Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Annu Rev Physiol ; 74: 177-98, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22035346

RESUMO

The well-being of the intestine and its host requires that this organ execute its complex function amid colonization by a large and diverse microbial community referred to as the gut microbiota. A myriad of interacting mechanisms of mucosal immunity permit the gut to corral the microbiota in such a way as to maximize the benefits and to minimize the danger of living in close proximity to this large microbial biomass. Toll-like receptors and Nod-like receptors, collectively referred to as pattern recognition receptors (PRRs), recognize a variety of microbial components and, hence, play a central role in governing the interface between host and microbiota. This review examines mechanisms by which PRR-microbiota interactions are regulated so as to allow activation of host defense when necessary while preventing excessive inflammation, which can have a myriad of negative consequences for the host. Analysis of published studies performed in human subjects and a variety of murine disease models reveals the central theme that PRRs play a key role in maintaining a healthful stable relationship between the intestine and its microbiota. In contrast, although select genetic ablations of PRR signaling may protect against some chronic diseases, the overriding theme of studies performed to date is that perturbations of PRR-microbiota interactions are more likely to promote disease states associated with inflammation.


Assuntos
Trato Gastrointestinal/microbiologia , Metagenoma/fisiologia , Receptores Toll-Like/fisiologia , Animais , Colite/microbiologia , Colite/fisiopatologia , Escherichia coli/fisiologia , Neoplasias Gastrointestinais/fisiopatologia , Trato Gastrointestinal/imunologia , Homeostase/fisiologia , Humanos , Doenças Metabólicas/microbiologia , Doenças Metabólicas/fisiopatologia , Proteínas Adaptadoras de Sinalização NOD/fisiologia , Receptores de Reconhecimento de Padrão/fisiologia , Transdução de Sinais/fisiologia
2.
Arthritis Rheum ; 65(4): 1064-73, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23280250

RESUMO

OBJECTIVE: Lipocalin 2 (LCN-2) is an innate immune protein that is expressed by a variety of cells and is highly up-regulated during several pathologic conditions, including immune complex (IC)-mediated inflammatory/autoimmune disorders. However, the function of LCN-2 during IC-mediated inflammation is largely unknown. Therefore, this study was undertaken to investigate the role of LCN-2 in IC-mediated diseases. METHODS: The up-regulation of LCN-2 was determined by enzyme-linked immunosorbent assay in 3 different mouse models of IC-mediated autoimmune disease: systemic lupus erythematosus, collagen-induced arthritis, and serum-transfer arthritis. The in vivo role of LCN-2 during IC-mediated inflammation was investigated using LCN-2-knockout mice and their wild-type littermates. RESULTS: LCN-2 levels were significantly elevated in all 3 of the autoimmune disease models. Further, in an acute skin inflammation model, LCN-2-knockout mice exhibited a 50% reduction in inflammation, with histopathologic analysis revealing notably reduced immune cell infiltration as compared to wild-type mice. Administration of recombinant LCN-2 to LCN-2-knockout mice restored inflammation to levels observed in wild-type mice. Neutralization of LCN-2 using a monoclonal antibody significantly reduced inflammation in wild-type mice. In contrast, LCN-2-knockout mice developed more severe serum-induced arthritis compared to wild-type mice. Histologic analysis revealed extensive tissue and bone destruction, with significantly reduced neutrophil infiltration but considerably more macrophage migration, in LCN-2-knockout mice compared to wild-type mice. CONCLUSION: These results demonstrate that LCN-2 may regulate immune cell recruitment to the site of inflammation, a process essential for the controlled initiation, perpetuation, and resolution of inflammatory processes. Thus, LCN-2 may present a promising target in the treatment of IC-mediated inflammatory/autoimmune diseases.


Assuntos
Proteínas de Fase Aguda/imunologia , Complexo Antígeno-Anticorpo/imunologia , Artrite Experimental/imunologia , Dermatite/imunologia , Inflamação/imunologia , Lipocalinas/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Proteínas Oncogênicas/imunologia , Doença Aguda , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/metabolismo , Animais , Complexo Antígeno-Anticorpo/metabolismo , Artrite Experimental/metabolismo , Doença Crônica , Dermatite/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Inflamação/metabolismo , Lipocalina-2 , Lipocalinas/genética , Lipocalinas/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Regulação para Cima
3.
J Immunol ; 189(4): 1911-9, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22786765

RESUMO

Various states of inflammation, including sepsis, are associated with hypoferremia, which limits iron availability to pathogens and reduces iron-mediated oxidative stress. Lipocalin 2 (Lcn2; siderocalin, 24p3) plays a central role in iron transport. Accordingly, Lcn2-deficient (Lcn2KO) mice exhibit elevated intracellular labile iron. In this study, we report that LPS induced systemic Lcn2 by 150-fold in wild-type mice at 24 h. Relative to wild-type littermates, Lcn2KO mice were markedly more sensitive to endotoxemia, exhibiting elevated indices of organ damage (transaminasemia, lactate dehydrogenase) and increased mortality. Such exacerbated endotoxemia was associated with substantially increased caspase-3 cleavage and concomitantly elevated immune cell apoptosis. Furthermore, cells from Lcn2KO mice were hyperresponsive to LPS ex vivo, exhibiting elevated cytokine secretion. Additionally, Lcn2KO mice exhibited delayed LPS-induced hypoferremia despite normal hepatic hepcidin expression and displayed decreased levels of the tissue redox state indicators cysteine and glutathione in liver and plasma. Desferroxamine, an iron chelator, significantly protects Lcn2KO mice from LPS-induced toxicity, including mortality, suggesting that Lcn2 may act as an antioxidant in vivo by regulating iron homeostasis. Thus, Lcn2-mediated regulation of labile iron protects the host against sepsis. Its small size and simple structure may make Lcn2 a deployable treatment for sepsis.


Assuntos
Proteínas de Fase Aguda/metabolismo , Homeostase/fisiologia , Ferro/metabolismo , Lipocalinas/metabolismo , Proteínas Oncogênicas/metabolismo , Sepse/metabolismo , Proteínas de Fase Aguda/deficiência , Proteínas de Fase Aguda/imunologia , Animais , Apoptose/fisiologia , Endotoxinas/toxicidade , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Immunoblotting , Lipocalina-2 , Lipocalinas/imunologia , Masculino , Camundongos , Camundongos Knockout , Proteínas Oncogênicas/deficiência , Proteínas Oncogênicas/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sepse/induzido quimicamente , Sepse/imunologia
4.
Gut ; 61(3): 373-84, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21646247

RESUMO

BACKGROUND: The extent to which numerous strains of genetically engineered mice, including mice lacking Toll-like receptor 5 (T5KO), display colitis is environment dependent. Gut microbiota underlie much of the variation in phenotype. Accordingly, embryonic rederivation of T5KO mice ameliorated their spontaneous colitis despite only partially correcting elevated proinflammatory gene expression. It was postulated that endogenous anti-inflammatory pathways mediated the absence of overt inflammation in these mice when their gut microbiota were reset. Consequently, it was hypothesised that neutralisation of the anti-inflammatory cytokine interleukin 10 (IL-10) might induce uniform colitis in T5KO mice, and thus provide a practical means to study mechanisms underlying their inflammation. METHODS: Two distinct strains of non-colitic T5KO mice, as well as mice lacking MyD88, Toll-like receptor 4 (TLR4), IL-1 receptor (IL-1R) and various double knockouts (DKOs) were treated weekly for 4 weeks with 1 mg/mouse of IL-10 receptor neutralising antibody (IL-10R mAb) and colitis assayed 1 week later. The composition of the caecal microbiota was determined by 454 pyrosequencing of 16S rRNA genes. RESULTS: Anti-IL-10R mAb treatment led to severe uniform intestinal inflammation in both strains of T5KO mice. Such neutralisation of IL-10 signalling did not cause colitis in wild-type littermates nor mice lacking TLR4, MyD88 or IL-1R. The susceptibility of T5KO mice to this colitis model was not rescued by absence of TLR4 in that T4/T5 DKO mice displayed severe colitis in response to anti-IL-10R mAb treatment. IL-1ß signalling was crucial for this colitis model in that IL-1R/T5 DKOs were completely protected from colitis in response to IL-10R mAb treatment. Lastly, it was observed that blockade of IL-10R function was associated with changes in the composition of gut microbiota, which were observed in mice that were susceptible and resistant to IL-10R mAb-induced colitis. CONCLUSION: Regardless of whether they harbour a colitogenic microbiota, loss of TLR5 predisposes mice to colitis triggered by immune dysregulation via an IL-1ß-dependent pathway.


Assuntos
Colite Ulcerativa/imunologia , Interleucina-1beta/fisiologia , Receptor 5 Toll-Like/deficiência , Animais , Anticorpos Monoclonais/imunologia , Ceco/microbiologia , Colite Ulcerativa/genética , Colite Ulcerativa/microbiologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Perfilação da Expressão Gênica/métodos , Masculino , Metagenoma/imunologia , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide , Receptores de Interleucina-10/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor 5 Toll-Like/imunologia
5.
Eur J Immunol ; 40(12): 3528-34, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21072873

RESUMO

The fact that some TLR-based vaccine adjuvants maintain function in TLR-deficient hosts highlights that their mechanism of function remains incompletely understood. Thus, we examined the ability of flagellin to induce cytokines and elicit/promote murine antibody responses upon deletion of the flagellin receptors TLR5 and/or NLRC4 (also referred to as IPAF) using a prime/boost regimen. In TLR5-KO mice, flagellin failed to induce NF-κB-regulated cytokines such as keratinocyte-derived chemokine (CXCL1) but induced WT levels of the inflammasome cytokine IL-18 (IL-1F4). Conversely, in NLRC4-KO mice, flagellin induced keratinocyte-derived chemokine, but not IL-18, whereas TLR5/NLRC4-DKO lacked induction of all cytokines measured. Flagellin/ovalbumin treatment resulted in high-antibody titers to both flagellin and ovalbumin in WT, TLR5-KO and DKO mice but did not elicit antibodies to either in TLR5/NLRC4-DKO mice. Thus, flagellin's ability to elicit/promote humoral immunity requires a germ-line-encoded receptor capable of recognizing this molecule. Such promotion of adaptive immunity can be effectively driven by either TLR5-mediated activation of NF-κB or NLRC4-mediated activation of the inflammasome.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Flagelina/administração & dosagem , Imunidade Humoral , Macrófagos/metabolismo , Receptor 5 Toll-Like/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/imunologia , Linhagem Celular , Quimiocina CXCL1/metabolismo , Flagelina/farmacologia , Interações Hospedeiro-Patógeno , Imunidade Humoral/genética , Imunidade Inata , Imunização , Inflamassomos , Interleucina-18/genética , Interleucina-18/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/metabolismo , Transdução de Sinais , Receptor 5 Toll-Like/genética , Receptor 5 Toll-Like/imunologia
6.
J Clin Invest ; 117(12): 3909-21, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18008007

RESUMO

Activation of TLRs by bacterial products results in rapid activation of genes encoding products designed to protect the host from perturbing microbes. In the intestine, which is colonized by a large and diverse population of commensal bacteria, TLR signaling may not function in a simple on/off mode. Here, we show that the flagellin receptor TLR5 has an essential and nonredundant role in protecting the gut from enteric microbes. Mice lacking TLR5 (TLR5KO mice) developed spontaneous colitis, as assessed by well-defined clinical, serologic, and histopathologic indicators of this disorder. Compared with WT littermates, TLR5KO mice that had not yet developed robust colitis exhibited decreased intestinal expression of TLR5-regulated host defense genes despite having an increased bacterial burden in the colon. In contrast, such TLR5KO mice displayed markedly increased colonic expression of hematopoietic-derived proinflammatory cytokines, suggesting that elevated levels of bacterial products may result in activation of other TLRs that drive colitis in TLR5KO mice. In accordance, deletion of TLR4 rescued the colitis of TLR5KO mice in that mice lacking both TLR4 and TLR5 also had elevated bacterial loads in the colon but lacked immunological, histopathological, and clinical evidence of colitis. That an engineered innate immune deficiency ultimately results in spontaneous intestinal inflammation supports the notion that an innate immune deficiency might underlie some instances of inflammatory bowel disease.


Assuntos
Colite/genética , Deleção de Genes , Imunidade Inata/genética , Doenças Inflamatórias Intestinais/genética , Receptor 5 Toll-Like/genética , Animais , Bactérias/imunologia , Colite/imunologia , Colite/microbiologia , Colite/patologia , Colo/imunologia , Colo/patologia , Citocinas/genética , Citocinas/imunologia , Flagelina/imunologia , Mediadores da Inflamação/imunologia , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/patologia , Camundongos , Camundongos Knockout , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Receptor 5 Toll-Like/imunologia
7.
Infect Immun ; 76(3): 1276-81, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18195036

RESUMO

The recognition of flagellin by Toll-like receptor 5 (TLR5) is the dominant means by which model intestinal epithelia activate proinflammatory gene expression in response to Salmonella enterica. The role of the flagellin-TLR5 interaction in vivo has been addressed primarily via studies that use flagellar mutants. Such studies suggest that host recognition of flagellin promotes rapid neutrophil recruitment that protects the host from this pathogen. However, these works do not directly address the role of TLR5 and are subject to the caveat that flagellar mutations may broadly affect Salmonella gene expression. Thus, we examined the role of the flagellin-TLR5 interaction via the use of TLR5-deficient (TLR5KO) mice. We utilized both the traditional model of murine Salmonella infection, wherein low-dose oral infection of mice with Salmonella enterica subsp. enterica serovar Typhimurium results in systemic typhoid-like disease, and a more recently characterized model in which mice are pretreated with streptomycin to result in gut-restricted acute enteritis. In the enteritis model, TLR5KO mice had more severe gut pathology, thus "phenocopying" previous results obtained with Salmonella mutants. In contrast, TLR5KO mice were resistant to Salmonella-induced typhoid-like disease. However, such resistance was not specific for flagellated serovar Typhimurium, but rather, TLR5KO mice were also resistant to challenges by flagellin-deficient serovar Typhimurium. Such resistance associated with elevations in the microbiota was ablated by antibiotic pretreatment and correlated with basal elevations in intestinal host defense gene expression. All together, these results indicate that the resistance of TLR5KO mice to Salmonella-induced typhoid-like illness resulted from alterations in their basal phenotype rather than from the lack of TLR5 ligation during the infection per se.


Assuntos
Imunidade Inata , Salmonelose Animal/genética , Salmonella typhimurium/patogenicidade , Receptor 5 Toll-Like/deficiência , Receptor 5 Toll-Like/imunologia , Animais , Anticorpos Antibacterianos/sangue , Enterite/genética , Ensaio de Imunoadsorção Enzimática , Fezes/química , Imunoglobulina A/análise , Imunoglobulina A/sangue , Imunoglobulina G/análise , Imunoglobulina G/sangue , Intestinos/química , Intestinos/imunologia , Intestinos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/análise , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sepse/genética , Análise de Sobrevida
8.
Curr Protoc Immunol ; 104: 15.25.1-15.25.14, 2014 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-24510619

RESUMO

Inflammatory bowel diseases (IBD), mainly comprising ulcerative colitis and Crohn's Disease, are complex and multifactorial diseases with unknown etiology. For the past 20 years, to study human IBD mechanistically, a number of murine models of colitis have been developed. These models are indispensable tools to decipher underlying mechanisms of IBD pathogenesis as well as to evaluate a number of potential therapeutics. Among various chemically induced colitis models, the dextran sulfate sodium (DSS)-induced colitis model is widely used because of its simplicity and many similarities with human ulcerative colitis. This model has both advantages and disadvantages that must be considered when employed. This protocol describes the DSS-induced colitis model, focusing on details and factors that could affect DSS-induced pathology.


Assuntos
Colite/imunologia , Sulfato de Dextrana/administração & dosagem , Modelos Animais de Doenças , Doenças Inflamatórias Intestinais/imunologia , Animais , Biomarcadores/química , Biomarcadores/metabolismo , Peso Corporal , Colite/induzido quimicamente , Fezes/química , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
9.
Adv Immunol ; 116: 93-112, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23063074

RESUMO

The mammalian intestine harbors trillions of microbes collectively known as the microbiota, which can be viewed as an anaerobic metabolic organ that benefits the host in a number of ways. The homeostasis of this large microbial biomass is a prerequisite to maintaining host health by maximizing symbiotic interrelations and minimizing the risk of living in a close relationship. The cooperation between the innate and adaptive immune systems of the host maintains homeostasis of the microbiota. The dysregulation/alteration of microbiota in various immunodeficiency states including both innate and adaptive deficiency results in metabolic disease. This review examines the influence of microbiota on host metabolic health in immunologically altered mice. Accumulated data from a variety of immune-deficient murine models indicate that altered microbiota can play a key role in origination of metabolic diseases through the following potential mechanisms: (i) increasing calorie extraction resulting in adiposity, (ii) inducing low-grade chronic inflammation in the gut directly or increasing systemic loads of microbial ligands via leaky guts, (iii) generating toxic metabolites from dietary components, and (iv) inducing a switch from pro-metabolic to pro-immune phenotype that drives malabsorption of lipids resulting in muscle wastage and weight loss-particularly upon states of adaptive immune deficiency. Further, these murine models demonstrate that altered microbiota is not purely a consequence of metabolic disease but plays a key role in driving this disorder.


Assuntos
Modelos Animais de Doenças , Trato Gastrointestinal/microbiologia , Doenças Metabólicas/microbiologia , Metagenoma , Camundongos , Animais , Bactérias/imunologia , Trato Gastrointestinal/imunologia , Humanos , Doenças Metabólicas/imunologia
10.
Cell Host Microbe ; 12(2): 139-52, 2012 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-22863420

RESUMO

Colitis results from breakdown of homeostasis between intestinal microbiota and the mucosal immune system, with both environmental and genetic influencing factors. Flagellin receptor TLR5-deficient mice (T5KO) display elevated intestinal proinflammatory gene expression and colitis with incomplete penetrance, providing a genetically sensitized system to study the contribution of microbiota to driving colitis. Both colitic and noncolitic T5KO exhibited transiently unstable microbiotas, with lasting differences in colitic T5KO, while their noncolitic siblings stabilized their microbiotas to resemble wild-type mice. Transient high levels of proteobacteria, especially enterobacteria species including E. coli, observed in close proximity to the gut epithelium were a striking feature of colitic microbiota. A Crohn's disease-associated E. coli strain induced chronic colitis in T5KO, which persisted well after the exogenously introduced bacterial species had been eliminated. Thus, an innate immune deficiency can result in unstable gut microbiota associated with low-grade inflammation, and harboring proteobacteria can drive and/or instigate chronic colitis.


Assuntos
Colite/microbiologia , Trato Gastrointestinal/microbiologia , Proteobactérias/fisiologia , Receptor 5 Toll-Like/imunologia , Animais , Colite/genética , Colite/imunologia , Enterobacteriaceae/fisiologia , Feminino , Trato Gastrointestinal/imunologia , Humanos , Masculino , Metagenoma , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor 5 Toll-Like/deficiência , Receptor 5 Toll-Like/genética
11.
J Inflamm (Lond) ; 8(1): 2, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21314942

RESUMO

BACKGROUND: Toll-like receptor-4 (TLR4) triggers inflammatory signaling in response to microbial lipoploysaccharide. It has been reported that loss of TLR4 protected against saturated fat-induced inflammation and insulin resistance. It is not known whether loss of TLR4 function offers protection against trans fat (TF) induced obesity, inflammation, and insulin resistance. We investigated whether mice with loss of function mutation in TLR4 were resistant to TF-induced pathologies such as obesity, inflammation, hyperglycemia, and hyperinsulinemia. METHODS: C57BL/6j and C57BL/10 mice were cross bred to generate TLR4 mutant and wild type (WT). TLR4 mutant (n = 12) and WT (n = 12) mice were fed either low fat (LF) (13.5% fat energy) or high TF diets (60% fat energy) for 12 weeks. In vitro experiments were conducted on mouse macrophage cells (RAW 264.7 and J774A.1) to investigate whether elaidic (trans 18:1) or oleic acid (cis 18:1) would upregulate inflammatory markers. RESULTS: TLR4 mutant mice were ~26.4% heavier than WT mice. In both genotypes, mice that received TF diet were significantly heavier than those mice that received LF diet (P < 0.01). TLR4 mutant mice compared to WT mice had significantly higher fasting blood glucose, serum insulin, insulin resistance, serum leptin, and serum cholesterol when they received TF diet (P < 0.05). No upregulation of iNOS or COX2 in response to either elaidic or oleic acid in macrophage cells was observed. CONCLUSIONS: Loss of function mutation in TLR4 not only did not protect mice from TF-induced obesity, hyperglycemia, hyperinsulinemia, and hypercholesterolemia but also exacerbated the above pathologies suggesting that functional TLR4 is necessary in attenuating TF-induced deleterious effects. It is likely that TF induces pathologies through pathways independent of TLR4.

12.
Nutr Metab (Lond) ; 8(1): 16, 2011 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-21388548

RESUMO

BACKGROUND AND OBJECTIVE: Systemic chronic inflammation is linked to metabolic syndrome, type-2 diabetes, and heart disease. Lipopolysaccharide (LPS), a Gram negative microbial product, triggers inflammation through toll-like-receptor-4 (TLR-4) signaling. It has been reported that dietary fatty acids also modulate inflammation through TLR-4. We investigated whether fish oil (FO) rich diet in comparison to saturated fat (SF) rich diet would confer protection from pathologies induced by LPS. METHODS: Twenty C57BL/6 mice were divided into two groups. One group received FO-diet and other received SF-diet ad libitum for 60 days. Diets were isocaloric containing 45% energy from fat. After 60-days of feeding, blood was collected after overnight fast. Mice were allowed to recover for 4-days, fasted for 5-hours, challenged with 100 ng/mL of LPS intraperitonially, and bled after 2-hours. After 7-days of recuperation, mice were challenged with 500 ng/mL of LPS intraperitonially and observed for physical health. RESULTS: Food intake was similar in FO- and SF-fed mice. FO-fed mice compared to SF-fed mice had significantly less body weight gain (P = 0.005), epididymal fat weight (P = 0.005), fasting blood glucose (70.8 vs 83.3 ng/dL; P < 0.05), HOMA-IR (5.0 vs 13.6; P < 0.019), and serum cholesterol (167 vs 94 mg/dL; P < 0.05). When challenged with LPS, FO-fed mice had significantly lower serum IL-1ß compared to SF-fed mice (2.0 vs 30.0 pg/mL; P < 0.001). After LPS-challenge, SF-fed mice had higher mortality, lost more body weight, and had greater decrease in blood glucose compared to FO-fed mice. CONCLUSION: Overall, FO-diet compared to SF-diet offered protection against deleterious effects of LPS in mice.

13.
Science ; 328(5975): 228-31, 2010 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-20203013

RESUMO

Metabolic syndrome is a group of obesity-related metabolic abnormalities that increase an individual's risk of developing type 2 diabetes and cardiovascular disease. Here, we show that mice genetically deficient in Toll-like receptor 5 (TLR5), a component of the innate immune system that is expressed in the gut mucosa and that helps defend against infection, exhibit hyperphagia and develop hallmark features of metabolic syndrome, including hyperlipidemia, hypertension, insulin resistance, and increased adiposity. These metabolic changes correlated with changes in the composition of the gut microbiota, and transfer of the gut microbiota from TLR5-deficient mice to wild-type germ-free mice conferred many features of metabolic syndrome to the recipients. Food restriction prevented obesity, but not insulin resistance, in the TLR5-deficient mice. These results support the emerging view that the gut microbiota contributes to metabolic disease and suggest that malfunction of the innate immune system may promote the development of metabolic syndrome.


Assuntos
Fenômenos Fisiológicos Bacterianos , Imunidade Inata , Intestinos/microbiologia , Síndrome Metabólica/etiologia , Receptor 5 Toll-Like/metabolismo , Animais , Glicemia/análise , Distribuição da Gordura Corporal , Peso Corporal , Restrição Calórica , Gorduras na Dieta/administração & dosagem , Feminino , Vida Livre de Germes , Hiperfagia/etiologia , Resistência à Insulina , Mucosa Intestinal/imunologia , Masculino , Síndrome Metabólica/imunologia , Síndrome Metabólica/microbiologia , Camundongos , Camundongos Knockout , Obesidade/etiologia , Obesidade/imunologia , Obesidade/microbiologia , Obesidade/prevenção & controle , Receptor 5 Toll-Like/deficiência , Receptor 5 Toll-Like/genética
15.
Semin Immunopathol ; 30(1): 11-21, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18066550

RESUMO

The intestine is normally colonized by a large and diverse commensal microbiota and is occasionally exposed to a variety of potential pathogens. In recent years, there has been substantial progress made in identifying molecular mechanisms that normally serve to protect the intestine from such enteric bacteria and which may go awry in chronic idiopathic inflammatory diseases of the gut. One specific molecular interaction that appears to play a key role in governing bacterial-intestinal interactions is that of the bacterial protein flagellin with toll-like receptor 5. This article reviews studies performed in vitro, in mice, and in humans that indicate an important role for the flagellin-TLR5 interaction in regulating both the innate and adaptive immune responses in the intestine.


Assuntos
Colite/imunologia , Flagelina/imunologia , Trato Gastrointestinal/imunologia , Receptor 5 Toll-Like/metabolismo , Animais , Colite/metabolismo , Flagelina/metabolismo , Trato Gastrointestinal/metabolismo , Humanos , Imunidade Inata , Polimorfismo Genético , Salmonelose Animal/imunologia , Transdução de Sinais , Receptor 5 Toll-Like/genética , Receptor 5 Toll-Like/imunologia
16.
J Immunol ; 180(12): 8280-5, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18523294

RESUMO

Sudden exposure of human populations to chemicals, pathogens, or radiation has the potential to result in substantial morbidity. A potential means of rapidly protecting such populations might be to activate innate host defense pathways, which can provide broad protection against a variety of insults. However, innate immune activators can, by themselves, result in severe inflammatory pathology, which in large part is driven by hemopoietic-derived cytokines such as TNF-alpha. We reasoned that, because it preferentially activates epithelial cells, the TLR5 agonist flagellin might not induce severe inflammatory pathology and yet be an ideal agent to provide such non-specific protection, particularly at the mucosal surfaces that serve as a front line of host defense. In accordance, we observed that systemic treatment of mice with purified flagellin did not induce the serologic, histopathologic, and clinical hallmarks of inflammation that are induced by LPS but yet protected mice against chemicals, pathogens, and ionizing radiation. Flagellin-elicited radioprotection required TLR5, the TLR signaling adaptor MyD88, and was effective if given between 2 h before to 4 h after exposure to irradiation. Flagellin-elicited radioprotection was, in part, mediated via effects on cells in bone marrow but yet rescued mortality without a pronounced rescue of radiation-induced anemia or leukopenia. Thus, systemic administration of flagellin may be a relatively safe means of providing temporary non-specific protection against a variety of challenges.


Assuntos
Sulfato de Dextrana/administração & dosagem , Flagelina/administração & dosagem , Raios gama , Protetores contra Radiação/administração & dosagem , Infecções por Rotavirus/prevenção & controle , Salmonelose Animal/prevenção & controle , Administração Oral , Animais , Citocinas/biossíntese , Sulfato de Dextrana/efeitos adversos , Flagelina/uso terapêutico , Raios gama/efeitos adversos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/efeitos da radiação , Injeções Intraperitoneais , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/efeitos adversos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/fisiologia , Protetores contra Radiação/uso terapêutico , Infecções por Rotavirus/imunologia , Salmonelose Animal/imunologia , Receptor 5 Toll-Like/deficiência , Receptor 5 Toll-Like/genética , Receptor 5 Toll-Like/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa