Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Mol Cell ; 77(4): 810-824.e8, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31901447

RESUMO

Lipid droplets (LDs) provide a reservoir for triacylglycerol storage and are a central hub for fatty acid trafficking and signaling in cells. Lipolysis promotes mitochondrial biogenesis and oxidative metabolism via a SIRT1/PGC-1α/PPARα-dependent pathway through an unknown mechanism. Herein, we identify that monounsaturated fatty acids (MUFAs) allosterically activate SIRT1 toward select peptide-substrates such as PGC-1α. MUFAs enhance PGC-1α/PPARα signaling and promote oxidative metabolism in cells and animal models in a SIRT1-dependent manner. Moreover, we characterize the LD protein perilipin 5 (PLIN5), which is known to enhance mitochondrial biogenesis and function, to be a fatty-acid-binding protein that preferentially binds LD-derived monounsaturated fatty acids and traffics them to the nucleus following cAMP/PKA-mediated lipolytic stimulation. Thus, these studies identify the first-known endogenous allosteric modulators of SIRT1 and characterize a LD-nuclear signaling axis that underlies the known metabolic benefits of MUFAs and PLIN5.


Assuntos
Ácidos Graxos Monoinsaturados/metabolismo , Gotículas Lipídicas/química , Perilipina-5/metabolismo , Sirtuína 1/metabolismo , Regulação Alostérica , Animais , Transporte Biológico , Linhagem Celular , Células Cultivadas , Dieta , Ácidos Graxos/metabolismo , Lipase/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Azeite de Oliva , Perilipina-5/fisiologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Transcrição Gênica
2.
Cell Commun Signal ; 20(1): 4, 2022 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-34998412

RESUMO

BACKGROUND: Research is revealing the complex coordination between cell signaling systems as they adapt to genetic and epigenetic changes. Tools to uncover these highly complex functional linkages will play an important role in advancing more efficacious disease treatments. Current tumor cell signal transduction research is identifying coordination between receptor types, receptor families, and transduction pathways to maintain tumor cell viability despite challenging tumor microenvironment conditions. METHODS: In this report, coactivated abnormal levels of signaling activity for c-Met and HER family receptors in live tumor cells were measured by a new clinical test to identify a subpopulation of breast cancer patients that could be responsive to combined targeted therapies. The CELsignia Multi-Pathway Signaling Function (CELsignia) Test uses an impedance biosensor to quantify an individual patient's ex vivo live tumor cell signaling response in real-time to specific HER family and c-Met co-stimulation and targeted therapies. RESULTS: The test identified breast tumors with hyperactive HER1, HER2, HER3/4, and c-Met coordinated signaling that express otherwise normal amounts of these receptors. The supporting data of the pre-clinical verification of this test included analyses of 79 breast cancer patients' cell response to HER and c-Met agonists. The signaling results were confirmed using clinically approved matching targeted drugs, and combinations of targeted drugs in addition to correlative mouse xenograft tumor response to HER and c-Met targeted therapies. CONCLUSIONS: The results of this study demonstrated the potential benefit of a functional test for identifying a subpopulation of breast cancer patients with coordinated abnormal HER and c-Met signaling for a clinical trial testing combination targeted therapy. Video Abstract.


Assuntos
Neoplasias da Mama , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Receptor ErbB-2/metabolismo , Transdução de Sinais , Microambiente Tumoral
3.
Hepatology ; 62(3): 964-7, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25854913

RESUMO

Hepatic steatosis is defined by the accumulation of lipid droplets (LDs). Once thought to be only inert energy storage depots, LDs are increasingly recognized as organelles that have important functions in hepatocytes beyond lipid storage. The lipid and protein composition of LDs is highly dynamic and influences their intrinsic metabolism and signaling properties, which ultimately links them to the changes in hepatic function. This concise review highlights recent discoveries in LD biology and unique aspects of hepatic LDs and their role in liver disease.


Assuntos
Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Gotículas Lipídicas/metabolismo , Metabolismo dos Lipídeos/fisiologia , Progressão da Doença , Humanos , Testes de Função Hepática , Prognóstico
4.
Cell Metab ; 2(2): 131-40, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16098830

RESUMO

Reducing obesity requires an elevation of energy expenditure and/or a suppression of food intake. Here we show that enhancing hepatic glycolysis reduces body weight and adiposity in obese mice. Overexpression of glucokinase or 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase is used to increase hepatic glycolysis. Either of the two treatments produces similar increases in rates of fatty acid oxidation in extrahepatic tissues, i.e., skeletal muscle, leading to an elevation of energy expenditure. However, only 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase overexpression causes a suppression of food intake and a decrease in hypothalamic neuropeptide Y expression, contributing to a more pronounced reduction of body weight with this treatment. Furthermore, the two treatments cause differential lipid profiles due to opposite effects on hepatic lipogenesis, associated with distinct phosphorylation states of carbohydrate response element binding protein and AMP-activated protein kinase. The step at which hepatic glycolysis is enhanced dramatically influences overall whole-body energy balance and lipid profiles.


Assuntos
Metabolismo Energético , Glicólise , Hepatócitos/metabolismo , Metabolismo dos Lipídeos , Obesidade/metabolismo , Tecido Adiposo/metabolismo , Animais , Peso Corporal , Células Cultivadas , Ingestão de Alimentos , Glucoquinase/genética , Glucoquinase/metabolismo , Glucose/metabolismo , Hepatócitos/citologia , Homeostase , Insulina/sangue , Masculino , Camundongos , Camundongos Endogâmicos , Fosfofrutoquinase-2/genética , Fosfofrutoquinase-2/metabolismo , Ratos
5.
Adv Enzyme Regul ; 46: 72-88, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16860376

RESUMO

Fructose-2,6-bisphosphate (F26P2) was identified as a regulator of glucose metabolism over 25 years ago. A truly bifunctional enzyme, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (6PFK2/FBP2), with two active sites synthesizes F26P2 from fructose-6-phosphate (F6P) and ATP or degrades F26P2 to F6P and Pi. In the classic view, F26P2 regulates glucose metabolism by allosteric effects on 6-phosphofructo-1-kinase (6PFK1, activation) and fructose-1,6-bisphosphatase (FBPase, inhibition). When levels of F26P2 are high, glycolysis is enhanced and gluconeogenesis is inhibited. In this regard, altering levels of F26P2 via 6PFK2/FBP2 overexpression has been used for metabolic modulation, and has been shown capable of restoring euglycemia in rodent models of diabetes. Recently, a number of novel observations have suggested that F26P2 has much broader effects on the enzymes of glucose metabolism. This is evidenced by the effects of F26P2 on the gene expression of two key glucose metabolic enzymes, glucokinase (GK) and glucose-6-phosphatase (G6Pase). When levels of F26P2 are elevated in the liver, the gene expression and protein amount of GK is increased whereas G6Pase is decreased. These coordinated changes in GK and G6Pase protein illustrate how F26P2 regulates glucose metabolism. F26P2 also affects the gene expression of enzymes related to lipid metabolism. When F26P2 levels are elevated in liver, the expression of two key lipogenic enzymes, acetyl-CoA carboxylase 1 (ACC1) and fatty acid synthase (FAS) is reduced, contributing to a unique coordinated decrease in lipogenesis. When combined, F26P2 effects on glucose and lipid metabolism provide cooperative regulation of fuel metabolism. The regulatory roles for F26P2 have also expanded to transcription factors, as well as certain key proteins (enzymes) of signaling and/or energy sensoring. Although some effects may be secondary to changes in metabolite levels, high levels of F26P2 have been shown to regulate protein amount and/or phosphorylation state of hepatic nuclear factor 1-alpha (HNF1alpha), carbohydrate response element binding protein (ChREBP), peroxisome proliferators-activated receptor alpha (PPARalpha), and peroxisome proliferators-activated receptor gamma co-activator 1beta (PGC1beta), as well as Akt and AMP-activated protein kinase (AMPK). Importantly, changes in these transcription factors, signaling proteins, and sensor proteins are produced in a way that appropriately coordinates whole body fuel metabolism.


Assuntos
Frutosedifosfatos/metabolismo , Gluconeogênese/fisiologia , Glicólise/fisiologia , Regulação Alostérica/fisiologia , Animais , Masculino , Camundongos , Ratos
6.
Cell Rep ; 15(2): 349-59, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-27050511

RESUMO

Metabolism is a highly integrated process that is coordinately regulated between tissues and within individual cells. FoxO proteins are major targets of insulin action and contribute to the regulation of gluconeogenesis, glycolysis, and lipogenesis in the liver. However, the mechanisms by which FoxO proteins exert these diverse effects in an integrated fashion remain poorly understood. We report that FoxO proteins also exert important effects on intrahepatic lipolysis and fatty acid oxidation via the regulation of adipose triacylglycerol lipase (ATGL), which mediates the first step in lipolysis, and its inhibitor, the G0/S1 switch 2 gene (G0S2). We also find that ATGL-dependent lipolysis plays a critical role in mediating diverse effects of FoxO proteins in the liver, including effects on gluconeogenic, glycolytic, and lipogenic gene expression and metabolism. These results indicate that intrahepatic lipolysis plays a critical role in mediating and integrating the regulation of glucose and lipid metabolism downstream of FoxO proteins.


Assuntos
Tecido Adiposo/metabolismo , Proteína Forkhead Box O1/metabolismo , Glucose/metabolismo , Lipase/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Homeostase , Humanos , Lipase/genética , Metabolismo dos Lipídeos/genética , Lipogênese , Masculino , Camundongos Transgênicos , Modelos Biológicos , Oxirredução
8.
Am J Physiol Endocrinol Metab ; 291(3): E536-43, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16621898

RESUMO

Hepatic insulin resistance is one of the characteristics of type 2 diabetes and contributes to the development of hyperglycemia. How changes in hepatic glucose flux lead to insulin resistance is not clearly defined. We determined the effects of decreasing the levels of hepatic fructose 2,6-bisphosphate (F26P(2)), a key regulator of glucose metabolism, on hepatic glucose flux in the normal 129J mice. Upon adenoviral overexpression of a kinase activity-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase, the enzyme that determines F26P(2) level, hepatic F26P(2) levels were decreased twofold compared with those of control virus-treated mice in basal state. In addition, under hyperinsulinemic conditions, hepatic F26P(2) levels were much lower than those of the control. The decrease in F26P(2) leads to the elevation of basal and insulin-suppressed hepatic glucose production. Also, the efficiency of insulin to suppress hepatic glucose production was decreased (63.3 vs. 95.5% suppression of the control). At the molecular level, a decrease in insulin-stimulated Akt phosphorylation was consistent with hepatic insulin resistance. In the low hepatic F26P(2) states, increases in both gluconeogenesis and glycogenolysis in the liver are responsible for elevations of hepatic glucose production and thereby contribute to the development of hyperglycemia. Additionally, the increased hepatic gluconeogenesis was associated with the elevated mRNA levels of peroxisome proliferator-activated receptor-gamma coactivator-1alpha and phosphoenolpyruvate carboxykinase. This study provides the first in vivo demonstration showing that decreasing hepatic F26P(2) levels leads to increased gluconeogenesis in the liver. Taken together, the present study demonstrates that perturbation of glucose flux in the liver plays a predominant role in the development of a diabetic phenotype, as characterized by hepatic insulin resistance.


Assuntos
Frutosedifosfatos/metabolismo , Glucose/metabolismo , Hiperglicemia/metabolismo , Resistência à Insulina/fisiologia , Fígado/metabolismo , Adenoviridae/genética , Animais , Glicemia/metabolismo , Expressão Gênica/genética , Gluconeogênese/genética , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Glucose-6-Fosfatase/genética , Glicogenólise/genética , Hiperglicemia/genética , Insulina/farmacologia , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos , Mutação/genética , Proteína Oncogênica v-akt/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Fosfofrutoquinase-2/genética , Fosfofrutoquinase-2/metabolismo , Transativadores/genética , Fatores de Transcrição , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA