Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Transl Med ; 15(723): eade8460, 2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-37992151

RESUMEN

Despite their high degree of effectiveness in the management of psychiatric conditions, exposure to antipsychotic drugs, including olanzapine and risperidone, is frequently associated with substantial weight gain and the development of diabetes. Even before weight gain, a rapid rise in circulating leptin concentrations can be observed in most patients taking antipsychotic drugs. To date, the contribution of this hyperleptinemia to weight gain and metabolic deterioration has not been defined. Here, with an established mouse model that recapitulates antipsychotic drug-induced obesity and insulin resistance, we not only confirm that hyperleptinemia occurs before weight gain but also demonstrate that hyperleptinemia contributes directly to the development of obesity and associated metabolic disorders. By suppressing the rise in leptin through the use of a monoclonal leptin-neutralizing antibody, we effectively prevented weight gain, restored glucose tolerance, and preserved adipose tissue and liver function in antipsychotic drug-treated mice. Mechanistically, suppressing excess leptin resolved local tissue and systemic inflammation typically associated with antipsychotic drug treatment. We conclude that hyperleptinemia is a key contributor to antipsychotic drug-associated weight gain and metabolic deterioration. Leptin suppression may be an effective approach to reducing the undesirable side effects of antipsychotic drugs.


Asunto(s)
Antipsicóticos , Enfermedades Metabólicas , Humanos , Ratones , Animales , Antipsicóticos/efectos adversos , Leptina/metabolismo , Obesidad/metabolismo , Aumento de Peso
2.
Nat Metab ; 5(1): 147-164, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36593271

RESUMEN

Leptin acts on hypothalamic neurons expressing agouti-related protein (AgRP) or pro-opiomelanocortin (POMC) to suppress appetite and increase energy expenditure, but the intracellular mechanisms that modulate central leptin signalling are not fully understood. Here we show that growth factor receptor-bound protein 10 (Grb10), an adaptor protein that binds to the insulin receptor and negatively regulates its signalling pathway, can interact with the leptin receptor and enhance leptin signalling. Ablation of Grb10 in AgRP neurons promotes weight gain, while overexpression of Grb10 in AgRP neurons reduces body weight in male and female mice. In parallel, deletion or overexpression of Grb10 in POMC neurons exacerbates or attenuates diet-induced obesity, respectively. Consistent with its role in leptin signalling, Grb10 in AgRP and POMC neurons enhances the anorexic and weight-reducing actions of leptin. Grb10 also exaggerates the inhibitory effects of leptin on AgRP neurons via ATP-sensitive potassium channel-mediated currents while facilitating the excitatory drive of leptin on POMC neurons through transient receptor potential channels. Our study identifies Grb10 as a potent leptin sensitizer that contributes to the maintenance of energy homeostasis by enhancing the response of AgRP and POMC neurons to leptin.


Asunto(s)
Leptina , Proopiomelanocortina , Ratones , Masculino , Femenino , Animales , Proteína Relacionada con Agouti/metabolismo , Leptina/metabolismo , Proopiomelanocortina/metabolismo , Proteína Adaptadora GRB10/metabolismo , Pérdida de Peso
3.
Sci Adv ; 8(38): eabq1799, 2022 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-36129988

RESUMEN

Pancreatic ß cell failure is a hallmark of diabetes. However, the causes of ß cell failure remain incomplete. Here, we report the identification of tetranectin (TN), an adipose tissue-enriched secretory molecule, as a negative regulator of insulin secretion in ß cells in diabetes. TN expression is stimulated by high glucose in adipocytes via the p38 MAPK/TXNIP/thioredoxin/OCT4 signaling pathway, and elevated serum TN levels are associated with diabetes. TN treatment greatly exacerbates hyperglycemia in mice and suppresses glucose-stimulated insulin secretion in islets. Conversely, knockout of TN or neutralization of TN function notably improves insulin secretion and glucose tolerance in high-fat diet-fed mice. Mechanistically, TN binds with high selectivity to ß cells and inhibits insulin secretion by blocking L-type Ca2+ channels. Our study uncovers an adipocyte-ß cell cross-talk that contributes to ß cell dysfunction in diabetes and suggests that neutralization of TN levels may provide a new treatment strategy for type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Adipocitos/metabolismo , Animales , Glucosa/metabolismo , Insulina/metabolismo , Secreción de Insulina , Lectinas Tipo C , Ratones , Tiorredoxinas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
Commun Biol ; 5(1): 771, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35915318

RESUMEN

A unique feature of the liver is its high regenerative capacity, which is essential to maintain liver homeostasis. However, key regulators of liver regeneration (LR) remain ill-defined. Here, we identify hepatic miR-182-5p as a key regulator of LR. Suppressing miR-182-5p, whose expression is significantly induced in the liver of mice post two-thirds partial hepatectomy (PH), abrogates PH-induced LR in mice. In contrast, liver-specific overexpression of miR-182-5p promotes LR in mice with PH. Overexpression of miR-182-5p failed to promote proliferation in hepatocytes, but stimulates proliferation when hepatocytes are cocultured with stellate cells. Mechanistically, miR-182-5p stimulates Cyp7a1-mediated cholic acid production in hepatocytes, which promotes hedgehog (Hh) ligand production in stellate cells, leading to the activation of Hh signaling in hepatocytes and consequent cell proliferation. Collectively, our study identified miR-182-5p as a critical regulator of LR and uncovers a Cyp7a1/cholic acid-dependent mechanism by which hepatocytes crosstalk to stellate cells to facilitate LR.


Asunto(s)
Regeneración Hepática , MicroARNs , Animales , Ácido Cólico/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Hepatocitos/metabolismo , Regeneración Hepática/genética , Ratones , MicroARNs/genética , MicroARNs/metabolismo
6.
Protein Cell ; 13(2): 90-101, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34374004

RESUMEN

The cyclic GMP-AMP (cGAMP) synthase (cGAS) has been identified as a cytosolic double stranded DNA sensor that plays a pivotal role in the type I interferon and inflammation responses via the STING-dependent signaling pathway. In the past several years, a growing body of evidence has revealed that cGAS is also localized in the nucleus where it is associated with distinct nuclear substructures such as nucleosomes, DNA replication forks, the double-stranded breaks, and centromeres, suggesting that cGAS may have other functions in addition to its role in DNA sensing. However, while the innate immune function of cGAS is well established, the non-canonical nuclear function of cGAS remains poorly understood. Here, we review our current understanding of the complex nature of nuclear cGAS and point to open questions on the novel roles and the mechanisms of action of this protein as a key regulator of cell nuclear function, beyond its well-established role in dsDNA sensing and innate immune response.


Asunto(s)
Núcleo Celular/inmunología , Inmunidad Innata , Nucleotidiltransferasas/inmunología , Transducción de Señal/inmunología , Núcleo Celular/genética , Humanos , Nucleotidiltransferasas/genética , Transducción de Señal/genética
7.
J Clin Invest ; 131(24)2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34730111

RESUMEN

Dysregulation in adipokine biosynthesis and function contributes to obesity-induced metabolic diseases. However, the identities and functions of many of the obesity-induced secretory molecules remain unknown. Here, we report the identification of leucine-rich alpha-2-glycoprotein 1 (LRG1) as an obesity-associated adipokine that exacerbates high fat diet-induced hepatosteatosis and insulin resistance. Serum levels of LRG1 were markedly elevated in obese humans and mice compared with their respective controls. LRG1 deficiency in mice greatly alleviated diet-induced hepatosteatosis, obesity, and insulin resistance. Mechanistically, LRG1 bound with high selectivity to the liver and promoted hepatosteatosis by increasing de novo lipogenesis and suppressing fatty acid ß-oxidation. LRG1 also inhibited hepatic insulin signaling by downregulating insulin receptor substrates 1 and 2. Our study identified LRG1 as a key molecule that mediates the crosstalk between adipocytes and hepatocytes in diet-induced hepatosteatosis and insulin resistance. Suppressing LRG1 expression and function may be a promising strategy for the treatment of obesity-related metabolic diseases.


Asunto(s)
Adipoquinas/metabolismo , Hígado Graso/metabolismo , Glicoproteínas/metabolismo , Resistencia a la Insulina , Obesidad/metabolismo , Adipoquinas/genética , Animales , Ácidos Grasos/genética , Ácidos Grasos/metabolismo , Hígado Graso/genética , Glicoproteínas/genética , Humanos , Ratones , Ratones Noqueados , Obesidad/genética , Oxidación-Reducción
8.
J Mol Cell Biol ; 13(10): 728-738, 2021 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-34665236

RESUMEN

The cyclic GMP‒AMP synthase (cGAS)‒stimulator of interferon genes (STING) signaling pathway senses the presence of cytosolic DNA and, in turn, triggers downstream signaling to induce the expression of inflammatory and type I interferon genes in immune cells. Whereas the innate immune function of the cGAS‒STING pathway is well studied over the past years, emerging evidence suggests that this signaling pathway may have additional functions beyond innate immune surveillance. Consistent with this notion, dysregulation of the cGAS‒STING signaling pathway in adipocytes, hepatocytes, and renal proximal tubule epithelial cells are associated with metabolic dysfunction, impaired energy homeostasis, and kidney diseases. In this review, we summarize current understanding of the cGAS‒STING pathway in several metabolic diseases such as obesity, insulin resistance, alcoholic and nonalcoholic fatty liver diseases, as well as acute kidney injury and chronic kidney disease. We also review the interaction between the cGAS‒STING pathway and lipid metabolism. Lastly, we discuss potential mechanisms by which cGAS‒STING signaling regulates metabolism and point toward future avenues of research targeting the cGAS‒STING pathway as possible means to treat common metabolic disorders.


Asunto(s)
Lesión Renal Aguda , Interferón Tipo I , Humanos , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Transducción de Señal
9.
JCI Insight ; 6(17)2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34264867

RESUMEN

A dynamically regulated microenvironment, which is mediated by crosstalk between adipocytes and neighboring cells, is critical for adipose tissue homeostasis and function. However, information on key molecules and/or signaling pathways regulating the crosstalk remains limited. In this study, we identify adipocyte miRNA-182-5p (miR-182-5p) as a crucial antiobesity molecule that stimulated beige fat thermogenesis by promoting the crosstalk between adipocytes and macrophages. miR-182-5p was highly enriched in thermogenic adipocytes, and its expression was markedly stimulated by cold exposure in mice. In contrast, miR-182-5p expression was significantly reduced in adipose tissues of obese humans and mice. Knockout of miR-185-5p decreased cold-induced beige fat thermogenesis whereas overexpression of miR-185-5p increased beiging and thermogenesis in mice. Mechanistically, miR-182-5p promoted FGF21 expression and secretion in adipocytes by suppressing nuclear receptor subfamily 1 group D member 1 (Nr1d1) at 5'-UTR, which in turn stimulates acetylcholine synthesis and release in macrophages. Increased acetylcholine expression activated the nicotine acetylcholine receptor in adipocytes, which stimulated PKA signaling and consequent thermogenic gene expression. Our study reveals a key role of the miR-182-5p/FGF21/acetylcholine/acetylcholine receptor axis that mediates the crosstalk between adipocytes and macrophages to promote beige fat thermogenesis. Activation of the miR-182-5p-induced signaling pathway in adipose tissue may be an effective approach to ameliorate obesity and associated metabolic diseases.


Asunto(s)
Acetilcolina/genética , Adipocitos/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Macrófagos/metabolismo , MicroARNs/genética , Obesidad/genética , Termogénesis/genética , Acetilcolina/biosíntesis , Adipocitos/patología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Modelos Animales de Enfermedad , Factores de Crecimiento de Fibroblastos/biosíntesis , Macrófagos/patología , Ratones , Ratones Noqueados , MicroARNs/biosíntesis , Obesidad/metabolismo , Obesidad/patología , Transducción de Señal
10.
Nat Commun ; 12(1): 326, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436607

RESUMEN

Adipose tissue-resident T cells have been recognized as a critical regulator of thermogenesis and energy expenditure, yet the underlying mechanisms remain unclear. Here, we show that high-fat diet (HFD) feeding greatly suppresses the expression of disulfide-bond A oxidoreductase-like protein (DsbA-L), a mitochondria-localized chaperone protein, in adipose-resident T cells, which correlates with reduced T cell mitochondrial function. T cell-specific knockout of DsbA-L enhances diet-induced thermogenesis in brown adipose tissue (BAT) and protects mice from HFD-induced obesity, hepatosteatosis, and insulin resistance. Mechanistically, DsbA-L deficiency in T cells reduces IFN-γ production and activates protein kinase A by reducing phosphodiesterase-4D expression, leading to increased BAT thermogenesis. Taken together, our study uncovers a mechanism by which T cells communicate with brown adipocytes to regulate BAT thermogenesis and whole-body energy homeostasis. Our findings highlight a therapeutic potential of targeting T cells for the treatment of over nutrition-induced obesity and its associated metabolic diseases.


Asunto(s)
Dieta Alta en Grasa , Glutatión Transferasa/deficiencia , Interferón gamma/biosíntesis , Linfocitos T/metabolismo , Termogénesis , Adipocitos Marrones/efectos de los fármacos , Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Regulación hacia Abajo/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Conducta Alimentaria , Glutatión Transferasa/metabolismo , Resistencia a la Insulina , Interferón gamma/administración & dosificación , Interferón gamma/farmacología , Masculino , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Obesidad/genética , Obesidad/patología , Linfocitos T/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo , Termogénesis/efectos de los fármacos , Termogénesis/genética , Proteína Desacopladora 1/metabolismo
11.
Commun Biol ; 3(1): 257, 2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32444826

RESUMEN

Obesity is a global epidemic that is caused by excessive energy intake or inefficient energy expenditure. Brown or beige fat dissipates energy as heat through non-shivering thermogenesis by their high density of mitochondria. However, how the mitochondrial stress-induced signal is coupled to the cellular thermogenic program remains elusive. Here, we show that mitochondrial DNA escape-induced activation of the cGAS-STING pathway negatively regulates thermogenesis in fat-specific DsbA-L knockout mice, a model of adipose tissue mitochondrial stress. Conversely, fat-specific overexpression of DsbA-L or knockout of STING protects mice against high-fat diet-induced obesity. Mechanistically, activation of the cGAS-STING pathway in adipocytes activated phosphodiesterase PDE3B/PDE4, leading to decreased cAMP levels and PKA signaling, thus reduced thermogenesis. Our study demonstrates that mitochondrial stress-activated cGAS-STING pathway functions as a sentinel signal that suppresses thermogenesis in adipose tissue. Targeting adipose cGAS-STING pathway may thus be a potential therapeutic strategy to counteract overnutrition-induced obesity and its associated metabolic diseases.


Asunto(s)
Glutatión Transferasa/fisiología , Proteínas de la Membrana/metabolismo , Mitocondrias/patología , Nucleotidiltransferasas/metabolismo , Obesidad/etiología , Hipernutrición/complicaciones , Termogénesis , Adipocitos/metabolismo , Adipocitos/patología , Animales , Dieta Alta en Grasa , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Nucleotidiltransferasas/genética , Obesidad/metabolismo , Obesidad/patología , Estrés Fisiológico
12.
Cells ; 8(10)2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31554182

RESUMEN

Adiponectin is an adipokine with anti-insulin resistance and anti-inflammatory functions. It exists in serum predominantly in three multimeric complexes: the trimer, hexamer, and high-molecular-weight forms. Although recent studies indicate that adiponectin promotes wound healing in rodents, its role in the wound healing process in humans is unknown. This study investigated the expression levels of adiponectin in adipose tissue and serum of women who experienced either normal or delayed wound healing after abdominal plastic surgery. We found that obese women with delayed healing had slightly lower total adiponectin levels in their adipose tissue compared with women with normal healing rates. Among the different isoforms of adiponectin, levels of the trimer forms were significantly reduced in adipose tissue, but not the serum, of obese women with delayed healing compared to women who healed normally. This study provides clinical evidence for a potential role of low-molecular-weight oligomers of adiponectin in the wound healing process as well as implications for an autocrine and/or paracrine mechanism of adiponectin action in adipose tissues.


Asunto(s)
Adiponectina/fisiología , Obesidad/fisiopatología , Cicatrización de Heridas/fisiología , Adiponectina/sangre , Adiponectina/genética , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Adulto , Anciano , Comunicación Autocrina/fisiología , Estudios de Casos y Controles , Femenino , Humanos , Persona de Mediana Edad , Obesidad/sangre , Obesidad/complicaciones , Obesidad/genética , Comunicación Paracrina/fisiología , Isoformas de Proteínas/sangre , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Factores de Tiempo , Adulto Joven
13.
J Mol Cell Biol ; 11(9): 781-790, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31220300

RESUMEN

Increasing brown and beige fat thermogenesis have an anti-obesity effect and thus great metabolic benefits. However, the molecular mechanisms regulating brown and beige fat thermogenesis remain to be further elucidated. We recently found that fat-specific knockout of Rheb promoted beige fat thermogenesis. In the current study, we show that Rheb has distinct effects on thermogenic gene expression in brown and beige fat. Fat-specific knockout of Rheb decreased protein kinase A (PKA) activity and thermogenic gene expression in brown adipose tissue of high-fat diet-fed mice. On the other hand, overexpression of Rheb activated PKA and increased uncoupling protein 1 expression in brown adipocytes. Mechanistically, Rheb overexpression in brown adipocytes increased Notch expression, leading to disassociation of the regulatory subunit from the catalytic subunit of PKA and subsequent PKA activation. Our study demonstrates that Rheb, by selectively modulating thermogenic gene expression in brown and beige adipose tissues, plays an important role in regulating energy homeostasis.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Termogénesis , Adipocitos/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Metabolismo Energético , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Modelos Biológicos , Obesidad/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro/genética , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
14.
Diabetes ; 68(6): 1099-1108, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31109939

RESUMEN

It has been appreciated for many years that there is a strong association between metabolism and immunity in advanced metazoan organisms. Distinct immune signatures and signaling pathways have been found not only in immune but also in metabolic cells. The newly discovered DNA-sensing cGAS-cGAMP-STING pathway mediates type I interferon inflammatory responses in immune cells to defend against viral and bacterial infections. Recent studies show that this pathway is also activated by host DNA aberrantly localized in the cytosol, contributing to increased sterile inflammation, insulin resistance, and the development of nonalcoholic fatty liver disease (NAFLD). Potential interactions of the cGAS-cGAMP-STING pathway with mTORC1 signaling, autophagy, and apoptosis have been reported, suggesting an important role of the cGAS-cGAMP-STING pathway in the networking and coordination of these important biological processes. However, the regulation, mechanism of action, and tissue-specific role of the cGAS-cGAMP-STING signaling pathway in metabolic disorders remain largely elusive. It is also unclear whether targeting this signaling pathway is effective for the prevention and treatment of obesity-induced metabolic diseases. Answers to these questions would provide new insights for developing effective therapeutic interventions for metabolic diseases such as insulin resistance, NAFLD, and type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/inmunología , Resistencia a la Insulina/inmunología , Proteínas de la Membrana/inmunología , Enfermedad del Hígado Graso no Alcohólico/inmunología , Nucleótidos Cíclicos/inmunología , Nucleotidiltransferasas/inmunología , Obesidad/inmunología , Apoptosis/inmunología , Autofagia/inmunología , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Inflamación , Interferón Tipo I/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas de la Membrana/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/metabolismo , Obesidad/metabolismo , Transducción de Señal
15.
Proc Natl Acad Sci U S A ; 114(46): 12196-12201, 2017 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-29087318

RESUMEN

Chronic inflammation in adipose tissue plays a key role in obesity-induced insulin resistance. However, the mechanisms underlying obesity-induced inflammation remain elusive. Here we show that obesity promotes mtDNA release into the cytosol, where it triggers inflammatory responses by activating the DNA-sensing cGAS-cGAMP-STING pathway. Fat-specific knockout of disulfide-bond A oxidoreductase-like protein (DsbA-L), a chaperone-like protein originally identified in the mitochondrial matrix, impaired mitochondrial function and promoted mtDNA release, leading to activation of the cGAS-cGAMP-STING pathway and inflammatory responses. Conversely, fat-specific overexpression of DsbA-L protected mice against high-fat diet-induced activation of the cGAS-cGAMP-STING pathway and inflammation. Taken together, we identify DsbA-L as a key molecule that maintains mitochondrial integrity. DsbA-L deficiency promotes inflammation and insulin resistance by activating the cGAS-cGAMP-STING pathway. Our study also reveals that, in addition to its well-characterized roles in innate immune surveillance, the cGAS-cGAMP-STING pathway plays an important role in mediating obesity-induced metabolic dysfunction.


Asunto(s)
ADN Mitocondrial/metabolismo , Glutatión Transferasa/genética , Resistencia a la Insulina , Proteínas de la Membrana/genética , Nucleotidiltransferasas/genética , Obesidad/genética , Células 3T3-L1 , Adipocitos/metabolismo , Adipocitos/patología , Animales , Dieta Alta en Grasa/efectos adversos , Regulación de la Expresión Génica , Glutatión Transferasa/deficiencia , Humanos , Inflamación , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Nucleotidiltransferasas/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Cultivo Primario de Células , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal
17.
Diabetes ; 66(5): 1198-1213, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28242620

RESUMEN

Beiging of white adipose tissue has potential antiobesity and antidiabetes effects, yet the underlying signaling mechanisms remain to be fully elucidated. Here we show that adipose-specific knockout of Rheb, an upstream activator of mechanistic target of rapamycin complex 1 (mTORC1), protects mice from high-fat diet-induced obesity and insulin resistance. On the one hand, Rheb deficiency in adipose tissue reduced mTORC1 signaling, increased lipolysis, and promoted beiging and energy expenditure. On the other hand, overexpression of Rheb in primary adipocytes significantly inhibited CREB phosphorylation and uncoupling protein 1 (UCP1) expression. Mechanistically, fat-specific knockout of Rheb increased cAMP levels, cAMP-dependent protein kinase (PKA) activity, and UCP1 expression in subcutaneous white adipose tissue. Interestingly, treating primary adipocytes with rapamycin only partially alleviated the suppressing effect of Rheb on UCP1 expression, suggesting the presence of a novel mechanism underlying the inhibitory effect of Rheb on thermogenic gene expression. Consistent with this notion, overexpression of Rheb stabilizes the expression of cAMP-specific phosphodiesterase 4D5 (PDE4D5) in adipocytes, whereas knockout of Rheb greatly reduced cellular levels of PDE4D5 concurrently with increased cAMP levels, PKA activation, and UCP1 expression. Taken together, our findings reveal Rheb as an important negative regulator of beige fat development and thermogenesis. In addition, Rheb is able to suppress the beiging effect through an mTORC1-independent mechanism.


Asunto(s)
Adipocitos/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Metabolismo Energético/genética , Resistencia a la Insulina/genética , Proteínas de Unión al GTP Monoméricas/genética , Neuropéptidos/genética , Obesidad/genética , Tejido Adiposo Beige/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Dieta Alta en Grasa , Regulación hacia Abajo , Regulación de la Expresión Génica , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Complejos Multiproteicos/metabolismo , Fosforilación , Proteína Homóloga de Ras Enriquecida en el Cerebro , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Proteína Desacopladora 1/genética
18.
FASEB J ; 31(6): 2314-2326, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28232481

RESUMEN

Hepatic insulin resistance and hepatosteatosis in diet-induced obesity are associated with various metabolic diseases, yet the underlying mechanisms remain to be fully elucidated. Here we show that the expression levels of the disulfide-bond A oxidoreductase-like protein (DsbA-L) are significantly reduced in the liver of obese mice and humans. Liver-specific knockout or adenovirus-mediated overexpression of DsbA-L exacerbates or alleviates, respectively, high-fat diet-induced mitochondrial dysfunction, hepatosteatosis, and insulin resistance in mice. Mechanistically, we found that DsbA-L is localized in mitochondria and that its deficiency is associated with impairment of maximum respiratory capacity, elevated cellular oxidative stress, and increased JNK activity. Our results identify DsbA-L as a critical regulator of mitochondrial function, and its down-regulation in the liver may contribute to obesity-induced hepatosteatosis and whole body insulin resistance.-Chen, H., Bai, J., Dong, F., Fang, H., Zhang, Y., Meng, W., Liu, B., Luo, Y., Liu, M., Bai, Y., Abdul-Ghani, M. A., Li, R., Wu, J., Zeng, R., Zhou, Z., Dong, L. Q., Liu, F. Hepatic DsbA-L protects mice from diet-induced hepatosteatosis and insulin resistance.


Asunto(s)
Grasas de la Dieta/efectos adversos , Hígado Graso/inducido químicamente , Glutatión Transferasa/metabolismo , Resistencia a la Insulina/fisiología , Hígado/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Regulación Enzimológica de la Expresión Génica , Técnica de Clampeo de la Glucosa , Glutatión Transferasa/genética , Hepatocitos , Humanos , Hígado/enzimología , Masculino , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/metabolismo , Estrés Oxidativo , Consumo de Oxígeno
19.
Cell Metab ; 19(6): 967-80, 2014 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-24746805

RESUMEN

Identification of key regulators of lipid metabolism and thermogenic functions has important therapeutic implications for the current obesity and diabetes epidemic. Here, we show that Grb10, a direct substrate of mechanistic/mammalian target of rapamycin (mTOR), is expressed highly in brown adipose tissue, and its expression in white adipose tissue is markedly induced by cold exposure. In adipocytes, mTOR-mediated phosphorylation at Ser501/503 switches the binding preference of Grb10 from the insulin receptor to raptor, leading to the dissociation of raptor from mTOR and downregulation of mTOR complex 1 (mTORC1) signaling. Fat-specific disruption of Grb10 increased mTORC1 signaling in adipose tissues, suppressed lipolysis, and reduced thermogenic function. The effects of Grb10 deficiency on lipolysis and thermogenesis were diminished by rapamycin administration in vivo. Our study has uncovered a unique feedback mechanism regulating mTORC1 signaling in adipose tissues and identified Grb10 as a key regulator of adiposity, thermogenesis, and energy expenditure.


Asunto(s)
Metabolismo Energético/fisiología , Proteína Adaptadora GRB10/metabolismo , Lipólisis/fisiología , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Termogénesis/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Células Cultivadas , Frío , Respuesta al Choque por Frío , Diabetes Mellitus , Retroalimentación Fisiológica , Proteína Adaptadora GRB10/biosíntesis , Resistencia a la Insulina , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Complejos Multiproteicos/antagonistas & inhibidores , Obesidad , Fosfatidilinositol 3-Quinasas , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-akt , Receptor de Insulina/metabolismo , Proteína Reguladora Asociada a mTOR , Transducción de Señal/fisiología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
20.
Environ Toxicol ; 25(3): 272-83, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19434695

RESUMEN

Sulfur dioxide (SO(2)) is a ubiquitous air pollutant that is present in low concentrations in the urban air, and in higher concentrations in the working environment. In the present study, male Wistar rats were housed in exposure chambers and treated with 14.00 +/- 1.01, 28.00 +/- 1.77 and 56.00 +/- 3.44 mg m(-3) SO(2) for 6 h/day for 7 days, while control group was exposed to filtered air in the same condition. The mRNA and protein levels of proto-oncogenes (c-fos, c-jun, c-myc, and Ki-ras) and tumor suppressor genes (p53, Rb, and p16) were analyzed in lungs using a real-time reverse transcription-polymerase chain reaction (real-time RT-PCR) assay and Western blot analysis. The results showed that mRNA and protein levels of c-fos, c-jun, c-myc, Ki-ras, and p53 in lungs were increased in a dose-dependent manner, while mRNA and protein levels of Rb and p16 were decreased in lungs of rats after SO(2) inhalation. These results lead to a conclusion that SO(2) exposure could activate expressions of proto-oncogenes and suppress expressions of tumor suppressor genes, which might relate to the molecular mechanism of cocarcinogenic properties and potential carcinogenic effects of SO(2). According to previous studies, the results also indicated that promoter genes of apoptosis and tumor suppressor genes could produce apoptotic signals to antagonize the growth signals that arise from oncogenes. Understanding its molecular controls will benefit development of treatments for many diseases.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Expresión Génica/efectos de los fármacos , Genes Supresores de Tumor , Exposición por Inhalación/efectos adversos , Pulmón/efectos de los fármacos , Proto-Oncogenes , Dióxido de Azufre/toxicidad , Animales , Western Blotting , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Pulmón/metabolismo , Masculino , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...