Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancer Prev Res (Phila) ; 13(1): 25-40, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31771941

RESUMO

Tightly regulated immune responses must occur in the intestine to avoid unwanted inflammation, which may cause chronic sequela leading to diseases such as colorectal cancer. Toll-like receptors play an important role in preventing aberrant immune responses in the intestine by sensing endogenous commensal microbiota and delivering important regulatory signals to the tissue. However, the role that specific innate receptors may play in the development of chronic inflammation and their impact on the composition of the colonic microbiota is not well understood. Using a model of inflammation-induced colorectal cancer, we found that Lactobacillus species are lost more quickly in wild-type (WT) mice than TLR6-deficient mice resulting in overall differences in bacterial composition. Despite the longer retention of Lactobacillus, the TLR6-deficient mice presented with more tumors and a worse overall outcome. Restoration of the lost Lactobacillus species suppressed inflammation, reduced tumor number, and prevented change in the abundance of Proteobacteria only when given to WT mice, indicating the effect of these Lactobacillus are TLR6 dependent. We found that the TLR6-dependent effects of Lactobacillus could be dissociated from one another via the involvement of IL10, which was necessary to dampen the inflammatory microenvironment, but had no effect on bacterial composition. Altogether, these data suggest that innate immune signals can shape the composition of the microbiota under chronic inflammatory conditions, bias the cytokine milieu of the tissue microenvironment, and influence the response to microbiota-associated therapies.


Assuntos
Colite/imunologia , Neoplasias Colorretais/imunologia , Microbioma Gastrointestinal/imunologia , Neoplasias Experimentais/imunologia , Receptor 6 Toll-Like/deficiência , Animais , Azoximetano/toxicidade , Colite/induzido quimicamente , Colite/genética , Colite/microbiologia , Colo/imunologia , Colo/microbiologia , Colo/patologia , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/genética , Neoplasias Colorretais/microbiologia , Sulfato de Dextrana/toxicidade , Feminino , Humanos , Imunidade Inata , Interleucina-10/administração & dosagem , Interleucina-10/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Lactobacillus/imunologia , Camundongos , Camundongos Knockout , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/genética , Neoplasias Experimentais/microbiologia , Probióticos/administração & dosagem , Proteobactérias/imunologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor 6 Toll-Like/genética
2.
IUBMB Life ; 71(2): 152-165, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30466159

RESUMO

It is now well appreciated that the human microbiome plays a significant role in a number of processes in the body, significantly affecting its metabolic, inflammatory, and immune homeostasis. Recent research has revealed that almost every mucosal surface in the human body is associated with a resident commensal microbiome of its own. While the gut microbiome and its role in regulation of host metabolism along with its alteration in a disease state has been well studied, there is a lacuna in understanding the resident microbiota of other mucosal surfaces. Among these, the scientific information on the role of lung microbiota in pulmonary diseases is currently severely limited. Historically, lungs have been considered to be sterile and lung diseases have only been studied in the context of bacterial pathogenesis. Recently however, studies have revealed a resilient microbiome in the upper and lower respiratory tracts and there is increased evidence on its central role in respiratory diseases. Knowledge of lung microbiome and its metabolic fallout (local and systemic) is still in its nascent stages and attracting immense interest in recent times. In this review, we will provide a perspective on lung-associated metabolic disorders defined for lung diseases (e.g., chronic obstructive pulmonary disease, asthma, and respiratory depression due to infection) and correlate it with lung microbial perturbation. Such perturbations may be due to altered biochemical or metabolic stress as well. Finally, we will draw evidence from microbiome and classical microbiology literature to demonstrate how specific lung morbidities associate with specific metabolic characteristics of the disease, and with the role of microbiome in this context. © 2018 IUBMB Life, 71(1):152-165, 2019.


Assuntos
Asma/metabolismo , Fibrose Cística/metabolismo , Neoplasias Pulmonares/metabolismo , Pneumonia Bacteriana/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Actinobacteria/imunologia , Actinobacteria/metabolismo , Actinobacteria/patogenicidade , Asma/imunologia , Asma/microbiologia , Asma/patologia , Fibrose Cística/imunologia , Fibrose Cística/microbiologia , Fibrose Cística/patologia , Firmicutes/imunologia , Firmicutes/metabolismo , Firmicutes/patogenicidade , Homeostase/imunologia , Humanos , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/microbiologia , Neoplasias Pulmonares/patologia , Microbiota/imunologia , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/patologia , Proteobactérias/imunologia , Proteobactérias/metabolismo , Proteobactérias/patogenicidade , Doença Pulmonar Obstrutiva Crônica/imunologia , Doença Pulmonar Obstrutiva Crônica/microbiologia , Doença Pulmonar Obstrutiva Crônica/patologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/microbiologia , Mucosa Respiratória/patologia
3.
Mucosal Immunol ; 11(6): 1591-1605, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30115998

RESUMO

Human mucosal-associated invariant T (MAIT) cell receptors (TCRs) recognize bacterial riboflavin pathway metabolites through the MHC class 1-related molecule MR1. However, it is unclear whether MAIT cells discriminate between many species of the human microbiota. To address this, we developed an in vitro functional assay through human T cells engineered for MAIT-TCRs (eMAIT-TCRs) stimulated by MR1-expressing antigen-presenting cells (APCs). We then screened 47 microbiota-associated bacterial species from different phyla for their eMAIT-TCR stimulatory capacities. Only bacterial species that encoded the riboflavin pathway were stimulatory for MAIT-TCRs. Most species that were high stimulators belonged to Bacteroidetes and Proteobacteria phyla, whereas low/non-stimulator species were primarily Actinobacteria or Firmicutes. Activation of MAIT cells by high- vs low-stimulating bacteria also correlated with the level of riboflavin they secreted or after bacterial infection of macrophages. Remarkably, we found that human T-cell subsets can also present riboflavin metabolites to MAIT cells in a MR1-restricted fashion. This T-T cell-mediated signaling also induced IFNγ, TNF and granzyme B from MAIT cells, albeit at lower level than professional APC. These findings suggest that MAIT cells can discriminate and categorize complex human microbiota through computation of TCR signals depending on antigen load and presenting cells, and fine-tune their functional responses.


Assuntos
Bacteroidetes/imunologia , Macrófagos/imunologia , Microbiota/imunologia , Células T Invariantes Associadas à Mucosa/imunologia , Proteobactérias/imunologia , Riboflavina/metabolismo , Apresentação de Antígeno , Antígenos de Bactérias/imunologia , Células Cultivadas , Engenharia Genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Interferon gama/metabolismo , Ativação Linfocitária , Macrófagos/microbiologia , Antígenos de Histocompatibilidade Menor/metabolismo , Células T Invariantes Associadas à Mucosa/microbiologia , Receptores de Antígenos de Linfócitos T/genética , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
4.
JCI Insight ; 3(5)2018 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-29515035

RESUMO

Premature infants are at high risk for developing bronchopulmonary dysplasia (BPD), characterized by chronic inflammation and inhibition of lung development, which we have recently identified as being modulated by microRNAs (miRNAs) and alterations in the airway microbiome. Exosomes and exosomal miRNAs may regulate cell differentiation and tissue and organ development. We discovered that tracheal aspirates from infants with severe BPD had increased numbers of, but smaller, exosomes compared with term controls. Similarly, bronchoalveolar lavage fluid from hyperoxia-exposed mice (an animal model of BPD) and supernatants from hyperoxia-exposed human bronchial epithelial cells (in vitro model of BPD) had increased exosomes compared with air controls. Next, in a prospective cohort study of tracheal aspirates obtained at birth from extremely preterm infants, utilizing independent discovery and validation cohorts, we identified unbiased exosomal miRNA signatures predictive of severe BPD. The strongest signal of reduced miR-876-3p in BPD-susceptible compared with BPD-resistant infants was confirmed in the animal model and in vitro models of BPD. In addition, based on our recent discovery of increased Proteobacteria in the airway microbiome being associated with BPD, we developed potentially novel in vivo and in vitro models for BPD combining Proteobacterial LPS and hyperoxia exposure. Addition of LPS led to a larger reduction in exosomal miR 876-3p in both hyperoxia and normoxia compared with hyperoxia alone, thus indicating a potential mechanism by which alterations in microbiota can suppress miR 876-3p. Gain of function of miR 876-3p improved the alveolar architecture in the in vivo BPD model, demonstrating a causal link between miR 876-3p and BPD. In summary, we provide evidence for the strong predictive biomarker potential of miR 876-3p in severe BPD. We also provide insights on the pathogenesis of neonatal lung disease, as modulated by hyperoxia and microbial product-induced changes in exosomal miRNA 876-3p, which could be targeted for future therapeutic development.


Assuntos
Células Epiteliais Alveolares/imunologia , Displasia Broncopulmonar/diagnóstico , Exossomos/metabolismo , Lactente Extremamente Prematuro/imunologia , MicroRNAs/metabolismo , Células Epiteliais Alveolares/citologia , Células Epiteliais Alveolares/microbiologia , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Líquido da Lavagem Broncoalveolar/citologia , Displasia Broncopulmonar/imunologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem Celular , Modelos Animais de Doenças , Exossomos/genética , Exossomos/imunologia , Feminino , Humanos , Hiperóxia/imunologia , Recém-Nascido de Peso Extremamente Baixo ao Nascer/imunologia , Recém-Nascido , Lipopolissacarídeos/imunologia , Masculino , Camundongos , MicroRNAs/genética , MicroRNAs/imunologia , Microbiota/imunologia , Prognóstico , Estudos Prospectivos , Proteobactérias/imunologia , Índice de Gravidade de Doença
5.
Cell Host Microbe ; 23(3): 302-311.e3, 2018 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-29478774

RESUMO

Serum immunoglobulin A (IgA) antibodies are readily detected in mice and people, but the mechanisms underlying the induction of serum IgA and its role in host protection remain uncertain. We report that select commensal bacteria induce several facets of systemic IgA-mediated immunity. Exposing conventional mice to a unique but natural microflora that included several members of the Proteobacteria phylum led to T cell-dependent increases in serum IgA levels and the induction of large numbers of IgA-secreting plasma cells in the bone marrow. The resulting serum IgA bound to a restricted collection of bacterial taxa, and antigen-specific serum IgA antibodies were readily induced after intestinal colonization with the commensal bacterium Helicobacter muridarum. Finally, movement to a Proteobacteria-rich microbiota led to serum IgA-mediated resistance to polymicrobial sepsis. We conclude that commensal microbes overtly influence the serum IgA repertoire, resulting in constitutive protection against bacterial sepsis.


Assuntos
Microbioma Gastrointestinal/imunologia , Imunoglobulina A/sangue , Imunoglobulina A/imunologia , Microbiota/imunologia , Sepse/imunologia , Sepse/microbiologia , Simbiose , Animais , Antígenos de Bactérias/sangue , Antígenos de Bactérias/imunologia , Linfócitos B/imunologia , Bactérias/classificação , Bactérias/genética , Bactérias/imunologia , Medula Óssea , Feminino , Mucosa Intestinal/microbiologia , Intestinos/imunologia , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmócitos/imunologia , Proteobactérias/classificação , Proteobactérias/imunologia , Proteobactérias/fisiologia , RNA Ribossômico 16S/genética , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA