Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Syst Biol ; 20(4): 338-361, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38467837

RESUMO

Microbial biochemistry is central to the pathophysiology of inflammatory bowel diseases (IBD). Improved knowledge of microbial metabolites and their immunomodulatory roles is thus necessary for diagnosis and management. Here, we systematically analyzed the chemical, ecological, and epidemiological properties of ~82k metabolic features in 546 Integrative Human Microbiome Project (iHMP/HMP2) metabolomes, using a newly developed methodology for bioactive compound prioritization from microbial communities. This suggested >1000 metabolic features as potentially bioactive in IBD and associated ~43% of prevalent, unannotated features with at least one well-characterized metabolite, thereby providing initial information for further characterization of a significant portion of the fecal metabolome. Prioritized features included known IBD-linked chemical families such as bile acids and short-chain fatty acids, and less-explored bilirubin, polyamine, and vitamin derivatives, and other microbial products. One of these, nicotinamide riboside, reduced colitis scores in DSS-treated mice. The method, MACARRoN, is generalizable with the potential to improve microbial community characterization and provide therapeutic candidates.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Humanos , Animais , Camundongos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/metabolismo , Metaboloma , Ácidos e Sais Biliares
2.
Cell Host Microbe ; 30(9): 1295-1310.e8, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-35985335

RESUMO

The intestinal epithelium plays critical roles in sensing and integrating dietary and microbial signals. How microbiota and intestinal epithelial cell (IEC) interactions regulate host physiology in the proximal small intestine, particularly the duodenum, is unclear. Using single-cell RNA sequencing of duodenal IECs under germ-free (GF) and different conventional microbiota compositions, we show that specific microbiota members alter epithelial homeostasis by increasing epithelial turnover rate, crypt proliferation, and major histocompatibility complex class II (MHCII) expression. Microbiome profiling identified Faecalibaculum rodentium as a key species involved in this regulation. F. rodentium decreases enterocyte expression of retinoic-acid-producing enzymes Adh1, Aldh1a1, and Rdh7, reducing retinoic acid signaling required to maintain certain intestinal eosinophil populations. Eosinophils suppress intraepithelial-lymphocyte-mediated production of interferon-γ that regulates epithelial cell function. Thus, we identify a retinoic acid-eosinophil-interferon-γ-dependent circuit by which the microbiota modulates duodenal epithelial homeostasis.


Assuntos
Eosinófilos , Tretinoína , Citrobacter rodentium , Células Epiteliais/metabolismo , Firmicutes , Homeostase , Interferon gama/metabolismo , Mucosa Intestinal/metabolismo , Tretinoína/metabolismo
3.
Immunohorizons ; 4(1): 23-32, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31980480

RESUMO

Tuft cells are an epithelial cell type critical for initiating type 2 immune responses to parasites and protozoa in the small intestine. To respond to these stimuli, intestinal tuft cells use taste chemosensory signaling pathways, but the role of taste receptors in type 2 immunity is poorly understood. In this study, we show that the taste receptor TAS1R3, which detects sweet and umami in the tongue, also regulates tuft cell responses in the distal small intestine. BALB/c mice, which have an inactive form of TAS1R3, as well as Tas1r3-deficient C57BL6/J mice both have severely impaired responses to tuft cell-inducing signals in the ileum, including the protozoa Tritrichomonas muris and succinate. In contrast, TAS1R3 is not required to mount an immune response to the helminth Heligmosomoides polygyrus, which infects the proximal small intestine. Examination of uninfected Tas1r3-/- mice revealed a modest reduction in the number of tuft cells in the proximal small intestine but a severe decrease in the distal small intestine at homeostasis. Together, these results suggest that TAS1R3 influences intestinal immunity by shaping the epithelial cell landscape at steady-state.


Assuntos
Células Epiteliais/imunologia , Mucosa Intestinal/imunologia , Intestino Delgado/imunologia , Receptores Acoplados a Proteínas G/imunologia , Receptores Acoplados a Proteínas G/metabolismo , Animais , Células Epiteliais/metabolismo , Microbioma Gastrointestinal , Homeostase , Íleo/imunologia , Íleo/parasitologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitologia , Intestino Delgado/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nematospiroides dubius/imunologia , Receptores Acoplados a Proteínas G/deficiência , Infecções por Strongylida/imunologia , Infecções por Strongylida/parasitologia , Paladar/fisiologia , Tritrichomonas/imunologia
4.
Gastroenterology ; 158(5): 1359-1372.e9, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31917258

RESUMO

BACKGROUND & AIMS: Intestinal microbes and their metabolites affect the development of colorectal cancer (CRC). Short-chain fatty acids are metabolites generated by intestinal microbes from dietary fiber. We investigated the mechanisms by which free fatty acid receptor 2 (FFAR2), a receptor for short-chain fatty acids that can affect the composition of the intestinal microbiome, contributes to the pathogenesis of CRC. METHODS: We performed studies with ApcMin/+ mice, ApcMin/+Ffar2-/- mice, mice with conditional disruption of Ffar2 in dendritic cells (DCs) (Ffar2fl/flCD11c-Cre mice), ApcMin/+Ffar2fl/flCD11c-Cre mice, and Ffar2fl/fl mice (controls); some mice were given dextran sodium sulfate to induce colitis, with or without a FFAR2 agonist or an antibody against interleukin 27 (IL27). Colon and tumor tissues were analyzed by histology, quantitative polymerase chain reaction, and 16S ribosomal RNA gene sequencing; lamina propria and mesenteric lymph node tissues were analyzed by RNA sequencing and flow cytometry. Intestinal permeability was measured after gavage with fluorescently labeled dextran. We collected data on colorectal tumors from The Cancer Genome Atlas. RESULTS: ApcMin/+Ffar2-/- mice developed significantly more spontaneous colon tumors than ApcMin/+ mice and had increased gut permeability before tumor development, associated with reduced expression of E-cadherin. Colon tumors from ApcMin/+Ffar2-/- mice had a higher number of bacteria than tumors from ApcMin/+ mice, as well as higher frequencies of CD39+CD8+ T cells and exhausted or dying T cells. DCs from ApcMin/+Ffar2-/- mice had an altered state of activation, increased death, and higher production of IL27. Administration of an antibody against IL27 reduced the numbers of colon tumors in ApcMin/+ mice with colitis. Frequencies of CD39+CD8+ T cells and IL27+ DCs were increased in colon lamina propria from Ffar2fl/flCD11c-Cre mice with colitis compared with control mice or mice without colitis. ApcMin/+Ffar2fl/flCD11c-Cre mice developed even more tumors than ApcMin/+Ffar2fl/fl mice, and their tumors had even higher numbers of IL27+ DCs. ApcMin/+ mice with colitis given the FFAR2 agonist developed fewer colon tumors, with fewer IL27+ DCs, than mice not given the agonist. DCs incubated with the FFAR2 agonist no longer had gene expression patterns associated with activation or IL27 production. CONCLUSIONS: Loss of FFAR2 promotes colon tumorigenesis in mice by reducing gut barrier integrity, increasing tumor bacterial load, promoting exhaustion of CD8+ T cells, and overactivating DCs, leading to their death. Antibodies against IL27 and an FFAR2 agonist reduce tumorigenesis in mice and might be developed for the treatment of CRC.


Assuntos
Colite/patologia , Neoplasias do Colo/imunologia , Células Dendríticas/imunologia , Microbioma Gastrointestinal/imunologia , Interleucinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinogênese/imunologia , Colite/induzido quimicamente , Colite/imunologia , Colo/efeitos dos fármacos , Colo/microbiologia , Colo/patologia , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Células Dendríticas/metabolismo , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Progressão da Doença , Ácidos Graxos não Esterificados/metabolismo , Feminino , Humanos , Interleucinas/imunologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Knockout , Permeabilidade , Cultura Primária de Células , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética
5.
Immunity ; 51(5): 871-884.e6, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31628054

RESUMO

Group 3 innate lymphoid cells (ILC3s) sense environmental signals that are critical for gut homeostasis and host defense. However, the metabolite-sensing G-protein-coupled receptors that regulate colonic ILC3s remain poorly understood. We found that colonic ILC3s expressed Ffar2, a microbial metabolite-sensing receptor, and that Ffar2 agonism promoted ILC3 expansion and function. Deficiency of Ffar2 in ILC3s decreased their in situ proliferation and ILC3-derived interleukin-22 (IL-22) production. This led to impaired gut epithelial function characterized by altered mucus-associated proteins and antimicrobial peptides and increased susceptibility to colonic injury and bacterial infection. Ffar2 increased IL-22+ CCR6+ ILC3s and influenced ILC3 abundance in colonic lymphoid tissues. Ffar2 agonism differentially activated AKT or ERK signaling and increased ILC3-derived IL-22 via an AKT and STAT3 axis. Our findings suggest that Ffar2 regulates colonic ILC3 proliferation and function, and they identify an ILC3-receptor signaling pathway modulating gut homeostasis and pathogen defense.


Assuntos
Imunidade Inata , Imunidade nas Mucosas , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Linfócitos/imunologia , Linfócitos/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Biomarcadores , Citocinas/metabolismo , Suscetibilidade a Doenças , Microbioma Gastrointestinal/imunologia , Expressão Gênica , Humanos , Imunomodulação , Mucosa Intestinal/patologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt , Receptores de Superfície Celular/agonistas , Fator de Transcrição STAT3/metabolismo
6.
Emerg Microbes Infect ; 8(1): 827-840, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31169073

RESUMO

Campylobacter showae a bacterium historically linked to gingivitis and periodontitis, has recently been associated with inflammatory bowel disease and colorectal cancer. Our aim was to generate genome sequences for new clinical C. showae strains and identify functional properties explaining their pathogenic potential. Eight C. showae genomes were assessed, four strains isolated from inflamed gut tissues from paediatric Crohn's disease patients, three strains from colonic adenomas, and one from a gastroenteritis patient stool. Genome assemblies were analyzed alongside the only 3 deposited C. showae genomes. The pangenome from these 11 strains consisted of 4686 unique protein families, and the core genome size was estimated at 1050 ± 15 genes with each new genome contributing an additional 206 ± 16 genes. Functional assays indicated that colonic strains segregated into 2 groups: adherent/invasive vs. non-adherent/non-invasive strains. The former possessed Type IV secretion machinery and S-layer proteins, while the latter contained Cas genes and other CRISPR associated proteins. Comparison of gene profiles with strains in Human Microbiome Project metagenomes showed that gut-derived isolates share genes specific to tongue dorsum and supragingival plaque counterparts. Our findings indicate that C. showae strains are phenotypically and genetically diverse and suggest that secretion systems may play an important role in virulence potential.


Assuntos
Infecções por Campylobacter/microbiologia , Campylobacter/genética , Genoma Bacteriano , Proteínas de Bactérias/genética , Campylobacter/classificação , Campylobacter/isolamento & purificação , Campylobacter/patogenicidade , Doença de Crohn/microbiologia , Gastroenterite/microbiologia , Genômica , Humanos , Fenótipo , Filogenia , Virulência , Fatores de Virulência/genética
7.
Elife ; 82019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30666959

RESUMO

Inflammatory bowel disease (IBD) is driven by dysfunction between host genetics, the microbiota, and immune system. Knowledge gaps remain regarding how IBD genetic risk loci drive gut microbiota changes. The Crohn's disease risk allele ATG16L1 T300A results in abnormal Paneth cells due to decreased selective autophagy, increased cytokine release, and decreased intracellular bacterial clearance. To unravel the effects of ATG16L1 T300A on the microbiota and immune system, we employed a gnotobiotic model using human fecal transfers into ATG16L1 T300A knock-in mice. We observed increases in Bacteroides ovatus and Th1 and Th17 cells in ATG16L1 T300A mice. Association of altered Schaedler flora mice with B. ovatus specifically increased Th17 cells selectively in ATG16L1 T300A knock-in mice. Changes occur before disease onset, suggesting that ATG16L1 T300A contributes to dysbiosis and immune infiltration prior to disease symptoms. Our work provides insight for future studies on IBD subtypes, IBD patient treatment and diagnostics.


Assuntos
Proteínas Relacionadas à Autofagia/genética , Doença de Crohn/genética , Doença de Crohn/microbiologia , Microbioma Gastrointestinal , Células Th1/citologia , Células Th17/citologia , Alelos , Animais , Bacteroides , Disbiose/genética , Disbiose/microbiologia , Transplante de Microbiota Fecal , Fezes/microbiologia , Técnicas de Introdução de Genes , Genótipo , Humanos , Sistema Imunitário , Camundongos , Polimorfismo Genético , Risco , Células Th1/microbiologia , Células Th17/microbiologia
8.
Science ; 357(6356): 1156-1160, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28912244

RESUMO

Growing evidence suggests that microbes can influence the efficacy of cancer therapies. By studying colon cancer models, we found that bacteria can metabolize the chemotherapeutic drug gemcitabine (2',2'-difluorodeoxycytidine) into its inactive form, 2',2'-difluorodeoxyuridine. Metabolism was dependent on the expression of a long isoform of the bacterial enzyme cytidine deaminase (CDDL), seen primarily in Gammaproteobacteria. In a colon cancer mouse model, gemcitabine resistance was induced by intratumor Gammaproteobacteria, dependent on bacterial CDDL expression, and abrogated by cotreatment with the antibiotic ciprofloxacin. Gemcitabine is commonly used to treat pancreatic ductal adenocarcinoma (PDAC), and we hypothesized that intratumor bacteria might contribute to drug resistance of these tumors. Consistent with this possibility, we found that of the 113 human PDACs that were tested, 86 (76%) were positive for bacteria, mainly Gammaproteobacteria.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/microbiologia , Desoxicitidina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/microbiologia , Animais , Neoplasias do Colo/microbiologia , Desoxicitidina/uso terapêutico , Gammaproteobacteria/isolamento & purificação , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mycoplasma hyorhinis/isolamento & purificação , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/microbiologia , Gencitabina , Neoplasias Pancreáticas
9.
Proc Natl Acad Sci U S A ; 114(1): 142-147, 2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-27980034

RESUMO

Hosts and their microbes have established a sophisticated communication system over many millennia. Within mammalian hosts, this dynamic cross-talk is essential for maintaining intestinal homeostasis. In a genetically susceptible host, dysbiosis of the gut microbiome and dysregulated immune responses are central to the development of inflammatory bowel disease (IBD). Previous surveys of stool from the T-bet-/-Rag2-/- IBD mouse model revealed microbial features that discriminate between health and disease states. Enterobacteriaceae expansion and increased gene abundances for benzoate degradation, two-component systems, and bacterial motility proteins pointed to the potential involvement of a catecholamine-mediated bacterial signaling axis in colitis pathogenesis. Enterobacteriaceae sense and respond to microbiota-generated signals and host-derived catecholamines through the two-component quorum-sensing Escherichia coli regulators B and C (QseBC) system. On signal detection, QseC activates a cascade to induce virulence gene expression. Although a single pathogen has not been identified as a causative agent in IBD, adherent-invasive Escherichia coli (AIEC) have been implicated. Flagellar expression is necessary for the IBD-associated AIEC strain LF82 to establish colonization. Thus, we hypothesized that qseC inactivation could reduce LF82's virulence, and found that an absence of qseC leads to down-regulated flagellar expression and motility in vitro and reduced colonization in vivo. We extend these findings on the potential of QseC-based IBD therapeutics to three preclinical IBD models, wherein we observe that QseC blockade can effectively modulate colitogenic microbiotas to reduce intestinal inflammation. Collectively, our data support a role for QseC-mediated bacterial signaling in IBD pathogenesis and indicate that QseC inhibition may be a useful microbiota-targeted approach for disease management.


Assuntos
Colite/patologia , Colite/terapia , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/genética , Escherichia coli/metabolismo , Percepção de Quorum/efeitos dos fármacos , Animais , Catecolaminas/metabolismo , Colite/microbiologia , Flagelos/genética , Flagelos/metabolismo , Microbioma Gastrointestinal , Regulação Bacteriana da Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Sulfonamidas/farmacologia , Virulência/genética
10.
Science ; 351(6279): 1329-33, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26847546

RESUMO

The intestinal epithelium forms an essential barrier between a host and its microbiota. Protozoa and helminths are members of the gut microbiota of mammals, including humans, yet the many ways that gut epithelial cells orchestrate responses to these eukaryotes remain unclear. Here we show that tuft cells, which are taste-chemosensory epithelial cells, accumulate during parasite colonization and infection. Disruption of chemosensory signaling through the loss of TRMP5 abrogates the expansion of tuft cells, goblet cells, eosinophils, and type 2 innate lymphoid cells during parasite colonization. Tuft cells are the primary source of the parasite-induced cytokine interleukin-25, which indirectly induces tuft cell expansion by promoting interleukin-13 production by innate lymphoid cells. Our results identify intestinal tuft cells as critical sentinels in the gut epithelium that promote type 2 immunity in response to intestinal parasites.


Assuntos
Células Quimiorreceptoras/imunologia , Enteropatias Parasitárias/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/parasitologia , Microbiota/imunologia , Canais de Cátion TRPM/imunologia , Animais , Quinases Semelhantes a Duplacortina , Eosinófilos/imunologia , Células Caliciformes/imunologia , Helmintíase/imunologia , Helmintíase/parasitologia , Helmintos/imunologia , Imunidade nas Mucosas , Interleucina-13/imunologia , Interleucina-17/imunologia , Enteropatias Parasitárias/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Serina-Treonina Quinases/imunologia , Infecções por Protozoários/imunologia , Infecções por Protozoários/parasitologia , Transdução de Sinais , Paladar , Transducina/genética , Transducina/imunologia , Tritrichomonas/imunologia
11.
Cell Rep ; 12(2): 244-57, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-26146082

RESUMO

Our study reveals a non-canonical role for CCL2 in modulating non-macrophage, myeloid-derived suppressor cells (MDSCs) and shaping a tumor-permissive microenvironment during colon cancer development. We found that intratumoral CCL2 levels increased in patients with colitis-associated colorectal cancer (CRC), adenocarcinomas, and adenomas. Deletion of CCL2 blocked progression from dysplasia to adenocarcinoma and reduced the number of colonic MDSCs in a spontaneous mouse model of colitis-associated CRC. In a transplantable mouse model of adenocarcinoma and an APC-driven adenoma model, CCL2 fostered MDSC accumulation in evolving colonic tumors and enhanced polymorphonuclear (PMN)-MDSC immunosuppressive features. Mechanistically, CCL2 regulated T cell suppression of PMN-MDSCs in a STAT3-mediated manner. Furthermore, CCL2 neutralization decreased tumor numbers and MDSC accumulation and function. Collectively, our experiments support that perturbing CCL2 and targeting MDSCs may afford therapeutic opportunities for colon cancer interception and prevention.


Assuntos
Adenocarcinoma/patologia , Quimiocina CCL2/metabolismo , Neoplasias do Colo/patologia , Adenocarcinoma/etiologia , Adenocarcinoma/metabolismo , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Transformação Celular Neoplásica , Quimiocina CCL2/antagonistas & inibidores , Quimiocina CCL2/genética , Colite/complicações , Colite/patologia , Neoplasias do Colo/etiologia , Neoplasias do Colo/metabolismo , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Células Mieloides/citologia , Células Mieloides/metabolismo , Neutrófilos/citologia , Neutrófilos/metabolismo , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Fator de Transcrição STAT3/metabolismo , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética
12.
Proc Natl Acad Sci U S A ; 112(25): 7803-8, 2015 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-26056274

RESUMO

Beneficial microbes that target molecules and pathways, such as oxidative stress, which can negatively affect both host and microbiota, may hold promise as an inflammatory bowel disease therapy. Prior work showed that a five-strain fermented milk product (FMP) improved colitis in T-bet(-/-) Rag2(-/-) mice. By varying the number of strains used in the FMP, we found that Lactococcus lactis I-1631 was sufficient to ameliorate colitis. Using comparative genomic analyses, we identified genes unique to L. lactis I-1631 involved in oxygen respiration. Respiration of oxygen results in reactive oxygen species (ROS) generation. Also, ROS are produced at high levels during intestinal inflammation and cause tissue damage. L. lactis I-1631 possesses genes encoding enzymes that detoxify ROS, such as superoxide dismutase (SodA). Thus, we hypothesized that lactococcal SodA played a role in attenuating colitis. Inactivation of the sodA gene abolished L. lactis I-1631's beneficial effect in the T-bet(-/-) Rag2(-/-) model. Similar effects were obtained in two additional colonic inflammation models, Il10(-/-) mice and dextran sulfate sodium-treated mice. Efforts to understand how a lipophobic superoxide anion (O2 (-)) can be detoxified by cytoplasmic lactoccocal SodA led to the finding that host antimicrobial-mediated lysis is a prerequisite for SodA release and SodA's extracytoplasmic O2 (-) scavenging. L. lactis I-1631 may represent a promising vehicle to deliver antioxidant, colitis-attenuating SodA to the inflamed intestinal mucosa, and host antimicrobials may play a critical role in mediating SodA's bioaccessibility.


Assuntos
Colite/metabolismo , Lactococcus lactis/metabolismo , Muramidase/metabolismo , Superóxido Dismutase/metabolismo , Animais , Colite/enzimologia , Colite/microbiologia , Mucosa Intestinal/enzimologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Camundongos , Espécies Reativas de Oxigênio/metabolismo
13.
ISME J ; 8(7): 1403-17, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24500617

RESUMO

Dysregulated immune responses to gut microbes are central to inflammatory bowel disease (IBD), and gut microbial activity can fuel chronic inflammation. Examining how IBD-directed therapies influence gut microbiomes may identify microbial community features integral to mitigating disease and maintaining health. However, IBD patients often receive multiple treatments during disease flares, confounding such analyses. Preclinical models of IBD with well-defined disease courses and opportunities for controlled treatment exposures provide a valuable solution. Here, we surveyed the gut microbiome of the T-bet(-/-) Rag2(-/-) mouse model of colitis during active disease and treatment-induced remission. Microbial features modified among these conditions included altered potential for carbohydrate and energy metabolism and bacterial pathogenesis, specifically cell motility and signal transduction pathways. We also observed an increased capacity for xenobiotics metabolism, including benzoate degradation, a pathway linking host adrenergic stress with enhanced bacterial virulence, and found decreased levels of fecal dopamine in active colitis. When transferred to gnotobiotic mice, gut microbiomes from mice with active disease versus treatment-induced remission elicited varying degrees of colitis. Thus, our study provides insight into specific microbial clades and pathways associated with health, active disease and treatment interventions in a mouse model of colitis.


Assuntos
Colite/microbiologia , Trato Gastrointestinal/microbiologia , Doenças Inflamatórias Intestinais/microbiologia , Microbiota/genética , Animais , Antibacterianos/farmacologia , Ácido Benzoico/metabolismo , Metabolismo dos Carboidratos , Movimento Celular , Colite/tratamento farmacológico , Colite/genética , Colite/patologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Dopamina/metabolismo , Metabolismo Energético , Humanos , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/microbiologia , Inflamação/patologia , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Camundongos , Camundongos Knockout , Microbiota/efeitos dos fármacos , Filogenia , RNA Ribossômico 16S/classificação , RNA Ribossômico 16S/genética , Indução de Remissão , Transdução de Sinais , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética
14.
Cell Host Microbe ; 14(2): 207-15, 2013 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-23954159

RESUMO

Increasing evidence links the gut microbiota with colorectal cancer. Metagenomic analyses indicate that symbiotic Fusobacterium spp. are associated with human colorectal carcinoma, but whether this is an indirect or causal link remains unclear. We find that Fusobacterium spp. are enriched in human colonic adenomas relative to surrounding tissues and in stool samples from colorectal adenoma and carcinoma patients compared to healthy subjects. Additionally, in the Apc(Min/+) mouse model of intestinal tumorigenesis, Fusobacterium nucleatum increases tumor multiplicity and selectively recruits tumor-infiltrating myeloid cells, which can promote tumor progression. Tumors from Apc(Min/+) mice exposed to F. nucleatum exhibit a proinflammatory expression signature that is shared with human fusobacteria-positive colorectal carcinomas. However, unlike other bacteria linked to colorectal carcinoma, F. nucleatum does not exacerbate colitis, enteritis, or inflammation-associated intestinal carcinogenesis. Collectively, these data suggest that, through recruitment of tumor-infiltrating immune cells, fusobacteria generate a proinflammatory microenvironment that is conducive for colorectal neoplasia progression.


Assuntos
Carcinogênese/imunologia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/microbiologia , Fusobacterium nucleatum/imunologia , Fusobacterium nucleatum/patogenicidade , Adenoma/imunologia , Adenoma/microbiologia , Adenoma/patologia , Animais , Neoplasias Colorretais/patologia , Citocinas/biossíntese , Modelos Animais de Doenças , Humanos , Leucócitos/imunologia , Camundongos
15.
N Engl J Med ; 369(6): 517-28, 2013 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-23924002

RESUMO

BACKGROUND: Immunosuppression is associated with a variety of idiopathic clinical syndromes that may have infectious causes. It has been hypothesized that the cord colitis syndrome, a complication of umbilical-cord hematopoietic stem-cell transplantation, is infectious in origin. METHODS: We performed shotgun DNA sequencing on four archived, paraffin-embedded endoscopic colon-biopsy specimens obtained from two patients with cord colitis. Computational subtraction of human and known microbial sequences and assembly of residual sequences into a bacterial draft genome were performed. We used polymerase-chain-reaction (PCR) assays and fluorescence in situ hybridization to determine whether the corresponding bacterium was present in additional patients and controls. RESULTS: DNA sequencing of the biopsy specimens revealed more than 2.5 million sequencing reads that did not match known organisms. These sequences were computationally assembled into a 7.65-Mb draft genome showing a high degree of homology with genomes of bacteria in the bradyrhizobium genus. The corresponding newly discovered bacterium was provisionally named Bradyrhizobium enterica. PCR identified B. enterica nucleotide sequences in biopsy specimens from all three additional patients with cord colitis whose samples were tested, whereas B. enterica sequences were absent in samples obtained from healthy controls and patients with colon cancer or graft-versus-host disease. CONCLUSIONS: We assembled a novel bacterial draft genome from the direct sequencing of tissue specimens from patients with cord colitis. Association of these sequences with cord colitis suggests that B. enterica may be an opportunistic human pathogen. (Funded by the National Cancer Institute and others.)


Assuntos
Bradyrhizobium/genética , Colite/microbiologia , Colo/microbiologia , Sangue Fetal , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Infecções Oportunistas/microbiologia , Biópsia , Bradyrhizobium/classificação , Bradyrhizobium/isolamento & purificação , Colite/imunologia , Neoplasias do Colo/microbiologia , DNA Bacteriano/análise , Diarreia/microbiologia , Feminino , Genoma Bacteriano , Doença Enxerto-Hospedeiro/microbiologia , Humanos , Hospedeiro Imunocomprometido , Masculino , Inclusão em Parafina , Filogenia , Reação em Cadeia da Polimerase , Análise de Sequência de DNA
16.
Science ; 341(6145): 569-73, 2013 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-23828891

RESUMO

Regulatory T cells (Tregs) that express the transcription factor Foxp3 are critical for regulating intestinal inflammation. Candidate microbe approaches have identified bacterial species and strain-specific molecules that can affect intestinal immune responses, including species that modulate Treg responses. Because neither all humans nor mice harbor the same bacterial strains, we posited that more prevalent factors exist that regulate the number and function of colonic Tregs. We determined that short-chain fatty acids, gut microbiota-derived bacterial fermentation products, regulate the size and function of the colonic Treg pool and protect against colitis in a Ffar2-dependent manner in mice. Our study reveals that a class of abundant microbial metabolites underlies adaptive immune microbiota coadaptation and promotes colonic homeostasis and health.


Assuntos
Bactérias/metabolismo , Colo/microbiologia , Ácidos Graxos Voláteis/metabolismo , Homeostase , Metagenoma , Linfócitos T Reguladores/fisiologia , Animais , Colite/metabolismo , Proteínas de Ligação a DNA/genética , Ácidos Graxos Voláteis/administração & dosagem , Fermentação , Vida Livre de Germes , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Linfócitos T Reguladores/transplante
17.
J Immunol ; 190(8): 3977-84, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23479229

RESUMO

CD8(+) T cells are fundamental for immune-mediated clearance of viral infections and contribute to immune pathology in autoimmune diseases such as type 1 diabetes. To execute these functions, CD8(+) T cells must differentiate into CTLs, a process that is precisely regulated by a variety of cytokines, costimulatory molecules, and transcription factors. IL-21 is an IL-2 family cytokine and a growth factor for multiple lymphocyte effector lineages, including cytotoxic CD8(+) T cells. Recent studies demonstrate that loss of IL-21 signaling results in reduced viral clearance in models of lymphocytic choriomeningitis virus infection, and also protection from type 1 diabetes in the NOD model. This is most likely the result of impaired CD8(+) CTL function in the absence of IL-21 signaling. Currently, the mechanisms by which IL-21 promotes CTL differentiation in CD8(+) T cells remain unclear, particularly the identity of the relevant transcription factor(s). We show that IL-21 promotes CTL function in vitro and killing of pancreatic islets in vivo via the use of transgenic mice expressing IL-21 in pancreatic ß cells. We demonstrate that IL-21 induces the expression of the transcription factor T-bet in CD8(+) T cells, predominantly via STAT1, and that T-bet is required for the induction of cytolytic molecules, including perforin and granzyme B in response to IL-21. Finally, we show that IL-21-induced CTL function is T-bet dependent, as T-bet deficiency results in defective IL-21-dependent cytotoxicity in CD8(+) T cells in vitro and in vivo. Thus, IL-21 drives CD8(+) CTL differentiation via the actions of the transcription factor T-bet.


Assuntos
Citotoxicidade Imunológica , Interleucinas/fisiologia , Proteínas com Domínio T/fisiologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Transferência Adotiva , Animais , Células Cultivadas , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
18.
Genome Res ; 22(2): 292-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22009990

RESUMO

The tumor microenvironment of colorectal carcinoma is a complex community of genomically altered cancer cells, nonneoplastic cells, and a diverse collection of microorganisms. Each of these components may contribute to carcinogenesis; however, the role of the microbiota is the least well understood. We have characterized the composition of the microbiota in colorectal carcinoma using whole genome sequences from nine tumor/normal pairs. Fusobacterium sequences were enriched in carcinomas, confirmed by quantitative PCR and 16S rDNA sequence analysis of 95 carcinoma/normal DNA pairs, while the Bacteroidetes and Firmicutes phyla were depleted in tumors. Fusobacteria were also visualized within colorectal tumors using FISH. These findings reveal alterations in the colorectal cancer microbiota; however, the precise role of Fusobacteria in colorectal carcinoma pathogenesis requires further investigation.


Assuntos
Neoplasias Colorretais/microbiologia , Fusobacterium/genética , Genoma Bacteriano , Fusobacterium/classificação , Fusobacterium/patogenicidade , Humanos , Intestino Grosso/microbiologia , Metagenoma/genética , Filogenia , RNA Ribossômico 16S/genética , Análise de Sequência de DNA
19.
Proc Natl Acad Sci U S A ; 107(42): 18132-7, 2010 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-20921388

RESUMO

Intestinal health requires the coexistence of eukaryotic self with the gut microbiota and dysregulated host-microbial interactions can result in intestinal inflammation. Here, we show that colitis improved in T-bet(-/-)Rag2(-/-) mice that consumed a fermented milk product containing Bifidobacterium animalis subsp. lactis DN-173 010 strain. A decrease in cecal pH and alterations in short chain fatty acid profiles occurred with consumption, and there were concomitant increases in the abundance of select lactate-consuming and butyrate-producing bacteria. These metabolic shifts created a nonpermissive environment for the Enterobacteriaceae recently identified as colitogenic in a T-bet(-/-)Rag2(-/-) ulcerative colitis mouse model. In addition, 16S rRNA-based analysis of the T-bet(-/-)Rag2(-/-) fecal microbiota suggest that the structure of the endogenous gut microbiota played a key role in shaping the host response to the bacterial strains studied herein. We have identified features of the gut microbiota, at the membership and functional level, associated with response to this B. lactis-containing fermented milk product, and therefore this model provides a framework for evaluating and optimizing probiotic-based functional foods.


Assuntos
Bifidobacterium/fisiologia , Colite/microbiologia , Enterobacteriaceae/patogenicidade , Inflamação/prevenção & controle , Leite , Animais , Fermentação , Camundongos , Camundongos Knockout
20.
Cell Host Microbe ; 8(3): 292-300, 2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20833380

RESUMO

Disruption of homeostasis between the host immune system and the intestinal microbiota leads to inflammatory bowel disease (IBD). Whether IBD is instigated by individual species or disruptions of entire microbial communities remains controversial. We characterized the fecal microbial communities in the recently described T-bet(-/-) ×Rag2(-/-) ulcerative colitis (TRUC) model driven by T-bet deficiency in the innate immune system. 16S rRNA-based analysis of TRUC and Rag2(-/-) mice revealed distinctive communities that correlate with host genotype. The presence of Klebsiella pneumoniae and Proteus mirabilis correlates with colitis in TRUC animals, and these TRUC-derived strains can elicit colitis in Rag2(-/-) and WT adults but require a maternally transmitted endogenous microbial community for maximal intestinal inflammation. Cross-fostering experiments indicated a role for these organisms in maternal transmission of disease. Our findings illustrate how gut microbial communities work in concert with specific culturable colitogenic agents to cause IBD.


Assuntos
Colite Ulcerativa/microbiologia , Colite Ulcerativa/patologia , Enterobacteriaceae/fisiologia , Intestinos/microbiologia , Klebsiella pneumoniae/isolamento & purificação , Interações Microbianas , Proteus mirabilis/isolamento & purificação , Animais , Colite Ulcerativa/terapia , Colo/microbiologia , Colo/patologia , Proteínas de Ligação a DNA/genética , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Enterobacteriaceae/isolamento & purificação , Fezes/microbiologia , Feminino , Hibridização in Situ Fluorescente , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Proteínas com Domínio T/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA