Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
1.
Nat Immunol ; 16(5): 525-33, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25848866

RESUMO

Innate immunological signals induced by pathogen- and/or damage-associated molecular patterns are essential for adaptive immune responses, but it is unclear if the brain has a role in this process. Here we found that while the abundance of tumor-necrosis factor (TNF) quickly increased in the brain of mice following bacterial infection, intra-brain delivery of TNF mimicked bacterial infection to rapidly increase the number of peripheral lymphocytes, especially in the spleen and fat. Studies of various mouse models revealed that hypothalamic responses to TNF were accountable for this increase in peripheral lymphocytes in response to bacterial infection. Finally, we found that hypothalamic induction of lipolysis mediated the brain's action in promoting this increase in the peripheral adaptive immune response. Thus, the brain-fat axis is important for rapid linkage of innate immunity to adaptive immunity.


Assuntos
Tecido Adiposo/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Hipotálamo/imunologia , Listeriose/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/administração & dosagem , Imunidade Adaptativa , Animais , Linfócitos T CD4-Positivos/microbiologia , Linfócitos T CD8-Positivos/microbiologia , Contagem de Células , Células Cultivadas , Ácidos Graxos/sangue , Hipotálamo/microbiologia , Imunidade Inata , Lipólise/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/líquido cefalorraquidiano
2.
Nature ; 560(7719): E33, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29950723

RESUMO

The microarray data generated and analysed in this Article have been uploaded to the Gene Expression Omnibus (GEO) under accession number GSE113383 . Accordingly, the 'Data availability' section of the Methods of the original Article has been rephrased online.

3.
BMC Genomics ; 24(1): 525, 2023 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-37670254

RESUMO

BACKGROUND: The incidence of kidney disease caused by thyroid cancer is rising worldwide. Observational studies cannot recognize whether thyroid cancer is independently associated with kidney disease. We performed the Mendelian randomization (MR) approach to genetically investigate the causality of thyroid cancer on immunoglobulin A nephropathy (IgAN). METHODS AND RESULTS: We explored the causal effect of thyroid cancer on IgAN by MR analysis. Fifty-two genetic loci and single nucleotide polymorphisms were related to thyroid cancer. The primary approach in this MR analysis was the inverse variance weighted (IVW) method, and MR‒Egger was the secondary method. Weighted mode and penalized weighted median were used to analyze the sensitivity. In this study, the random-effect IVW models showed the causal impact of genetically predicted thyroid cancer across the IgAN risk (OR, 1.191; 95% CI, 1.131-1.253, P < 0.001). Similar results were also obtained in the weighted mode method (OR, 1.048; 95% CI, 0.980-1.120, P = 0.179) and penalized weighted median (OR, 1.185; 95% CI, 1.110-1.264, P < 0.001). However, the MR‒Egger method revealed that thyroid cancer decreased the risk of IgAN, but this difference was not significant (OR, 0.948; 95% CI, 0.855-1.051, P = 0.316). The leave-one-out sensitivity analysis did not reveal the driving influence of any individual SNP on the association between thyroid cancer and IgAN. CONCLUSION: The IVW model indicated a significant causality of thyroid cancer with IgAN. However, MR‒Egger had a point estimation in the opposite direction. According to the MR principle, the evidence of this study did not support a stable significant causal association between thyroid cancer and IgAN. The results still need to be confirmed by future studies.


Assuntos
Glomerulonefrite por IGA , Neoplasias da Glândula Tireoide , Humanos , Análise da Randomização Mendeliana , Loci Gênicos , Polimorfismo de Nucleotídeo Único
4.
Cell ; 135(1): 61-73, 2008 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-18854155

RESUMO

Overnutrition is associated with chronic inflammation in metabolic tissues. Whether metabolic inflammation compromises the neural regulatory systems and therefore promotes overnutrition-associated diseases remains unexplored. Here we show that a mediator of metabolic inflammation, IKKbeta/NF-kappaB, normally remains inactive although enriched in hypothalamic neurons. Overnutrition atypically activates hypothalamic IKKbeta/NF-kappaB at least in part through elevated endoplasmic reticulum stress in the hypothalamus. While forced activation of hypothalamic IKKbeta/NF-kappaB interrupts central insulin/leptin signaling and actions, site- or cell-specific suppression of IKKbeta either broadly across the brain or locally within the mediobasal hypothalamus, or specifically in hypothalamic AGRP neurons significantly protects against obesity and glucose intolerance. The molecular mechanisms involved include regulation by IKKbeta/NF-kappaB of SOCS3, a core inhibitor of insulin and leptin signaling. Our results show that the hypothalamic IKKbeta/NF-kappaB program is a general neural mechanism for energy imbalance underlying obesity and suggest that suppressing hypothalamic IKKbeta/NF-kappaB may represent a strategy to combat obesity and related diseases.


Assuntos
Metabolismo Energético , Hipotálamo/fisiopatologia , Proteínas I-kappa B/metabolismo , NF-kappa B/metabolismo , Hipernutrição/fisiopatologia , Transdução de Sinais , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Retículo Endoplasmático/metabolismo , Humanos , Hipotálamo/imunologia , Imunidade Inata , Resistência à Insulina , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Obesidade/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/metabolismo
5.
Nature ; 548(7665): 52-57, 2017 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-28746310

RESUMO

It has been proposed that the hypothalamus helps to control ageing, but the mechanisms responsible remain unclear. Here we develop several mouse models in which hypothalamic stem/progenitor cells that co-express Sox2 and Bmi1 are ablated, as we observed that ageing in mice started with a substantial loss of these hypothalamic cells. Each mouse model consistently displayed acceleration of ageing-like physiological changes or a shortened lifespan. Conversely, ageing retardation and lifespan extension were achieved in mid-aged mice that were locally implanted with healthy hypothalamic stem/progenitor cells that had been genetically engineered to survive in the ageing-related hypothalamic inflammatory microenvironment. Mechanistically, hypothalamic stem/progenitor cells contributed greatly to exosomal microRNAs (miRNAs) in the cerebrospinal fluid, and these exosomal miRNAs declined during ageing, whereas central treatment with healthy hypothalamic stem/progenitor cell-secreted exosomes led to the slowing of ageing. In conclusion, ageing speed is substantially controlled by hypothalamic stem cells, partially through the release of exosomal miRNAs.


Assuntos
Envelhecimento/genética , Envelhecimento/fisiologia , Exossomos/genética , Hipotálamo/citologia , Hipotálamo/fisiologia , Longevidade/fisiologia , MicroRNAs/genética , Células-Tronco Neurais/fisiologia , Envelhecimento/líquido cefalorraquidiano , Envelhecimento/patologia , Animais , Microambiente Celular , Exossomos/metabolismo , Hipotálamo/patologia , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Inflamação , Longevidade/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/líquido cefalorraquidiano , MicroRNAs/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/transplante , Complexo Repressor Polycomb 1/deficiência , Proteínas Proto-Oncogênicas/deficiência , Fatores de Transcrição SOXB1/deficiência , Fatores de Tempo
6.
Proc Natl Acad Sci U S A ; 117(33): 20149-20158, 2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32747560

RESUMO

The C2 domain containing protein extended synaptotagmin (E-Syt) plays important roles in both lipid homeostasis and the intracellular signaling; however, its role in physiology remains largely unknown. Here, we show that hypothalamic E-Syt3 plays a critical role in diet-induced obesity (DIO). E-Syt3 is characteristically expressed in the hypothalamic nuclei. Whole-body or proopiomelanocortin (POMC) neuron-specific ablation of E-Syt3 ameliorated DIO and related comorbidities, including glucose intolerance and dyslipidemia. Conversely, overexpression of E-Syt3 in the arcuate nucleus moderately promoted food intake and impaired energy expenditure, leading to increased weight gain. Mechanistically, E-Syt3 ablation led to increased processing of POMC to α-melanocyte-stimulating hormone (α-MSH), increased activities of protein kinase C and activator protein-1, and enhanced expression of prohormone convertases. These findings reveal a previously unappreciated role for hypothalamic E-Syt3 in DIO and related metabolic disorders.


Assuntos
Regulação da Expressão Gênica/fisiologia , Obesidade/induzido quimicamente , Obesidade/genética , Sinaptotagminas/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Predisposição Genética para Doença , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Pró-Proteína Convertase 1/genética , Pró-Proteína Convertase 1/metabolismo , Pró-Proteína Convertase 2/genética , Pró-Proteína Convertase 2/metabolismo , Sinaptotagminas/genética
7.
BMC Cardiovasc Disord ; 21(1): 530, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34749646

RESUMO

BACKGROUND: Estimated plasma volume status (ePVS) has been reported that associated with poor prognosis in heart failure patients. However, no researchinvestigated the association of ePVS and prognosis in patients with acute myocardial infarction (AMI). Therefore, we aimed to determine the association between ePVS and in-hospital mortality in AMI patients. METHODS AND RESULTS: We extracted AMI patients data from MIMIC-III database. A generalized additive model and logistic regression model were used to demonstrate the association between ePVS levels and in-hospital mortality in AMI patients. Kaplan-Meier survival analysis was used to pooled the in-hospital mortality between the various group. ROC curve analysis were used to assessed the discrimination of ePVS for predicting in-hospital mortality. 1534 eligible subjects (1004 males and 530 females) with an average age of 67.36 ± 0.36 years old were included in our study finally. 136 patients (73 males and 63 females) died in hospital, with the prevalence of in-hospital mortality was 8.9%. The result of the Kaplan-Meier analysis showed that the high-ePVS group (ePVS ≥ 5.28 mL/g) had significant lower survival possibility in-hospital admission compared with the low-ePVS group (ePVS < 5.28 mL/g). In the unadjusted model, high-level of ePVS was associated with higher OR (1.09; 95% CI 1.06-1.12; P < 0.001) compared with low-level of ePVS. After adjusted the vital signs data, laboratory data, and treatment, high-level of ePVS were also associated with increased OR of in-hospital mortality, 1.06 (95% CI 1.03-1.09; P < 0.001), 1.05 (95% CI 1.01-1.08; P = 0.009), 1.04 (95% CI 1.01-1.07; P = 0.023), respectively. The ROC curve indicated that ePVS has acceptable discrimination for predicting in-hospital mortality. The AUC value was found to be 0.667 (95% CI 0.653-0.681). CONCLUSION: Higher ePVS values, calculated simply from Duarte's formula (based on hemoglobin/hematocrit) was associated with poor prognosis in AMI patients. EPVS is a predictor for predicting in-hospital mortality of AMI, and could help refine risk stratification.


Assuntos
Mortalidade Hospitalar , Infarto do Miocárdio/fisiopatologia , Volume Plasmático , Idoso , Bases de Dados Factuais , Feminino , Hematócrito , Hemoglobinas/análise , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/mortalidade , Prognóstico , Curva ROC
8.
J Biol Chem ; 294(13): 4946-4955, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30709906

RESUMO

Hypoxia-inducible factor-2α (HIF2α) is a nuclear transcription factor that plays a critical role in cell survival including metabolic adaptation under hypoxia as well as normoxia, but whether HIF2α contributes to the control of whole-body metabolic balance is unclear. In this study, we found that the hypothalamic HIF2α protein level rapidly increases in young mice that are centrally stimulated with insulin. However, this insulin-induced HIF2α up-regulation is substantially attenuated in mice of advanced age. This attenuation is comparable with the effect of high-calorie feeding in young mice. Of note, unlike high-calorie feeding conditions, age-dependent HIF2α attenuation occurs without impaired activation of the hypothalamic IR/IRS-2/AKT/FOXO1 pathway in response to insulin. Molecular and physiological analyses revealed that hypothalamic HIF2α contributes to the action of central insulin in regulation of proopiomelanocortin (Pomc) gene expression and food intake. HIF2α knockout in POMC neurons led to age-dependent excess weight gain and fat increase, a phenotype that was associated with a mild degree of glucose intolerance and insulin resistance. In conclusion, hypothalamic HIF2α responds to insulin, and the up-regulation is involved in adaptive metabolic regulation as age increases, whereas impairment of HIF2α in the hypothalamus contributes to weight gain and glucose disorders in age-dependent manners.


Assuntos
Envelhecimento/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Intolerância à Glucose/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Transdução de Sinais , Envelhecimento/genética , Envelhecimento/patologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Regulação da Expressão Gênica , Intolerância à Glucose/genética , Intolerância à Glucose/patologia , Hipotálamo/patologia , Insulina/genética , Camundongos , Camundongos Transgênicos , Pró-Opiomelanocortina/biossíntese , Pró-Opiomelanocortina/genética
9.
Pacing Clin Electrophysiol ; 43(9): 908-912, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32459008

RESUMO

BACKGROUND: Contraction of the esophagus was observed during cryoablation for paroxysmal atrial fibrillation (PAF). The purpose of this study is to investigate the mechanism of esophageal contraction and the correlation between the contraction and esophageal thermal lesions. METHODS: This prospective study enrolled 64 patients with PAF undergoing second-generation cryoballoon (CB2) ablation for pulmonary vein isolation (PVI). During PVI for the left inferior pulmonary vein, contrast esophagography was performed before and during cryoablation. The sample population was divided into two groups: A (31 patients) and B (33 patients). Group A consisted of patients in whom the distal half of the CB was in proximity to the esophagus, while for group B the esophagus was away from the distal half of the CB. Esophageal contraction was recorded as a variation in the width of the esophageal lumen during PVI. Postablation esophageal endoscopy was done on all patients. RESULTS: The reduction in the width of the esophageal lumen in group A was greater than in group B during freezing (40.12 ± 23.24% vs 8.14 ± 10.35%, P < .001). Following endoscopy, no apparent esophageal lesion was detected in all patients. CONCLUSION: The extent of esophageal contraction is correlated with the positioning of the esophagus at the distal half of the CB. The findings of this study indicate that esophageal contraction during freezing may be a self-protective mechanism.


Assuntos
Fibrilação Atrial/cirurgia , Criocirurgia/métodos , Esôfago/lesões , Veias Pulmonares/cirurgia , Esofagoscopia , Esôfago/diagnóstico por imagem , Feminino , Fluoroscopia , Humanos , Complicações Intraoperatórias/diagnóstico por imagem , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos
10.
Sensors (Basel) ; 20(22)2020 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-33207544

RESUMO

Motion capture data are widely used in different research fields such as medical, entertainment, and industry. However, most motion researches using motion capture data are carried out in the time-domain. To understand human motion complexities, it is necessary to analyze motion data in the frequency-domain. In this paper, to analyze human motions, we present a framework to transform motions into the instantaneous frequency-domain using the Hilbert-Huang transform (HHT). The empirical mode decomposition (EMD) that is a part of HHT decomposes nonstationary and nonlinear signals captured from the real-world experiments into pseudo monochromatic signals, so-called intrinsic mode function (IMF). Our research reveals that the multivariate EMD can decompose complicated human motions into a finite number of nonlinear modes (IMFs) corresponding to distinct motion primitives. Analyzing these decomposed motions in Hilbert spectrum, motion characteristics can be extracted and visualized in instantaneous frequency-domain. For example, we apply our framework to (1) a jump motion, (2) a foot-injured gait, and (3) a golf swing motion.

11.
Nature ; 497(7448): 211-6, 2013 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-23636330

RESUMO

Ageing is a result of gradual and overall functional deteriorations across the body; however, it is unknown whether an individual tissue primarily works to mediate the ageing progress and control lifespan. Here we show that the hypothalamus is important for the development of whole-body ageing in mice, and that the underlying basis involves hypothalamic immunity mediated by IκB kinase-ß (IKK-ß), nuclear factor κB (NF-κB) and related microglia-neuron immune crosstalk. Several interventional models were developed showing that ageing retardation and lifespan extension are achieved in mice by preventing ageing-related hypothalamic or brain IKK-ß and NF-κB activation. Mechanistic studies further revealed that IKK-ß and NF-κB inhibit gonadotropin-releasing hormone (GnRH) to mediate ageing-related hypothalamic GnRH decline, and GnRH treatment amends ageing-impaired neurogenesis and decelerates ageing. In conclusion, the hypothalamus has a programmatic role in ageing development via immune-neuroendocrine integration, and immune inhibition or GnRH restoration in the hypothalamus/brain represent two potential strategies for optimizing lifespan and combating ageing-related health problems.


Assuntos
Envelhecimento/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/fisiologia , Quinase I-kappa B/metabolismo , NF-kappa B/metabolismo , Envelhecimento/efeitos dos fármacos , Envelhecimento/genética , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Cognição/efeitos dos fármacos , Feminino , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Hormônio Liberador de Gonadotropina/farmacologia , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/enzimologia , Quinase I-kappa B/deficiência , Quinase I-kappa B/genética , Longevidade/efeitos dos fármacos , Longevidade/genética , Longevidade/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/enzimologia , Microglia/fisiologia , Neurogênese , Reprodução/fisiologia
12.
J Biol Chem ; 291(29): 15131-42, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27226618

RESUMO

Leptin and TNFα can individually work in the brain to affect blood pressure; however, it remains unknown whether these two cytokines might have an interactive role in this process and, if so, how. In this work, we found that leptin stimulation led to TNFα production under both in vitro and in vivo conditions, and diurnal fluctuation of leptin concentrations in the cerebrospinal fluid predicted the circadian changes of TNFα gene expression in the hypothalamus. Signaling analysis showed that leptin stimulation led to a rapid and strong STAT3 activation followed by a second-phase moderate STAT3 activation, which was selectively abolished by anti-inflammatory chemical PS1145 or TNFα antagonist WP9QY. Physiological study in normal mice revealed that diurnal rise of blood pressure was abrogated following central administration of PS1145 or a leptin receptor antagonist. Central TNFα pretreatment was found to potentiate the effect of leptin in elevating blood pressure in normal mice. In pathophysiology, dietary obesity mimicked TNFα pretreatment in promoting leptin-induced blood pressure rise, and this effect was blocked by central treatment with either PS1145 or WP9QY. Hence, central leptin employs TNFα to mediate the diurnal blood pressure elevation in physiology while enhancement of this mechanism can contribute to hypertension development.


Assuntos
Pressão Sanguínea/fisiologia , Ritmo Circadiano/fisiologia , Hipertensão/fisiopatologia , Leptina/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Células HEK293 , Humanos , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiologia , Técnicas In Vitro , Leptina/líquido cefalorraquidiano , Leptina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/fisiopatologia , Fosforilação/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/genética
13.
Am J Physiol Endocrinol Metab ; 312(3): E161-E174, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27894066

RESUMO

Hypothalamic inflammation was recently found to mediate obesity-related hypertension, but the responsible upstream mediators remain unexplored. In this study, we show that dietary obesity is associated with extracellular release of mitochondrial DNA (mtDNA) into the cerebrospinal fluid and that central delivery of mtDNA mimics transforming growth factor-ß (TGFß) excess to activate downstream signaling pathways. Physiological study reveals that central administration of mtDNA or TGFß is sufficient to cause hypertension in mice. Knockout of the TGFß receptor in proopiomelanocortin neurons counteracts the hypertensive effect of not only TGFß but also mtDNA excess, while the hypertensive action of central mtDNA can be blocked pharmacologically by a TGFß receptor antagonist or genetically by TGFß receptor knockout. Finally, we confirm that obesity-induced hypertension can be reversed through central treatment with TGFß receptor antagonist. In conclusion, circulating mtDNA in the brain employs neural TGFß pathway to mediate a central inflammatory mechanism of obesity-related hypertension.


Assuntos
Pressão Sanguínea/imunologia , DNA Mitocondrial/imunologia , Hipertensão/imunologia , Hipotálamo/imunologia , Obesidade/imunologia , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Fator de Crescimento Transformador beta/imunologia , Animais , Benzamidas/farmacologia , Western Blotting , DNA Mitocondrial/líquido cefalorraquidiano , DNA Mitocondrial/metabolismo , DNA Mitocondrial/farmacologia , Dieta Hiperlipídica , Dioxóis/farmacologia , Hipertensão/etiologia , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neurônios/imunologia , Neurônios/metabolismo , Obesidade/complicações , Pró-Opiomelanocortina/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/imunologia , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/imunologia , Terceiro Ventrículo , Fator de Crescimento Transformador beta1/farmacologia
14.
EMBO J ; 32(11): 1529-42, 2013 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-23584532

RESUMO

Lifespan of C. elegans is affected by the nervous system; however, the underlying neural integration still remains unclear. In this work, we targeted an antagonistic neural system consisting of low-oxygen sensing BAG neurons and high-oxygen sensing URX neurons. While ablation of BAG neurons increases lifespan of C. elegans, ablation of URX neurons decreases lifespan. Genetic analysis revealed that BAG and URX neurons counterbalance each other via different guanylate cyclases (GCYs) to control lifespan balance. Lifespan-modulating effects of GCYs in these neurons are independent of the actions from insulin/IGF-1 signalling, germline signalling, sensory perception, or dietary restriction. Given the known gas-sensing property of these neurons, we profiled that lifespan of C. elegans is promoted under moderately low oxygen (4-12%) or moderately high carbon dioxide (5%) but inhibited under high-level oxygen (40%); however, these pro-longevity and anti-longevity effects are counteracted, respectively, by BAG and URX neurons via different GCYs. In conclusion, BAG and URX neurons work as a neural-regulatory system to counterbalance each other via different GCYs to control lifespan homeostasis.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Guanilato Ciclase/genética , Longevidade/genética , Neurônios/fisiologia , Animais , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/metabolismo , Dióxido de Carbono/fisiologia , Sobrevivência Celular , Regulação Enzimológica da Expressão Gênica , Guanilato Ciclase/metabolismo , Homeostase , Humanos , Modelos Biológicos , Mutação , Neurônios/enzimologia , Especificidade de Órgãos , Organismos Geneticamente Modificados , Oxigênio/fisiologia , Transdução de Sinais/fisiologia
15.
Clin Sci (Lond) ; 131(3): 211-223, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28057892

RESUMO

Hypertension is a major health problem with great consequences for public health. Despite its role as the primary cause of significant morbidity and mortality associated with cardiovascular disease, the pathogenesis of essential hypertension remains largely unknown. The central nervous system (CNS) in general, and the hypothalamus in particular, are intricately involved in the development and maintenance of hypertension. Over the last several decades, the understanding of the brain's role in the development of hypertension has dramatically increased. This brief review is to summarize the neural mechanisms of hypertension with a focus on neuroendocrine and neurotransmitter involvement, highlighting recent findings that suggest that hypothalamic inflammation disrupts key signalling pathways to affect the central control of blood pressure, and therefore suggesting future development of interventional strategies that exploit recent findings pertaining to the hypothalamic control of blood pressure as well as the inflammatory-sympathetic mechanisms involved in hypertension.


Assuntos
Pressão Sanguínea , Hipertensão/etiologia , Doenças Hipotalâmicas/complicações , Hipotálamo/fisiologia , Inflamação/complicações , Animais , Humanos , Hipertensão/terapia , Terapia de Alvo Molecular
16.
FASEB J ; 30(1): 214-29, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26373801

RESUMO

The purpose of this study was to determine the role of canonical transient receptor potential 3 (TRPC3) channel in allergen-induced airway disease (AIAD) and its underlying signaling mechanisms. The procedures included (1) intravenous injection of lentiviral TRPC3 channel or nonsilencing short hairpin ribonucleic acid (shRNA) to make the channel knockdown (KD) or control mice, (2) allergen sensitization/challenge to induce AIAD, (3) patch-clamp recording and Ca(2+) imaging to examine the channel activity, and (4) gene manipulations and other methods to determine the underlying signaling mechanisms. The findings are that (1) intravenous or intranasal delivery of TRPC3 channel lentiviral shRNAs or blocker 1-[4-[(2,3,3-trichloro-1-oxo-2-propen-1-yl)amino]phenyl]-5-(trifluoromethyl)-1H-pyrazole-4-carboxylic acid prevents AIAD in mice, (2) TRPC3 channel KD and overexpression, respectively, blocks and augments protein kinase C-α/nuclear factor of κ light polypeptide gene enhancer in B-cell inhibitor-α (PKC-α/IκB-α)-mediated or calcineurin/IκB-ß-dependent, NF-κB-dependent allergen-induced airway smooth muscle cell (ASMC) hyperproliferation and cyclin D1 (an important cell proliferation molecule) induction, and (3) the changes of the major molecules of the PKC-α/IκBα- and calcineurin/IκB-ß-dependent NF-κB signaling pathways are also observed in asthmatic human ASMCs. The conclusions are that TRPC3 channels plays an essential role in AIAD via the PKC-α/IκB-α- and calcineurin/IκB-ß-dependent NF-κB signaling pathways, and lentiviral shRNA or inhibitor of TRPC3 channels may become novel and effective treatments for AIAD.


Assuntos
NF-kappa B/metabolismo , Hipersensibilidade Respiratória/metabolismo , Canais de Cátion TRPC/genética , Potenciais de Ação , Animais , Calcineurina/metabolismo , Sinalização do Cálcio , Proliferação de Células , Células Cultivadas , Terapia Genética , Quinase I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Camundongos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/genética , Proteína Quinase C/metabolismo , Pirazóis/farmacologia , Pirazóis/uso terapêutico , Hipersensibilidade Respiratória/tratamento farmacológico , Hipersensibilidade Respiratória/terapia , Sistemas do Segundo Mensageiro , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/metabolismo
17.
Am J Physiol Endocrinol Metab ; 311(1): E32-41, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27166279

RESUMO

Interdisciplinary studies in the research fields of endocrinology and immunology show that obesity-associated overnutrition leads to neuroinflammatory molecular changes, in particular in the hypothalamus, chronically causing various disorders known as elements of metabolic syndrome. In this process, neural or hypothalamic inflammation impairs the neuroendocrine and autonomic regulation of the brain over blood pressure and glucose homeostasis as well as insulin secretion, and elevated sympathetic activation has been appreciated as a critical mediator. This review describes the involved physiology and mechanisms, with a focus on glucose and blood pressure balance, and suggests that neuroinflammation employs the autonomic nervous system to mediate the development of diabetes and hypertension.


Assuntos
Diabetes Mellitus/metabolismo , Hipertensão/metabolismo , Hipotálamo/metabolismo , Síndrome Metabólica/metabolismo , Obesidade/metabolismo , Hipernutrição/metabolismo , Sistema Nervoso Simpático/metabolismo , Animais , Sistema Nervoso Autônomo/imunologia , Sistema Nervoso Autônomo/metabolismo , Sistema Nervoso Autônomo/fisiopatologia , Glicemia/metabolismo , Pressão Sanguínea/fisiologia , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Diabetes Mellitus/imunologia , Glucose/metabolismo , Homeostase , Humanos , Hipertensão/imunologia , Hipertensão/fisiopatologia , Hipotálamo/imunologia , Hipotálamo/fisiopatologia , Inflamação , Insulina/metabolismo , Secreção de Insulina , Síndrome Metabólica/imunologia , Obesidade/imunologia , Hipernutrição/imunologia , Sistema Nervoso Simpático/imunologia , Sistema Nervoso Simpático/fisiopatologia
19.
PLoS Biol ; 9(7): e1001112, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21814490

RESUMO

Hypoxia-inducible factor (HIF) is a nuclear transcription factor that responds to environmental and pathological hypoxia to induce metabolic adaptation, vascular growth, and cell survival. Here we found that HIF subunits and HIF2α in particular were normally expressed in the mediobasal hypothalamus of mice. Hypothalamic HIF was up-regulated by glucose to mediate the feeding control of hypothalamic glucose sensing. Two underlying molecular pathways were identified, including suppression of PHDs by glucose metabolites to prevent HIF2α degradation and the recruitment of AMPK and mTOR/S6K to regulate HIF2α protein synthesis. HIF activation was found to directly control the transcription of POMC gene. Genetic approach was then employed to develop conditional knockout mice with HIF inhibition in POMC neurons, revealing that HIF loss-of-function in POMC neurons impaired hypothalamic glucose sensing and caused energy imbalance to promote obesity development. The metabolic effects of HIF in hypothalamic POMC neurons were independent of leptin signaling or pituitary ACTH pathway. Hypothalamic gene delivery of HIF counteracted overeating and obesity under conditions of nutritional excess. In conclusion, HIF controls hypothalamic POMC gene to direct the central nutrient sensing in regulation of energy and body weight balance.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Metabolismo Energético , Glucose/farmacologia , Hipotálamo/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pró-Opiomelanocortina/genética , Animais , Dieta Hiperlipídica , Ingestão de Alimentos , Feminino , Fumaratos/farmacologia , Regulação da Expressão Gênica , Genes Reporter , Glucose/fisiologia , Hipotálamo/citologia , Leptina/farmacologia , Leptina/fisiologia , Luciferases/biossíntese , Luciferases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Obesidade/metabolismo , Obesidade/terapia , Hipófise/metabolismo , Pró-Opiomelanocortina/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Regiões Promotoras Genéticas , Succinatos/farmacologia
20.
Proc Natl Acad Sci U S A ; 108(7): 2939-44, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21282643

RESUMO

Chronic endoplasmic reticulum (ER) stress was recently revealed to affect hypothalamic neuroendocrine pathways that regulate feeding and body weight. However, it remains unexplored whether brain ER stress could use a neural route to rapidly cause the peripheral disorders that underlie the development of type 2 diabetes (T2D) and the metabolic syndrome. Using a pharmacologic model that delivered ER stress inducer thapsigargin into the brain, this study demonstrated that a short-term brain ER stress over 3 d was sufficient to induce glucose intolerance, systemic and hepatic insulin resistance, and blood pressure (BP) increase. The collection of these changes was accompanied by elevated sympathetic tone and prevented by sympathetic suppression. Molecular studies revealed that acute induction of metabolic disorders via brain ER stress was abrogated by NF-κB inhibition in the hypothalamus. Therapeutic experiments further revealed that acute inhibition of brain ER stress with tauroursodeoxycholic acid (TUDCA) partially reversed obesity-associated metabolic and blood pressure disorders. In conclusion, ER stress in the brain represents a mediator of the sympathetic disorders that underlie the development of insulin resistance syndrome and T2D.


Assuntos
Pressão Sanguínea/fisiologia , Diabetes Mellitus Tipo 2/etiologia , Retículo Endoplasmático/patologia , Hipotálamo/fisiopatologia , Insulina/metabolismo , Sistemas Neurossecretores/fisiologia , Estresse Fisiológico/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Western Blotting , Peso Corporal , Ingestão de Alimentos , Retículo Endoplasmático/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Intolerância à Glucose/induzido quimicamente , Proteínas de Fluorescência Verde , Hipotálamo/efeitos dos fármacos , Imunoprecipitação , Resistência à Insulina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/antagonistas & inibidores , Sistemas Neurossecretores/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estresse Fisiológico/efeitos dos fármacos , Ácido Tauroquenodesoxicólico/farmacologia , Telemetria , Tapsigargina/toxicidade
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa