Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Protein Cell ; 13(6): 394-421, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-33826123

RESUMO

Obesity and aging are two important epidemic factors for metabolic syndrome and many other health issues, which contribute to devastating diseases such as cardiovascular diseases, stroke and cancers. The brain plays a central role in controlling metabolic physiology in that it integrates information from other metabolic organs, sends regulatory projections and orchestrates the whole-body function. Emerging studies suggest that brain dysfunction in sensing various internal cues or processing external cues may have profound effects on metabolic and other physiological functions. This review highlights brain dysfunction linked to genetic mutations, sex, brain inflammation, microbiota, stress as causes for whole-body pathophysiology, arguing brain dysfunction as a root cause for the epidemic of aging and obesity-related disorders. We also speculate key issues that need to be addressed on how to reveal relevant brain dysfunction that underlines the development of these disorders and diseases in order to develop new treatment strategies against these health problems.


Assuntos
Envelhecimento , Hipotálamo , Encéfalo/metabolismo , Metabolismo Energético , Humanos , Hipotálamo/metabolismo , Obesidade/metabolismo
2.
Life Sci Alliance ; 5(3)2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34862272

RESUMO

Murine neural stem cells (NSCs) were recently shown to release piRNA-containing exosomes/microvesicles (Ex/Mv) for exerting antiviral immunity, but it remains unknown if these Ex/Mv could target SARS-CoV-2 and whether the PIWI-piRNA system is important for these antiviral actions. Here, using in vitro infection models, we show that hypothalamic NSCs (htNSCs) Ex/Mv provided an innate immunity protection against SARS-CoV-2. Importantly, enhanced antiviral actions were achieved by using induced Ex/Mv that were derived from induced htNSCs through twice being exposed to several RNA fragments of SARS-CoV-2 genome, a process that was designed not to involve protein translation of these RNA fragments. The increased antiviral effects of these induced Ex/Mv were associated with increased expression of piRNA species some of which could predictably target SARS-CoV-2 genome. Knockout of piRNA-interacting protein PIWIL2 in htNSCs led to reductions in both innate and induced antiviral effects of Ex/Mv in targeting SARS-CoV-2. Taken together, this study demonstrates a case suggesting Ex/Mv from certain cell types have innate and adaptive immunity against SARS-CoV-2, and the PIWI-piRNA system is important for these antiviral actions.


Assuntos
Proteínas Argonautas/metabolismo , COVID-19/imunologia , COVID-19/metabolismo , Micropartículas Derivadas de Células/metabolismo , Exossomos , RNA Interferente Pequeno/metabolismo , RNA/metabolismo , SARS-CoV-2 , Células A549 , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , Genoma Viral , Humanos , Hipotálamo/metabolismo , Sistema Imunitário , Imunidade Inata , Técnicas In Vitro , Camundongos
3.
Handb Clin Neurol ; 181: 311-322, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34238467

RESUMO

Over the past decade, hypothalamic microinflammation has been studied and appreciated as a core mechanism involved in the advancement of metabolic syndrome and aging. Accumulating evidence suggests that atypical microinflammatory insults disturb hypothalamic regulation resulting in metabolic imbalance and aging progression, establishing a common causality for these two pathophysiologic statuses. Studies have causally linked these changes to activation of key proinflammatory pathways, especially NF-κB signaling within the hypothalamus, which leads to hypothalamic neuronal dysregulation, astrogliosis, microgliosis, and loss of adult hypothalamic neural stem/progenitor cells. While hypothalamic microinflammation is a complex, multifaceted process, initial work has been done to reveal how it contributes to the pathogenesis of metabolic syndrome and aging, and studies inhibiting hypothalamic microinflammation through targeting proinflammatory signaling pathways have shown to be beneficial against these disorders and diseases. In this chapter, we provide a broad overview on hypothalamic microinflammation, focusing on its features, inducers, and shared pathogenic roles in metabolic syndrome and aging.


Assuntos
Hipotálamo , Síndrome Metabólica , Adulto , Envelhecimento , Humanos , Inflamação , Neurônios , Transdução de Sinais
4.
Life Sci Alliance ; 3(12)2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33087487

RESUMO

This study reports that parathymosin (PTMS) is secreted by hypothalamic stem/progenitor cells (htNSC) to inhibit senescence of recipient cells such as fibroblasts. Upon release, PTMS is rapidly transferred into the nuclei of various cell types, including neuronal GT1-7 cells and different peripheral cells, and it is effectively transferred into neuronal nuclei in various brain regions in vivo. Notably, brain neurons also produce and release PTMS, and because neuronal populations are large, they are important for maintaining PTMS in the cerebrospinal fluid which is further transferable into the blood. Compared with several other brain regions, the hypothalamus is stronger for long-distance PTMS transfer, supporting a key hypothalamic role in this function. In physiology, aging is associated with declines in PTMS production and transfer in the brain, and ptms knockdown in the hypothalamus versus hippocampus were studied showing different contributions to neurobehavioral physiology. In conclusion, the brain is an endocrine organ through secretion and nuclear transfer of PTMS, and the hypothalamus-brain orchestration of this function is protective in physiology and counteractive against aging-related disorders.


Assuntos
Secreções Corporais/metabolismo , Hipotálamo/metabolismo , Timosina/análogos & derivados , Animais , Encéfalo/metabolismo , Glândulas Endócrinas/metabolismo , Fibroblastos/metabolismo , Hipocampo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Células-Tronco/metabolismo , Timosina/metabolismo , Timosina/fisiologia
5.
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
6.
Mol Metab ; 32: 56-68, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32029230

RESUMO

OBJECTIVE: The glycoprotein hormone erythropoietin (EPO) is required for erythropoiesis, and the kidney is the primary site of adult EPO synthesis. Limited evidence has suggested that EPO could be detectable in the brain under certain conditions, but it remains unknown if the brain might have its own EPO system for biological functions that are independent of peripheral EPO production and action. We performed this study to address this question using mice under normal conditions versus pathophysiological conditions including aging and dietary obesity. METHODS: EPO expression was measured in different brain regions as well as in the cerebrospinal fluid. Hypothalamic ventricular EPO was administered to physiologically examine possible therapeutic effects on the conditions of aging and dietary obesity. Body weight, body composition, insulin tolerance, and glucose tolerance were measured to assess the central effects of EPO on metabolic physiology, and muscle strength and histology were analyzed to assess the central effects of EPO on muscle function. In addition, ß2-adrenergic receptor knockout bone marrow transplant was employed to determine the potential role of bone marrow in linking the brain to some of these peripheral functions. RESULTS: This study revealed that EPO is expressed in the ventromedial hypothalamus in addition to a few other brain regions and is present in the cerebrospinal fluid. Unlike blood EPO concentration, which increased with aging and dietary obesity, hypothalamic EPO decreased in these disease conditions. Therapeutically, aged mice were chronically treated with EPO in the hypothalamic ventricle, showing an increase in lean mass, while body weight and fat mass decreased as a result of a moderate reduction of food intake. Both muscle and metabolic functions were improved by this central treatment, and mechanistically, adrenergic signals to the bone marrow played a role in conveying hypothalamic EPO to these peripheral actions. Dietary obesity was also studied, showing that hypothalamic EPO treatment caused a reduction in food intake and obesity, leading to improved metabolic functions related to decreased fat as well as increased lean mass. CONCLUSIONS: Hypothalamic EPO plays a role in the central regulation of muscle and metabolic physiology, while its decline contributes to aging and obesity physiology in a manner that is independent of peripheral EPO.


Assuntos
Eritropoetina/metabolismo , Hipotálamo/metabolismo , Músculos/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Eritropoetina/administração & dosagem , Teste de Tolerância a Glucose , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/induzido quimicamente , Obesidade/metabolismo
7.
Elife ; 92020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-32081132

RESUMO

We report that mouse hypothalamic stem/progenitor cells produce multiple pancreatic, gastrointestinal and hypothalamic peptides in addition to exosomes. Through cell sorting and selection according to insulin promoter activity, we generated a subpopulation(s) of these cells which formed 3D spherical structure with combined features of hypothalamic neurospheres and pancreatic islets. Through testing streptozotocin-induced pancreatic islet disruption and fatal diabetes, we found that peripheral implantation of these spheres in mice led to remarkable improvements in general health and survival in addition to a moderate antidiabetic effect, and notably these pro-survival versus metabolic effects were dissociable to a significant extent. Mechanistically, secretion of exosomes by these spheres was essential for enhancing survival while production of insulin was important for the antidiabetic effect. In summary, hypothalamic neural stem/progenitor cells comprise subpopulations with multifaceted secretion, and their derived hypothalamic islets can be implanted peripherally to enhance general health and survival together with an antidiabetic benefit.


Assuntos
Transplante de Células , Diabetes Mellitus Experimental/terapia , Hipotálamo/metabolismo , Células-Tronco Neurais/metabolismo , Animais , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Análise de Sobrevida
8.
Cell Metab ; 31(3): 534-548.e5, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32004475

RESUMO

Age-dependent loss of hypothalamic neural stem cells (htNSCs) is important for the pathological consequences of aging; however, it is unclear what drives the senescence of htNSCs. Here, we report that a long non-coding RNA, Hnscr, is abundantly expressed in the htNSCs of young mice but decreases markedly in middle-aged mice. We show that depletion of Hnscr is sufficient to drive the senescence of htNSCs and aging-like phenotypes in mice. Mechanistically, Hnscr binds to Y-box protein 1 (YB-1) to prevent its degradation and thus the attenuation of transcription of the senescence marker gene p16INK4A. Through molecular docking, we discovered that a naturally occurring small compound, theaflavin 3-gallate, can mimic the activity of Hnscr. Treatment of middle-aged mice with theaflavin 3-gallate reduced the senescence of htNSCs while improving aging-associated pathology. These results point to a mediator of the aging process and one that can be pharmacologically targeted to improve aging-related outcomes.


Assuntos
Envelhecimento/fisiologia , Senescência Celular , Hipotálamo/citologia , Células-Tronco Neurais/citologia , Animais , Biflavonoides/química , Biflavonoides/farmacologia , Catequina/química , Catequina/farmacologia , Senescência Celular/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Ácido Gálico/análogos & derivados , Ácido Gálico/química , Ácido Gálico/farmacologia , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Fenótipo , Ligação Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Proteólise/efeitos dos fármacos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteína 1 de Ligação a Y-Box/metabolismo
9.
Cell Metab ; 30(1): 19-35, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31269425

RESUMO

Under conditions leading to aging and metabolic syndrome, the hypothalamus atypically undergoes proinflammatory signaling activation leading to a chronic and stable background inflammation, referred to as "hypothalamic microinflammation." Through the past decade of research, progress has been made to causally link this hypothalamic inflammation to the mechanism of aging as well as metabolic syndrome, promoting the "hypothalamic microinflammation" theory, which helps characterize the consensus of these epidemic health problems. In general, it is consistently appreciated that hypothalamic microinflammation emerges during the early stages of aging and metabolic syndrome and evolves to be multifaceted and advanced alongside disease progression, while inhibition of key inflammatory components in the hypothalamus has a broad range of effects in counteracting these disorders. Herein, focusing on aging and metabolic syndrome, this writing aims to provide an overview of and insights into the mediators, signaling components, cellular impacts, and physiological significance of this hypothalamic microinflammation.


Assuntos
Hipotálamo/metabolismo , Doenças Metabólicas/metabolismo , Envelhecimento/fisiologia , Animais , Metabolismo Energético/fisiologia , Humanos , Síndrome Metabólica/metabolismo
10.
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
11.
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
12.
Cell Metab ; 25(5): 1091-1102.e4, 2017 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-28467927

RESUMO

Central regulation of metabolic physiology is mediated critically through neuronal functions; however, whether astrocytes are also essential remains unclear. Here we show that the high-order processes of astrocytes in the mediobasal hypothalamus displayed shortening in fasting and elongation in fed status. Chronic overnutrition and astrocytic IKKß/NF-κB upregulation similarly impaired astrocytic plasticity, leading to sustained shortening of high-order processes. In physiology, astrocytic IKKß/NF-κB upregulation resulted in early-onset effects, including glucose intolerance and blood pressure rise, and late-onset effects, including body weight and fat gain. Appropriate inhibition in astrocytic IKKß/NF-κB protected against chronic overnutrition impairing astrocytic plasticity and these physiological functions. Mechanistically, astrocytic regulation of hypothalamic extracellular GABA level and therefore BDNF expression were found partly accountable. Hence, astrocytic process plasticity and IKKß/NF-κB play significant roles in central control of blood glucose, blood pressure, and body weight as well as the central induction of these physiological disorders leading to disease.


Assuntos
Astrócitos/metabolismo , Glicemia/metabolismo , Pressão Sanguínea , Hipotálamo/fisiologia , Quinase I-kappa B/metabolismo , NF-kappa B/metabolismo , Animais , Astrócitos/patologia , Peso Corporal , Células Cultivadas , Jejum , Intolerância à Glucose/metabolismo , Intolerância à Glucose/fisiopatologia , Células HEK293 , Humanos , Hipotálamo/fisiopatologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Obesidade/fisiopatologia
13.
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
14.
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
15.
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
16.
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
17.
Nat Commun ; 6: 6704, 2015 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-25848677

RESUMO

Metabolic homeostasis is regulated by the brain, but whether this regulation involves learning and memory of metabolic information remains unexplored. Here we use a calorie-based, taste-independent learning/memory paradigm to show that Drosophila form metabolic memories that help in balancing food choice with caloric intake; however, this metabolic learning or memory is lost under chronic high-calorie feeding. We show that loss of individual learning/memory-regulating genes causes a metabolic learning defect, leading to elevated trehalose and lipid levels. Importantly, this function of metabolic learning requires not only the mushroom body but also the hypothalamus-like pars intercerebralis, while NF-κB activation in the pars intercerebralis mimics chronic overnutrition in that it causes metabolic learning impairment and disorders. Finally, we evaluate this concept of metabolic learning/memory in mice, suggesting that the hypothalamus is involved in a form of nutritional learning and memory, which is critical for determining resistance or susceptibility to obesity. In conclusion, our data indicate that the brain, and potentially the hypothalamus, direct metabolic learning and the formation of memories, which contribute to the control of systemic metabolic homeostasis.


Assuntos
Encéfalo/metabolismo , Ingestão de Energia , Metabolismo dos Lipídeos , Memória/fisiologia , Corpos Pedunculados/metabolismo , NF-kappa B/metabolismo , Trealose/metabolismo , Animais , Comportamento Animal , Drosophila , Homeostase , Hipotálamo/metabolismo , Aprendizagem/fisiologia , Camundongos , Hipernutrição/metabolismo , Hipernutrição/psicologia
18.
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
19.
Trends Neurosci ; 38(1): 36-44, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25458920

RESUMO

Chronic microinflammation is a hallmark of many aging-related neurodegenerative diseases as well as metabolic syndrome-driven diseases. Recent research indicates that chronic caloric excess can lead to hypothalamic microinflammation, which in turn participates in the development and progression of metabolic syndrome disorders such as obesity, glucose intolerance, and hypertension. Additionally, it was recently shown that increasing age after young adulthood can cause hypothalamic microinflammation independently of nutritional status, mediating a central mechanism of systemic aging. Taken together, these findings suggest that the hypothalamus has a fundamental role, via hypothalamic microinflammation, in translating overnutrition and aging into complex outcomes. Here we summarize recent work and suggest a conceptual model in which hypothalamic microinflammation is a common mediator of metabolic syndrome and aging.


Assuntos
Envelhecimento/imunologia , Hipotálamo/imunologia , Síndrome Metabólica/imunologia , Animais , Humanos , Neuroimunomodulação
20.
Nat Med ; 20(9): 1001-8, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25086906

RESUMO

The brain, in particular the hypothalamus, plays a role in regulating glucose homeostasis; however, it remains unclear whether this organ is causally and etiologically involved in the development of diabetes. Here, we found that hypothalamic transforming growth factor-ß (TGF-ß) production is excessive under conditions of not only obesity but also aging, which are two general etiological factors of type 2 diabetes. Pharmacological and genetic approaches revealed that central TGF-ß excess caused hyperglycemia and glucose intolerance independent of a change in body weight. Further, using cell-specific genetic analyses in vivo, we found that astrocytes and proopiomelanocortin neurons are responsible for the production and prodiabetic effect of central TGF-ß, respectively. Mechanistically, TGF-ß excess induced a hypothalamic RNA stress response, resulting in accelerated mRNA decay of IκBα, an inhibitor of proinflammatory nuclear factor-κB. These results reveal an atypical, mRNA metabolism-driven hypothalamic nuclear factor-κB activation, a mechanism that links obesity as well as aging to hypothalamic inflammation and ultimately to type 2 diabetes.


Assuntos
Envelhecimento/fisiologia , Diabetes Mellitus/fisiopatologia , Obesidade/fisiopatologia , RNA/metabolismo , Estresse Fisiológico , Fator de Crescimento Transformador beta/metabolismo , Astrócitos/metabolismo , Diabetes Mellitus/etiologia , Diabetes Mellitus/metabolismo , Teste de Tolerância a Glucose , Humanos , Hipotálamo/fisiopatologia , NF-kappa B/metabolismo , Pró-Opiomelanocortina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA