bile acids inhibit TH17 cell function, an activity that may be relevant to the pathophysiology of inflammatory disorders such as inflammatory bowel disease.


Subject(s)
Bacteria , Bile Acids and Salts , Inflammatory Bowel Diseases , Bacteria/metabolism , Cell Differentiation , Gastrointestinal Tract/microbiology , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Interleukin-17 , Lithocholic Acid/metabolism , Lithocholic Acid/pharmacology , Th17 Cells
7.
Nat Chem Biol ; 17(10): 1046-1056, 2021 10.
Article in English | MEDLINE | ID: mdl-34552222

ABSTRACT

Human-associated microorganisms play a vital role in human health, and microbial imbalance has been linked to a wide range of disease states. In this Review, we explore recent efforts to progress from correlative studies that identify microorganisms associated with human disease to experiments that establish causal relationships between microbial products and host phenotypes. We propose that successful efforts to uncover phenotypes often follow a chain of evidence that proceeds from (1) association studies; to (2) observations in germ-free animals and antibiotic-treated animals and humans; to (3) fecal microbiota transplants (FMTs); to (4) identification of strains; and then (5) molecules that elicit a phenotype. Using this experimental 'funnel' as our guide, we explore how the microbiota contributes to metabolic disorders and hypertension, infections, and neurological conditions. We discuss the potential to use FMTs and microbiota-inspired therapies to treat human disease as well as the limitations of these approaches.


Subject(s)
Communicable Diseases/microbiology , Host Microbial Interactions/physiology , Microbiota/physiology , Animals , Anti-Infective Agents/pharmacology , Fecal Microbiota Transplantation , Germ-Free Life , Humans
8.
Nat Metab ; 3(9): 1228-1241, 2021 09.
Article in English | MEDLINE | ID: mdl-34552267

ABSTRACT

Primary sclerosing cholangitis (PSC) is a chronic cholestatic liver disease of unknown aetiology for which there are no approved therapeutic options. Patients with PSC display changes in gut microbiota and in bile acid (BA) composition; however, the contribution of these alterations to disease pathogenesis remains controversial. Here we identify a role for microbiota-dependent changes in BA synthesis that modulates PSC pathophysiology. In a genetic mouse model of PSC, we show that loss of microbiota-mediated negative feedback control of BA synthesis results in increased hepatic BA concentrations, disruption of bile duct barrier function and, consequently, fatal liver injury. We further show that these changes are dependent on decreased BA signalling to the farnesoid X receptor, which modulates the activity of the rate-limiting enzyme in BA synthesis, CYP7A1. Moreover, patients with advanced stages of PSC show suppressed BA synthesis as measured by serum C4 levels, which is associated with poor disease prognosis. Our preclinical data highlight the microbiota-dependent dynamics of BA metabolism in cholestatic liver disease, which could be important for future therapies targeting BA and gut microbiome interactions, and identify C4 as a potential biomarker to functionally stratify patients with PSC and predict disease outcomes.


Subject(s)
Bile Acids and Salts/metabolism , Cholestasis/metabolism , Gastrointestinal Microbiome , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Anti-Bacterial Agents/administration & dosage , Cholangitis, Sclerosing/metabolism , Cholangitis, Sclerosing/pathology , Humans , Liver/metabolism , Mice , Prognosis , ATP-Binding Cassette Sub-Family B Member 4
9.
Cell Host Microbe ; 29(9): 1366-1377.e9, 2021 09 08.
Article in English | MEDLINE | ID: mdl-34416161

ABSTRACT

Bile acids act as signaling molecules that regulate immune homeostasis, including the differentiation of CD4+ T cells into distinct T cell subsets. The bile acid metabolite isoallolithocholic acid (isoalloLCA) enhances the differentiation of anti-inflammatory regulatory T cells (Treg cells) by facilitating the formation of a permissive chromatin structure in the promoter region of the transcription factor forkhead box P3 (Foxp3). Here, we identify gut bacteria that synthesize isoalloLCA from 3-oxolithocholic acid and uncover a gene cluster responsible for the conversion in members of the abundant human gut bacterial phylum Bacteroidetes. We also show that the nuclear hormone receptor NR4A1 is required for the effect of isoalloLCA on Treg cells. Moreover, the levels of isoalloLCA and its biosynthetic genes are significantly reduced in patients with inflammatory bowel diseases, suggesting that isoalloLCA and its bacterial producers may play a critical role in maintaining immune homeostasis in humans.


Subject(s)
Bacteroidetes/metabolism , Bile Acids and Salts/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phenanthrenes/metabolism , T-Lymphocytes, Regulatory/immunology , Cell Differentiation/physiology , Chromatin/metabolism , Forkhead Transcription Factors/genetics , Humans , Inflammatory Bowel Diseases/pathology , Multigene Family/genetics , Promoter Regions, Genetic/genetics , Signal Transduction/physiology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/cytology
10.
ACS Chem Biol ; 16(8): 1401-1412, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34279901

ABSTRACT

Bile acids play crucial roles in host physiology by acting both as detergents that aid in digestion and as signaling molecules that bind to host receptors. Gut bacterial bile salt hydrolase (BSH) enzymes perform the gateway reaction leading to the conversion of host-produced primary bile acids into bacterially modified secondary bile acids. Small molecule probes that target BSHs will help elucidate the causal roles of these metabolites in host physiology. We previously reported the development of a covalent BSH inhibitor with low gut permeability. Here, we build on our previous findings and describe the development of a second-generation gut-restricted BSH inhibitor with enhanced potency, reduced off-target effects, and durable in vivo efficacy. Structure-activity relationship (SAR) studies focused on the bile acid core identified a compound, AAA-10, containing a C3-sulfonated lithocholic acid scaffold and an alpha-fluoromethyl ketone warhead as a potent pan-BSH inhibitor. This compound inhibits BSH activity in mouse and human fecal slurry, bacterial cultures, and purified BSH proteins and displays reduced toxicity against mammalian cells compared to first generation compounds. Oral administration of AAA-10 to wild-type mice for 5 days resulted in a decrease in the abundance of the secondary bile acids deoxycholic acid (DCA) and lithocholic acid (LCA) in the mouse GI tract with low systemic exposure of AAA-10, demonstrating that AAA-10 is an effective tool for inhibiting BSH activity and modulating bile acid pool composition in vivo.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Gastrointestinal Microbiome/drug effects , Lithocholic Acid/analogs & derivatives , Lithocholic Acid/pharmacology , Animals , Bacteria/drug effects , Bile Acids and Salts/metabolism , Cell Line, Tumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/toxicity , Feces/chemistry , Feces/enzymology , Humans , Lithocholic Acid/toxicity , Male , Mice, Inbred C57BL , Molecular Structure , Structure-Activity Relationship
11.
Bio Protoc ; 11(6)2021 Mar 20.
Article in English | MEDLINE | ID: mdl-33855108

ABSTRACT

The activation of the Takeda G-protein receptor 5 (TGR5, also known as the G protein-coupled bile acid receptor 1, GPBAR1) in enteroendocrine L-cells results in secretion of the anti-diabetic hormone Glucagon-Like Peptide 1 (GLP-1) into systemic circulation. Consequently, recent research has focused on identification and development of TGR5 agonists as type 2 diabetes therapeutics. However, the clinical application of TGR5 agonists has been hampered by side effects of these compounds that primarily result from their absorption into circulation. Here we describe an in vitro screening protocol to evaluate the TGR5 agonism, GLP-1 secretion, and gut-restricted properties of small molecules. The protocol involves differentiating gut epithelial and endocrine cells together in transwells to assess both the pharmacodynamics of TGR5 agonists and the toxicity of compounds to the intestinal monolayer. As a proof of concept, we demonstrate the use of the protocol in evaluating properties of naturally occurring bile acid metabolites that are potent TGR5 agonists. This protocol is adapted from Chaudhari et al. (2021).

12.
Cell Host Microbe ; 29(3): 408-424.e7, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33434516

ABSTRACT

Bariatric surgery is the most effective treatment for type 2 diabetes and is associated with changes in gut metabolites. Previous work uncovered a gut-restricted TGR5 agonist with anti-diabetic properties-cholic acid-7-sulfate (CA7S)-that is elevated following sleeve gastrectomy (SG). Here, we elucidate a microbiome-dependent pathway by which SG increases CA7S production. We show that a microbial metabolite, lithocholic acid (LCA), is increased in murine portal veins post-SG and by activating the vitamin D receptor, induces hepatic mSult2A1/hSULT2A expression to drive CA7S production. An SG-induced shift in the microbiome increases gut expression of the bile acid transporters Asbt and Ostα, which in turn facilitate selective transport of LCA across the gut epithelium. Cecal microbiota transplant from SG animals is sufficient to recreate the pathway in germ-free (GF) animals. Activation of this gut-liver pathway leads to CA7S synthesis and GLP-1 secretion, causally connecting a microbial metabolite with the improvement of diabetic phenotypes.


Subject(s)
Bariatric Surgery , Gastrointestinal Microbiome/physiology , Liver/metabolism , Animals , Diabetes Mellitus, Type 2 , Gastrectomy , Germ-Free Life , Glucagon-Like Peptide 1 , Hep G2 Cells , Humans , Ileum/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Calcitriol/genetics , Sulfotransferases/metabolism
13.
Nat Chem Biol ; 17(1): 20-29, 2021 01.
Article in English | MEDLINE | ID: mdl-32747812

ABSTRACT

Bariatric surgery, the most effective treatment for obesity and type 2 diabetes, is associated with increased levels of the incretin hormone glucagon-like peptide-1 (GLP-1) and changes in levels of circulating bile acids. The levels of individual bile acids in the gastrointestinal (GI) tract after surgery have, however, remained largely unstudied. Using ultra-high performance liquid chromatography-mass spectrometry-based quantification, we observed an increase in an endogenous bile acid, cholic acid-7-sulfate (CA7S), in the GI tract of both mice and humans after sleeve gastrectomy. We show that CA7S is a Takeda G-protein receptor 5 (TGR5) agonist that increases Tgr5 expression and induces GLP-1 secretion. Furthermore, CA7S administration increases glucose tolerance in insulin-resistant mice in a TGR5-dependent manner. CA7S remains gut restricted, minimizing off-target effects previously observed for TGR5 agonists absorbed into the circulation. By studying changes in individual metabolites after surgery, the present study has revealed a naturally occurring TGR5 agonist that exerts systemic glucoregulatory effects while remaining confined to the gut.


Subject(s)
Anti-Obesity Agents/pharmacology , Bariatric Surgery/methods , Cholic Acid/pharmacology , Obesity/surgery , Receptors, G-Protein-Coupled/genetics , Animals , Anti-Obesity Agents/metabolism , Bile/chemistry , Bile/metabolism , Caco-2 Cells , Cholic Acid/biosynthesis , Colon/metabolism , Gene Expression Regulation , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Glucose Tolerance Test , HEK293 Cells , Humans , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/genetics , Obesity/metabolism , Obesity/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Sulfates
14.
15.
Nat Chem Biol ; 16(3): 318-326, 2020 03.
Article in English | MEDLINE | ID: mdl-32042200

ABSTRACT

Bile salt hydrolase (BSH) enzymes are widely expressed by human gut bacteria and catalyze the gateway reaction leading to secondary bile acid formation. Bile acids regulate key metabolic and immune processes by binding to host receptors. There is an unmet need for a potent tool to inhibit BSHs across all gut bacteria to study the effects of bile acids on host physiology. Here, we report the development of a covalent pan-inhibitor of gut bacterial BSHs. From a rationally designed candidate library, we identified a lead compound bearing an alpha-fluoromethyl ketone warhead that modifies BSH at the catalytic cysteine residue. This inhibitor abolished BSH activity in conventional mouse feces. Mice gavaged with a single dose of this compound displayed decreased BSH activity and decreased deconjugated bile acid levels in feces. Our studies demonstrate the potential of a covalent BSH inhibitor to modulate bile acid composition in vivo.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Gastrointestinal Microbiome/physiology , Amidohydrolases/physiology , Animals , Bacteria/enzymology , Bile Acids and Salts/metabolism , Drug Design , Female , Humans , Male , Mice , Mice, Inbred C57BL , Small Molecule Libraries
16.
Nature ; 576(7785): 143-148, 2019 12.
Article in English | MEDLINE | ID: mdl-31776512

ABSTRACT

Bile acids are abundant in the mammalian gut, where they undergo bacteria-mediated transformation to generate a large pool of bioactive molecules. Although bile acids are known to affect host metabolism, cancer progression and innate immunity, it is unknown whether they affect adaptive immune cells such as T helper cells that express IL-17a (TH17 cells) or regulatory T cells (Treg cells). Here we screen a library of bile acid metabolites and identify two distinct derivatives of lithocholic acid (LCA), 3-oxoLCA and isoalloLCA, as T cell regulators in mice. 3-OxoLCA inhibited the differentiation of TH17 cells by directly binding to the key transcription factor retinoid-related orphan receptor-γt (RORγt) and isoalloLCA increased the differentiation of Treg cells through the production of mitochondrial reactive oxygen species (mitoROS), which led to increased expression of FOXP3. The isoalloLCA-mediated enhancement of Treg cell differentiation required an intronic Foxp3 enhancer, the conserved noncoding sequence (CNS) 3; this represents a mode of action distinct from that of previously identified metabolites that increase Treg cell differentiation, which require CNS1. The administration of 3-oxoLCA and isoalloLCA to mice reduced TH17 cell differentiation and increased Treg cell differentiation, respectively, in the intestinal lamina propria. Our data suggest mechanisms through which bile acid metabolites control host immune responses, by directly modulating the balance of TH17 and Treg cells.


Subject(s)
Cell Differentiation/drug effects , Lithocholic Acid/pharmacology , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects , Animals , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Lithocholic Acid/chemistry , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
17.
Elife ; 72018 07 17.
Article in English | MEDLINE | ID: mdl-30014852

ABSTRACT

The human gut microbiota impacts host metabolism and has been implicated in the pathophysiology of obesity and metabolic syndromes. However, defining the roles of specific microbial activities and metabolites on host phenotypes has proven challenging due to the complexity of the microbiome-host ecosystem. Here, we identify strains from the abundant gut bacterial phylum Bacteroidetes that display selective bile salt hydrolase (BSH) activity. Using isogenic strains of wild-type and BSH-deleted Bacteroides thetaiotaomicron, we selectively modulated the levels of the bile acid tauro-ß-muricholic acid in monocolonized gnotobiotic mice. B. thetaiotaomicron BSH mutant-colonized mice displayed altered metabolism, including reduced weight gain and respiratory exchange ratios, as well as transcriptional changes in metabolic, circadian rhythm, and immune pathways in the gut and liver. Our results demonstrate that metabolites generated by a single microbial gene and enzymatic activity can profoundly alter host metabolism and gene expression at local and organism-level scales.


Subject(s)
Amidohydrolases/metabolism , Bacteroides thetaiotaomicron/enzymology , Gastrointestinal Tract/microbiology , Host Microbial Interactions , Taurocholic Acid/analogs & derivatives , Amidohydrolases/genetics , Animals , Bacteroides thetaiotaomicron/genetics , Bacteroides thetaiotaomicron/isolation & purification , Body Weight , Circadian Rhythm , Gene Expression Profiling , Germ-Free Life , Immunity , Intestines/physiology , Liver/physiology , Metabolism , Mice , Respiration , Taurocholic Acid/metabolism
18.
Cell Host Microbe ; 20(6): 709-715, 2016 Dec 14.
Article in English | MEDLINE | ID: mdl-27916477

ABSTRACT

Renal disease is growing in prevalence and has striking co-morbidities with metabolic and cardiovascular disease. Indoxyl sulfate (IS) is a toxin that accumulates in plasma when kidney function declines and contributes to the progression of chronic kidney disease. IS derives exclusively from the gut microbiota. Bacterial tryptophanases convert tryptophan to indole, which is absorbed and modified by the host to produce IS. Here, we identify a widely distributed family of tryptophanases in the gut commensal Bacteroides and find that deleting this gene eliminates the production of indole in vitro. By altering the status or abundance of the Bacteroides tryptophanase, we can modulate IS levels in gnotobiotic mice and in the background of a conventional murine gut community. Our results demonstrate that it is possible to control host IS levels by targeting the microbiota and suggest a possible strategy for treating renal disease.


Subject(s)
Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/microbiology , Indican/metabolism , Indican/toxicity , Animal Feed , Animals , Bacteria/drug effects , Bacteria/enzymology , Bacteria/genetics , Bacteria/metabolism , Bacteroides/enzymology , Bacteroides/genetics , Diet , Disease Models, Animal , Disease Progression , Gastrointestinal Microbiome/genetics , Genetic Engineering , Germ-Free Life/drug effects , Humans , Indoles/metabolism , Metagenome , Mice , Microbiota/genetics , Renal Insufficiency, Chronic , Toxins, Biological/biosynthesis , Toxins, Biological/urine , Tryptophan/metabolism , Tryptophanase/metabolism
19.
ACS Chem Neurosci ; 7(10): 1463-1468, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27501251

ABSTRACT

A novel family of small molecule inhibitors of voltage-gated sodium channels (NaVs) based on the structure of batrachotoxin (BTX), a well-known channel agonist, is described. Protein mutagenesis and electrophysiology experiments reveal the binding site as the inner pore region of the channel, analogous to BTX, alkaloid toxins, and local anesthetics. Homology modeling of the eukaryotic channel based on recent crystallographic analyses of bacterial NaVs suggests a mechanism of action for ion conduction block.


Subject(s)
Batrachotoxins/analysis , Batrachotoxins/pharmacology , Sodium Channel Blockers/pharmacology , Animals , Batrachotoxins/chemical synthesis , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Models, Molecular , Molecular Structure , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mutation , Patch-Clamp Techniques , Rats , Sodium Channel Blockers/chemical synthesis , Sodium Channels/genetics , Sodium Channels/metabolism , Structure-Activity Relationship
20.
Nat Chem Biol ; 11(9): 685-90, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26192599

ABSTRACT

The gut bile acid pool is millimolar in concentration, varies widely in composition among individuals and is linked to metabolic disease and cancer. Although these molecules are derived almost exclusively from the microbiota, remarkably little is known about which bacterial species and genes are responsible for their biosynthesis. Here we report a biosynthetic pathway for the second most abundant class in the gut, 3ß-hydroxy(iso)-bile acids, whose levels exceed 300 µM in some humans and are absent in others. We show, for the first time, that iso-bile acids are produced by Ruminococcus gnavus, a far more abundant commensal than previously known producers, and that the iso-bile acid pathway detoxifies deoxycholic acid and thus favors the growth of the keystone genus Bacteroides. By revealing the biosynthetic genes for an abundant class of bile acids, our work sets the stage for predicting and rationally altering the composition of the bile acid pool.


Subject(s)
3-alpha-Hydroxysteroid Dehydrogenase (B-Specific)/metabolism , Bacterial Proteins/metabolism , Bile Acids and Salts/biosynthesis , Ruminococcus/metabolism , 3-alpha-Hydroxysteroid Dehydrogenase (B-Specific)/genetics , Bacterial Proteins/genetics , Bacteroides/drug effects , Bacteroides/growth & development , Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , Bile Acids and Salts/pharmacology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Kinetics , Microbial Sensitivity Tests , Microbiota/physiology , NAD/metabolism , NADP/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ruminococcus/isolation & purification , Stereoisomerism , Symbiosis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...