Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Host Microbe ; 31(10): 1639-1654.e10, 2023 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-37776864

RESUMO

During intestinal inflammation, host nutritional immunity starves microbes of essential micronutrients, such as iron. Pathogens scavenge iron using siderophores, including enterobactin; however, this strategy is counteracted by host protein lipocalin-2, which sequesters iron-laden enterobactin. Although this iron competition occurs in the presence of gut bacteria, the roles of commensals in nutritional immunity involving iron remain unexplored. Here, we report that the gut commensal Bacteroides thetaiotaomicron acquires iron and sustains its resilience in the inflamed gut by utilizing siderophores produced by other bacteria, including Salmonella, via a secreted siderophore-binding lipoprotein XusB. Notably, XusB-bound enterobactin is less accessible to host sequestration by lipocalin-2 but can be "re-acquired" by Salmonella, allowing the pathogen to evade nutritional immunity. Because the host and pathogen have been the focus of studies of nutritional immunity, this work adds commensal iron metabolism as a previously unrecognized mechanism modulating the host-pathogen interactions and nutritional immunity.


Assuntos
Infecções por Salmonella , Sideróforos , Humanos , Lipocalina-2/metabolismo , Sideróforos/metabolismo , Enterobactina/metabolismo , Bactérias/metabolismo , Ferro/metabolismo
2.
bioRxiv ; 2023 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-37425782

RESUMO

During intestinal inflammation, host nutritional immunity starves microbes of essential micronutrients such as iron. Pathogens scavenge iron using siderophores, which is counteracted by the host using lipocalin-2, a protein that sequesters iron-laden siderophores, including enterobactin. Although the host and pathogens compete for iron in the presence of gut commensal bacteria, the roles of commensals in nutritional immunity involving iron remain unexplored. Here, we report that the gut commensal Bacteroides thetaiotaomicron acquires iron in the inflamed gut by utilizing siderophores produced by other bacteria including Salmonella, via a secreted siderophore-binding lipoprotein termed XusB. Notably, XusB-bound siderophores are less accessible to host sequestration by lipocalin-2 but can be "re-acquired" by Salmonella , allowing the pathogen to evade nutritional immunity. As the host and pathogen have been the focus of studies of nutritional immunity, this work adds commensal iron metabolism as a previously unrecognized mechanism modulating the interactions between pathogen and host nutritional immunity.

3.
Infect Immun ; 90(3): e0001322, 2022 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-35100011

RESUMO

Research on Brucella pathogenesis has focused primarily on its ability to cause persistent intracellular infection of the mononuclear phagocyte system. At these sites, Brucella abortus evades innate immunity, which results in low-level inflammation and chronic infection of phagocytes. In contrast, the host response in the placenta during infection is characterized by severe inflammation and extensive extracellular replication of B. abortus. Despite the importance of reproductive disease caused by Brucella infection, our knowledge of the mechanisms involved in placental inflammation and abortion is limited. To understand the immune responses specifically driving placental pathology, we modeled placental B. abortus infection in pregnant mice. B. abortus infection caused an increase in the production of tumor necrosis factor alpha (TNF-α), specifically in the placenta. We found that placental expression levels of Tnfa and circulating TNF-α were dependent on the induction of endoplasmic reticulum stress and the B. abortus type IV secretion system (T4SS) effector protein VceC. Blockade of TNF-α reduced placental inflammation and improved fetal viability in mice. This work sheds light on a tissue-specific response of the placenta to B. abortus infection that may be important for bacterial transmission via abortion in the natural host species.


Assuntos
Brucelose Bovina , Brucelose , Animais , Brucella abortus/fisiologia , Brucelose/microbiologia , Bovinos , Feminino , Inflamação , Camundongos , Placenta , Gravidez , Fator de Necrose Tumoral alfa/metabolismo
4.
Lancet Oncol ; 23(3): e116-e128, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35090673

RESUMO

Sporadic colorectal cancer has traditionally been viewed as a malignancy of older individuals. However, as the global prevalence of the disease diagnosed in younger individuals (<50 years) is expected to increase within the next decade, greater recognition is now being given to early-onset colorectal cancer. The cause of the predicted rise in prevalence is largely unknown and probably multifactorial. In this Series paper, we discuss the potential underlying causes of early-onset colorectal cancer, the role of energy balance, biological and genomic mechanisms (including microbiome aspects), and the treatment of early-onset colorectal cancer. We have specifically considered the psychosocial challenges of being diagnosed with colorectal cancer at younger age and the potential financial toxicity that might ensue. This Series paper brings a comprehensive review based on the existing data in the hopes of optimising the overall outcomes for patients with early-onset colorectal cancer.


Assuntos
Neoplasias Colorretais , Idade de Início , Neoplasias Colorretais/epidemiologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/terapia , Genômica , Humanos , Prevalência
5.
Science ; 373(6556): 813-818, 2021 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-34385401

RESUMO

A Western-style, high-fat diet promotes cardiovascular disease, in part because it is rich in choline, which is converted to trimethylamine (TMA) by the gut microbiota. However, whether diet-induced changes in intestinal physiology can alter the metabolic capacity of the microbiota remains unknown. Using a mouse model of diet-induced obesity, we show that chronic exposure to a high-fat diet escalates Escherichia coli choline catabolism by altering intestinal epithelial physiology. A high-fat diet impaired the bioenergetics of mitochondria in the colonic epithelium to increase the luminal bioavailability of oxygen and nitrate, thereby intensifying respiration-dependent choline catabolism of E. coli In turn, E. coli choline catabolism increased levels of circulating trimethlamine N-oxide, which is a potentially harmful metabolite generated by gut microbiota.


Assuntos
Colo/fisiologia , Dieta Hiperlipídica , Escherichia coli/metabolismo , Mucosa Intestinal/fisiologia , Metilaminas/metabolismo , Animais , Hipóxia Celular , Colina/administração & dosagem , Colina/metabolismo , Colo/citologia , Metabolismo Energético , Células Epiteliais/fisiologia , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Fezes/microbiologia , Microbioma Gastrointestinal , Inflamação , Mucosa Intestinal/metabolismo , Masculino , Metilaminas/sangue , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Nitratos/metabolismo , Obesidade , Consumo de Oxigênio
6.
Cell Host Microbe ; 29(6): 851-853, 2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-34111391

RESUMO

In this issue of Cell Host & Microbe,Xing et al. (2021) show that gut microbiota confer resistance to colitis and colon cancer by stimulating IL-6 and IL-1ß production and Th17 cell expansion. Their findings reveal that even a single bacterial strain, Odoribacter splanchnicus, can confer protective immunity against cancer.


Assuntos
Colite , Microbiota , Neoplasias , Bacteroidetes , Humanos , Imunidade Inata , Células Th17
7.
Dis Model Mech ; 14(5)2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-34060626

RESUMO

Landmark discoveries in the gut microbiome field have paved the way for new research aimed at illuminating the influence of microbiota in colorectal cancer. A major challenge is to account for the effect of inherently variable environmental factors on the host and the gut microbiome, while concurrently determining their contribution to carcinogenesis. Here, we briefly discuss the role of the gut microbial community in colorectal cancer and elaborate on the recent insight that environmental factors related to a Western diet and lifestyle may drive the bloom of tumorigenic members of the gut microbiota. We also discuss how future research focused on untangling host-microbe interactions in the colon may influence medical insights that relate to the prevention and treatment of colorectal cancer.


Assuntos
Neoplasias Colorretais/complicações , Disbiose/complicações , Estilo de Vida , Neoplasias Colorretais/microbiologia , Neoplasias Colorretais/terapia , Disbiose/microbiologia , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/patologia , Interações Hospedeiro-Patógeno , Humanos
8.
Infect Immun ; 88(7)2020 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-32122941

RESUMO

In high-income countries, the leading causes of death are noncommunicable diseases (NCDs), such as obesity, cancer, and cardiovascular disease. An important feature of most NCDs is inflammation-induced gut dysbiosis characterized by a shift in the microbial community structure from obligate to facultative anaerobes such as Proteobacteria This microbial imbalance can contribute to disease pathogenesis by either a depletion in or the production of microbiota-derived metabolites. However, little is known about the mechanism by which inflammation-mediated changes in host physiology disrupt the microbial ecosystem in our large intestine leading to disease. Recent work by our group suggests that during gut homeostasis, epithelial hypoxia derived from peroxisome proliferator-activated receptor γ (PPAR-γ)-dependent ß-oxidation of microbiota-derived short-chain fatty acids limits oxygen availability in the colon, thereby maintaining a balanced microbial community. During inflammation, disruption in gut anaerobiosis drives expansion of facultative anaerobic Enterobacteriaceae, regardless of their pathogenic potential. Therefore, our research group is currently exploring the concept that dysbiosis-associated expansion of Enterobacteriaceae can be viewed as a microbial signature of epithelial dysfunction and may play a greater role in different models of NCDs, including diet-induced obesity, atherosclerosis, and inflammation-associated colorectal cancer.


Assuntos
Disbiose , Metabolismo Energético , Microbioma Gastrointestinal , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Doenças não Transmissíveis/epidemiologia , Animais , Colo/metabolismo , Colo/microbiologia , Suscetibilidade a Doenças , Enterobacteriaceae/metabolismo , Humanos , Obesidade/complicações , Obesidade/metabolismo , Oxirredução , Medição de Risco , Fatores de Risco
9.
mBio ; 10(5)2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31575772

RESUMO

Intestinal inflammation is a risk factor for colorectal cancer formation, but the underlying mechanisms remain unknown. Here, we investigated whether colitis alters the colonic microbiota to enhance its cancer-inducing activity. Colitis increased epithelial oxygenation in the colon of mice and drove an expansion of Escherichia coli within the gut-associated microbial community through aerobic respiration. An aerobic expansion of colibactin-producing E. coli was required for the cancer-inducing activity of this pathobiont in a mouse model of colitis-associated colorectal cancer formation. We conclude that increased epithelial oxygenation in the colon is associated with an expansion of a prooncogenic driver species, thereby increasing the cancer-inducing activity of the microbiota.IMPORTANCE One of the environmental factors important for colorectal cancer formation is the gut microbiota, but the habitat filters that control its cancer-inducing activity remain unknown. Here, we show that chemically induced colitis elevates epithelial oxygenation in the colon, thereby driving an expansion of colibactin-producing Escherichia coli, a prooncogenic driver species. These data suggest that elevated epithelial oxygenation is a potential risk factor for colorectal cancer formation because the consequent changes in the gut habitat escalate the cancer-inducing activity of the microbiota.


Assuntos
Carcinogênese , Colite/microbiologia , Neoplasias Colorretais/microbiologia , Infecções por Escherichia coli/complicações , Microbioma Gastrointestinal , Oxigênio/metabolismo , Aerobiose , Animais , Colite/induzido quimicamente , Colite/complicações , Sulfato de Dextrana , Escherichia coli , Infecções por Escherichia coli/microbiologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/metabolismo , Policetídeos/metabolismo
10.
J Exp Med ; 216(10): 2378-2393, 2019 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-31358565

RESUMO

Chronic inflammation and gut microbiota dysbiosis, in particular the bloom of genotoxin-producing E. coli strains, are risk factors for the development of colorectal cancer. Here, we sought to determine whether precision editing of gut microbiota metabolism and composition could decrease the risk for tumor development in mouse models of colitis-associated colorectal cancer (CAC). Expansion of experimentally introduced E. coli strains in the azoxymethane/dextran sulfate sodium colitis model was driven by molybdoenzyme-dependent metabolic pathways. Oral administration of sodium tungstate inhibited E. coli molybdoenzymes and selectively decreased gut colonization with genotoxin-producing E. coli and other Enterobacteriaceae. Restricting the bloom of Enterobacteriaceae decreased intestinal inflammation and reduced the incidence of colonic tumors in two models of CAC, the azoxymethane/dextran sulfate sodium colitis model and azoxymethane-treated, Il10-deficient mice. We conclude that metabolic targeting of protumoral Enterobacteriaceae during chronic inflammation is a suitable strategy to prevent the development of malignancies arising from gut microbiota dysbiosis.


Assuntos
Colite/microbiologia , Neoplasias Colorretais/microbiologia , Disbiose/microbiologia , Microbioma Gastrointestinal , Neoplasias Experimentais/microbiologia , Animais , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/genética , Sulfato de Dextrana/toxicidade , Disbiose/induzido quimicamente , Disbiose/genética , Escherichia coli/crescimento & desenvolvimento , Interleucina-10/deficiência , Camundongos , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/genética
11.
Nat Commun ; 10(1): 1826, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-31015451

RESUMO

The bacterial pathogen Shigella flexneri causes 270 million cases of bacillary dysentery (blood in stool) worldwide every year, resulting in more than 200,000 deaths. A major challenge in combating bacillary dysentery is the lack of a small-animal model that recapitulates the symptoms observed in infected individuals, including bloody diarrhea. Here, we show that similar to humans, infant rabbits infected with S. flexneri experience severe inflammation, massive ulceration of the colonic mucosa, and bloody diarrhea. T3SS-dependent invasion of epithelial cells is necessary and sufficient for mediating immune cell infiltration and vascular lesions. However, massive ulceration of the colonic mucosa, bloody diarrhea, and dramatic weight loss are strictly contingent on the ability of the bacteria to spread from cell to cell. The infant rabbit model features bacterial dissemination as a critical determinant of S. flexneri pathogenesis and provides a unique small-animal model for research and development of therapeutic interventions.


Assuntos
Diarreia/patologia , Disenteria Bacilar/patologia , Hemorragia Gastrointestinal/patologia , Shigella flexneri/patogenicidade , Sistemas de Secreção Tipo III/imunologia , Animais , Animais Recém-Nascidos/microbiologia , Colo/microbiologia , Colo/patologia , Diarreia/microbiologia , Modelos Animais de Doenças , Disenteria Bacilar/microbiologia , Células Epiteliais/microbiologia , Feminino , Hemorragia Gastrointestinal/microbiologia , Células HT29 , Humanos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Gravidez , Coelhos
12.
Mucosal Immunol ; 11(5): 1299-1305, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29743614

RESUMO

A balanced gut microbiota is important for human health, but the mechanisms that maintain homeostasis are incompletely understood. Recent insights suggest the host plays a key role in shaping its gut microbiota to be beneficial. While host control in the small intestine curbs bacterial numbers to avoid competition for simple sugars and amino acids, the host limits oxygen availability in the large intestine to obtain microbial fermentation products from fiber. Epithelial cells are major players in imposing ecological control mechanisms, which involves the release of antimicrobial peptides by small-intestinal Paneth cells and maintenance of luminal anaerobiosis by epithelial hypoxia in the colon. Harnessing these epithelial control mechanisms for therapeutic means could provide a novel lynchpin for strategies to remediate dysbiosis.


Assuntos
Microbioma Gastrointestinal/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , Microbiota/imunologia , Animais , Ecologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Homeostase/imunologia , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Intestinos/imunologia , Intestinos/microbiologia
13.
Cell Host Microbe ; 23(2): 266-273.e4, 2018 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-29447698

RESUMO

Salmonella enterica serovar (S.) Typhi is an extraintestinal pathogen that evolved from Salmonella serovars causing gastrointestinal disease. Compared with non-typhoidal Salmonella serovars, the genomes of typhoidal serovars contain various loss-of-function mutations. However, the contribution of these genetic differences to this shift in pathogen ecology remains unknown. We show that the ydiQRSTD operon, which is deleted in S. Typhi, enables S. Typhimurium to utilize microbiota-derived butyrate during gastrointestinal disease. Unexpectedly, genetic ablation of butyrate utilization reduces S. Typhimurium epithelial invasion and attenuates intestinal inflammation. Deletion of ydiD renders S. Typhimurium sensitive to butyrate-mediated repression of invasion gene expression. Combined with the gain of virulence-associated (Vi) capsular polysaccharide and loss of very-long O-antigen chains, two features characteristic of S. Typhi, genetic ablation of butyrate utilization abrogates S. Typhimurium-induced intestinal inflammation. Thus, the transition from a gastrointestinal to an extraintestinal pathogen involved discrete genetic changes, providing insights into pathogen evolution and emergence.


Assuntos
Butiratos/metabolismo , Colite/patologia , Intoxicação Alimentar por Salmonella/patologia , Salmonella typhi/genética , Salmonella typhimurium/genética , Animais , Linhagem Celular Tumoral , Clostridium/isolamento & purificação , Clostridium/patogenicidade , Colite/microbiologia , Escherichia coli , Feminino , Humanos , Intestinos/microbiologia , Intestinos/patologia , Camundongos , Camundongos Endogâmicos CBA , Intoxicação Alimentar por Salmonella/microbiologia , Salmonella typhi/patogenicidade , Salmonella typhimurium/patogenicidade , Sistemas de Secreção Tipo III/genética
14.
Science ; 357(6351): 570-575, 2017 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-28798125

RESUMO

Perturbation of the gut-associated microbial community may underlie many human illnesses, but the mechanisms that maintain homeostasis are poorly understood. We found that the depletion of butyrate-producing microbes by antibiotic treatment reduced epithelial signaling through the intracellular butyrate sensor peroxisome proliferator-activated receptor γ (PPAR-γ). Nitrate levels increased in the colonic lumen because epithelial expression of Nos2, the gene encoding inducible nitric oxide synthase, was elevated in the absence of PPAR-γ signaling. Microbiota-induced PPAR-γ signaling also limits the luminal bioavailability of oxygen by driving the energy metabolism of colonic epithelial cells (colonocytes) toward ß-oxidation. Therefore, microbiota-activated PPAR-γ signaling is a homeostatic pathway that prevents a dysbiotic expansion of potentially pathogenic Escherichia and Salmonella by reducing the bioavailability of respiratory electron acceptors to Enterobacteriaceae in the lumen of the colon.


Assuntos
Disbiose/metabolismo , Disbiose/microbiologia , Enterobacteriaceae/patogenicidade , Microbioma Gastrointestinal , Óxido Nítrico Sintase Tipo II/metabolismo , PPAR gama/metabolismo , Proteína 4 Semelhante a Angiopoietina/genética , Anilidas/farmacologia , Animais , Antibacterianos/farmacologia , Butiratos/metabolismo , Células CACO-2 , Clostridium/efeitos dos fármacos , Clostridium/metabolismo , Colite/metabolismo , Colite/microbiologia , Colo/metabolismo , Colo/microbiologia , Disbiose/induzido quimicamente , Disbiose/genética , Enterobacteriaceae/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Feminino , Expressão Gênica , Homeostase , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nitratos/metabolismo , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/genética , Oxirredução , PPAR gama/antagonistas & inibidores , PPAR gama/genética , Transdução de Sinais , Estreptomicina/farmacologia
15.
Int J Med Microbiol ; 306(8): 604-610, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27760693

RESUMO

Salmonella enterica serotype Typhimurium is able to expand in the lumen of the inflamed intestine through mechanisms that have not been fully resolved. Here we utilized streptomycin-pretreated mice and dextran sodium sulfate (DSS)-treated mice to investigate how pathways for S. Typhimurium iron acquisition contribute to pathogen expansion in the inflamed intestine. Competitive infection with an iron uptake-proficient S. Typhimurium strain and mutant strains lacking tonB feoB, feoB, tonB or iroN in streptomycin pretreated mice demonstrated that ferric iron uptake requiring IroN and TonB conferred a fitness advantage during growth in the inflamed intestine. However, the fitness advantage conferred by ferrous iron uptake mechanisms was independent of inflammation and was only apparent in models where the normal microbiota composition had been disrupted by antibiotic treatment.


Assuntos
Gastroenterite/microbiologia , Intestinos/microbiologia , Ferro/metabolismo , Redes e Vias Metabólicas/genética , Infecções por Salmonella/microbiologia , Salmonella typhimurium/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bovinos , Modelos Animais de Doenças , Feminino , Masculino , Camundongos Endogâmicos C57BL , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
16.
Microbiol Spectr ; 4(2)2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-27227304

RESUMO

Many bacterial pathogens can cause acute infections that are cleared with the onset of adaptive immunity, but a subset of these pathogens can establish persistent, and sometimes lifelong, infections. While bacteria that cause chronic infections are phylogenetically diverse, they share common features in their interactions with the host that enable a protracted period of colonization. This article will compare the persistence strategies of two chronic pathogens from the Proteobacteria, Brucella abortus and Salmonella enterica serovar Typhi, to consider how these two pathogens, which are very different at the genomic level, can utilize common strategies to evade immune clearance to cause chronic intracellular infections of the mononuclear phagocyte system.


Assuntos
Bactérias/patogenicidade , Infecções Bacterianas/microbiologia , Animais , Anticorpos Antibacterianos/imunologia , Bactérias/imunologia , Infecções Bacterianas/imunologia , Doença Crônica , Citocinas/imunologia , Interações Hospedeiro-Parasita , Humanos , Evasão da Resposta Imune , Sistema Imunitário , Imunidade Inata , Macrófagos/imunologia
17.
Nature ; 532(7599): 394-7, 2016 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-27007849

RESUMO

Endoplasmic reticulum (ER) stress is a major contributor to inflammatory diseases, such as Crohn disease and type 2 diabetes. ER stress induces the unfolded protein response, which involves activation of three transmembrane receptors, ATF6, PERK and IRE1α. Once activated, IRE1α recruits TRAF2 to the ER membrane to initiate inflammatory responses via the NF-κB pathway. Inflammation is commonly triggered when pattern recognition receptors (PRRs), such as Toll-like receptors or nucleotide-binding oligomerization domain (NOD)-like receptors, detect tissue damage or microbial infection. However, it is not clear which PRRs have a major role in inducing inflammation during ER stress. Here we show that NOD1 and NOD2, two members of the NOD-like receptor family of PRRs, are important mediators of ER-stress-induced inflammation in mouse and human cells. The ER stress inducers thapsigargin and dithiothreitol trigger production of the pro-inflammatory cytokine IL-6 in a NOD1/2-dependent fashion. Inflammation and IL-6 production triggered by infection with Brucella abortus, which induces ER stress by injecting the type IV secretion system effector protein VceC into host cells, is TRAF2, NOD1/2 and RIP2-dependent and can be reduced by treatment with the ER stress inhibitor tauroursodeoxycholate or an IRE1α kinase inhibitor. The association of NOD1 and NOD2 with pro-inflammatory responses induced by the IRE1α/TRAF2 signalling pathway provides a novel link between innate immunity and ER-stress-induced inflammation.


Assuntos
Estresse do Retículo Endoplasmático , Inflamação/metabolismo , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Transdução de Sinais , Animais , Proteínas da Membrana Bacteriana Externa/metabolismo , Brucella abortus/imunologia , Brucella abortus/patogenicidade , Linhagem Celular , Ditiotreitol/farmacologia , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/patologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Endorribonucleases/antagonistas & inibidores , Feminino , Humanos , Imunidade Inata , Inflamação/induzido quimicamente , Interleucina-6/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD1/imunologia , Proteína Adaptadora de Sinalização NOD2/imunologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Receptores de Reconhecimento de Padrão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator 2 Associado a Receptor de TNF/metabolismo , Ácido Tauroquenodesoxicólico/farmacologia , Tapsigargina/farmacologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA