Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 161(7): 1576-1591, 2015 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-26091038

RESUMO

The synthesis of type I collagen, the main component of bone matrix, precedes the expression of Runx2, the earliest determinant of osteoblast differentiation. We hypothesized that the energetic needs of osteoblasts might explain this apparent paradox. We show here that glucose, the main nutrient of osteoblasts, is transported in these cells through Glut1, whose expression precedes that of Runx2. Glucose uptake favors osteoblast differentiation by suppressing the AMPK-dependent proteasomal degradation of Runx2 and promotes bone formation by inhibiting another function of AMPK. While RUNX2 cannot induce osteoblast differentiation when glucose uptake is compromised, raising blood glucose levels restores collagen synthesis in Runx2-null osteoblasts and initiates bone formation in Runx2-deficient embryos. Moreover, RUNX2 favors Glut1 expression, and this feedforward regulation between RUNX2 and Glut1 determines the onset of osteoblast differentiation during development and the extent of bone formation throughout life. These results reveal an unexpected intricacy between bone and glucose metabolism.


Assuntos
Diferenciação Celular , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Glucose/metabolismo , Osteoblastos/metabolismo , Osteogênese , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/genética , Sequência de Aminoácidos , Animais , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Homeostase , Camundongos , Osteoblastos/citologia , Alinhamento de Sequência , Crânio/citologia
2.
Nature ; 543(7645): 385-390, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28273060

RESUMO

Bone has recently emerged as a pleiotropic endocrine organ that secretes at least two hormones, FGF23 and osteocalcin, which regulate kidney function and glucose homeostasis, respectively. These findings have raised the question of whether other bone-derived hormones exist and what their potential functions are. Here we identify, through molecular and genetic analyses in mice, lipocalin 2 (LCN2) as an osteoblast-enriched, secreted protein. Loss- and gain-of-function experiments in mice demonstrate that osteoblast-derived LCN2 maintains glucose homeostasis by inducing insulin secretion and improves glucose tolerance and insulin sensitivity. In addition, osteoblast-derived LCN2 inhibits food intake. LCN2 crosses the blood-brain barrier, binds to the melanocortin 4 receptor (MC4R) in the paraventricular and ventromedial neurons of the hypothalamus and activates an MC4R-dependent anorexigenic (appetite-suppressing) pathway. These results identify LCN2 as a bone-derived hormone with metabolic regulatory effects, which suppresses appetite in a MC4R-dependent manner, and show that the control of appetite is an endocrine function of bone.


Assuntos
Regulação do Apetite/fisiologia , Osso e Ossos/metabolismo , Lipocalina-2/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Osso e Ossos/citologia , AMP Cíclico/metabolismo , Ingestão de Alimentos/fisiologia , Feminino , Fator de Crescimento de Fibroblastos 23 , Glucose/metabolismo , Homeostase , Hipotálamo/citologia , Hipotálamo/metabolismo , Insulina/metabolismo , Resistência à Insulina , Secreção de Insulina , Masculino , Camundongos , Neurônios/metabolismo , Obesidade/metabolismo , Osteoblastos/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Magreza/metabolismo
4.
Am J Physiol Endocrinol Metab ; 317(2): E185-E193, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30964706

RESUMO

Weight regain after weight loss is a well-described phenomenon in both humans and animal models of obesity. Reduced energy expenditure and increased caloric intake are considered the main drivers of weight regain. We hypothesized that adipose tissue with obesity memory (OM) has a tissue-autonomous lipolytic defect, allowing for increased efficiency of lipid storage. We utilized a mouse model of diet-induced obesity, which was subjected to 60% caloric restriction to achieve lean body weight, followed by a short period of high-fat diet (HFD) rechallenge. Age-matched lean mice fed HFD for the first time were used as the control group. Upon rechallenge with HFD, mice with OM had higher respiratory exchange ratios than lean mice with no OM despite comparable body weight, suggesting higher utilization of glucose over fatty acid oxidation. White adipose tissue explants with OM had comparable lipolytic response after caloric restriction; however, reduced functional lipolytic response to norepinephrine was noted as early as 5 days after rechallenge with HFD and was accompanied by reduction in hormone-sensitive lipase serine phosphorylation. The relative lipolytic defect was associated with increased expression of inflammatory genes and a decrease in adrenergic receptor genes, most notably Adrb3. Taken together, white adipose tissue of lean mice with OM shows increased sensitization to HFD compared with white adipose tissue with no OM, rendering it resistant to catecholamine-induced lipolysis. This relative lipolytic defect is tissue-autonomous and could play a role in the rapid weight regain observed after weight loss.


Assuntos
Tecido Adiposo Branco/metabolismo , Lipólise/fisiologia , Aumento de Peso/fisiologia , Animais , Dieta Hiperlipídica , Gorduras na Dieta/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Lipólise/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/genética , Obesidade/metabolismo , Receptores Adrenérgicos beta 3/genética , Receptores Adrenérgicos beta 3/metabolismo , Esterol Esterase/genética , Esterol Esterase/metabolismo , Aumento de Peso/genética
5.
J Biol Chem ; 290(41): 24772-83, 2015 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-26306048

RESUMO

We previously reported that the skeletal muscle-specific overexpression of Fyn in mice resulted in a severe muscle wasting phenotype despite the activation of mTORC1 signaling. To investigate the bases for the loss of muscle fiber mass, we examined the relationship between Fyn activation of mTORC1, JNK, and endoplasmic reticulum stress. Overexpression of Fyn in skeletal muscle in vivo and in HEK293T cells in culture resulted in the activation of IRE1α and JNK, leading to increased cell death. Fyn synergized with the general endoplasmic reticulum stress inducer thapsigargin, resulting in the activation of IRE1α and further accelerated cell death. Moreover, inhibition of mTORC1 with rapamycin suppressed IRE1α activation and JNK phosphorylation, resulting in protecting cells against Fyn- and thapsigargin-induced cell death. Moreover, rapamycin treatment in vivo reduced the skeletal muscle IRE1α activation in the Fyn-overexpressing transgenic mice. Together, these data demonstrate the presence of a Fyn-induced endoplasmic reticulum stress that occurred, at least in part, through the activation of mTORC1, as well as subsequent activation of the IRE1α-JNK pathway driving cell death.


Assuntos
Endorribonucleases/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Células HEK293 , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Músculo Esquelético/citologia , Proteínas Proto-Oncogênicas c-fyn/genética , Transdução de Sinais/efeitos dos fármacos , Tapsigargina/farmacologia
6.
EMBO Rep ; 14(9): 795-803, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23907538

RESUMO

Macroautophagy (MA) regulates cellular quality control and energy balance. For example, loss of MA in aP2-positive adipocytes converts white adipose tissue (WAT) into brown adipose tissue (BAT)-like, enhancing BAT function and thereby insulin sensitivity. However, whether MA regulates early BAT development is unknown. We report that deleting Atg7 in myogenic Myf5+ progenitors inhibits MA in Myf5-cell-derived BAT and muscle. Knock out (KO) mice have defective BAT differentiation and function. Surprisingly, their body temperature is higher due to WAT lipolysis-driven increases in fatty acid oxidation in 'Beige' cells in inguinal WAT, BAT and muscle. KO mice also present impaired muscle differentiation, reduced muscle mass and glucose intolerance. Our studies show that ATG7 in Myf5+ progenitors is required to maintain energy and glucose homeostasis through effects on BAT and muscle development. Decreased MA in myogenic progenitors with age and/or overnutrition might contribute to the metabolic defects and sarcopenia observed in these conditions.


Assuntos
Tecido Adiposo Marrom/metabolismo , Autofagia , Metabolismo Energético , Glucose/metabolismo , Homeostase , Músculo Esquelético/metabolismo , Fator Regulador Miogênico 5/metabolismo , Tecido Adiposo Marrom/crescimento & desenvolvimento , Animais , Proteína 7 Relacionada à Autofagia , Diferenciação Celular , Ácidos Graxos/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Músculo Esquelético/crescimento & desenvolvimento , Fator Regulador Miogênico 5/genética , Células-Tronco/citologia , Células-Tronco/metabolismo
7.
Am J Physiol Regul Integr Comp Physiol ; 307(10): R1251-9, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25231351

RESUMO

Pompe disease is due to a deficiency in acid-α-glucosidase (GAA) and results in debilitating skeletal muscle wasting, characterized by the accumulation of glycogen and autophagic vesicles. Given the role of lysosomes as a platform for mTORC1 activation, we examined mTORC1 activity in models of Pompe disease. GAA-knockdown C2C12 myoblasts and GAA-deficient human skin fibroblasts of infantile Pompe patients were found to have decreased mTORC1 activation. Treatment with the cell-permeable leucine analog L-leucyl-L-leucine methyl ester restored mTORC1 activation. In vivo, Pompe mice also displayed reduced basal and leucine-stimulated mTORC1 activation in skeletal muscle, whereas treatment with a combination of insulin and leucine normalized mTORC1 activation. Chronic leucine feeding restored basal and leucine-stimulated mTORC1 activation, while partially protecting Pompe mice from developing kyphosis and the decline in muscle mass. Leucine-treated Pompe mice showed increased spontaneous activity and running capacity, with reduced muscle protein breakdown and glycogen accumulation. Together, these data demonstrate that GAA deficiency results in reduced mTORC1 activation that is partly responsible for the skeletal muscle wasting phenotype. Moreover, mTORC1 stimulation by dietary leucine supplementation prevented some of the detrimental skeletal muscle dysfunction that occurs in the Pompe disease mouse model.


Assuntos
Suplementos Nutricionais , Dipeptídeos/farmacologia , Doença de Depósito de Glicogênio Tipo II/tratamento farmacológico , Complexos Multiproteicos/metabolismo , Músculo Esquelético/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , alfa-Glucosidases/deficiência , Animais , Linhagem Celular , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Glicogênio/metabolismo , Doença de Depósito de Glicogênio Tipo II/enzimologia , Doença de Depósito de Glicogênio Tipo II/genética , Doença de Depósito de Glicogênio Tipo II/patologia , Doença de Depósito de Glicogênio Tipo II/fisiopatologia , Humanos , Insulina/farmacologia , Cifose/enzimologia , Cifose/patologia , Cifose/fisiopatologia , Cifose/prevenção & controle , Lisossomos/efeitos dos fármacos , Lisossomos/enzimologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Atrofia Muscular/enzimologia , Atrofia Muscular/patologia , Atrofia Muscular/fisiopatologia , Atrofia Muscular/prevenção & controle , Mioblastos/efeitos dos fármacos , Mioblastos/enzimologia , Interferência de RNA , Transfecção , alfa-Glucosidases/genética
8.
Nat Commun ; 14(1): 38, 2023 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-36596796

RESUMO

Recent studies implicate macrophages in regulation of thermogenic, sympathetic neuron-mediated norepinephrine (NE) signaling in adipose tissues, but understanding of such non-classical macrophage activities is incomplete. Here we show that male mice lacking the allograft inflammatory factor-1 (AIF1) protein resist high fat diet (HFD)-induced obesity and hyperglycemia. We link this phenotype to higher adipose NE levels that stem from decreased monoamine oxidase A (MAOA) expression and NE clearance by AIF1-deficient macrophages, and find through reciprocal bone marrow transplantation that donor Aif1-/- vs WT genotype confers the obesity phenotype in mice. Interestingly, human sequence variants near the AIF1 locus associate with obesity and diabetes; in adipose samples from participants with obesity, we observe direct correlation of AIF1 and MAOA transcript levels. These findings identify AIF1 as a regulator of MAOA expression in macrophages and catecholamine activity in adipose tissues - limiting energy expenditure and promoting energy storage - and suggest how it might contribute to human obesity.


Assuntos
Tecido Adiposo , Catecolaminas , Obesidade , Animais , Humanos , Masculino , Camundongos , Tecido Adiposo/metabolismo , Adiposidade , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Catecolaminas/metabolismo , Dieta Hiperlipídica/efeitos adversos , Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Norepinefrina/metabolismo , Obesidade/genética , Obesidade/metabolismo
9.
Cell Metab ; 5(5): 371-81, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17488639

RESUMO

Mice null for Fyn (a member of the Src family of nonreceptor tyrosine kinases) display a reduced percentage of adipose mass associated with decreased adipocyte cell size. In parallel, there is a substantial reduction in fasting plasma glucose, insulin, triglycerides, and free fatty acids concomitant with decreased intrahepatocellular and intramyocellular lipid accumulation. Importantly, the Fyn null mice exhibit improved glucose tolerance resulting from increased peripheral tissue (adipose and skeletal muscle) insulin sensitivity with a very small effect in the liver. Moreover, whole-body, adipose, and skeletal muscle fatty acid uptake and oxidation are increased along with AMP kinase activation and acetyl-CoA carboxylase inhibition. Together, these data demonstrate crosstalk between Src-family kinase activity and fatty acid oxidation and show that the loss of Fyn markedly improves peripheral tissue insulin sensitivity by relieving a selective negative modulation of AMP kinase activity in adipose tissue and skeletal muscle.


Assuntos
Tecido Adiposo/metabolismo , Ácidos Graxos/metabolismo , Insulina/metabolismo , Músculo Esquelético/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Adenilato Quinase/metabolismo , Animais , Western Blotting , Calorimetria Indireta , Glucose , Camundongos , Oxirredução , Proteínas Proto-Oncogênicas c-fyn/genética , Tomografia Computadorizada por Raios X
10.
Cell Metab ; 5(2): 151-6, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17276357

RESUMO

Recent studies have demonstrated a strong relationship between aging-associated reductions in mitochondrial function, dysregulated intracellular lipid metabolism, and insulin resistance. Given the important role of the AMP-activated protein kinase (AMPK) in the regulation of fat oxidation and mitochondrial biogenesis, we examined AMPK activity in young and old rats and found that acute stimulation of AMPK-alpha(2) activity by 5'-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside (AICAR) and exercise was blunted in skeletal muscle of old rats. Furthermore, mitochondrial biogenesis in response to chronic activation of AMPK with beta-guanidinopropionic acid (beta-GPA) feeding was also diminished in old rats. These results suggest that aging-associated reductions in AMPK activity may be an important contributing factor in the reduced mitochondrial function and dysregulated intracellular lipid metabolism associated with aging.


Assuntos
Envelhecimento , Mitocôndrias/enzimologia , Complexos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Guanidinas/administração & dosagem , Guanidinas/farmacologia , Masculino , Mitocôndrias/efeitos dos fármacos , Condicionamento Físico Animal , Propionatos/administração & dosagem , Propionatos/farmacologia , Ratos , Ratos Endogâmicos F344 , Ribonucleotídeos/farmacologia
11.
Am J Physiol Endocrinol Metab ; 302(5): E532-9, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22185839

RESUMO

Conventional (whole body) CYP2E1 knockout mice displayed protection against high-fat diet-induced weight gain, obesity, and hyperlipidemia with increased energy expenditure despite normal food intake and spontaneous locomotor activity. In addition, the CYP2E1 knockout mice displayed a marked improvement in glucose tolerance on both normal chow and high-fat diets. Euglycemic-hyperinsulinemic clamps demonstrated a marked protection against high-fat diet-induced insulin resistance in CYP2E1 knockout mice, with enhanced adipose tissue glucose uptake and insulin suppression of hepatic glucose output. In parallel, adipose tissue was protected against high-fat diet-induced proinflammatory cytokine production. Taken together, these data demonstrate that the CYP2E1 deletion protects mice against high-fat diet-induced insulin resistance with improved glucose homeostasis in vivo.


Assuntos
Citocromo P-450 CYP2E1/fisiologia , Dieta Hiperlipídica/efeitos adversos , Resistência à Insulina , Obesidade/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Transporte Biológico , Citocromo P-450 CYP2E1/genética , Citocinas/sangue , Fígado Gorduroso/etiologia , Fígado Gorduroso/patologia , Fígado Gorduroso/prevenção & controle , Glucose/metabolismo , Intolerância à Glucose/sangue , Intolerância à Glucose/etiologia , Intolerância à Glucose/prevenção & controle , Hiperlipidemias/sangue , Hiperlipidemias/etiologia , Hiperlipidemias/prevenção & controle , Insulina/metabolismo , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Terapia de Alvo Molecular , Fibras Musculares Esqueléticas/metabolismo , Obesidade/etiologia , Obesidade/fisiopatologia , Obesidade/prevenção & controle , Transdução de Sinais
12.
Sci Adv ; 8(17): eabm7012, 2022 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-35476450

RESUMO

The immune checkpoint B7-H3 (CD276) is a member of the B7 family that has been studied in the tumor microenvironment and immunotherapy, but its potential role in metabolism remains largely unknown. Here, we show that B7-H3 is highly expressed in mouse and human adipose tissue at steady state, with the highest levels in adipocyte progenitor cells. B7-H3 is rapidly down-regulated upon the initiation of adipocyte differentiation. Combined RNA sequencing and metabolic studies reveal that B7-H3 stimulates glycolytic and mitochondrial activity of adipocyte progenitors. Loss of B7-H3 in progenitors results in impaired oxidative metabolism program and increased lipid accumulation in derived adipocytes. Consistent with these observations, mice knocked out for B7-H3 develop spontaneous obesity, metabolic dysfunction, and adipose tissue inflammation. Our results reveal an unexpected metabolic role for B7-H3 in adipose tissue and open potential new avenues for the treatment of metabolic diseases by targeting the B7-H3 pathway.

13.
Elife ; 112022 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-35254259

RESUMO

Cholinergic and sympathetic counter-regulatory networks control numerous physiological functions, including learning/memory/cognition, stress responsiveness, blood pressure, heart rate, and energy balance. As neurons primarily utilize glucose as their primary metabolic energy source, we generated mice with increased glycolysis in cholinergic neurons by specific deletion of the fructose-2,6-phosphatase protein TIGAR. Steady-state and stable isotope flux analyses demonstrated increased rates of glycolysis, acetyl-CoA production, acetylcholine levels, and density of neuromuscular synaptic junction clusters with enhanced acetylcholine release. The increase in cholinergic signaling reduced blood pressure and heart rate with a remarkable resistance to cold-induced hypothermia. These data directly demonstrate that increased cholinergic signaling through the modulation of glycolysis has several metabolic benefits particularly to increase energy expenditure and heat production upon cold exposure.


Assuntos
Acetilcolina , Junção Neuromuscular , Acetilcolina/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Colinérgicos/metabolismo , Camundongos , Músculo Esquelético/metabolismo , Junção Neuromuscular/fisiologia , Monoéster Fosfórico Hidrolases/metabolismo , Termogênese
14.
Mol Metab ; 48: 101227, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33812059

RESUMO

OBJECTIVE: Liver glycogen levels are dynamic and highly regulated by nutrient availability as the levels decrease during fasting and are restored during the feeding cycle. However, feeding in the presence of fructose in water suppresses glycogen accumulation in the liver by upregulating the expression of the glucose-6-phosphatase catalytic subunit (G6pc) gene, although the exact mechanism is unknown. We generated liver-specific knockout MED13 mice that lacked the transcriptional Mediator complex kinase module to examine its effect on the transcriptional activation of inducible target gene expression, such as the ChREBP- and FOXO1-dependent control of the G6pc gene promoter. METHODS: The relative changes in liver expression of lipogenic and gluconeogenic genes as well as glycogen levels were examined in response to feeding standard low-fat laboratory chow supplemented with water or water containing sucrose or fructose in control (Med13fl/fl) and liver-specific MED13 knockout (MED13-LKO) mice. RESULTS: Although MED13 deficiency had no significant effect on constitutive gene expression, all the dietary inducible gene transcripts were significantly reduced despite the unchanged insulin sensitivity in the MED13-LKO mice compared to that in the control mice. G6pc gene transcription displayed the most significant difference between the Med13 fl/fl and MED13-LKO mice, particularly when fed fructose. Following fasting that depleted liver glycogen, feeding induced the restoration of glycogen levels except in the presence of fructose. MED13 deficiency rescued the glycogen accumulation defect in the presence of fructose. This resulted from the suppression of G6pc expression and thus G6PC enzymatic activity. Among two transcriptional factors that regulate G6pc gene expression, FOXO1 binding to the G6pc promoter was not affected, whereas ChREBP binding was dramatically reduced in MED13-LKO hepatocytes. In addition, there was a marked suppression of FOXO1 and ChREBP-ß transcriptional activities in MED13-LKO hepatocytes. CONCLUSIONS: Taken together, our data suggest that the kinase module of the Mediator complex is necessary for the transcriptional activation of metabolic genes such as G6pc and has an important role in regulating glycogen levels in the liver through altering transcription factor binding and activity at the G6pc promoter.


Assuntos
Domínio Catalítico/genética , Frutose/metabolismo , Glucose-6-Fosfatase/química , Glucose-6-Fosfatase/metabolismo , Glicogênio Hepático/biossíntese , Fígado/metabolismo , Complexo Mediador/metabolismo , Transdução de Sinais/genética , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Células Cultivadas , Ativação Enzimática/genética , Jejum , Frutose/farmacologia , Expressão Gênica , Gluconeogênese/efeitos dos fármacos , Gluconeogênese/genética , Glucose-6-Fosfatase/genética , Hepatócitos/metabolismo , Resistência à Insulina/genética , Lipogênese/efeitos dos fármacos , Lipogênese/genética , Masculino , Complexo Mediador/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
15.
Cell Metab ; 25(1): 182-196, 2017 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-28011087

RESUMO

The transcription factor EB (TFEB) is an essential component of lysosomal biogenesis and autophagy for the adaptive response to food deprivation. To address the physiological function of TFEB in skeletal muscle, we have used muscle-specific gain- and loss-of-function approaches. Here, we show that TFEB controls metabolic flexibility in muscle during exercise and that this action is independent of peroxisome proliferator-activated receptor-γ coactivator1α (PGC1α). Indeed, TFEB translocates into the myonuclei during physical activity and regulates glucose uptake and glycogen content by controlling expression of glucose transporters, glycolytic enzymes, and pathways related to glucose homeostasis. In addition, TFEB induces the expression of genes involved in mitochondrial biogenesis, fatty acid oxidation, and oxidative phosphorylation. This coordinated action optimizes mitochondrial substrate utilization, thus enhancing ATP production and exercise capacity. These findings identify TFEB as a critical mediator of the beneficial effects of exercise on metabolism.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Metabolismo , Condicionamento Físico Animal , Adenilato Quinase/metabolismo , Animais , Autofagia/genética , Núcleo Celular/metabolismo , Metabolismo Energético/genética , Genes Mitocondriais , Genoma , Glucose/metabolismo , Homeostase/genética , Insulina/metabolismo , Metabolismo/genética , Camundongos Knockout , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Biogênese de Organelas , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Transporte Proteico , Transdução de Sinais/genética
16.
Shock ; 45(6): 677-85, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26682946

RESUMO

Lipopolysaccharide (LPS) is known to impair insulin-stimulated muscle glucose uptake (MGU). We determined if increased glucose transport (GLUT4) or phosphorylation capacity (hexokinase II; HKII) could overcome the impairment in MGU. We used mice that overexpressed GLUT4 (GLUT4) or HKII (HK) in skeletal muscle. Studies were performed in conscious, chronically catheterized (carotid artery and jugular vein) mice. Mice received an intravenous bolus of either LPS (10 µg/g body weight) or vehicle (VEH). After 5 h, a hyperinsulinemic-euglycemic clamp was performed. As MGU is also dependent on cardiovascular function that is negatively affected by LPS, cardiac function was assessed using echocardiography. LPS decreased whole body glucose disposal and MGU in wild-type (WT) and HK mice. In contrast, the decrease was attenuated in GLUT4 mice. Although membrane-associated GLUT4 was increased in VEH-treated GLUT4 mice, LPS impaired membrane-associated GLUT4 in GLUT4 mice to the same level as LPS-treated WT mice. This suggested that overexpression of GLUT4 had further benefits beyond preserving transport activity. In fact, GLUT4 overexpression attenuated the LPS-induced decrease in cardiac function. The maintenance of MGU in GLUT4 mice following LPS was accompanied by sustained anaerobic glycolytic flux as suggested by increased muscle Pdk4 expression, and elevated lactate availability. Thus, enhanced glucose transport, but not phosphorylation capacity, ameliorates LPS-induced impairments in MGU. This benefit is mediated by long-term adaptations to the overexpression of GLUT4 that sustain muscle anaerobic glycolytic flux and cardiac function in response to LPS.


Assuntos
Glicemia/metabolismo , Insulina/metabolismo , Lipopolissacarídeos/metabolismo , Músculo Esquelético/metabolismo , Fosforilação , Animais , Modelos Animais de Doenças , Transportador de Glucose Tipo 4/metabolismo , Glicogênio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Musculares/metabolismo
17.
Front Physiol ; 6: 393, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26733885

RESUMO

Muscle wasting that occurs during aging or from disease pathology presents with an accumulation of lipid species termed ceroid or lipofuscin. This unique species of lipid has been characterized in various cell types but its properties and organization in skeletal muscle remains unclear. Using immunofluorescence and transmission electron microscopy, we were able to visualize and characterize an atypical lipid storing organelle in skeletal muscle. White myofibers contain two organelles at each pole of the myonuclei and red myofibers contain many of these structures in and around the perinuclear space. These organelles contain markers for late endosomes, are morphologically similar to multivesicular bodies, store lipid, and hypertrophy in aged muscle and a model of muscle wasting with an accumulation of large amounts of lipofuscin. Rapamycin treatment reduces the multivesicular body hypertrophy, restores late endosomal protein markers, and also increases the number and intensity of lipofuscin deposits. Together, these data demonstrate for the first time a perinuclear organelle in skeletal muscle that hypertrophies in muscle wasting phenotypes and is involved in endocytic lipid storage.

18.
Diabetes ; 64(7): 2636-45, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25732192

RESUMO

Adenylyl cyclase type 5 knockout (AC5KO) mice have increased longevity and share a similar phenotype with calorie-restricted wild-type (WT) mice. To determine the in vivo metabolic properties of AC5 deficiency, we compared the effects of standard diet (SD) and high-fat diet (HFD) on obesity, energy balance, glucose regulation, and insulin sensitivity. AC5KO mice on SD had reduced body weight and adiposity compared with WT mice. Blood cholesterol and triglyceride levels were also significantly reduced in AC5KO mice. Indirect calorimetry demonstrated increased oxygen consumption, respiratory exchange ratio, and energy expenditure in AC5KO compared with WT mice on both SD and HFD. AC5KO mice also displayed improved glucose tolerance and increased whole-body insulin sensitivity, accompanied by decreased liver glycogen stores. Euglycemic-hyperinsulinemic clamp studies confirmed the marked improvement of glucose homeostasis and insulin sensitivity in AC5KO mice primarily through increased insulin sensitivity in skeletal muscle. Moreover, the genes involved in mitochondrial biogenesis and function were significantly increased in AC5KO skeletal muscle. These data demonstrate that deficiency of AC5 protects against obesity, glucose intolerance, and insulin resistance, supporting AC5 as a potential novel therapeutic target for treatment of obesity and diabetes.


Assuntos
Adenilil Ciclases/deficiência , Resistência à Insulina , Obesidade/prevenção & controle , Inibidores de Adenilil Ciclases , Adenilil Ciclases/fisiologia , Animais , Dieta Hiperlipídica , Metabolismo Energético , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/metabolismo
19.
PLoS One ; 9(2): e89199, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586592

RESUMO

Mice treated with the antidepressant trans-2-phenylcyclopropylamine (2-PCPA) were protected against diet-induced-obesity, and adiposity was reversed in pre-established diet-induced obese mice. Contrary to a recent report that inhibition of lysine-specific demethylase-1 by 2-PCPA results in increased energy expenditure, long-term 2-PCPA treatment had no such effect but its protection against obesity was associated with increased spontaneous locomotor activity, Moreover, pair feeding to assure equal caloric intake in wild type mice as well as in genetic hyperphagic mice (ob/ob) also resulted in weight reduction in 2-PCPA treated mice that correlated with increased activity but no change in energy expenditure. Similarly, short-term intraperitoneal injections of 2-PCPA did not affect food intake but caused a substantial increase in locomotor activity in the light cycle that correlated with increased energy expenditure, whereas activity and energy expenditure were unchanged in the dark cycle. Lastly, 2-PCPA was also effective in reducing obesity in genetic UCP1 null mice. These data suggest that 2-PCPA can reduce obesity by decreasing food intake in the long term while increasing activity in the short-term. However, the protective and weight loss effects of 2-PCPA are independent of UCP1-regulated thermogenesis or basal energy expenditure.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Ingestão de Alimentos/efeitos dos fármacos , Obesidade/prevenção & controle , Tranilcipromina/uso terapêutico , Animais , Composição Corporal/efeitos dos fármacos , Ingestão de Energia/efeitos dos fármacos , Teste de Tolerância a Glucose , Masculino , Camundongos , Camundongos Obesos , Obesidade/tratamento farmacológico , Obesidade/etiologia , Tranilcipromina/farmacologia
20.
Cell Rep ; 9(5): 1574-1583, 2014 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-25466256

RESUMO

Adipose tissue inflammation is one pathway shown to mediate insulin resistance in obese humans and rodents. Obesity induces dynamic cellular changes in adipose tissue to increase proinflammatory cytokines and diminish anti-inflammatory cytokines. However, we have found that anti-inflammatory interleukin-13 (IL-13) is unexpectedly induced in adipose tissue of obese humans and high-fat diet (HFD)-fed mice, and the source of IL-13 is primarily the adipocyte. Moreover, HFD-induced proinflammatory cytokines such as tumor necrosis factor alpha (TNF-α) and IL-1ß mediate IL-13 production in adipocytes in an IKKß-dependent manner. In contrast, adipocyte-specific IKKß-deficient mice show diminished IL-13 expression and enhanced inflammation after HFD feeding, resulting in a worsening of the insulin-resistant state. Together these data demonstrate that although IKKß activates the expression of proinflammatory mediators, in adipocytes, IKKß signaling also induces the expression of the anti-inflammatory cytokine IL-13, which plays a unique protective role by limiting adipose tissue inflammation and insulin resistance.


Assuntos
Adipócitos/enzimologia , Tecido Adiposo/metabolismo , Quinase I-kappa B/metabolismo , Interleucina-13/fisiologia , Comunicação Parácrina , Adipócitos/imunologia , Tecido Adiposo/imunologia , Animais , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Epididimo/metabolismo , Retroalimentação Fisiológica , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/imunologia , Masculino , Camundongos Knockout , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa