Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 7199, 2024 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-38532110

RESUMO

Development and progression of malignancies are accompanied and influenced by alterations in the surrounding immune microenvironment. Understanding the cellular and molecular interactions between immune cells and cancer cells has not only provided important fundamental insights into the disease, but has also led to the development of new immunotherapies. The C-type lectin Dendritic Cell ImmunoReceptor (DCIR) is primarily expressed by myeloid cells and is an important regulator of immune homeostasis, as demonstrated in various autoimmune, infectious and inflammatory contexts. Yet, the impact of DCIR on cancer development remains largely unknown. Analysis of available transcriptomic data of colorectal cancer (CRC) patients revealed that high DCIR gene expression is associated with improved patients' survival, immunologically "hot" tumors and high immunologic constant of rejection, thus arguing for a protective and immunoregulatory role of DCIR in CRC. In line with these correlative data, we found that deficiency of DCIR1, the murine homologue of human DCIR, leads to the development of significantly larger tumors in an orthotopic murine model of CRC. This phenotype is accompanied by an altered phenotype of tumor-associated macrophages (TAMs) and a reduction in the percentage of activated effector CD4+ and CD8+ T cells in CRC tumors of DCIR1-deficient mice. Overall, our results show that DCIR promotes antitumor immunity in CRC, making it an attractive target for the future development of immunotherapies to fight the second deadliest cancer in the world.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias Colorretais , Animais , Humanos , Camundongos , Linfócitos T CD8-Positivos/metabolismo , Neoplasias Colorretais/metabolismo , Células Dendríticas , Imunidade , Lectinas Tipo C/metabolismo , Microambiente Tumoral
2.
J Immunol ; 207(7): 1857-1870, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34479945

RESUMO

The lungs harbor multiple resident microbial communities, otherwise known as the microbiota. There is an emerging interest in deciphering whether the pulmonary microbiota modulate local immunity, and whether this knowledge could shed light on mechanisms operating in the response to respiratory pathogens. In this study, we investigate the capacity of a pulmonary Lactobacillus strain to modulate the lung T cell compartment and assess its prophylactic potential upon infection with Mycobacterium tuberculosis, the etiological agent of tuberculosis. In naive mice, we report that a Lactobacillus murinus (Lagilactobacillus murinus) strain (CNCM I-5314) increases the presence of lung Th17 cells and of a regulatory T cell (Treg) subset known as RORγt+ Tregs. In particular, intranasal but not intragastric administration of CNCM I-5314 increases the expansion of these lung leukocytes, suggesting a local rather than systemic effect. Resident Th17 and RORγt+ Tregs display an immunosuppressive phenotype that is accentuated by CNCM I-5314. Despite the well-known ability of M. tuberculosis to modulate lung immunity, the immunomodulatory effect by CNCM I-5314 is dominant, as Th17 and RORγt+ Tregs are still highly increased in the lung at 42-d postinfection. Importantly, CNCM I-5314 administration in M. tuberculosis-infected mice results in reduction of pulmonary inflammation, without increasing M. tuberculosis burden. Collectively, our findings provide evidence for an immunomodulatory capacity of CNCM I-5314 at steady state and in a model of chronic inflammation in which it can display a protective role, suggesting that L. murinus strains found in the lung may shape local T cells in mice and, perhaps, in humans.


Assuntos
Lactobacillus/fisiologia , Pulmão/imunologia , Mycobacterium tuberculosis/fisiologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Tuberculose Pulmonar/imunologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Humanos , Pulmão/microbiologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia
3.
Mol Cell ; 73(6): 1282-1291.e8, 2019 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-30792174

RESUMO

Toxin-antitoxin (TA) systems regulate fundamental cellular processes in bacteria and represent potential therapeutic targets. We report a new RES-Xre TA system in multiple human pathogens, including Mycobacterium tuberculosis. The toxin, MbcT, is bactericidal unless neutralized by its antitoxin MbcA. To investigate the mechanism, we solved the 1.8 Å-resolution crystal structure of the MbcTA complex. We found that MbcT resembles secreted NAD+-dependent bacterial exotoxins, such as diphtheria toxin. Indeed, MbcT catalyzes NAD+ degradation in vitro and in vivo. Unexpectedly, the reaction is stimulated by inorganic phosphate, and our data reveal that MbcT is a NAD+ phosphorylase. In the absence of MbcA, MbcT triggers rapid M. tuberculosis cell death, which reduces mycobacterial survival in macrophages and prolongs the survival of infected mice. Our study expands the molecular activities employed by bacterial TA modules and uncovers a new class of enzymes that could be exploited to treat tuberculosis and other infectious diseases.


Assuntos
Antitoxinas/metabolismo , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Macrófagos/microbiologia , Mycobacterium tuberculosis/enzimologia , Fosforilases/metabolismo , Sistemas Toxina-Antitoxina , Tuberculose/microbiologia , Animais , Antibióticos Antituberculose/farmacologia , Antitoxinas/química , Antitoxinas/genética , Carga Bacteriana , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Células Cultivadas , Modelos Animais de Doenças , Feminino , Interações Hospedeiro-Patógeno , Humanos , Cinética , Macrófagos/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos SCID , Camundongos Transgênicos , Viabilidade Microbiana , Modelos Moleculares , Mycobacterium smegmatis/enzimologia , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/patogenicidade , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidade , NAD/metabolismo , Fosforilases/química , Fosforilases/genética , Conformação Proteica , Sistemas Toxina-Antitoxina/genética , Tuberculose/tratamento farmacológico
4.
Front Immunol ; 9: 2656, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30487801

RESUMO

Tuberculosis (TB), caused by the airborne bacterial pathogen Mycobacterium tuberculosis, remains a major source of morbidity and mortality worldwide. So far, the study of host-pathogen interactions in TB has mostly focused on the physiology and virulence of the pathogen, as well as, on the various innate and adaptive immune compartments of the host. Microbial organisms endogenous to our body, the so-called microbiota, interact not only with invading pathogens, but also with our immune system. Yet, the impact of the microbiota on host defense against M. tuberculosis remains poorly understood. In order to address this question, we adapted a robust and reproducible mouse model of microbial dysbiosis based on a combination of wide-spectrum antibiotics. We found that microbiota dysbiosis resulted in an increased early colonization of the lungs by M. tuberculosis during the first week of infection, correlating with an altered diversity of the gut microbiota during this time period. At the cellular level, no significant difference in the recruitment of conventional myeloid cells, including macrophages, dendritic cells and neutrophils, to the lungs could be detected during the first week of infection between microbiota-competent and -deficient mice. At the molecular level, microbiota depletion did not impact the global production of pro-inflammatory cytokines, such as interferon (IFN)γ, tumor necrosis factor (TNF)α and interleukin (IL)-1ß in the lungs. Strikingly, a reduced number of mucosal-associated invariant T (MAIT) cells, a population of innate-like lymphocytes whose development is known to depend on the host microbiota, was observed in the lungs of the antibiotics-treated animals after 1week of infection. These cells produced less IL-17A in antibiotics-treated mice. Notably, dysbiosis correction through the inoculation of a complex microbiota in antibiotics-treated animals reversed these phenotypes and improved the ability of MAIT cells to proliferate. Altogether, our results demonstrate that the host microbiota contributes to early protection of lung colonization by M. tuberculosis, possibly through sustaining the function(s) of MAIT cells. Our study calls for a better understanding of the impact of the microbiota on host-pathogen interactions in TB. Ultimately, this study may help to develop novel therapeutic approaches based on the use of beneficial microbes, or components thereof, to boost anti-mycobacterial immunity.


Assuntos
Pulmão , Microbiota/imunologia , Células T Invariantes Associadas à Mucosa/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose Pulmonar , Animais , Citocinas/imunologia , Disbiose/imunologia , Disbiose/microbiologia , Disbiose/patologia , Feminino , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Células T Invariantes Associadas à Mucosa/patologia , Mycobacterium tuberculosis/patogenicidade , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/patologia , Tuberculose Pulmonar/prevenção & controle
5.
Am J Respir Crit Care Med ; 197(6): 801-813, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29161093

RESUMO

RATIONALE: In addition to their well-known function as antibody-producing cells, B lymphocytes can markedly influence the course of infectious or noninfectious diseases via antibody-independent mechanisms. In tuberculosis (TB), B cells accumulate in lungs, yet their functional contribution to the host response remains poorly understood. OBJECTIVES: To document the role of B cells in TB in an unbiased manner. METHODS: We generated the transcriptome of B cells isolated from Mycobacterium tuberculosis (Mtb)-infected mice and validated the identified key pathways using in vitro and in vivo assays. The obtained data were substantiated using B cells from pleural effusion of patients with TB. MEASUREMENTS AND MAIN RESULTS: B cells isolated from Mtb-infected mice displayed a STAT1 (signal transducer and activator of transcription 1)-centered signature, suggesting a role for IFNs in B-cell response to infection. B cells stimulated in vitro with Mtb produced type I IFN, via a mechanism involving the innate sensor STING (stimulator of interferon genes), and antagonized by MyD88 (myeloid differentiation primary response 88) signaling. In vivo, B cells expressed type I IFN in the lungs of Mtb-infected mice and, of clinical relevance, in pleural fluid from patients with TB. Type I IFN expression by B cells induced an altered polarization of macrophages toward a regulatory/antiinflammatory profile in vitro. In vivo, increased provision of type I IFN by B cells in a murine model of B cell-restricted Myd88 deficiency correlated with an enhanced accumulation of regulatory/antiinflammatory macrophages in Mtb-infected lungs. CONCLUSIONS: Type I IFN produced by Mtb-stimulated B cells favors macrophage polarization toward a regulatory/antiinflammatory phenotype during Mtb infection.


Assuntos
Linfócitos B/metabolismo , Interferon Tipo I/metabolismo , Macrófagos/metabolismo , Tuberculose/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Pulmão/metabolismo , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis , Transdução de Sinais , Baço/metabolismo , Baço/microbiologia
6.
Proc Natl Acad Sci U S A ; 114(4): E540-E549, 2017 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-28069953

RESUMO

Immune response against pathogens is a tightly regulated process that must ensure microbial control while preserving integrity of the infected organs. Tuberculosis (TB) is a paramount example of a chronic infection in which antimicrobial immunity is protective in the vast majority of infected individuals but can become detrimental if not finely tuned. Here, we report that C-type lectin dendritic cell (DC) immunoreceptor (DCIR), a key component in DC homeostasis, is required to modulate lung inflammation and bacterial burden in TB. DCIR is abundantly expressed in pulmonary lesions in Mycobacterium tuberculosis-infected nonhuman primates during both latent and active disease. In mice, we found that DCIR deficiency impairs STAT1-mediated type I IFN signaling in DCs, leading to increased production of IL-12 and increased differentiation of T lymphocytes toward Th1 during infection. As a consequence, DCIR-deficient mice control M. tuberculosis better than WT animals but also develop more inflammation characterized by an increased production of TNF and inducible NOS (iNOS) in the lungs. Altogether, our results reveal a pathway by which a C-type lectin modulates the equilibrium between infection-driven inflammation and pathogen's control through sustaining type I IFN signaling in DCs.


Assuntos
Células Dendríticas/imunologia , Interferon Tipo I/imunologia , Lectinas Tipo C/imunologia , Tuberculose/imunologia , Animais , Feminino , Lectinas Tipo C/genética , Macaca mulatta , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Fator de Transcrição STAT1/imunologia , Transdução de Sinais
7.
Gut ; 66(5): 872-885, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-26838600

RESUMO

OBJECTIVE: To identify a causal mechanism responsible for the enhancement of insulin resistance and hyperglycaemia following periodontitis in mice fed a fat-enriched diet. DESIGN: We set-up a unique animal model of periodontitis in C57Bl/6 female mice by infecting the periodontal tissue with specific and alive pathogens like Porphyromonas gingivalis (Pg), Fusobacterium nucleatum and Prevotella intermedia. The mice were then fed with a diabetogenic/non-obesogenic fat-enriched diet for up to 3 months. Alveolar bone loss, periodontal microbiota dysbiosis and features of glucose metabolism were quantified. Eventually, adoptive transfer of cervical (regional) and systemic immune cells was performed to demonstrate the causal role of the cervical immune system. RESULTS: Periodontitis induced a periodontal microbiota dysbiosis without mainly affecting gut microbiota. The disease concomitantly impacted on the regional and systemic immune response impairing glucose metabolism. The transfer of cervical lymph-node cells from infected mice to naive recipients guarded against periodontitis-aggravated metabolic disease. A treatment with inactivated Pg prior to the periodontal infection induced specific antibodies against Pg and protected the mouse from periodontitis-induced dysmetabolism. Finally, a 1-month subcutaneous chronic infusion of low rates of lipopolysaccharides from Pg mimicked the impact of periodontitis on immune and metabolic parameters. CONCLUSIONS: We identified that insulin resistance in the high-fat fed mouse is enhanced by pathogen-induced periodontitis. This is caused by an adaptive immune response specifically directed against pathogens and associated with a periodontal dysbiosis.


Assuntos
Imunidade Adaptativa , Infecções por Bacteroidaceae/complicações , Disbiose/imunologia , Resistência à Insulina/imunologia , Periodontite/imunologia , Periodontite/prevenção & controle , Porphyromonas gingivalis , Animais , Transplante de Células , Dieta Hiperlipídica , Modelos Animais de Doenças , Disbiose/microbiologia , Disbiose/prevenção & controle , Feminino , Gengiva/microbiologia , Hiperglicemia/imunologia , Hiperglicemia/microbiologia , Interferon gama/sangue , Interleucina-6/sangue , Lipopolissacarídeos/imunologia , Linfonodos/citologia , Linfócitos , Camundongos , Camundongos Endogâmicos C57BL , Microbiota , Periodontite/microbiologia , Periodontite/patologia , Porphyromonas gingivalis/imunologia , Distribuição Aleatória , Baço/citologia , Vacinação
8.
Gut ; 66(2): 258-269, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-26565000

RESUMO

OBJECTIVE: The gut-brain axis is considered as a major regulatory checkpoint in the control of glucose homeostasis. The detection of nutrients and/or hormones in the duodenum informs the hypothalamus of the host's nutritional state. This process may occur via hypothalamic neurons modulating central release of nitric oxide (NO), which in turn controls glucose entry into tissues. The enteric nervous system (ENS) modulates intestinal contractions in response to various stimuli, but the importance of this interaction in the control of glucose homeostasis via the brain is unknown. We studied whether apelin, a bioactive peptide present in the gut, regulates ENS-evoked contractions, thereby identifying a new physiological partner in the control of glucose utilisation via the hypothalamus. DESIGN: We measured the effect of apelin on electrical and mechanical duodenal responses via telemetry probes and isotonic sensors in normal and obese/diabetic mice. Changes in hypothalamic NO release, in response to duodenal contraction modulated by apelin, were evaluated in real time with specific amperometric probes. Glucose utilisation in tissues was measured with orally administrated radiolabeled glucose. RESULTS: In normal and obese/diabetic mice, glucose utilisation is improved by the decrease of ENS/contraction activities in response to apelin, which generates an increase in hypothalamic NO release. As a consequence, glucose entry is significantly increased in the muscle. CONCLUSIONS: Here, we identify a novel mode of communication between the intestine and the hypothalamus that controls glucose utilisation. Moreover, our data identified oral apelin administration as a novel potential target to treat metabolic disorders.


Assuntos
Adipocinas/farmacologia , Sistema Nervoso Entérico/efeitos dos fármacos , Glucose/metabolismo , Hipotálamo/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Contração Muscular/efeitos dos fármacos , Animais , Apelina , Técnicas Biossensoriais , Diabetes Mellitus/fisiopatologia , Duodeno/efeitos dos fármacos , Duodeno/metabolismo , Sistema Nervoso Entérico/fisiologia , Motilidade Gastrointestinal/efeitos dos fármacos , Homeostase , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso/fisiologia , Óxido Nítrico/metabolismo , Obesidade/fisiopatologia , Telemetria
9.
Am J Physiol Gastrointest Liver Physiol ; 310(11): G1091-101, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27033119

RESUMO

Periodontitis and type 2 diabetes are connected pandemic diseases, and both are risk factors for cardiovascular complications. Nevertheless, the molecular factors relating these two chronic pathologies are poorly understood. We have shown that, in response to a long-term fat-enriched diet, mice present particular gut microbiota profiles related to three metabolic phenotypes: diabetic-resistant (DR), intermediate (Inter), and diabetic-sensitive (DS). Moreover, many studies suggest that a dysbiosis of periodontal microbiota could be associated with the incidence of metabolic and cardiac diseases. We investigated whether periodontitis together with the periodontal microbiota may also be associated with these different cardiometabolic phenotypes. We report that the severity of glucose intolerance is related to the severity of periodontitis and cardiac disorders. In detail, alveolar bone loss was more accentuated in DS than Inter, DR, and normal chow-fed mice. Molecular markers of periodontal inflammation, such as TNF-α and plasminogen activator inhibitor-1 mRNA levels, correlated positively with both alveolar bone loss and glycemic index. Furthermore, the periodontal microbiota of DR mice was dominated by the Streptococcaceae family of the phylum Firmicutes, whereas the periodontal microbiota of DS mice was characterized by increased Porphyromonadaceae and Prevotellaceae families. Moreover, in DS mice the periodontal microbiota was indicated by an abundance of the genera Prevotella and Tannerella, which are major periodontal pathogens. PICRUSt analysis of the periodontal microbiome highlighted that prenyltransferase pathways follow the cardiometabolic adaptation to a high-fat diet. Finally, DS mice displayed a worse cardiac phenotype, percentage of fractional shortening, heart rhythm, and left ventricle weight-to-tibia length ratio than Inter and DR mice. Together, our data show that periodontitis combined with particular periodontal microbiota and microbiome is associated with metabolic adaptation to a high-fat diet related to the severity of cardiometabolic alteration.


Assuntos
Adaptação Fisiológica , Doenças Cardiovasculares/metabolismo , Dieta Hiperlipídica , Intolerância à Glucose , Microbiota , Periodontite/microbiologia , Função Ventricular , Animais , Doenças Cardiovasculares/complicações , Doenças Cardiovasculares/microbiologia , Dimetilaliltranstransferase/metabolismo , Disbiose/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Periodontite/complicações , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Prevotella/isolamento & purificação , Streptococcaceae/isolamento & purificação , Fator de Necrose Tumoral alfa/metabolismo
10.
J Neurosci ; 32(35): 11970-9, 2012 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-22933782

RESUMO

Hormones such as leptin and ghrelin can rapidly rewire hypothalamic feeding circuits when injected into rodent brains. These experimental manipulations suggest that the hypothalamus might reorganize continually in adulthood to integrate the metabolic status of the whole body. In this study, we examined whether hypothalamic plasticity occurs in naive animals according to their nutritional conditions. For this purpose, we fed mice with a short-term high-fat diet (HFD) and assessed brain remodeling through its molecular and functional signature. We found that HFD for 3 d rewired the hypothalamic arcuate nucleus, increasing the anorexigenic tone due to activated pro-opiomelanocortin (POMC) neurons. We identified the polysialic acid molecule (PSA) as a mediator of the diet-induced rewiring of arcuate POMC. Moreover, local pharmacological inhibition and genetic disruption of the PSA signaling limits the behavioral and metabolic adaptation to HFD, as treated mice failed to normalize energy intake and showed increased body weight gain after the HFD challenge. Altogether, these findings reveal the existence of physiological hypothalamic rewiring involved in the homeostatic response to dietary fat. Furthermore, defects in the hypothalamic plasticity-driven adaptive response to HFD are obesogenic and could be involved in the development of metabolic diseases.


Assuntos
Adaptação Fisiológica/fisiologia , Núcleo Arqueado do Hipotálamo/fisiologia , Gorduras na Dieta/administração & dosagem , Pró-Opiomelanocortina/fisiologia , Ácidos Siálicos/fisiologia , Animais , Ingestão de Energia/genética , Metabolismo Energético/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Rede Nervosa/fisiologia , Plasticidade Neuronal/genética , Técnicas de Cultura de Órgãos , Pró-Opiomelanocortina/metabolismo , Sialiltransferases/deficiência , Sialiltransferases/genética , Transdução de Sinais/genética , Aumento de Peso/genética
11.
Antioxid Redox Signal ; 15(6): 1477-96, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21395477

RESUMO

AIMS: Apelin and its receptor have emerged as promising targets for the treatment of insulin resistance. Indeed, peripheral administration of apelin stimulates glucose utilization and insulin sensitivity via a nitric oxide (NO) pathway. In addition to being expressed on peripheral metabolically active adipose tissues, apelin is also found in the brain. However, no data are available on the role of central effects of apelin on metabolic control. We studied glucose metabolism in response to acute and chronic intracerebroventricular (i.c.v.) injection of apelin performed in normal and obese/diabetic mice. RESULTS: We demonstrate that i.c.v. injection of apelin into fed mice improves glucose control via NO-dependent mechanisms. These results have been strengthened by transgenic (eNOS-KO mice), pharmacological (L-NMMA i.c.v. treated mice), and real-time measurement of NO release with amperometric probes detection. High-fat diet-fed mice displayed a severely blunted response to i.c.v. apelin associated with a lack of NO response by the hypothalamus. Moreover, central administration of high dose apelin in fasted normal mice provoked hyperinsulinemia, hyperglycemia, glucose intolerance, and insulin resistance. CONCLUSION: These data provide compelling evidence that central apelin participates in the regulation of glucose homeostasis and suggest a novel pathophysiological mechanism involved in the transition from normal to diabetic state.


Assuntos
Glucose/metabolismo , Homeostase , Hipotálamo/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Óxido Nítrico/metabolismo , Adipocinas , Animais , Apelina , Vias Biossintéticas , Encéfalo/metabolismo , Ritmo Circadiano , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Infusões Intraventriculares , Insulina/metabolismo , Resistência à Insulina , Secreção de Insulina , Peptídeos e Proteínas de Sinalização Intercelular/administração & dosagem , Peptídeos e Proteínas de Sinalização Intercelular/sangue , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo
12.
J Lipid Res ; 52(6): 1247-1255, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21421848

RESUMO

Autotaxin (ATX) is a secreted lysophospholipase D that generates the lipid mediator lysophosphatidic acid (LPA). ATX is secreted by adipose tissue and its expression is enhanced in obese/insulin-resistant individuals. Here, we analyzed the specific contribution of adipose-ATX to fat expansion associated with nutritional obesity and its consequences on plasma LPA levels. We established ATX(F/F)/aP2-Cre (FATX-KO) transgenic mice carrying a null ATX allele specifically in adipose tissue. FATX-KO mice and their control littermates were fed either a normal or a high-fat diet (HFD) (45% fat) for 13 weeks. FATX-KO mice showed a strong decrease (up to 90%) in ATX expression in white and brown adipose tissue, but not in other ATX-expressing organs. This was associated with a 38% reduction in plasma LPA levels. When fed an HFD, FATX-KO mice showed a higher fat mass and a higher adipocyte size than control mice although food intake was unchanged. This was associated with increased expression of peroxisome proliferator-activated receptor (PPAR)γ2 and of PPAR-sensitive genes (aP2, adiponectin, leptin, glut-1) in subcutaneous white adipose tissue, as well as in an increased tolerance to glucose. These results show that adipose-ATX is a negative regulator of fat mass expansion in response to an HFD and contributes to plasma LPA levels.


Assuntos
Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Adiposidade , Modelos Animais de Doenças , Lisofosfolipídeos , Complexos Multienzimáticos , Obesidade/metabolismo , PPAR gama/metabolismo , Fosfodiesterase I , Pirofosfatases , Adipócitos/citologia , Adipócitos/metabolismo , Tecido Adiposo Marrom/fisiopatologia , Tecido Adiposo Branco/fisiopatologia , Animais , Glicemia/análise , Tamanho Celular , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/metabolismo , Feminino , Efeito Fundador , Deleção de Genes , Teste de Tolerância a Glucose , Insulina/sangue , Lisofosfolipídeos/sangue , Masculino , Camundongos , Camundongos Knockout , Complexos Multienzimáticos/deficiência , Complexos Multienzimáticos/genética , Obesidade/genética , Obesidade/fisiopatologia , PPAR gama/antagonistas & inibidores , PPAR gama/genética , Fosfodiesterase I/deficiência , Fosfodiesterase I/genética , Diester Fosfórico Hidrolases , Pirofosfatases/deficiência , Pirofosfatases/genética
13.
Diabetes ; 60(3): 735-45, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21266325

RESUMO

OBJECTIVE: AMP-activated protein kinase (AMPK) signaling acts as a sensor of nutrients and hormones in the hypothalamus, thereby regulating whole-body energy homeostasis. Deletion of Ampkα2 in pro-opiomelanocortin (POMC) neurons causes obesity and defective neuronal glucose sensing. LKB1, the Peutz-Jeghers syndrome gene product, and Ca(2+)-calmodulin-dependent protein kinase kinase ß (CaMKKß) are key upstream activators of AMPK. This study aimed to determine their role in POMC neurons upon energy and glucose homeostasis regulation. RESEARCH DESIGN AND METHODS: Mice lacking either Camkkß or Lkb1 in POMC neurons were generated, and physiological, electrophysiological, and molecular biology studies were performed. RESULTS: Deletion of Camkkß in POMC neurons does not alter energy homeostasis or glucose metabolism. In contrast, female mice lacking Lkb1 in POMC neurons (PomcLkb1KO) display glucose intolerance, insulin resistance, impaired suppression of hepatic glucose production, and altered expression of hepatic metabolic genes. The underlying cellular defect in PomcLkb1KO mice involves a reduction in melanocortin tone caused by decreased α-melanocyte-stimulating hormone secretion. However, Lkb1-deficient POMC neurons showed normal glucose sensing, and body weight was unchanged in PomcLkb1KO mice. CONCLUSIONS: Our findings demonstrate that LKB1 in hypothalamic POMC neurons plays a key role in the central regulation of peripheral glucose metabolism but not body-weight control. This phenotype contrasts with that seen in mice lacking AMPK in POMC neurons with defects in body-weight regulation but not glucose homeostasis, which suggests that LKB1 plays additional functions distinct from activating AMPK in POMC neurons.


Assuntos
Glucose/metabolismo , Homeostase/genética , Hipotálamo/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Ativadas por AMP , Análise de Variância , Animais , Área Sob a Curva , Peso Corporal/genética , Contagem de Células , Ingestão de Alimentos/genética , Eletrofisiologia , Metabolismo Energético/genética , Feminino , Glucose/genética , Técnica Clamp de Glucose , Imuno-Histoquímica , Resistência à Insulina/genética , Masculino , Camundongos , Camundongos Transgênicos , Pró-Opiomelanocortina/genética , Proteínas Serina-Treonina Quinases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Estatísticas não Paramétricas
14.
Antioxid Redox Signal ; 14(3): 415-23, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20879900

RESUMO

Intestinal detection of nutrients is a crucial step to inform the whole body of the nutritional status. In this paradigm, peripheral information generated by nutrients is transferred to the brain, which in turn controls physiological functions, including glucose metabolism. Here, we investigated the effect of enteric glucose sensors stimulation on hypothalamic nitric oxide (NO) release in lean or in obese/diabetic (db/db) mice. By using specific NO amperometric probes implanted directly in the hypothalamus of mice, we demonstrated that NO release is stimulated in response to enteric glucose sensors activation in lean but not in db/db mice. Alteration of gut to hypothalamic NO signaling in db/db mice is associated with a drastic increase in inflammatory, oxidative/nitric oxide (iNOS, IL-1ß), and endoplasmic reticulum stress (CHOP, ATF4) genes expression in the jejunum. Although we could not exclude the importance of the hypothalamic inflammatory state in obese and diabetic mice, our results provide compelling evidence that enteric glucose sensors could be considered as potential targets for metabolic diseases.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Glucose/metabolismo , Hipotálamo/metabolismo , Inflamação/metabolismo , Jejuno/patologia , Camundongos Obesos , Óxido Nítrico/metabolismo , Animais , Biomarcadores/metabolismo , Inflamação/patologia , Jejuno/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
15.
Diabetes ; 57(10): 2603-12, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18519802

RESUMO

OBJECTIVE: Ingested glucose is detected by specialized sensors in the enteric/hepatoportal vein, which send neural signals to the brain, which in turn regulates key peripheral tissues. Hence, impairment in the control of enteric-neural glucose sensing could contribute to disordered glucose homeostasis. The aim of this study was to determine the cells in the brain targeted by the activation of the enteric glucose-sensing system. RESEARCH DESIGN AND METHODS: We selectively activated the axis in mice using a low-rate intragastric glucose infusion in wild-type and glucagon-like peptide-1 (GLP-1) receptor knockout mice, neuropeptide Y-and proopiomelanocortin-green fluorescent protein-expressing mice, and high-fat diet diabetic mice. We quantified the whole-body glucose utilization rate and the pattern of c-Fos positive in the brain. RESULTS: Enteric glucose increased muscle glycogen synthesis by 30% and regulates c-Fos expression in the brainstem and the hypothalamus. Moreover, the synthesis of muscle glycogen was diminished after central infusion of the GLP-1 receptor (GLP-1Rc) antagonist Exendin 9-39 and abolished in GLP-1Rc knockout mice. Gut-glucose-sensitive c-Fos-positive cells of the arcuate nucleus colocalized with neuropeptide Y-positive neurons but not with proopiomelanocortin-positive neurons. Furthermore, high-fat feeding prevented the enteric activation of c-Fos expression. CONCLUSIONS: We conclude that the gut-glucose sensor modulates peripheral glucose metabolism through a nutrient-sensitive mechanism, which requires brain GLP-1Rc signaling and is impaired during diabetes.


Assuntos
Sistema Nervoso Central/efeitos dos fármacos , Glucose/farmacologia , Receptores de Glucagon/fisiologia , Animais , Glicemia/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/fisiologia , Peptídeo 1 Semelhante ao Glucagon/sangue , Receptor do Peptídeo Semelhante ao Glucagon 1 , Glucose/administração & dosagem , Glicogênio/metabolismo , Imuno-Histoquímica , Insulina/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de Glucagon/metabolismo
16.
Endocrinology ; 149(10): 4768-77, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18556349

RESUMO

Glucagon-like peptide-1 (GLP-1) is a peptide released by the intestine and the brain. We previously demonstrated that brain GLP-1 increases glucose-dependent hyperinsulinemia and insulin resistance. These two features are major characteristics of the onset of type 2 diabetes. Therefore, we investigated whether blocking brain GLP-1 signaling would prevent high-fat diet (HFD)-induced diabetes in the mouse. Our data show that a 1-month chronic blockage of brain GLP-1 signaling by exendin-9 (Ex9), totally prevented hyperinsulinemia and insulin resistance in HFD mice. Furthermore, food intake was dramatically increased, but body weight gain was unchanged, showing that brain GLP-1 controlled energy expenditure. Thermogenesis, glucose utilization, oxygen consumption, carbon dioxide production, muscle glycolytic respiratory index, UCP2 expression in muscle, and basal ambulatory activity were all increased by the exendin-9 treatment. Thus, we have demonstrated that in response to a HFD, brain GLP-1 signaling induces hyperinsulinemia and insulin resistance and decreases energy expenditure by reducing metabolic thermogenesis and ambulatory activity.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Gorduras na Dieta/farmacologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Resistência à Insulina/fisiologia , Transdução de Sinais/fisiologia , Animais , Glicemia/metabolismo , Regulação da Temperatura Corporal/efeitos dos fármacos , Regulação da Temperatura Corporal/fisiologia , Tronco Encefálico/fisiologia , Dióxido de Carbono/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Intolerância à Glucose/tratamento farmacológico , Intolerância à Glucose/metabolismo , Hiperinsulinismo/tratamento farmacológico , Hiperinsulinismo/metabolismo , Canais Iônicos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/metabolismo , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Músculo Esquelético/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo III , Consumo de Oxigênio/efeitos dos fármacos , Consumo de Oxigênio/fisiologia , Fragmentos de Peptídeos/farmacologia , Resistência Física/efeitos dos fármacos , Resistência Física/fisiologia , Proglucagon/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Desacopladora 2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...