Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell ; 174(6): 1388-1405.e21, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30193112

RESUMO

Empiric probiotics are commonly consumed by healthy individuals as means of life quality improvement and disease prevention. However, evidence of probiotic gut mucosal colonization efficacy remains sparse and controversial. We metagenomically characterized the murine and human mucosal-associated gastrointestinal microbiome and found it to only partially correlate with stool microbiome. A sequential invasive multi-omics measurement at baseline and during consumption of an 11-strain probiotic combination or placebo demonstrated that probiotics remain viable upon gastrointestinal passage. In colonized, but not germ-free mice, probiotics encountered a marked mucosal colonization resistance. In contrast, humans featured person-, region- and strain-specific mucosal colonization patterns, hallmarked by predictive baseline host and microbiome features, but indistinguishable by probiotics presence in stool. Consequently, probiotics induced a transient, individualized impact on mucosal community structure and gut transcriptome. Collectively, empiric probiotics supplementation may be limited in universally and persistently impacting the gut mucosa, meriting development of new personalized probiotic approaches.


Assuntos
Microbioma Gastrointestinal , Probióticos/administração & dosagem , Adolescente , Adulto , Idoso , Animais , Bactérias/genética , Bactérias/isolamento & purificação , Fezes/microbiologia , Feminino , Mucosa Gástrica/microbiologia , Humanos , Mucosa Intestinal/microbiologia , Masculino , Metagenômica , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Efeito Placebo , Análise de Componente Principal , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo , Transcriptoma , Adulto Jovem
2.
Cell ; 174(6): 1406-1423.e16, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30193113

RESUMO

Probiotics are widely prescribed for prevention of antibiotics-associated dysbiosis and related adverse effects. However, probiotic impact on post-antibiotic reconstitution of the gut mucosal host-microbiome niche remains elusive. We invasively examined the effects of multi-strain probiotics or autologous fecal microbiome transplantation (aFMT) on post-antibiotic reconstitution of the murine and human mucosal microbiome niche. Contrary to homeostasis, antibiotic perturbation enhanced probiotics colonization in the human mucosa but only mildly improved colonization in mice. Compared to spontaneous post-antibiotic recovery, probiotics induced a markedly delayed and persistently incomplete indigenous stool/mucosal microbiome reconstitution and host transcriptome recovery toward homeostatic configuration, while aFMT induced a rapid and near-complete recovery within days of administration. In vitro, Lactobacillus-secreted soluble factors contributed to probiotics-induced microbiome inhibition. Collectively, potential post-antibiotic probiotic benefits may be offset by a compromised gut mucosal recovery, highlighting a need of developing aFMT or personalized probiotic approaches achieving mucosal protection without compromising microbiome recolonization in the antibiotics-perturbed host.


Assuntos
Antibacterianos/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Probióticos/administração & dosagem , Adolescente , Adulto , Idoso , Animais , Transplante de Microbiota Fecal , Fezes/microbiologia , Feminino , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Lactobacillus/efeitos dos fármacos , Lactobacillus/genética , Lactobacillus/isolamento & purificação , Lactococcus/genética , Lactococcus/isolamento & purificação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , RNA Ribossômico 16S/análise , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo , Adulto Jovem
3.
J Exp Med ; 215(2): 481-500, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29343501

RESUMO

Mucosal Langerhans cells (LCs) originate from pre-dendritic cells and monocytes. However, the mechanisms involved in their in situ development remain unclear. Here, we demonstrate that the differentiation of murine mucosal LCs is a two-step process. In the lamina propria, signaling via BMP7-ALK3 promotes translocation of LC precursors to the epithelium. Within the epithelium, TGF-ß1 finalizes LC differentiation, and ALK5 is crucial to this process. Moreover, the local microbiota has a major impact on the development of mucosal LCs, whereas LCs in turn maintain mucosal homeostasis and prevent tissue destruction. These results reveal the differential and sequential role of TGF-ß1 and BMP7 in LC differentiation and highlight the intimate interplay of LCs with the microbiota.


Assuntos
Proteína Morfogenética Óssea 7/imunologia , Células de Langerhans/imunologia , Microbiota/imunologia , Fator de Crescimento Transformador beta1/imunologia , Animais , Antígenos de Superfície/genética , Antígenos de Superfície/metabolismo , Proteína Morfogenética Óssea 7/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Humanos , Imunidade nas Mucosas , Células de Langerhans/citologia , Células de Langerhans/metabolismo , Lectinas Tipo C/deficiência , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Masculino , Lectinas de Ligação a Manose/deficiência , Lectinas de Ligação a Manose/genética , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucosa Bucal/citologia , Mucosa Bucal/imunologia , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Transdução de Sinais/imunologia , Células-Tronco/citologia , Células-Tronco/imunologia , Transcriptoma , Fator de Crescimento Transformador beta1/genética , Regulação para Cima
4.
Proc Natl Acad Sci U S A ; 114(3): E337-E346, 2017 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-28049839

RESUMO

The oral epithelium contributes to innate immunity and oral mucosal homeostasis, which is critical for preventing local inflammation and the associated adverse systemic conditions. Nevertheless, the mechanisms by which the oral epithelium maintains homeostasis are poorly understood. Here, we studied the role of growth arrest specific 6 (GAS6), a ligand of the TYRO3-AXL-MERTK (TAM) receptor family, in regulating oral mucosal homeostasis. Expression of GAS6 was restricted to the outer layers of the oral epithelium. In contrast to protein S, the other TAM ligand, which was constitutively expressed postnatally, expression of GAS6 initiated only 3-4 wk after birth. Further analysis revealed that GAS6 expression was induced by the oral microbiota in a myeloid differentiation primary response gene 88 (MyD88)-dependent fashion. Mice lacking GAS6 presented higher levels of inflammatory cytokines, elevated frequencies of neutrophils, and up-regulated activity of enzymes, generating reactive nitrogen species. We also found an imbalance in Th17/Treg ratio known to control tissue homeostasis, as Gas6-deficient dendritic cells preferentially secreted IL-6 and induced Th17 cells. As a result of this immunological shift, a significant microbial dysbiosis was observed in Gas6-/- mice, because anaerobic bacteria largely expanded by using inflammatory byproducts for anaerobic respiration. Using chimeric mice, we found a critical role for GAS6 in epithelial cells in maintaining oral homeostasis, whereas its absence in hematopoietic cells synergized the level of dysbiosis. We thus propose GAS6 as a key immunological regulator of host-commensal interactions in the oral epithelium.


Assuntos
Homeostase/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mucosa Bucal/metabolismo , Animais , Disbiose/metabolismo , Células Epiteliais/metabolismo , Imunidade Inata/imunologia , Inflamação/metabolismo , Interleucina-6 , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Neutrófilos/metabolismo , Proteína S/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , c-Mer Tirosina Quinase/metabolismo
5.
BMC Med ; 14(1): 83, 2016 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-27256449

RESUMO

HIV/AIDS causes severe dysfunction of the immune system through CD4+ T cell depletion, leading to dysregulation of both the adaptive and innate immune arms. A primary target for viral infection is the gastrointestinal tract, which is a reservoir of CD4+ T cells. In addition to being a major immune hub, the human gastrointestinal tract harbors trillions of commensal microorganisms, the microbiota, which have recently been shown to play critical roles in health. Alterations in the composition and function of microbiota have been implicated in a variety of 'multi-factorial' disorders, including infectious, autoimmune, metabolic, and neoplastic disorders. It is widely accepted that, in addition to its direct role in altering the gastrointestinal CD4+ T cell compartment, HIV infection is characterized by gut microbiota compositional and functional changes. Herein, we review such alterations and discuss their potential local and systemic effects on the HIV-positive host, as well as potential roles of novel microbiota-targeting treatments in modulating HIV progression and associated adverse systemic manifestations.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Microbioma Gastrointestinal/imunologia , Infecções por HIV/microbiologia , Linfócitos T CD4-Positivos/microbiologia , Infecções por HIV/virologia , Humanos
6.
Bioinform Biol Insights ; 10: 19-25, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27127406

RESUMO

The human intestinal microbiome is a microbial ecosystem that expresses as many as 100 times more genes than the human host, thereby constituting an important component of the human holobiome, which contributes to multiple health and disease processes. As most commensal species are difficult or impossible to culture, genomic characterization of microbiome composition and function, under various environmental conditions, comprises a central tool in understanding its roles in health and disease. The first decade of microbiome research was mainly characterized by usage of DNA sequencing-based 16S rDNA and shotgun metagenome sequencing, allowing for the elucidation of microbial composition and genome structure. Technological advances in RNA-seq have recently provided us with an ability to gain insight into the genes that are actively expressed in complex bacterial communities, enabling the elucidation of the functional changes that dictate the microbiome functions at given contexts, its interactions with the host, and functional alterations that accompany the conversion of a healthy microbiome toward a disease-driving configuration. Here, we highlight some of the key metatranscriptomics strategies that are implemented to determine microbiota gene expression and its regulation and discuss the advantages and potential challenges associated with these approaches.

7.
Cell ; 163(6): 1428-43, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26638072

RESUMO

Host-microbiome co-evolution drives homeostasis and disease susceptibility, yet regulatory principles governing the integrated intestinal host-commensal microenvironment remain obscure. While inflammasome signaling participates in these interactions, its activators and microbiome-modulating mechanisms are unknown. Here, we demonstrate that the microbiota-associated metabolites taurine, histamine, and spermine shape the host-microbiome interface by co-modulating NLRP6 inflammasome signaling, epithelial IL-18 secretion, and downstream anti-microbial peptide (AMP) profiles. Distortion of this balanced AMP landscape by inflammasome deficiency drives dysbiosis development. Upon fecal transfer, colitis-inducing microbiota hijacks this microenvironment-orchestrating machinery through metabolite-mediated inflammasome suppression, leading to distorted AMP balance favoring its preferential colonization. Restoration of the metabolite-inflammasome-AMP axis reinstates a normal microbiota and ameliorates colitis. Together, we identify microbial modulators of the NLRP6 inflammasome and highlight mechanisms by which microbiome-host interactions cooperatively drive microbial community stability through metabolite-mediated innate immune modulation. Therefore, targeted "postbiotic" metabolomic intervention may restore a normal microenvironment as treatment or prevention of dysbiosis-driven diseases.


Assuntos
Colo/imunologia , Colo/microbiologia , Inflamassomos/imunologia , Microbiota , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Animais , Peptídeos Catiônicos Antimicrobianos , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colo/metabolismo , Disbiose/metabolismo , Vida Livre de Germes , Doenças Inflamatórias Intestinais/induzido quimicamente , Doenças Inflamatórias Intestinais/tratamento farmacológico , Interleucina-18/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Superfície Celular/genética , Taurina/administração & dosagem
8.
Cell ; 163(5): 1079-1094, 2015 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-26590418

RESUMO

Elevated postprandial blood glucose levels constitute a global epidemic and a major risk factor for prediabetes and type II diabetes, but existing dietary methods for controlling them have limited efficacy. Here, we continuously monitored week-long glucose levels in an 800-person cohort, measured responses to 46,898 meals, and found high variability in the response to identical meals, suggesting that universal dietary recommendations may have limited utility. We devised a machine-learning algorithm that integrates blood parameters, dietary habits, anthropometrics, physical activity, and gut microbiota measured in this cohort and showed that it accurately predicts personalized postprandial glycemic response to real-life meals. We validated these predictions in an independent 100-person cohort. Finally, a blinded randomized controlled dietary intervention based on this algorithm resulted in significantly lower postprandial responses and consistent alterations to gut microbiota configuration. Together, our results suggest that personalized diets may successfully modify elevated postprandial blood glucose and its metabolic consequences. VIDEO ABSTRACT.


Assuntos
Algoritmos , Glicemia/análise , Diabetes Mellitus Tipo 2/sangue , Período Pós-Prandial , Diabetes Mellitus Tipo 2/dietoterapia , Diabetes Mellitus Tipo 2/microbiologia , Dieta para Diabéticos , Microbioma Gastrointestinal , Humanos , Smartphone
9.
Gut Microbes ; 6(2): 149-55, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25831243

RESUMO

Non-caloric artificial sweeteners (NAS) are common food supplements consumed by millions worldwide as means of combating weight gain and diabetes, by retaining sweet taste without increasing caloric intake. While they are considered safe, there is increasing controversy regarding their potential ability to promote metabolic derangements in some humans. We recently demonstrated that NAS consumption could induce glucose intolerance in mice and distinct human subsets, by functionally altering the gut microbiome. In this commentary, we discuss these findings in the context of previous and recent works demonstrating the effects of NAS on host health and the microbiome, and the challenges and open questions that need to be addressed in understanding the effects of NAS consumption on human health.


Assuntos
Dieta/métodos , Microbioma Gastrointestinal/efeitos dos fármacos , Edulcorantes/metabolismo , Animais , Intolerância à Glucose , Humanos , Camundongos
10.
Cell ; 159(3): 514-29, 2014 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-25417104

RESUMO

All domains of life feature diverse molecular clock machineries that synchronize physiological processes to diurnal environmental fluctuations. However, no mechanisms are known to cross-regulate prokaryotic and eukaryotic circadian rhythms in multikingdom ecosystems. Here, we show that the intestinal microbiota, in both mice and humans, exhibits diurnal oscillations that are influenced by feeding rhythms, leading to time-specific compositional and functional profiles over the course of a day. Ablation of host molecular clock components or induction of jet lag leads to aberrant microbiota diurnal fluctuations and dysbiosis, driven by impaired feeding rhythmicity. Consequently, jet-lag-induced dysbiosis in both mice and humans promotes glucose intolerance and obesity that are transferrable to germ-free mice upon fecal transplantation. Together, these findings provide evidence of coordinated metaorganism diurnal rhythmicity and offer a microbiome-dependent mechanism for common metabolic disturbances in humans with aberrant circadian rhythms, such as those documented in shift workers and frequent flyers.


Assuntos
Relógios Circadianos , Ritmo Circadiano , Intolerância à Glucose , Microbiota , Animais , Disbiose/microbiologia , Disbiose/fisiopatologia , Comportamento Alimentar , Homeostase , Humanos , Síndrome do Jet Lag/fisiopatologia , Doenças Metabólicas/microbiologia , Doenças Metabólicas/fisiopatologia , Camundongos , Obesidade/metabolismo , Sono
11.
Nature ; 514(7521): 181-6, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25231862

RESUMO

Non-caloric artificial sweeteners (NAS) are among the most widely used food additives worldwide, regularly consumed by lean and obese individuals alike. NAS consumption is considered safe and beneficial owing to their low caloric content, yet supporting scientific data remain sparse and controversial. Here we demonstrate that consumption of commonly used NAS formulations drives the development of glucose intolerance through induction of compositional and functional alterations to the intestinal microbiota. These NAS-mediated deleterious metabolic effects are abrogated by antibiotic treatment, and are fully transferrable to germ-free mice upon faecal transplantation of microbiota configurations from NAS-consuming mice, or of microbiota anaerobically incubated in the presence of NAS. We identify NAS-altered microbial metabolic pathways that are linked to host susceptibility to metabolic disease, and demonstrate similar NAS-induced dysbiosis and glucose intolerance in healthy human subjects. Collectively, our results link NAS consumption, dysbiosis and metabolic abnormalities, thereby calling for a reassessment of massive NAS usage.


Assuntos
Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Intolerância à Glucose/induzido quimicamente , Intolerância à Glucose/microbiologia , Microbiota/efeitos dos fármacos , Edulcorantes/efeitos adversos , Animais , Antibacterianos/farmacologia , Aspartame/efeitos adversos , Peso Corporal/efeitos dos fármacos , Dieta Hiperlipídica , Gorduras na Dieta/farmacologia , Fezes/microbiologia , Feminino , Vida Livre de Germes , Glucose/metabolismo , Intolerância à Glucose/metabolismo , Humanos , Masculino , Síndrome Metabólica/induzido quimicamente , Síndrome Metabólica/metabolismo , Síndrome Metabólica/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Sacarina/administração & dosagem , Sacarina/efeitos adversos , Sacarose/efeitos adversos , Sacarose/análogos & derivados , Relação Cintura-Quadril
12.
Nature ; 489(7414): 91-100, 2012 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-22955619

RESUMO

Transcription factors bind in a combinatorial fashion to specify the on-and-off states of genes; the ensemble of these binding events forms a regulatory network, constituting the wiring diagram for a cell. To examine the principles of the human transcriptional regulatory network, we determined the genomic binding information of 119 transcription-related factors in over 450 distinct experiments. We found the combinatorial, co-association of transcription factors to be highly context specific: distinct combinations of factors bind at specific genomic locations. In particular, there are significant differences in the binding proximal and distal to genes. We organized all the transcription factor binding into a hierarchy and integrated it with other genomic information (for example, microRNA regulation), forming a dense meta-network. Factors at different levels have different properties; for instance, top-level transcription factors more strongly influence expression and middle-level ones co-regulate targets to mitigate information-flow bottlenecks. Moreover, these co-regulations give rise to many enriched network motifs (for example, noise-buffering feed-forward loops). Finally, more connected network components are under stronger selection and exhibit a greater degree of allele-specific activity (that is, differential binding to the two parental alleles). The regulatory information obtained in this study will be crucial for interpreting personal genome sequences and understanding basic principles of human biology and disease.


Assuntos
DNA/genética , Enciclopédias como Assunto , Redes Reguladoras de Genes/genética , Genoma Humano/genética , Anotação de Sequência Molecular , Sequências Reguladoras de Ácido Nucleico/genética , Fatores de Transcrição/metabolismo , Alelos , Linhagem Celular , Fator de Transcrição GATA1/metabolismo , Perfilação da Expressão Gênica , Genômica , Humanos , Células K562 , Especificidade de Órgãos , Fosforilação/genética , Polimorfismo de Nucleotídeo Único/genética , Mapas de Interação de Proteínas , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Seleção Genética/genética , Sítio de Iniciação de Transcrição
13.
Recent Pat DNA Gene Seq ; 6(3): 180-8, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22762737

RESUMO

Our genes influence our athletic ability. However, the causal genetic factors and mechanisms, and the extent of their effects, remain largely elusive. Many studies investigate this association between specific genes and athletic performance. Such studies have increased in number over the past few years, as recent developments and patents in DNA sequencing have made large amounts of sequencing data available for such analysis. In this paper, we consider four of the most intensively studied genes in relation to athletic ability: angiotensin I-converting enzyme, alpha-actinin 3, peroxismose proliferator-activator receptor alpha and nitric oxide synthase 3. We investigate the connection between genotype and athletic phenotype in the context of these four genes in various sport fields and across different ethnicities and genders. We do an extensive literature survey on these genes and the polymorphisms (single nucleotide polymorphisms or indels) found to be associated with athletic performance. We also present, for each of these polymorphisms, the allele frequencies in the different ethnicities reported in the pilot phase of the 1000 Genomes Project - arguably the largest human genome-sequencing endeavor to date. We discuss the considerable success, and significant drawbacks, of past research along these lines, and propose interesting directions for future research.


Assuntos
Actinina/genética , Estudos de Associação Genética , Genoma Humano , Óxido Nítrico Sintase Tipo III/genética , PPAR alfa/genética , Peptidil Dipeptidase A/genética , Esportes/fisiologia , Alelos , Atletas , Etnicidade/genética , Frequência do Gene , Genótipo , Projeto Genoma Humano , Humanos , Polimorfismo de Nucleotídeo Único
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...