Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 15(6): e0234726, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32559205

RESUMO

Hepatocellular carcinoma (HCC), the most malignant form of primary liver cancer, is the fourth most prevalent cause of cancer mortality globally. It was recently discovered that the dietary fermentable fiber, inulin, can reprogram the murine liver to favor HCC development in a gut microbiota-dependent manner. Determining the molecular pathways that are either over expressed or repressed during inulin-induced HCC would provide a platform of potential therapeutic targets. In the present study, we have combined analysis of the novel inulin-induced HCC murine model and human HCC samples to identify differentially expressed genes (DEGs) in hepatocarcinogenesis. Hepatic transcriptome profiling revealed that there were 674 DEGs in HCC mice compared to mice safeguarded from HCC. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis uncovered enrichment in ECM-receptor interaction, steroid hormone biosynthesis, PPAR signaling pathway, focal adhesion and protein digestion and absorption during inulin-induced HCC. Tandem mass tag based quantitative, multiplexed proteomic analysis delineated 57 differentially expressed proteins, where the over-expressed proteins were associated with cell adhesion molecules, valine, leucine and isoleucine degradation and ECM-receptor interaction. After obtaining the human orthologs of the mouse genes, we did a comparison analysis to level 3 RNA-seq data found in the Cancer Genome Atlas (TCGA) database, corresponding to human HCC (n = 361) and healthy liver (n = 50) samples. Out of the 549 up-regulated and 68 down-regulated human orthologs identified, 142 genes (137 significantly over-expressed and 5 significantly under-expressed) were associated with human HCC. Using univariate survival analysis, we found 27 over-expressed genes involved in cell-cell adhesion and cell division that were associated with poor HCC patient survival. Overall, the genetic and proteomics signatures highlight potential underlying mechanisms in inulin-induced HCC and support that this murine HCC model is human relevant.


Assuntos
Carcinoma Hepatocelular/patologia , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/patologia , Animais , Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/mortalidade , Moléculas de Adesão Celular/metabolismo , Modelos Animais de Doenças , Ontologia Genética , Humanos , Insulina/toxicidade , Estimativa de Kaplan-Meier , Fígado/metabolismo , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Proteômica , Transdução de Sinais , Receptor 5 Toll-Like/deficiência , Receptor 5 Toll-Like/genética , Transcriptoma
2.
Am J Physiol Gastrointest Liver Physiol ; 318(5): G955-G965, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32200644

RESUMO

Functional fermentable fibers are considered essential for a healthy diet. Recently, we demonstrated that gut microbiota dysbiotic mice fed an inulin-containing diet (ICD) developed hepatocellular carcinoma (HCC) within 6 mo. In particular, a subset of Toll-like receptor 5-deficient (T5KO) mice prone to HCC exhibited rapid onset of hyperbilirubinemia (HB) and cholemia; these symptoms provide rationale that ICD induces cholestasis. Our objective in the present study was to determine whether inulin-fed T5KO-HB mice exhibit other known consequences of cholestasis, including essential fatty acid and fat-soluble vitamin deficiencies. Here, we measured hepatic fatty acids and serum vitamin A and D levels from wild-type (WT), T5KO low bilirubin (LB) and T5KO-HB mice fed ICD for 4 wk. Additionally, hepatic RNAseq and proteomics were performed to ascertain other metabolic alterations. Compared with WT and T5KO-LB, T5KO-HB mice exhibited steatorrhea, i.e., ~50% increase in fecal lipids. This could contribute to the significant reduction of linoleate in hepatic neutral lipids in T5KO-HB mice. Additionally, serum vitamins A and D were ~50% reduced in T5KO-HB mice, which was associated with metabolic compromises. Overall, our study highlights that fermentable fiber-induced cholestasis is further characterized by depletion of macro-and micronutrients.NEW & NOTEWORTHY Feeding a dietary, fermentable fiber diet to a subset of Toll-like receptor 5 deficient (T5KO) mice induces early onset hyperbilirubinemia and cholemia that later manifests to hepatocellular carcinoma (HCC). Our study highlights that fermentable fiber-induced cholestasis is characterized with modest macro- and micronutrient deficiencies that may further contribute to hepatic biliary disease. Compared with chemical induction, immunization, surgery, or genetic manipulation, these findings provide a novel approach to study the cholestatic subtype of HCC.


Assuntos
Fibras na Dieta , Fígado Gorduroso/metabolismo , Absorção Intestinal , Inulina , Metabolismo dos Lipídeos , Fígado/metabolismo , Síndromes de Malabsorção/metabolismo , Receptor 5 Toll-Like/deficiência , Deficiência de Vitamina A/metabolismo , Deficiência de Vitamina D/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Colestase/genética , Colestase/metabolismo , Colestase/patologia , Modelos Animais de Doenças , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Fermentação , Fígado/patologia , Síndromes de Malabsorção/genética , Síndromes de Malabsorção/patologia , Masculino , Camundongos Knockout , Receptor 5 Toll-Like/genética , Deficiência de Vitamina A/genética , Deficiência de Vitamina D/genética
3.
Chem Biol Interact ; 299: 102-110, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30508503

RESUMO

BACKGROUND: Liver injury is a serious threat for human health and life. Toll-like receptor 5 (TLR5) has reported to be a vital mediator in flagellin or tetrachloride (CCl4)-induced liver injury. However, the roles and etiology of TLR5 in hyperammonaemia (HA)-induced liver injury are poor defined. METHODS: HA rats were generated by intragastric administration using ammonium chloride solution. Liver status was assessed by haematoxylin and eosin (H&E) staining and measuring serum levels of liver injury markers. Immunohistochemistry (IHC) assay was used to visualize protein expression in tissues. Apoptotic index in tissues was determined by TUNEL assay. RT-qPCR assay was employed to test mRNA expression. Oxidative stress responses was assessed by detecting levels of reactive oxygen species (ROS) and related indicators. NF-κB activity was examined by TransAM NF-κB colorimetric kit. RESULTS: TLR5 was highly expressed in liver tissues of HA rats. TLR5 knockdown ameliorated HA-induced liver injury by inhibiting liver cell apoptosis. TLR5 depletion inhibited HA-induced pro-inflammatory cytokine expression in liver tissues, but had no effect on the infiltration of T and macrophage cells into liver tissues. TLR5 silencing impaired HA-induced oxidative stress responses in hepatocytes, but not in hepatic stellate cells (HSCs). TLR5 downregulation inhibited HA-induced activation on TLR5/NF-κB and TLR5/MAPK signaling pathways. CONCLUSION: TLR5 silencing reduced HA-induced liver injury by inhibiting hepatocyte apoptosis, oxidative stress and inflammation responses via inactivating NF-κB and MAPK signals, deepening our understanding on the molecular mechanism of HA-induced liver injury and providing a potential therapeutic target for alleviating liver injury.


Assuntos
Citocinas/metabolismo , Hiperamonemia/patologia , Hepatopatias/patologia , Estresse Oxidativo , Transdução de Sinais , Receptor 5 Toll-Like/genética , Alanina Transaminase/sangue , Cloreto de Amônio/toxicidade , Animais , Apoptose/efeitos dos fármacos , Aspartato Aminotransferases/sangue , Células Estreladas do Fígado/citologia , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Hiperamonemia/complicações , Hiperamonemia/veterinária , Fígado/metabolismo , Fígado/patologia , Hepatopatias/etiologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 5 Toll-Like/deficiência
4.
Osteoarthritis Cartilage ; 27(1): 129-139, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30240938

RESUMO

OBJECTIVE: Metabolic syndrome is characterized by obesity, hyperglycemia, hypertension, insulin resistance, and dyslipidemia. Metabolic syndrome is associated with osteoarthritis (OA), but it is unclear if the association is attributable to increased mechanical loading on joints caused by obesity or other aspects of metabolic syndrome. Here we examined the effects of altered metabolism, obesity, and the gut microbiome on load-induced OA. DESIGN: Cartilage damage was induced through cyclic compressive loading in four groups of adult male mice: Toll-like receptor-5 deficient (TLR5KO) mice that develop metabolic syndrome due to alterations in the gut microbiome, TLR5KO mice submitted to chronic antibiotics to prevent metabolic syndrome (TLR5KOΔMicrobiota), C57BL/6J mice fed a high fat diet to cause obesity (HFD), and untreated C57BL/6J mice (WT). Loading was applied for 2 weeks (n = 10-11/group) or 6 weeks (n = 10-11/group). RESULTS: After 2 weeks of loading, cartilage damage (OARSI score) was not different among groups. After 6 weeks of loading, HFD mice had increased load-induced cartilage damage, while TLR5KO mice had cartilage damage comparable to WT mice. TLR5KOΔMicrobiota mice had less cartilage damage than other groups. HFD mice had elevated serum inflammatory markers. Each group had a distinct gut microbiome composition. CONCLUSIONS: Severe obesity increased load-induced cartilage damage, while milder changes in adiposity/metabolic syndrome seen in TLR5KO mice did not. Furthermore, the effects of systemic inflammation/obesity on cartilage damage depend on the duration of mechanical loading. Lastly, reduced cartilage damage in the TLR5KOΔMicrobiota mice suggests that the gut microbiome may influence cartilage pathology.


Assuntos
Artrite Experimental/etiologia , Microbioma Gastrointestinal , Síndrome Metabólica/complicações , Obesidade/complicações , Osteoartrite/etiologia , Tecido Adiposo/patologia , Animais , Artrite Experimental/microbiologia , Artrite Experimental/patologia , Biomarcadores/sangue , Índice de Massa Corporal , Cartilagem Articular/patologia , Citocinas/sangue , Mediadores da Inflamação/sangue , Lipopolissacarídeos/sangue , Masculino , Síndrome Metabólica/sangue , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/sangue , Osteoartrite/microbiologia , Osteoartrite/patologia , Receptor 5 Toll-Like/deficiência , Receptor 5 Toll-Like/genética , Suporte de Carga/fisiologia
5.
PLoS One ; 13(4): e0195310, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29617463

RESUMO

BACKGROUND: Inability to maintain a stable and beneficial microbiota is associated with chronic gut inflammation, which classically manifests as colitis but may more commonly exist as low-grade inflammation that promotes metabolic syndrome. Alterations in microbiota, and associated inflammation, can originate from dysfunction in host proteins that manage the microbiota, such as the flagellin receptor TLR5. That the complete absence of a microbiota (i.e. germfree conditions) eliminates all evidence of inflammation in TLR5-deficient mice demonstrates that this model of gut inflammation is microbiota-dependent. We hypothesize that such microbiota dependency reflects an inability to manage pathobionts, such as Adherent-Invasive E. coli (AIEC). Herein, we examined the extent to which microbiota mismanagement and associated inflammation in TLR5-deficient mice would manifest in a limited and pathobiont-free microbiota. For this purpose, WT and TLR5-deficient mice were generated and maintained with the 8-member consortium of bacteria referred to as "Altered Schaedler Flora" (ASF). Such ASF animals were subsequently inoculated with AIEC reference strain LF82. Feces were assayed for bacterial loads, fecal lipopolysaccharide and flagellin loads, fecal inflammatory marker lipocalin-2 and microbiota composition. RESULTS: Relative to similarly maintained WT mice, mice lacking TLR5 (T5KO) did not display low-grade intestinal inflammation nor metabolic syndrome under ASF conditions. Concomitantly, the ASF microbial community was similar between WT and T5KO mice, while inoculation with AIEC strain LF82 resulted in alteration of the ASF community in T5KO mice compared to WT control animals. AIEC LF82 inoculation in ASF T5KO mice resulted in microbiota components having elevated levels of bioactive lipopolysaccharide and flagellin, a modest level of low-grade inflammation and increased adiposity. CONCLUSIONS: In a limited-complexity pathobiont-free microbiota, loss of the flagellin receptor TLR5 does not impact microbiota composition nor its ability to promote inflammation. Addition of AIEC to this ecosystem perturbs microbiota composition, increases levels of lipopolysaccharide and flagellin, but only modestly promotes gut inflammation and adiposity, suggesting that the phenotypes previously associated with loss of this innate immune receptor require disruption of complex microbiota.


Assuntos
Gastroenterite/imunologia , Gastroenterite/microbiologia , Microbioma Gastrointestinal/imunologia , Síndromes de Imunodeficiência/metabolismo , Síndromes de Imunodeficiência/microbiologia , Intestinos/imunologia , Intestinos/microbiologia , Animais , Escherichia coli , Transplante de Microbiota Fecal , Fezes/microbiologia , Feminino , Flagelina/metabolismo , Imunidade Inata , Lipopolissacarídeos/metabolismo , Masculino , Síndrome Metabólica/imunologia , Síndrome Metabólica/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor 5 Toll-Like/deficiência , Receptor 5 Toll-Like/genética
6.
Sci Rep ; 7: 42688, 2017 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-28202909

RESUMO

Toll-like receptors (TLR) provide a critical link between innate and adaptive immunity, both important players in atherosclerosis. Since evidence for the role of TLR5 is lacking, we aimed to establish this in the immune axis of atherosclerosis. We assessed the effect of the TLR5-specific ligand Flagellin on macrophage maturation and T-cell polarisation. Next, we generated TLR5-/-LDLr-/- chimeras to study the effect of hematopoietic TLR5 deficiency on atherosclerosis formation. Flagellin stimulation did not influence wildtype or TLR5-/- macrophage maturation. Only in wildtype macrophages, Flagellin exposure increased MCP-1 and IL6 expression. Flagellin alone reduced T-helper 1 proliferation, which was completely overruled in the presence of T-cell receptor activation. In vivo, hematopoietic TLR5 deficiency attenuated atherosclerotic lesion formation by ≈25% (1030*103 ± 63*103 vs. 792*103 ± 61*103 µm2; p = 0.013) and decreased macrophage area (81.3 ± 12.0 vs. 44.2 ± 6.6 µm2; p = 0.011). In TLR5-/- chimeric mice, we observed lower IL6 plasma levels (36.4 ± 5.6 vs. 15.1 ± 2.2 pg/mL; p = 0.003), lower (activated) splenic CD4+ T-cell content (32.3 ± 2.1 vs. 21.0 ± 1.2%; p = 0.0018), accompanied by impaired T-cell proliferative responses. In conclusion, hematopoietic TLR5 deficiency inhibits atherosclerotic lesion formation by attenuated macrophage accumulation and defective T-cell responsiveness.


Assuntos
Aterosclerose/etiologia , Aterosclerose/metabolismo , Leucócitos/imunologia , Leucócitos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Receptor 5 Toll-Like/deficiência , Animais , Aterosclerose/sangue , Aterosclerose/patologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Movimento Celular/genética , Movimento Celular/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Contagem de Leucócitos , Camundongos , Camundongos Knockout , Monócitos/imunologia , Monócitos/metabolismo , Placa Aterosclerótica/genética , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Subpopulações de Linfócitos T/citologia
7.
Biomed Pharmacother ; 81: 345-355, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27261612

RESUMO

Atherosclerosis is a specific form of an artery wall thickens, a syndrome affecting arterial blood vessels due to a chronic inflammatory response in the walls of arteries, which is promoted by fat accumulation. Toll-like receptors (TLRs) play prominent roles in inflammatory responses. And TLR5 is overexpressed in several diseases. Here in our study, we investigated the effect of TLR5 in high fat-induced atherosclerosis via NF-κB signaling pathway modulating pro-inflammatory cytokines releasing. Our results found that high fat induced atherosclerosis in wild type mice with fat accumulation and inflammatory response through NF-κB activation. Contrastly, TLR5 knockout mice displayed lower fat accumulation and ameliorated inflammation after high fat feeding with NF-κB inactivation. In addition, pterostilbene, as a natural dimethyl ether derivative of resveratrol mainly from blueberries, has diverse pharmacological activities, especially anti-inflammation. Our study also found that pterostilbene displayed inhibited role in suppressing inflammatory response through inactivating NF-κB signaling pathway regulated by TLR5 down-regulation in high fat-induced mice. Moreover, in vitro experiments of vascular smooth muscle cells (VSMCs) challenged with LPS or TNF-α, further indicated that NF-κB was involved in atherosclerosis progression, leading to high secretion of pro-inflammatory cytokines. However, VSMCs from TLR5 deficient mice inhibited phosphorylated levels of NF-κB signalilng pathway, finally resulting in down-regulation of inflammatory cytokines. Notably, pterostilbene also displayed suppressed role in inflammatory response via NF-κB inactivity in LPS or TNF-α-induced VSMCs by decreasing TLR5 expression. The results above indicated a novel therapeutic strategy of pterostilbene to protect against atherosclerosis via TLR5 regulation for clinic treatment in the future.


Assuntos
Aterosclerose/tratamento farmacológico , Dieta Hiperlipídica , Inflamação/tratamento farmacológico , NF-kappa B/metabolismo , Transdução de Sinais , Estilbenos/uso terapêutico , Receptor 5 Toll-Like/deficiência , Animais , Aterosclerose/complicações , Aterosclerose/metabolismo , Aterosclerose/patologia , Técnicas de Silenciamento de Genes , Inflamação/complicações , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estilbenos/farmacologia , Receptor 5 Toll-Like/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
8.
Sci Rep ; 6: 24199, 2016 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-27063435

RESUMO

Flagellin has the capacity to activate both Toll-like receptor 5 (TLR5) and Nod-like receptor C4 (NLRC4)/neuronal apoptosis inhibitory protein 5 (NAIP5) inflammasome signaling. We fused E7m (the inactivated E7 of human papillomavirus) to either end of the flagellin protein, and the resulting recombinant flagellin-E7m proteins (rFliCE7m and rE7mFliC) were used as immunogens. Both fusion proteins activated receptor signaling to different degrees. rE7mFliC-induced TLR5 activity was 10-fold higher than that of rFliCE7m, whereas rFliCE7m activated the NLRC4/NAIP5 pathway more strongly. Therefore, these recombinant proteins provided a tool to investigate which signaling pathway is critical for the induction of antigen-specific T cell responses and anti-tumor immunity. We demonstrated that rFliCE7m induced higher levels of E7-specific IFN-gamma-secreting cells and cytotoxic T lymphocytes (CTLs) than rE7mFliC, and a single injection with rFliCE7m but not rE7mFliC inhibited E7-expressing tumor growth in vivo. Furthermore, we confirmed that CD8(+) T cells played a major role in the anti-tumor immunity induced by rFliCE7m. These findings suggested that the NLRC4/NAIP5 intracellular signaling pathway was critical for the induction of anti-tumor immunity. These observations provide important information for the rational design of flagellin-based immunotherapy.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/metabolismo , Flagelina/metabolismo , Proteína Inibidora de Apoptose Neuronal/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Receptor 5 Toll-Like/metabolismo , Animais , Células da Medula Óssea/citologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Dicroísmo Circular , ELISPOT , Feminino , Flagelina/genética , Genes Reporter , Células HEK293 , Humanos , Imunidade Inata , Interferon gama/análise , Interferon gama/metabolismo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas E7 de Papillomavirus/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais/efeitos dos fármacos , Receptor 5 Toll-Like/deficiência , Receptor 5 Toll-Like/genética
9.
PLoS One ; 11(3): e0150943, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26950299

RESUMO

Over the past decade, emerging evidence has linked alterations in the gut microbial composition to a wide range of diseases including obesity, type 2 diabetes, and cardiovascular disease. Toll-like receptors (TLRs) are the major mediators for the interactions between gut microbiota and host innate immune system, which is involved in the localization and structuring of host gut microbiota. A previous study found that TLR5 deficient mice (TLR5KO1) had altered gut microbial composition which led to the development of metabolic syndrome including hyperlipidemia, hypertension, insulin resistance and increased adiposity. In the current study, a second TLR5-deficient mouse model was studied (TLR5KO2). TLR5 deficient mice did not manifest metabolic abnormalities related to the metabolic syndrome compared with littermate controls maintained on normal chow or after feeding a high fat diet. Analysis of the gut microbial composition of littermate TLR5KO2 and wild type mice revealed no significant difference in the overall microbiota structure between genotypes. However, the TLR5KO2 microbiota was distinctly different from that previously reported for TLR5KO1 mice with metabolic syndrome. We conclude that an altered composition of the microbiota in a given environment can result in metabolic syndrome, but it is not a consequence of TLR5 deficiency per se.


Assuntos
Meio Ambiente , Microbioma Gastrointestinal/genética , Deleção de Genes , Genótipo , Síndrome Metabólica/genética , Síndrome Metabólica/microbiologia , Receptor 5 Toll-Like/genética , Animais , Bactérias/classificação , Bactérias/genética , Glicemia/metabolismo , Composição Corporal/genética , Peso Corporal/genética , Ingestão de Alimentos/genética , Técnicas de Inativação de Genes , Interação Gene-Ambiente , Teste de Tolerância a Glucose , Imunidade Inata/genética , Insulina/sangue , Resistência à Insulina/genética , Lipídeos/sangue , Síndrome Metabólica/imunologia , Camundongos , Receptor 5 Toll-Like/deficiência
10.
Mediators Inflamm ; 2015: 852126, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26681840

RESUMO

BACKGROUND: This study aims to investigate the effect of feeding low-fat diet (LFD) to diet-induced obesity (DIO) mice lacking TLR5 (TLR5(-/-)), which have a tendency to develop glucose intolerance with increased adiposity, compared to that in C57BL/6 mice. RESULTS: TLR5(-/-) and C57BL/6 male mice were divided into three subgroups: (1) control, mice were fed a standard AIN-76A (fat: 11.5 kcal%) diet for 12 weeks; (2) DIO, mice were fed a 58 kcal% high-fat diet (HFD) for 12 weeks; and (3) diet, mice were fed a HFD for 8 weeks to induce obesity and then switched to a 10.5 kcal% LFD for 4 weeks. The glucose intolerance in DIO TLR5(-/-) mice was more significant than that in DIO C57BL/6 mice and was not attenuated by a switch to the LFD. Weight-reduction with LFD had significantly decreased the epididymal fat mass in C57BL/6 mice but not in TLR5(-/-) mice. In addition, the LFD-fed TLR5(-/-) mice showed significantly higher expression of ghrelin in the serum and resistin in the epididymal fat than that in C57BL/6 mice. CONCLUSIONS: This study demonstrated that TLR5 gene knockout impairs some effects of weight-reduction in DIO.


Assuntos
Dieta com Restrição de Gorduras , Obesidade/dietoterapia , Obesidade/imunologia , Receptor 5 Toll-Like/deficiência , Adiposidade , Animais , Citocinas/sangue , Dieta Hiperlipídica/efeitos adversos , Grelina/sangue , Intolerância à Glucose/etiologia , Intolerância à Glucose/imunologia , Intolerância à Glucose/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo , Resistina/metabolismo , Receptor 5 Toll-Like/genética , Aumento de Peso , Redução de Peso
11.
Cell Metab ; 22(6): 983-96, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26525535

RESUMO

The gut microbiota plays a key role in host metabolism. Toll-like receptor 5 (TLR5), a flagellin receptor, is required for gut microbiota homeostasis. Accordingly, TLR5-deficient (T5KO) mice are prone to develop microbiota-dependent metabolic syndrome. Here we observed that T5KO mice display elevated neutral lipids with a compositional increase of oleate [C18:1 (n9)] relative to wild-type littermates. Increased oleate contribution to hepatic lipids and liver SCD1 expression were both microbiota dependent. Analysis of short-chain fatty acids (SCFAs) and (13)C-acetate label incorporation revealed elevated SCFA in ceca and hepatic portal blood and increased liver de novo lipogenesis in T5KO mice. Dietary SCFAs further aggravated metabolic syndrome in T5KO mice. Deletion of hepatic SCD1 not only prevented hepatic neutral lipid oleate enrichment but also ameliorated metabolic syndrome in T5KO mice. Collectively, these results underscore the key role of the gut microbiota-liver axis in the pathogenesis of metabolic diseases.


Assuntos
Fígado/metabolismo , Síndrome Metabólica/patologia , Estearoil-CoA Dessaturase/metabolismo , Receptor 5 Toll-Like/genética , Animais , Peso Corporal , Restrição Calórica , Dieta Hiperlipídica , Ácidos Graxos Voláteis/sangue , Fezes/química , Feminino , Resistência à Insulina , Intestinos/microbiologia , Lipogênese , Espectroscopia de Ressonância Magnética , Masculino , Síndrome Metabólica/metabolismo , Camundongos , Camundongos Knockout , Microbiota , Ácido Oleico/metabolismo , Estearoil-CoA Dessaturase/deficiência , Estearoil-CoA Dessaturase/genética , Receptor 5 Toll-Like/deficiência , Regulação para Cima
12.
Gut Microbes ; 6(4): 279-83, 2015 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-26067589

RESUMO

Alterations in the gut microbiota have been implicated to play a role in potentiating inflammatory bowel diseases in both humans and mice. Mice lacking the flagellin receptor, toll-like receptor 5 (TLR5), are prone to develop spontaneous gut inflammation, but are significantly protected when treated with antibiotics or maintained in germ-free conditions. However, given that the incidence of spontaneous inflammation in TLR5KO mice is quite variable in conventional conditions (typically ∼10% show clear colitis), this result is far from definitive and does not rule out that TLR5KO mice might be prone to develop inflammation even in the absence of a microbiota. Herein, we demonstrate that neutralization of IL10 signaling induces colitis in 100% of TLR5KO mice which provide a more rigorous approach to evaluate the role of microbiota in gut inflammation. Mice treated with antibiotics or maintained in germ-free condition are substantially protected against IL-10R neutralization-induced colitis, underscoring that gut inflammation in TLR5KO mice is dependent upon the presence of a gut microbiota.


Assuntos
Colite/patologia , Microbioma Gastrointestinal , Receptor 5 Toll-Like/deficiência , Animais , Antibacterianos/administração & dosagem , Vida Livre de Germes , Interleucina-10/antagonistas & inibidores , Camundongos , Camundongos Knockout , Receptores de Interleucina-10/deficiência
13.
Clin Sci (Lond) ; 129(2): 187-98, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25757463

RESUMO

Myocardial ischaemia-reperfusion (MIR) triggers a sterile inflammatory response important for myocardial healing, but which may also contribute to adverse ventricular remodelling. Such inflammation is initiated by molecular danger signals released by damaged myocardium, which induce innate immune responses by activating toll-like receptors (TLRs). Detrimental roles have been recently reported for TLR2, TLR3 and TLR4. The role of other TLRs is unknown. We therefore evaluated the role of TLR5, expressed at high level in the heart, in the development of myocardial damage and inflammation acutely triggered by MIR. TLR5(-/-) and wild-type (WT) mice were exposed to MIR (30 min ischaemia, 2 h reperfusion). We measured infarct size, markers of cardiac oxidative stress, myocardial phosphorylation state of mitogen-activated protein (MAP) kinases and AKT, expression levels of chemokines and cytokines in the heart and plasma, as well as cardiac function by echography and conductance volumetry. TLR5-deficient mice had normal cardiac morphology and function under physiological conditions. After MIR, the absence of TLR5 promoted an increase in infarct size and myocardial oxidative stress. Lack of TLR5 fostered p38 phosphorylation, reduced AKT phosphorylation and markedly increased the expression of inflammatory cytokines, whereas it precipitated acute LV (left ventricle) dysfunction. Therefore, contrary to the detrimental roles of TLR2, TLR3 and TLR4 in the infarcted heart, TLR5 is important to limit myocardial damage, inflammation and functional compromise after MIR.


Assuntos
Mediadores da Inflamação/metabolismo , Inflamação/metabolismo , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/metabolismo , Receptor 5 Toll-Like/deficiência , Animais , Modelos Animais de Doenças , Genótipo , Imunidade Inata , Inflamação/genética , Inflamação/imunologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/genética , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/imunologia , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio/imunologia , Miocárdio/patologia , Estresse Oxidativo , Fenótipo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor 5 Toll-Like/genética , Disfunção Ventricular Esquerda/imunologia , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/patologia , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Cell Mol Immunol ; 12(6): 729-42, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25418468

RESUMO

Flagellin is a potent activator of a broad range of cell types that are involved in innate and adaptive immunity. Therefore, it is a good adjuvant candidate for vaccines, and it might function as a biological protectant against both major acute radiation syndrome during cancer radiotherapy and a mitigator of radiation emergencies. However, accumulating evidence has implicated flagellin in the occurrence of some inflammatory diseases, such as acute lung inflammation, cardiovascular collapse and inflammatory bowel disease. The aim of this study was to elucidate whether only flagellin-TLR5 signaling activation plays a role in the pathophysiology of liver or whether some other flagellin activity also contributes to liver injury either via bacterial infections or during clinical applications. Recombinant flagellin proteins with or without TLR5-stimulating activity were used to evaluate the role of flagellin-TLR5 signaling in liver injury in wild-type and TLR5 KO mice. Gross lesions and large areas of hepatocellular necrosis were observed in liver tissue 12 h after the intraperitoneal administration of 100 or 200 µg flagellin (FliC) in a dose- and time-dependent manner in wild-type mice, but not in TLR5 KO mice. Deletion of the N-terminal or TLR5 binding domain of flagellin inhibited flagellin-induced inflammatory responses and the subsequent acute liver function abnormality and damage. These data confirmed that flagellin is an essential determinant of liver injury and demonstrated that the over-activation of TLR5 signaling by high-dose flagellin caused acute inflammatory responses, neutrophil accumulation and oxidative stress in the liver, which contributes to the progression and severity of flagellin-induced liver injury.


Assuntos
Sequência de Aminoácidos , Doença Hepática Induzida por Substâncias e Drogas/imunologia , Flagelina/genética , Deleção de Sequência , Receptor 5 Toll-Like/genética , Animais , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/patologia , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Feminino , Flagelina/imunologia , Flagelina/farmacologia , Regulação da Expressão Gênica , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Infiltração de Neutrófilos , Estrutura Terciária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Salmonella enterica/química , Salmonella enterica/patogenicidade , Transdução de Sinais , Receptor 5 Toll-Like/deficiência , Receptor 5 Toll-Like/imunologia
15.
J Immunol ; 192(4): 1587-96, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24442437

RESUMO

Flagellin is a potent immunogen that activates the innate immune system via TLR5 and Naip5/6, and generates strong T and B cell responses. The adaptor protein MyD88 is critical for signaling by TLR5, as well as IL-1Rs and IL-18Rs, major downstream mediators of the Naip5/6 Nlrc4-inflammasome. In this study, we define roles of known flagellin receptors and MyD88 in Ab responses generated toward flagellin. We used mice genetically deficient in flagellin recognition pathways to characterize innate immune components that regulate isotype-specific Ab responses. Using purified flagellin from Salmonella, we dissected the contribution of innate flagellin recognition pathways to promote Ab responses toward flagellin and coadministered OVA in C57BL/6 mice. We demonstrate IgG2c responses toward flagellin were TLR5 and inflammasome dependent; IgG1 was the dominant isotype and partially TLR5 and inflammasome dependent. Our data indicate a substantial flagellin-specific IgG1 response was induced through a TLR5-, inflammasome-, and MyD88-independent pathway. IgA anti-FliC responses were TLR5 and MyD88 dependent and caspase-1 independent. Unlike C57BL/6 mice, flagellin-immunized A/J mice induced codominant IgG1 and IgG2a responses. Furthermore, MyD88-independent, flagellin-induced Ab responses were even more pronounced in A/J MyD88(-/-) mice, and IgA anti-FliC responses were suppressed by MyD88. Flagellin also worked as an adjuvant toward coadministered OVA, but it only promoted IgG1 anti-OVA responses. Our results demonstrate that a novel pathway for flagellin recognition contributes to Ab production. Characterization of this pathway will be useful for understanding immunity to flagellin and the rationale design of flagellin-based vaccines.


Assuntos
Flagelina/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Proteína Inibidora de Apoptose Neuronal/metabolismo , Receptor 5 Toll-Like/metabolismo , Animais , Caspase 1/deficiência , Caspase 1/genética , Caspase 1/metabolismo , Células Cultivadas , Flagelina/genética , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , Inflamassomos/metabolismo , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Proteína Inibidora de Apoptose Neuronal/deficiência , Proteína Inibidora de Apoptose Neuronal/genética , Ovalbumina , Receptores de IgG/metabolismo , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-18/metabolismo , Salmonella typhimurium/enzimologia , Salmonella typhimurium/genética , Receptor 5 Toll-Like/deficiência , Receptor 5 Toll-Like/genética
16.
Gut ; 63(7): 1069-80, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23896971

RESUMO

BACKGROUND: Inflammatory bowel disease (IBD) is driven by a seemingly aberrant immune response to the gut microbiota with disease development dictated by genetics and environmental factors. A model exemplifying this notion is our recent demonstration that colonisation of adherent-invasive Escherichia coli (AIEC) during microbiota acquisition drove chronic colitis in mice lacking the flagellin receptor TLR5 (T5KO). T5KO colitis persisted beyond AIEC clearance and requires TLR4 and the NLRC4 inflammasome. We hypothesised that AIEC instigates chronic inflammation by increasing microbial lipopolysaccharide (LPS) and flagellin levels. GOAL: Examine if transient AIEC colonisation lastingly alters levels of LPS and flagellin and changes microbiota composition. METHODS: Germ-free mice (wild type (WT) and T5KO) were inoculated with AIEC strain LF82 and placed in standard housing allowing a complex microbiota that eliminated AIEC in both mice strains. Faeces were assayed for the inflammatory marker, lipocalin-2, bacterial loads, and microbiota composition by pyrosequencing. Faecal LPS and flagellin bioactivity were measured via a cell-based reporter assay. RESULTS: Transient AIEC colonisation, in WT mice, did not alter inflammatory markers, bacterial loads, microbiota composition, nor its pro-inflammatory potential. By contrast, transient AIEC colonisation of T5KO mice drove chronic inflammation which correlated with microbiota components having higher levels of bioactive LPS and flagellin. Such AIEC-induced elevation of LPS and flagellin persisted well beyond AIEC clearance, required AIEC be flagellated, and was associated with alteration in microbiota species composition including a loss of species diversity. CONCLUSIONS: AIEC, and perhaps other pathobionts, may instigate chronic inflammation in susceptible hosts by altering the gut microbiota composition so as to give it an inherently greater ability to activate innate immunity/pro-inflammatory gene expression.


Assuntos
Colite/microbiologia , Colo/microbiologia , Infecções por Escherichia coli/complicações , Mucosa Intestinal/microbiologia , Microbiota , Receptor 5 Toll-Like/deficiência , Proteínas de Fase Aguda/metabolismo , Animais , Aderência Bacteriana/imunologia , Carga Bacteriana , Biomarcadores/metabolismo , Doença Crônica , Colite/imunologia , DNA Bacteriano/análise , Ensaio de Imunoadsorção Enzimática , Escherichia coli/genética , Escherichia coli/imunologia , Infecções por Escherichia coli/imunologia , Fezes/química , Fezes/microbiologia , Flagelina/metabolismo , Vida Livre de Germes , Imunidade Inata , Mucosa Intestinal/imunologia , Lipocalina-2 , Lipocalinas/metabolismo , Lipopolissacarídeos/metabolismo , Camundongos , Camundongos Knockout , Proteínas Oncogênicas/metabolismo , Análise de Sequência de DNA
18.
Cell Host Microbe ; 14(5): 571-81, 2013 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-24237702

RESUMO

Gut mucosal barrier breakdown and inflammation have been associated with high levels of flagellin, the principal bacterial flagellar protein. Although several gut commensals can produce flagella, flagellin levels are low in the healthy gut, suggesting the existence of control mechanisms. We find that mice lacking the flagellin receptor Toll-like receptor 5 (TLR5) exhibit a profound loss of flagellin-specific immunoglobulins (Igs) despite higher total Ig levels in the gut. Ribotyping of IgA-coated cecal microbiota showed Proteobacteria evading antibody coating in the TLR5(-/-) gut. A diversity of microbiome members overexpressed flagellar genes in the TLR5(-/-) host. Proteobacteria and Firmicutes penetrated small intestinal villi, and flagellated bacteria breached the colonic mucosal barrier. In vitro, flagellin-specific Ig inhibited bacterial motility and downregulated flagellar gene expression. Thus, innate-immunity-directed development of flagellin-specific adaptive immune responses can modulate the microbiome's production of flagella in a three-way interaction that helps to maintain mucosal barrier integrity and homeostasis.


Assuntos
Flagelina/imunologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Imunidade nas Mucosas , Locomoção , Microbiota/imunologia , Imunidade Adaptativa , Animais , Flagelos/imunologia , Imunidade Inata , Imunoglobulina A/imunologia , Camundongos , Camundongos Knockout , Receptor 5 Toll-Like/deficiência , Receptor 5 Toll-Like/imunologia
19.
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
20.
Immunity ; 36(2): 276-87, 2012 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-22306017

RESUMO

Microbial penetration of the intestinal epithelial barrier triggers inflammatory responses that include induction of the bactericidal C-type lectin RegIIIγ. Systemic administration of flagellin, a bacterial protein that stimulates Toll-like receptor 5 (TLR5), induces epithelial expression of RegIIIγ and protects mice from intestinal colonization with antibiotic-resistant bacteria. Flagellin-induced RegIIIγ expression is IL-22 dependent, but how TLR signaling leads to IL-22 expression is incompletely defined. By using conditional depletion of lamina propria dendritic cell (LPDC) subsets, we demonstrated that CD103(+)CD11b(+) LPDCs, but not monocyte-derived CD103(-)CD11b(+) LPDCs, expressed high amounts of IL-23 after bacterial flagellin administration and drove IL-22-dependent RegIIIγ production. Maximal expression of IL-23 subunits IL-23p19 and IL-12p40 occurred within 60 min of exposure to flagellin. IL-23 subsequently induced a burst of IL-22 followed by sustained RegIIIγ expression. Thus, CD103(+)CD11b(+) LPDCs, in addition to promoting long-term tolerance to ingested antigens, also rapidly produce IL-23 in response to detection of flagellin in the lamina propria.


Assuntos
Células Dendríticas/imunologia , Flagelina/imunologia , Interleucina-23/biossíntese , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Animais , Antígenos CD/metabolismo , Antígeno CD11b/metabolismo , Células Dendríticas/classificação , Flagelina/administração & dosagem , Imunidade Inata , Imunidade nas Mucosas , Cadeias alfa de Integrinas/metabolismo , Interleucina-23/deficiência , Interleucina-23/genética , Interleucinas/biossíntese , Interleucinas/deficiência , Interleucinas/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas a Pancreatite , Proteínas/genética , Transdução de Sinais/imunologia , Receptor 5 Toll-Like/deficiência , Receptor 5 Toll-Like/genética , Receptor 5 Toll-Like/metabolismo , Regulação para Cima , Interleucina 22
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA