Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(15): 3857-3876, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39059362

RESUMEN

The past 50 years of interdisciplinary research in humans and model organisms has delivered unprecedented insights into the mechanisms through which diet affects energy balance. However, translating these results to prevent and treat obesity and its associated diseases remains challenging. Given the vast scope of this literature, we focus this Review on recent conceptual advances in molecular nutrition targeting the management of energy balance, including emerging dietary and pharmaceutical interventions and their interactions with the human gut microbiome. Notably, multiple current dietary patterns of interest embrace moderate-to-high fat intake or prioritize the timing of eating over macronutrient intake. Furthermore, the rapid expansion of microbiome research findings has complicated multiple longstanding tenets of nutrition while also providing new opportunities for intervention. Continued progress promises more precise and reliable dietary recommendations that leverage our growing knowledge of the microbiome, the changing landscape of clinical interventions, and our molecular understanding of human biology.


Asunto(s)
Dieta , Microbioma Gastrointestinal , Obesidad , Humanos , Animales , Obesidad/metabolismo , Obesidad/microbiología , Metabolismo Energético
2.
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
3.
Cell ; 187(5): 1191-1205.e15, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38366592

RESUMEN

Carbohydrate intolerance, commonly linked to the consumption of lactose, fructose, or sorbitol, affects up to 30% of the population in high-income countries. Although sorbitol intolerance is attributed to malabsorption, the underlying mechanism remains unresolved. Here, we show that a history of antibiotic exposure combined with high fat intake triggered long-lasting sorbitol intolerance in mice by reducing Clostridia abundance, which impaired microbial sorbitol catabolism. The restoration of sorbitol catabolism by inoculation with probiotic Escherichia coli protected mice against sorbitol intolerance but did not restore Clostridia abundance. Inoculation with the butyrate producer Anaerostipes caccae restored a normal Clostridia abundance, which protected mice against sorbitol-induced diarrhea even when the probiotic was cleared. Butyrate restored Clostridia abundance by stimulating epithelial peroxisome proliferator-activated receptor-gamma (PPAR-γ) signaling to restore epithelial hypoxia in the colon. Collectively, these mechanistic insights identify microbial sorbitol catabolism as a potential target for approaches for the diagnosis, treatment, and prevention of sorbitol intolerance.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos , Microbioma Gastrointestinal , Sorbitol , Animales , Ratones , Antibacterianos/farmacología , Butiratos , Clostridium , Escherichia coli , Sorbitol/metabolismo
4.
Cell ; 184(7): 1740-1756.e16, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33705688

RESUMEN

The core symptoms of many neurological disorders have traditionally been thought to be caused by genetic variants affecting brain development and function. However, the gut microbiome, another important source of variation, can also influence specific behaviors. Thus, it is critical to unravel the contributions of host genetic variation, the microbiome, and their interactions to complex behaviors. Unexpectedly, we discovered that different maladaptive behaviors are interdependently regulated by the microbiome and host genes in the Cntnap2-/- model for neurodevelopmental disorders. The hyperactivity phenotype of Cntnap2-/- mice is caused by host genetics, whereas the social-behavior phenotype is mediated by the gut microbiome. Interestingly, specific microbial intervention selectively rescued the social deficits in Cntnap2-/- mice through upregulation of metabolites in the tetrahydrobiopterin synthesis pathway. Our findings that behavioral abnormalities could have distinct origins (host genetic versus microbial) may change the way we think about neurological disorders and how to treat them.


Asunto(s)
Microbioma Gastrointestinal , Locomoción , Conducta Social , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Modelos Animales de Enfermedad , Potenciales Postsinápticos Excitadores , Trasplante de Microbiota Fecal , Heces/microbiología , Limosilactobacillus reuteri/metabolismo , Limosilactobacillus reuteri/fisiología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/microbiología , Trastornos del Neurodesarrollo/patología , Trastornos del Neurodesarrollo/terapia , Análisis de Componente Principal , Agitación Psicomotora/patología , Transmisión Sináptica
5.
Cell ; 181(6): 1263-1275.e16, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32437658

RESUMEN

Very low-carbohydrate, high-fat ketogenic diets (KDs) induce a pronounced shift in metabolic fuel utilization that elevates circulating ketone bodies; however, the consequences of these compounds for host-microbiome interactions remain unknown. Here, we show that KDs alter the human and mouse gut microbiota in a manner distinct from high-fat diets (HFDs). Metagenomic and metabolomic analyses of stool samples from an 8-week inpatient study revealed marked shifts in gut microbial community structure and function during the KD. Gradient diet experiments in mice confirmed the unique impact of KDs relative to HFDs with a reproducible depletion of bifidobacteria. In vitro and in vivo experiments showed that ketone bodies selectively inhibited bifidobacterial growth. Finally, mono-colonizations and human microbiome transplantations into germ-free mice revealed that the KD-associated gut microbiota reduces the levels of intestinal pro-inflammatory Th17 cells. Together, these results highlight the importance of trans-kingdom chemical dialogs for mediating the host response to dietary interventions.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/fisiología , Intestinos/inmunología , Intestinos/microbiología , Células Th17/inmunología , Células Th17/fisiología , Adolescente , Adulto , Animales , Dieta Alta en Grasa/métodos , Dieta Cetogénica/métodos , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microbiota/inmunología , Microbiota/fisiología , Persona de Mediana Edad , Células Th17/microbiología , Adulto Joven
6.
Cell ; 174(2): 271-284.e14, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-29887373

RESUMEN

The small intestinal tuft cell-ILC2 circuit mediates epithelial responses to intestinal helminths and protists by tuft cell chemosensory-like sensing and IL-25-mediated activation of lamina propria ILC2s. Small intestine ILC2s constitutively express the IL-25 receptor, which is negatively regulated by A20 (Tnfaip3). A20 deficiency in ILC2s spontaneously triggers the circuit and, unexpectedly, promotes adaptive small-intestinal lengthening and remodeling. Circuit activation occurs upon weaning and is enabled by dietary polysaccharides that render mice permissive for Tritrichomonas colonization, resulting in luminal accumulation of acetate and succinate, metabolites of the protist hydrogenosome. Tuft cells express GPR91, the succinate receptor, and dietary succinate, but not acetate, activates ILC2s via a tuft-, TRPM5-, and IL-25-dependent pathway. Also induced by parasitic helminths, circuit activation and small intestinal remodeling impairs infestation by new helminths, consistent with the phenomenon of concomitant immunity. We describe a metabolic sensing circuit that may have evolved to facilitate mutualistic responses to luminal pathosymbionts.


Asunto(s)
Intestino Delgado/fisiología , Tritrichomonas/metabolismo , Acetatos/metabolismo , Animales , Fibras de la Dieta/metabolismo , Metabolismo Energético , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/parasitología , Interleucinas/genética , Interleucinas/metabolismo , Mucosa Intestinal/citología , Intestino Delgado/microbiología , Intestino Delgado/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microbiota , Plásmidos/genética , Plásmidos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Interleucina/metabolismo , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/metabolismo , Ácido Succínico/metabolismo , Canales Catiónicos TRPM/metabolismo , Tritrichomonas/crecimiento & desarrollo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo
7.
Nat Immunol ; 21(4): 422-433, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32205880

RESUMEN

A20 is an anti-inflammatory protein that is strongly linked to human disease. Here, we find that mice expressing three distinct targeted mutations of A20's zinc finger 7 (ZF7) ubiquitin-binding motif uniformly developed digit arthritis with features common to psoriatic arthritis, while mice expressing point mutations in A20's OTU or ZF4 motifs did not exhibit this phenotype. Arthritis in A20ZF7 mice required T cells and MyD88, was exquisitely sensitive to tumor necrosis factor and interleukin-17A, and persisted in germ-free conditions. A20ZF7 cells exhibited prolonged IκB kinase activity that drove exaggerated transcription of late-phase nuclear factor-κB response genes in vitro and in prediseased mouse paws in vivo. In addition, mice expressing double-mutant A20 proteins in A20's ZF4 and ZF7 motifs died perinatally with multi-organ inflammation. Therefore, A20's ZF4 and ZF7 motifs synergistically prevent inflammatory disease in a non-catalytic manner.


Asunto(s)
Artritis Psoriásica/metabolismo , Inflamación/metabolismo , Ubiquitina/metabolismo , Animales , Células Cultivadas , Interleucina-17 , Ratones , Ratones Endogámicos C57BL , Mutación/genética , FN-kappa B/metabolismo , Unión Proteica/fisiología , Transducción de Señal/fisiología , Transcripción Genética/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitinación/fisiología , Dedos de Zinc/fisiología
8.
Cell ; 163(5): 1051-1052, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26590412

RESUMEN

Zeevi et al. report that extensive monitoring of a human cohort for variations in dietary intake, lifestyle, host phenotype, and the gut microbiome has enabled the development of a machine-learning algorithm that accurately predicts the individual glycemic response to meals, providing an important first step toward personalized nutrition.


Asunto(s)
Algoritmos , Glucemia/análisis , Diabetes Mellitus Tipo 2/sangre , Periodo Posprandial , Humanos
9.
Immunity ; 53(2): 264-276, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32814025

RESUMEN

Emerging evidence suggests that the effect of dietary intake on human health and disease is linked to both the immune system and the microbiota. Yet, we lack an integrated mechanistic model for how these three complex systems relate, limiting our ability to understand and treat chronic and infectious disease. Here, we review recent findings at the interface of microbiology, immunology, and nutrition, with an emphasis on experimentally tractable models and hypothesis-driven mechanistic work. We outline emerging mechanistic concepts and generalizable approaches to bridge the gap between microbial ecology and molecular mechanism. These set the stage for a new era of precision human nutrition informed by a deep and comprehensive knowledge of the diverse cell types in and on the human body.


Asunto(s)
Dieta , Microbioma Gastrointestinal/fisiología , Inmunomodulación/fisiología , Humanos , Inmunidad/fisiología , Inflamación/patología
10.
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
11.
Artículo en Inglés | MEDLINE | ID: mdl-39107044

RESUMEN

Drugs represent our first, and sometimes last, line of defense for many diseases, yet despite decades of research we still do not fully understand why a given drug works in one patient and fails in the next. The human gut microbiome is one of the missing puzzle pieces, due to its ability to parallel and extend host pathways for drug metabolism, along with more complex host-microbiome interactions. Herein, we focus on the well-established links between the gut microbiome and drugs for heart disease and cancer, plus emerging data on neurological disease. We highlight the interdisciplinary methods that are available and how they can be used to address major remaining knowledge gaps, including the consequences of microbial drug metabolism for treatment outcomes. Continued progress in this area promises fundamental biological insights into humans and their associated microbial communities and strategies for leveraging the microbiome to improve the practice of medicine.

12.
Nature ; 595(7866): 272-277, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34163067

RESUMEN

Diet is a major factor that shapes the gut microbiome1, but the consequences of diet-induced changes in the microbiome for host pathophysiology remain poorly understood. We conducted a randomized human intervention study using a very-low-calorie diet (NCT01105143). Although metabolic health was improved, severe calorie restriction led to a decrease in bacterial abundance and restructuring of the gut microbiome. Transplantation of post-diet microbiota to mice decreased their body weight and adiposity relative to mice that received pre-diet microbiota. Weight loss was associated with impaired nutrient absorption and enrichment in Clostridioides difficile, which was consistent with a decrease in bile acids and was sufficient to replicate metabolic phenotypes in mice in a toxin-dependent manner. These results emphasize the importance of diet-microbiome interactions in modulating host energy balance and the need to understand the role of diet in the interplay between pathogenic and beneficial symbionts.


Asunto(s)
Bacterias/aislamiento & purificación , Bacterias/metabolismo , Restricción Calórica , Dieta Reductora , Microbioma Gastrointestinal/fisiología , Adiposidad , Animales , Bacterias/crecimiento & desarrollo , Bacterias/patogenicidad , Toxinas Bacterianas/metabolismo , Ácidos y Sales Biliares/metabolismo , Peso Corporal , Clostridioides difficile/crecimiento & desarrollo , Clostridioides difficile/aislamiento & purificación , Clostridioides difficile/metabolismo , Metabolismo Energético , Humanos , Absorción Intestinal , Masculino , Ratones , Nutrientes/metabolismo , Simbiosis , Pérdida de Peso
13.
PLoS Biol ; 21(4): e3002087, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37018375

RESUMEN

Aging is often accompanied by an increased risk of an array of diseases spanning the cardiovascular, nervous, and immune systems, among others. Despite remarkable progress in understanding the cellular and molecular mechanisms involved in aging, the role of the microbiome remains understudied. In this Essay, we highlight recent progress towards understanding if and how the microbiome contributes to aging and age-associated diseases. Furthermore, we discuss the need to consider sexually dimorphic phenotypes in the context of aging and the microbiome. We also highlight the broad implications for this emerging area of interdisciplinary research to address long-standing questions about host-microbiome interactions across the life span.


Asunto(s)
Microbiota , Microbiota/fisiología , Sistema Inmunológico
14.
PLoS Biol ; 21(5): e3002125, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37205710

RESUMEN

Human gut bacteria perform diverse metabolic functions with consequences for host health. The prevalent and disease-linked Actinobacterium Eggerthella lenta performs several unusual chemical transformations, but it does not metabolize sugars and its core growth strategy remains unclear. To obtain a comprehensive view of the metabolic network of E. lenta, we generated several complementary resources: defined culture media, metabolomics profiles of strain isolates, and a curated genome-scale metabolic reconstruction. Stable isotope-resolved metabolomics revealed that E. lenta uses acetate as a key carbon source while catabolizing arginine to generate ATP, traits which could be recapitulated in silico by our updated metabolic model. We compared these in vitro findings with metabolite shifts observed in E. lenta-colonized gnotobiotic mice, identifying shared signatures across environments and highlighting catabolism of the host signaling metabolite agmatine as an alternative energy pathway. Together, our results elucidate a distinctive metabolic niche filled by E. lenta in the gut ecosystem. Our culture media formulations, atlas of metabolomics data, and genome-scale metabolic reconstructions form a freely available collection of resources to support further study of the biology of this prevalent gut bacterium.


Asunto(s)
Actinobacteria , Microbioma Gastrointestinal , Humanos , Ratones , Animales , Biología de Sistemas , Ecosistema , Actinobacteria/metabolismo
15.
Mol Microbiol ; 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37712143

RESUMEN

Drugs intended to target mammalian cells can have broad off-target effects on the human gut microbiota with potential downstream consequences for drug efficacy and side effect profiles. Yet, despite a rich literature on antibiotic resistance, we still know very little about the mechanisms through which commensal bacteria evade non-antibiotic drugs. Here, we focus on statins, one of the most prescribed drug types in the world and an essential tool in the prevention and treatment of high circulating cholesterol levels. Prior work in humans, mice, and cell culture support an off-target effect of statins on human gut bacteria; however, the genetic determinants of statin sensitivity remain unknown. We confirmed that simvastatin inhibits the growth of diverse human gut bacterial strains grown in communities and in pure cultures. Drug sensitivity varied between phyla and was dose-dependent. We selected two representative simvastatin-sensitive species for more in-depth analysis: Eggerthella lenta (phylum: Actinobacteriota) and Bacteroides thetaiotaomicron (phylum: Bacteroidota). Transcriptomics revealed that both bacterial species upregulate genes in response to simvastatin that alter the cell membrane, including fatty acid biogenesis (E. lenta) and drug efflux systems (B. thetaiotaomicron). Transposon mutagenesis identified a key efflux system in B. thetaiotaomicron that enables growth in the presence of statins. Taken together, these results emphasize the importance of the bacterial cell membrane in countering the off-target effects of host-targeted drugs. Continued mechanistic dissection of the various mechanisms through which the human gut microbiota evades drugs will be essential to understand and predict the effects of drug administration in human cohorts and the potential downstream consequences for health and disease.

16.
Cell ; 134(5): 708-13, 2008 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-18775300

RESUMEN

Metagenomics seeks to characterize the composition of microbial communities, their operations, and their dynamically coevolving relationships with the habitats they occupy without having to culture community members. Uniting metagenomics with analyses of the products of microbial community metabolism (metabolomics) will shed light on how microbial communities function in a variety of environments, including the human body.


Asunto(s)
Genética Microbiana , Genómica , Metagenoma , Ecología , Humanos
17.
Proc Natl Acad Sci U S A ; 117(27): 16009-16018, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32571913

RESUMEN

Food and drug products contain diverse and abundant small-molecule additives (excipients) with unclear impacts on human physiology, drug safety, and response. Here, we evaluate their potential impact on intestinal drug absorption. By screening 136 unique compounds for inhibition of the key intestinal transporter OATP2B1 we identified and validated 24 potent OATP2B1 inhibitors, characterized by higher molecular weight and hydrophobicity compared to poor or noninhibitors. OATP2B1 inhibitors were also enriched for dyes, including 8 azo (R-N=N-R') dyes. Pharmacokinetic studies in mice confirmed that FD&C Red No. 40, a common azo dye excipient and a potent inhibitor of OATP2B1, decreased the plasma level of the OATP2B1 substrate fexofenadine, suggesting that FD&C Red No. 40 has the potential to block drug absorption through OATP2B1 inhibition in vivo. However, the gut microbiomes of multiple unrelated healthy individuals as well as diverse human gut bacterial isolates were capable of inactivating the identified azo dye excipients, producing metabolites that no longer inhibit OATP2B1 transport. These results support a beneficial role for the microbiome in limiting the unintended effects of food and drug additives in the intestine and provide a framework for the data-driven selection of excipients. Furthermore, the ubiquity and genetic diversity of gut bacterial azoreductases coupled to experiments in conventionally raised and gnotobiotic mice suggest that variations in gut microbial community structure may be less important to consider relative to the high concentrations of azo dyes in food products, which have the potential to saturate gut bacterial enzymatic activity.


Asunto(s)
Bacterias/metabolismo , Excipientes/metabolismo , Aditivos Alimentarios/metabolismo , Alimentos , Microbioma Gastrointestinal/fisiología , Absorción Intestinal/fisiología , Transportadores de Anión Orgánico/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Animales , Antialérgicos/metabolismo , Antialérgicos/farmacocinética , Compuestos Azo , Bacterias/aislamiento & purificación , Excipientes/farmacocinética , Femenino , Aditivos Alimentarios/farmacocinética , Antagonistas de los Receptores Histamínicos H1 no Sedantes/metabolismo , Antagonistas de los Receptores Histamínicos H1 no Sedantes/farmacocinética , Humanos , Absorción Intestinal/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Terfenadina/análogos & derivados , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
18.
Proc Natl Acad Sci U S A ; 117(50): 32029-32037, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33257580

RESUMEN

Disease tolerance, the capacity of tissues to withstand damage caused by a stimulus without a decline in host fitness, varies across tissues, environmental conditions, and physiologic states. While disease tolerance is a known strategy of host defense, its role in noninfectious diseases has been understudied. Here, we provide evidence that a thermogenic fat-epithelial cell axis regulates intestinal disease tolerance during experimental colitis. We find that intestinal disease tolerance is a metabolically expensive trait, whose expression is restricted to thermoneutral mice and is not transferable by the microbiota. Instead, disease tolerance is dependent on the adrenergic state of thermogenic adipocytes, which indirectly regulate tolerogenic responses in intestinal epithelial cells. Our work has identified an unexpected mechanism that controls intestinal disease tolerance with implications for colitogenic diseases.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Colitis/inmunología , Neoplasias del Colon/inmunología , Resistencia a la Enfermedad , Infecciones por Enterobacteriaceae/inmunología , Adipocitos/metabolismo , Tejido Adiposo Pardo/citología , Animales , Azoximetano/administración & dosificación , Comunicación Celular , Citrobacter rodentium/patogenicidad , Colitis/inducido químicamente , Colitis/microbiología , Colitis/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/patología , Sulfato de Dextran/toxicidad , Infecciones por Enterobacteriaceae/inducido químicamente , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/patología , Células Epiteliales/metabolismo , Femenino , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Masculino , Ratones , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Termogénesis/inmunología
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA