Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell ; 187(12): 2952-2968.e13, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38795705

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Glucocorticoides , Hidrógeno , Progestinas , Humanos , Progestinas/metabolismo , Hidrógeno/metabolismo , Femenino , Glucocorticoides/metabolismo , Embarazo , Animales , Familia de Multigenes , Heces/microbiología , Pregnanolona/metabolismo , Ratones
2.
Nature ; 603(7903): 907-912, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35296854

RESUMEN

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.


Asunto(s)
Bacterias , Ácidos y Sales Biliares , Enfermedades Inflamatorias del Intestino , Bacterias/metabolismo , Diferenciación Celular , Tracto Gastrointestinal/microbiología , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/microbiología , Interleucina-17 , Ácido Litocólico/metabolismo , Ácido Litocólico/farmacología , Células Th17
3.
Nat Chem Biol ; 20(4): 410-421, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38347214

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Mamíferos , Metaboloma , Sulfatos
4.
5.
Nature ; 576(7785): 143-148, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31776512

RESUMEN

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.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Ácido Litocólico/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Células Th17/efectos de los fármacos , Animales , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Ácido Litocólico/química , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células Th17/citología , Células Th17/inmunología , Células Th17/metabolismo
6.
Nat Chem Biol ; 17(10): 1046-1056, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34552222

RESUMEN

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.


Asunto(s)
Enfermedades Transmisibles/microbiología , Interacciones Microbiota-Huesped/fisiología , Microbiota/fisiología , Animales , Antiinfecciosos/farmacología , Trasplante de Microbiota Fecal , Vida Libre de Gérmenes , Humanos
7.
Nat Chem Biol ; 17(1): 20-29, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32747812

RESUMEN

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.


Asunto(s)
Fármacos Antiobesidad/farmacología , Cirugía Bariátrica/métodos , Ácido Cólico/farmacología , Obesidad/cirugía , Receptores Acoplados a Proteínas G/genética , Animales , Fármacos Antiobesidad/metabolismo , Bilis/química , Bilis/metabolismo , Células CACO-2 , Ácido Cólico/biosíntesis , Colon/metabolismo , Regulación de la Expresión Génica , Péptido 1 Similar al Glucagón/genética , Péptido 1 Similar al Glucagón/metabolismo , Prueba de Tolerancia a la Glucosa , Células HEK293 , Humanos , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Sulfatos
8.
Nat Chem Biol ; 16(3): 318-326, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32042200

RESUMEN

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.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Amidohidrolasas/metabolismo , Microbioma Gastrointestinal/fisiología , Amidohidrolasas/fisiología , Animales , Bacterias/enzimología , Ácidos y Sales Biliares/metabolismo , Diseño de Fármacos , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Bibliotecas de Moléculas Pequeñas
9.
Nature ; 505(7484): 559-63, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24336217

RESUMEN

Long-term dietary intake influences the structure and activity of the trillions of microorganisms residing in the human gut, but it remains unclear how rapidly and reproducibly the human gut microbiome responds to short-term macronutrient change. Here we show that the short-term consumption of diets composed entirely of animal or plant products alters microbial community structure and overwhelms inter-individual differences in microbial gene expression. The animal-based diet increased the abundance of bile-tolerant microorganisms (Alistipes, Bilophila and Bacteroides) and decreased the levels of Firmicutes that metabolize dietary plant polysaccharides (Roseburia, Eubacterium rectale and Ruminococcus bromii). Microbial activity mirrored differences between herbivorous and carnivorous mammals, reflecting trade-offs between carbohydrate and protein fermentation. Foodborne microbes from both diets transiently colonized the gut, including bacteria, fungi and even viruses. Finally, increases in the abundance and activity of Bilophila wadsworthia on the animal-based diet support a link between dietary fat, bile acids and the outgrowth of microorganisms capable of triggering inflammatory bowel disease. In concert, these results demonstrate that the gut microbiome can rapidly respond to altered diet, potentially facilitating the diversity of human dietary lifestyles.


Asunto(s)
Bacterias/genética , Bacterias/aislamiento & purificación , Dieta , Tracto Gastrointestinal/microbiología , Metagenoma , Microbiota , Adulto , Bacterias/efectos de los fármacos , Bacteroides/efectos de los fármacos , Bacteroides/genética , Bacteroides/aislamiento & purificación , Ácidos y Sales Biliares/análisis , Ácidos y Sales Biliares/metabolismo , Bilophila/efectos de los fármacos , Bilophila/genética , Bilophila/aislamiento & purificación , Carnivoría , Dieta/efectos adversos , Dieta Vegetariana , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Heces/química , Heces/microbiología , Femenino , Fermentación/efectos de los fármacos , Microbiología de Alimentos , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/virología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Herbivoria , Humanos , Enfermedades Inflamatorias del Intestino/microbiología , Masculino , Metagenoma/efectos de los fármacos , Metagenoma/genética , Microbiota/efectos de los fármacos , Microbiota/genética , Factores de Tiempo , Adulto Joven
10.
Nat Chem Biol ; 11(9): 685-90, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26192599

RESUMEN

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.


Asunto(s)
3-alfa-Hidroxiesteroide Deshidrogenasa (B-Específica)/metabolismo , Proteínas Bacterianas/metabolismo , Ácidos y Sales Biliares/biosíntesis , Ruminococcus/metabolismo , 3-alfa-Hidroxiesteroide Deshidrogenasa (B-Específica)/genética , Proteínas Bacterianas/genética , Bacteroides/efectos de los fármacos , Bacteroides/crecimiento & desarrollo , Ácidos y Sales Biliares/química , Ácidos y Sales Biliares/metabolismo , Ácidos y Sales Biliares/farmacología , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Humanos , Cinética , Pruebas de Sensibilidad Microbiana , Microbiota/fisiología , NAD/metabolismo , NADP/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ruminococcus/aislamiento & purificación , Estereoisomerismo , Simbiosis/fisiología
11.
Cell Host Microbe ; 30(4): 435-438, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35421338

RESUMEN

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.


Asunto(s)
Microbiota , Humanos
12.
Nat Microbiol ; 7(9): 1404-1418, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35982310

RESUMEN

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.


Asunto(s)
Bacteroides thetaiotaomicron , Vías Biosintéticas , Animales , Bacterias , Tracto Gastrointestinal , Humanos , Ratones , Sulfato Adenililtransferasa
13.
Sci Adv ; 8(34): eabo2794, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-36026454

RESUMEN

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.


Asunto(s)
Ácidos y Sales Biliares , Microbioma Gastrointestinal , Animales , Hígado , Micelas , Permeabilidad , Ratas
14.
Bio Protoc ; 11(6)2021 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-33855108

RESUMEN

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).

15.
ACS Chem Biol ; 16(8): 1401-1412, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34279901

RESUMEN

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.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Ácido Litocólico/análogos & derivados , Ácido Litocólico/farmacología , Animales , Bacterias/efectos de los fármacos , Ácidos y Sales Biliares/metabolismo , Línea Celular Tumoral , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/toxicidad , Heces/química , Heces/enzimología , Humanos , Ácido Litocólico/toxicidad , Masculino , Ratones Endogámicos C57BL , Estructura Molecular , Relación Estructura-Actividad
16.
Cell Host Microbe ; 29(3): 408-424.e7, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33434516

RESUMEN

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.


Asunto(s)
Cirugía Bariátrica , Microbioma Gastrointestinal/fisiología , Hígado/metabolismo , Animales , Diabetes Mellitus Tipo 2 , Gastrectomía , Vida Libre de Gérmenes , Péptido 1 Similar al Glucagón , Células Hep G2 , Humanos , Íleon/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Calcitriol/genética , Sulfotransferasas/metabolismo
17.
Cell Host Microbe ; 29(9): 1366-1377.e9, 2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34416161

RESUMEN

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.


Asunto(s)
Bacteroidetes/metabolismo , Ácidos y Sales Biliares/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Fenantrenos/metabolismo , Linfocitos T Reguladores/inmunología , Diferenciación Celular/fisiología , Cromatina/metabolismo , Factores de Transcripción Forkhead/genética , Humanos , Enfermedades Inflamatorias del Intestino/patología , Familia de Multigenes/genética , Regiones Promotoras Genéticas/genética , Transducción de Señal/fisiología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/citología
18.
Nat Metab ; 3(9): 1228-1241, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34552267

RESUMEN

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.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Colestasis/metabolismo , Microbioma Gastrointestinal , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Animales , Antibacterianos/administración & dosificación , Colangitis Esclerosante/metabolismo , Colangitis Esclerosante/patología , Humanos , Hígado/metabolismo , Ratones , Pronóstico , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
19.
Elife ; 72018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30014852

RESUMEN

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.


Asunto(s)
Amidohidrolasas/metabolismo , Bacteroides thetaiotaomicron/enzimología , Tracto Gastrointestinal/microbiología , Interacciones Microbiota-Huesped , Ácido Taurocólico/análogos & derivados , Amidohidrolasas/genética , Animales , Bacteroides thetaiotaomicron/genética , Bacteroides thetaiotaomicron/aislamiento & purificación , Peso Corporal , Ritmo Circadiano , Perfilación de la Expresión Génica , Vida Libre de Gérmenes , Inmunidad , Intestinos/fisiología , Hígado/fisiología , Metabolismo , Ratones , Respiración , Ácido Taurocólico/metabolismo
20.
Org Lett ; 9(21): 4363-6, 2007 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-17887696

RESUMEN

Rhodium-catalyzed reactions of sulfonate ester derivatives are biased strongly toward 1,6-insertion and thus offer a general method for assembling delta-sultones. Two protocols for staging this cyclization reaction are described, which capitalize on the unique ability of either diazo or iodonium ylide intermediates to form Rh-carbene species. The value of these heterocycles for fine chemicals synthesis is demonstrated in both reductive and oxidative reactions that make possible excision of the -SO3- moiety.


Asunto(s)
Compuestos Heterocíclicos/química , Naftalenosulfonatos/síntesis química , Rodio/química , Catálisis , Técnicas Químicas Combinatorias , Ciclización , Ésteres , Estructura Molecular , Naftalenosulfonatos/química , Estereoisomerismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA