Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 6540, 2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39095402

RESUMO

Foam cells in atheroma are engorged with lipid droplets (LDs) that contain esters of regulatory lipids whose metabolism remains poorly understood. LD-associated hydrolase (LDAH) has a lipase structure and high affinity for LDs of foam cells. Using knockout and transgenic mice of both sexes, here we show that LDAH inhibits atherosclerosis development and promotes stable lesion architectures. Broad and targeted lipidomic analyzes of primary macrophages and comparative lipid profiling of atheroma identified a broad impact of LDAH on esterified sterols, including natural liver X receptor (LXR) sterol ligands. Transcriptomic analyzes coupled with rescue experiments show that LDAH modulates the expression of prototypical LXR targets and leads macrophages to a less inflammatory phenotype with a profibrotic gene signature. These studies underscore the role of LDs as reservoirs and metabolic hubs of bioactive lipids, and suggest that LDAH favorably modulates macrophage activation and protects against atherosclerosis via lipolytic mobilization of regulatory sterols.


Assuntos
Aterosclerose , Gotículas Lipídicas , Receptores X do Fígado , Macrófagos , Camundongos Knockout , Animais , Aterosclerose/metabolismo , Aterosclerose/genética , Aterosclerose/prevenção & controle , Aterosclerose/patologia , Receptores X do Fígado/metabolismo , Receptores X do Fígado/genética , Camundongos , Masculino , Ligantes , Feminino , Gotículas Lipídicas/metabolismo , Macrófagos/metabolismo , Esteróis/metabolismo , Células Espumosas/metabolismo , Camundongos Transgênicos , Camundongos Endogâmicos C57BL , Humanos , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Ativação de Macrófagos , Esterol Esterase
2.
Nat Commun ; 12(1): 5296, 2021 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-34489478

RESUMO

The vascular endothelium is present within metabolic organs and actively regulates energy metabolism. Here we show osteocalcin, recognized as a bone-secreted metabolic hormone, is expressed in mouse primary endothelial cells isolated from heart, lung and liver. In human osteocalcin promoter-driven green fluorescent protein transgenic mice, green fluorescent protein signals are enriched in endothelial cells lining aorta, small vessels and capillaries and abundant in aorta, skeletal muscle and eye of adult mice. The depletion of lipoprotein receptor-related protein 1 induces osteocalcin through a Forkhead box O -dependent pathway in endothelial cells. Whereas depletion of osteocalcin abolishes the glucose-lowering effect of low-density lipoprotein receptor-related protein 1 depletion, osteocalcin treatment normalizes hyperglycemia in multiple mouse models. Mechanistically, osteocalcin receptor-G protein-coupled receptor family C group 6 member A and insulin-like-growth-factor-1 receptor are in the same complex with osteocalcin and required for osteocalcin-promoted insulin signaling pathway. Therefore, our results reveal an endocrine/paracrine role of endothelial cells in regulating insulin sensitivity, which may have therapeutic implications in treating diabetes and insulin resistance through manipulating vascular endothelium.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Glucose/metabolismo , Hiperglicemia/genética , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Osteocalcina/genética , Animais , Células Endoteliais/patologia , Endotélio Vascular/patologia , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Teste de Tolerância a Glucose , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Hiperglicemia/metabolismo , Hiperglicemia/patologia , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Masculino , Camundongos , Camundongos Knockout , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteocalcina/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
3.
J Family Med Prim Care ; 10(5): 2038-2040, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34195146

RESUMO

Chorioangioma is the most common benign non trophoblastic tumor of the placenta. It is a rare presentation with incidence of 0.6-1% of all pregnancies. It is associated with feto maternal complications like polyhydramnios, cervical incompetence, preterm labor, increased rate of cesarean delivery, abruptio placentae, malpresentation, postpartum hemorrhage, fetal growth restriction, fetal anemia, fetal thrombocytopenia, non immune hydrops, fetal cardiac failure, cerebral embolism, cerebral infarction, intrauterine fetal and neonatal death. Ultrasound is the gold standard for diagnosis . Here we present a case of giant chorioangioma of 6 * 5 cm with complication of polyhydramnios, preterm labor, abruptio placenta and placenta previa successfully managed with good maternal and fetal outcome.

4.
J Clin Exp Hepatol ; 10(6): 636-640, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33311899

RESUMO

Ovaries are a common niche for metastasis. Metastatic malignancies account for 5-30% of all ovarian malignancies. Hepatocellular carcinoma (HCC) is one of the rare malignancies to metastasize to the ovaries. Of all the variants of HCC, fibrolamellar HCC (FLHCC) variant is extremely uncommon and accounts for around 1% of all HCC cases. FLHCC metastasizing to ovaries, at presentation, is an exceptional occurrence. We present a case of a young female who presented with bilateral adnexal masses and was diagnosed as metastatic FLHCC on histopathological examination and confirmed by immunohistochemistry. In addition, a thorough literature review highlighting the previously reported cases is also presented.

5.
JCI Insight ; 52019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31265437

RESUMO

Hormones produced by the anterior pituitary gland regulate an array of important physiological functions, but pituitary hormone disorders are not fully understood. Herein we report that genetically-engineered mice with deletion of the hedgehog signaling receptor Patched1 by S100a4 promoter-driven Cre recombinase (S100a4-Cre;Ptch1fl/fl mutants) exhibit adult-onset hypogonadotropic hypogonadism and multiple pituitary hormone disorders. During the transition from puberty to adult, S100a4-Cre;Ptch1fl/fl mice of both sexes develop hypogonadism coupled with reduced gonadotropin levels. Their pituitary glands also display severe structural and functional abnormalities, as revealed by transmission electron microscopy and expression of key genes regulating pituitary endocrine functions. S100a4-Cre activity in the anterior pituitary gland is restricted to CD45+ cells of hematopoietic origin, including folliculo-stellate cells and other immune cell types, causing sex-specific changes in the expression of genes regulating the local microenvironment of the anterior pituitary. These findings provide in vivo evidence for the importance of pituitary hematopoietic cells in regulating fertility and endocrine function, in particular during sexual maturation and likely through sexually dimorphic mechanisms. These findings support a previously unrecognized role of hematopoietic cells in causing hypogonadotropic hypogonadism and provide inroads into the molecular and cellular basis for pituitary hormone disorders in humans.


Assuntos
Hipogonadismo/metabolismo , Integrases/metabolismo , Receptor Patched-1/metabolismo , Hipófise/metabolismo , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Animais , Epididimo/patologia , Feminino , Humanos , Hipogonadismo/genética , Hipogonadismo/patologia , Masculino , Camundongos , Camundongos Knockout , Ovário/patologia , Receptor Patched-1/genética , Adeno-Hipófise/metabolismo , Reprodução/fisiologia , Glândulas Seminais/patologia , Maturidade Sexual , Transdução de Sinais , Testículo , Testosterona/sangue , Útero/patologia
6.
Nat Metab ; 1(1): 70-85, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-31198906

RESUMO

Specific metabolic underpinnings of androgen receptor (AR)-driven growth in prostate adenocarcinoma (PCa) are largely undefined, hindering the development of strategies to leverage the metabolic dependencies of this disease when hormonal manipulations fail. Here we show that the mitochondrial pyruvate carrier (MPC), a critical metabolic conduit linking cytosolic and mitochondrial metabolism, is transcriptionally regulated by AR. Experimental MPC inhibition restricts proliferation and metabolic outputs of the citric acid cycle (TCA) including lipogenesis and oxidative phosphorylation in AR-driven PCa models. Mechanistically, metabolic disruption resulting from MPC inhibition activates the eIF2α/ATF4 integrated stress response (ISR). ISR signaling prevents cell cycle progression while coordinating salvage efforts, chiefly enhanced glutamine assimilation into the TCA, to regain metabolic homeostasis. We confirm that MPC function is operant in PCa tumors in-vivo using isotopomeric metabolic flux analysis. In turn, we apply a clinically viable small molecule targeting the MPC, MSDC0160, to pre-clinical PCa models and find that MPC inhibition suppresses tumor growth in hormone-responsive and castrate-resistant conditions. Collectively, our findings characterize the MPC as a tractable therapeutic target in AR-driven prostate tumors.


Assuntos
Mitocôndrias/metabolismo , Neoplasias da Próstata/etiologia , Neoplasias da Próstata/metabolismo , Ácido Pirúvico/metabolismo , Receptores Androgênicos/metabolismo , Animais , Transporte Biológico , Linhagem Celular Tumoral , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Glutamina/metabolismo , Humanos , Masculino , Redes e Vias Metabólicas , Camundongos , Camundongos Transgênicos , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/patologia , Neoplasias de Próstata Resistentes à Castração/etiologia , Neoplasias de Próstata Resistentes à Castração/metabolismo , Neoplasias de Próstata Resistentes à Castração/patologia , Ligação Proteica , Transdução de Sinais
7.
Diabetes ; 67(12): 2541-2553, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30002134

RESUMO

Chronic inflammation accompanies obesity and limits subcutaneous white adipose tissue (WAT) expandability, accelerating the development of insulin resistance and type 2 diabetes mellitus. MicroRNAs (miRNAs) influence expression of many metabolic genes in fat cells, but physiological roles in WAT remain poorly characterized. Here, we report that expression of the miRNA miR-30a in subcutaneous WAT corresponds with insulin sensitivity in obese mice and humans. To examine the hypothesis that restoration of miR-30a expression in WAT improves insulin sensitivity, we injected adenovirus (Adv) expressing miR-30a into the subcutaneous fat pad of diabetic mice. Exogenous miR-30a expression in the subcutaneous WAT depot of obese mice coupled improved insulin sensitivity and increased energy expenditure with decreased ectopic fat deposition in the liver and reduced WAT inflammation. High-throughput proteomic profiling and RNA-Seq suggested that miR-30a targets the transcription factor STAT1 to limit the actions of the proinflammatory cytokine interferon-γ (IFN-γ) that would otherwise restrict WAT expansion and decrease insulin sensitivity. We further demonstrated that miR-30a opposes the actions of IFN-γ, suggesting an important role for miR-30a in defending adipocytes against proinflammatory cytokines that reduce peripheral insulin sensitivity. Together, our data identify a critical molecular signaling axis, elements of which are involved in uncoupling obesity from metabolic dysfunction.


Assuntos
Resistência à Insulina/fisiologia , Fígado/metabolismo , MicroRNAs/metabolismo , Obesidade/metabolismo , Gordura Subcutânea/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Metabolismo Energético/fisiologia , Camundongos , MicroRNAs/genética , Obesidade/etiologia , Obesidade/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo
8.
Nat Med ; 23(12): 1444-1453, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29106398

RESUMO

Asprosin is a recently discovered fasting-induced hormone that promotes hepatic glucose production. Here we demonstrate that asprosin in the circulation crosses the blood-brain barrier and directly activates orexigenic AgRP+ neurons via a cAMP-dependent pathway. This signaling results in inhibition of downstream anorexigenic proopiomelanocortin (POMC)-positive neurons in a GABA-dependent manner, which then leads to appetite stimulation and a drive to accumulate adiposity and body weight. In humans, a genetic deficiency in asprosin causes a syndrome characterized by low appetite and extreme leanness; this is phenocopied by mice carrying similar mutations and can be fully rescued by asprosin. Furthermore, we found that obese humans and mice had pathologically elevated concentrations of circulating asprosin, and neutralization of asprosin in the blood with a monoclonal antibody reduced appetite and body weight in obese mice, in addition to improving their glycemic profile. Thus, in addition to performing a glucogenic function, asprosin is a centrally acting orexigenic hormone that is a potential therapeutic target in the treatment of both obesity and diabetes.


Assuntos
Regulação do Apetite/genética , Hipotálamo/metabolismo , Proteínas dos Microfilamentos/fisiologia , Fragmentos de Peptídeos/fisiologia , Hormônios Peptídicos/fisiologia , Adolescente , Adulto , Animais , Depressores do Apetite/metabolismo , Feminino , Fibrilina-1 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Neurônios/metabolismo , Fragmentos de Peptídeos/genética , Hormônios Peptídicos/genética , Ratos , Transdução de Sinais , Adulto Jovem
9.
Hepatology ; 66(2): 498-509, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28378930

RESUMO

The nuclear receptors farnesoid X receptor (FXR; NR1H4) and small heterodimer partner (SHP; NR0B2) play crucial roles in bile acid homeostasis. Global double knockout of FXR and SHP signaling (DKO) causes severe cholestasis and liver injury at early ages. Here, we report an unexpected beneficial impact on glucose and fatty acid metabolism in aged DKO mice, which show suppressed body weight gain and adiposity when maintained on normal chow. This phenotype was not observed in single Fxr or Shp knockouts. Liver-specific Fxr/Shp double knockout mice fully phenocopied the DKO mice, with lower hepatic triglyceride accumulation, improved glucose/insulin tolerance, and accelerated fatty acid use. In both DKO and liver-specific Fxr/Shp double knockout livers, these metabolic phenotypes were associated with altered expression of fatty acid metabolism and autophagy-machinery genes. Loss of the hepatic FXR/SHP axis reprogrammed white and brown adipose tissue gene expression to boost fatty acid usage. CONCLUSION: Combined deletion of the hepatic FXR/SHP axis improves glucose/fatty acid homeostasis in aged mice, reversing the aging phenotype of body weight gain, increased adiposity, and glucose/insulin tolerance, suggesting a central role of this axis in whole-body energy homeostasis. (Hepatology 2017;66:498-509).


Assuntos
Ácidos Graxos/metabolismo , Deleção de Genes , Regulação da Expressão Gênica , Homeostase/genética , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , Receptores Citoplasmáticos e Nucleares/genética , Envelhecimento/genética , Análise de Variância , Animais , Autofagia/genética , Células Cultivadas , Modelos Animais de Doenças , Glucose/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distribuição Aleatória
10.
Oman Med J ; 32(1): 73-76, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28042408

RESUMO

Pyomyoma is an unusual complication of leiomyoma, occurring most commonly in the postpartum, postabortal, and post-menopausal periods. It develops due to infection in necrotic foci within leiomyoma, which is more common during pregnancy due to rapid growth, and in postmenopausal women due to vascular insufficiency. Other contributing factors are curettage, gynecological surgery, cervical stenosis, immunodeficiency, and recently, uterine artery embolization. It presents with a typical triad of sepsis, leiomyoma, and absence of any apparent source of infection. We report a case of persistent postabortal fever in a 26-year-old female due to a pyomyoma, which resolved after a myomectomy. Pyomyoma may become life threatening in the event of intraperitoneal rupture resulting in pyoperitoneum and septic shock. Hence, gynecologists should consider this diagnosis in women with a leiomyoma and sepsis in the absence of any apparent source of infection.

11.
Cell ; 165(3): 566-79, 2016 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-27087445

RESUMO

Hepatic glucose release into the circulation is vital for brain function and survival during periods of fasting and is modulated by an array of hormones that precisely regulate plasma glucose levels. We have identified a fasting-induced protein hormone that modulates hepatic glucose release. It is the C-terminal cleavage product of profibrillin, and we name it Asprosin. Asprosin is secreted by white adipose, circulates at nanomolar levels, and is recruited to the liver, where it activates the G protein-cAMP-PKA pathway, resulting in rapid glucose release into the circulation. Humans and mice with insulin resistance show pathologically elevated plasma asprosin, and its loss of function via immunologic or genetic means has a profound glucose- and insulin-lowering effect secondary to reduced hepatic glucose release. Asprosin represents a glucogenic protein hormone, and therapeutically targeting it may be beneficial in type II diabetes and metabolic syndrome.


Assuntos
Jejum/metabolismo , Proteínas dos Microfilamentos/metabolismo , Fragmentos de Peptídeos/metabolismo , Hormônios Peptídicos/metabolismo , Tecido Adiposo Branco/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos/administração & dosagem , Ritmo Circadiano , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Jejum/sangue , Feminino , Retardo do Crescimento Fetal/metabolismo , Fibrilina-1 , Glucose/metabolismo , Humanos , Insulina/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Proteínas dos Microfilamentos/sangue , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/genética , Dados de Sequência Molecular , Fragmentos de Peptídeos/sangue , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Hormônios Peptídicos/sangue , Hormônios Peptídicos/química , Hormônios Peptídicos/genética , Progéria/metabolismo , Proteínas Recombinantes/administração & dosagem , Alinhamento de Sequência
12.
Cardiovasc Res ; 109(2): 294-304, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26487692

RESUMO

AIMS: Atherosclerosis development can be ameliorated by promoting reverse cholesterol transport (RCT) from arteries. The process involves cholesterol efflux from foam cells to extracellular acceptors such as apolipoprotein A-I (apoA-I) and high-density lipoprotein (HDL) that mediate transport to the liver. Perilipin-2 (PLIN2) is a lipid droplet (LD)-associated protein that in macrophages facilitates cholesterol storage and prevents efflux. We hypothesized that atheroprotection would be enhanced by concurrently targeting PLIN2 to increase the efflux capacity of foam cells and increasing plasma apoA-I and HDL. METHODS AND RESULTS: PLIN2-knockout and wild-type mice lacking apolipoprotein E (PLIN2(-/-)/apoE(-/-) and PLIN2(+/+)/apoE(-/-)) were treated with a helper-dependent adenoviral vector encoding human apoA-I (HDAd-AI) or with control empty vector. Treatment with HDAd-AI increased hepatic apoA-I production, plasma apoA-I and HDL-cholesterol (HDL-C), and apoA-I deposition in lesions to a similar extent in PLIN2(-/-)/apoE(-/-) and PLIN2(+/+)/apoE(-/-) mice. However, atherosclerosis development at the aortic sinus was considerably lower in HDAd-AI-treated PLIN2(-/-)/apoE(-/-) mice. A more stable lesion phenotype, with increased collagen content, was primarily associated to treatment with HDAd-AI, but was enhanced under PLIN2 deficiency. PLIN2 deficiency and apoA-I cumulatively reduced LDs and cholesterol ester content in cultured macrophages. Neutral lipid in atheroma was significantly reduced in HDAd-AI-treated PLIN2(-/-)/apoE(-/-) mice, and RCT from macrophages to feces was enhanced in PLIN2(-/-) macrophages. CONCLUSION: These studies demonstrate a mutually beneficial relationship between PLIN2 deficiency and elevated apoA-I/HDL-C in preventing atherosclerosis development. The data support that targeting foam cell components to mobilize cholesterol may be a promising strategy to enhance the atheroprotection of plasma cholesterol acceptors.


Assuntos
Aterosclerose/metabolismo , Colesterol/metabolismo , Células Espumosas/metabolismo , Perilipina-2/metabolismo , Animais , Apolipoproteína A-I/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerose/genética , Transporte Biológico/genética , Transporte Biológico/fisiologia , Ésteres do Colesterol/genética , Ésteres do Colesterol/metabolismo , Lipoproteínas HDL/metabolismo , Macrófagos/metabolismo , Camundongos Knockout , Perilipina-2/genética
13.
Nat Commun ; 6: 7339, 2015 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-26081744

RESUMO

Diseases of lipid metabolism are a major cause of human morbidity, but no animal model entirely recapitulates human lipoprotein metabolism. Here we develop a xenograft mouse model using hepatocytes from a patient with familial hypercholesterolaemia caused by loss-of-function mutations in the low-density lipoprotein receptor (LDLR). Like familial hypercholesterolaemia patients, our familial hypercholesterolaemia liver chimeric mice develop hypercholesterolaemia and a 'humanized' serum profile, including expression of the emerging drug targets cholesteryl ester transfer protein and apolipoprotein (a), for which no genes exist in mice. We go on to replace the missing LDLR in familial hypercholesterolaemia liver chimeric mice using an adeno-associated virus 9-based gene therapy and restore normal lipoprotein profiles after administration of a single dose. Our study marks the first time a human metabolic disease is induced in an experimental animal model by human hepatocyte transplantation and treated by gene therapy. Such xenograft platforms offer the ability to validate human experimental therapies and may foster their rapid translation into the clinic.


Assuntos
Modelos Animais de Doenças , Terapia Genética , Hiperlipoproteinemia Tipo II/terapia , Receptores de LDL/genética , Animais , Criança , Dependovirus , Feminino , Hepatócitos/transplante , Xenoenxertos , Humanos , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/metabolismo , Lipoproteínas/metabolismo
14.
Nat Cell Biol ; 15(6): 647-58, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23604321

RESUMO

The lysosomal-autophagic pathway is activated by starvation and plays an important role in both cellular clearance and lipid catabolism. However, the transcriptional regulation of this pathway in response to metabolic cues is uncharacterized. Here we show that the transcription factor EB (TFEB), a master regulator of lysosomal biogenesis and autophagy, is induced by starvation through an autoregulatory feedback loop and exerts a global transcriptional control on lipid catabolism via Ppargc1α and Ppar1α. Thus, during starvation a transcriptional mechanism links the autophagic pathway to cellular energy metabolism. The conservation of this mechanism in Caenorhabditis elegans suggests a fundamental role for TFEB in the evolution of the adaptive response to food deprivation. Viral delivery of TFEB to the liver prevented weight gain and metabolic syndrome in both diet-induced and genetic mouse models of obesity, suggesting a new therapeutic strategy for disorders of lipid metabolism.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Metabolismo dos Lipídeos , Síndrome Metabólica/metabolismo , Obesidade/metabolismo , Inanição/metabolismo , Animais , Autofagia/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/farmacologia , Caenorhabditis elegans/metabolismo , Linhagem Celular Tumoral , Metabolismo Energético , Retroalimentação Fisiológica , Regulação da Expressão Gênica , Células HeLa , Homeostase , Humanos , Fígado/metabolismo , Lisossomos/genética , Masculino , Síndrome Metabólica/genética , Síndrome Metabólica/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Obesidade/genética , PPAR alfa/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Inanição/genética , Transativadores/metabolismo , Fatores de Transcrição , Transcrição Gênica , Aumento de Peso
15.
J Biol Chem ; 285(52): 40581-92, 2010 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-20956520

RESUMO

The triterpenoid 2-Cyano-3,12-dioxooleana-1,9-dien-28-oic-acid (CDDO) and its methyl ester (CDDO-Me) are undergoing clinical trials in cancer and leukemia therapy. Here we report that CDDO-Me ameliorates diabetes in high fat diet-fed type 2 diabetic mice and in Lepr(db/db) mice. CDDO-Me reduces proinflammatory cytokine expression in these animals. Oral CDDO-Me administration reduces total body fat, plasma triglyceride, and free fatty acid levels. It also improves glucose tolerance and insulin tolerance tests. Its potent glucose-lowering activity results from enhanced insulin action. Hyperinsulinemic-euglycemic clamp reveals an increased glucose infusion rate required to maintain euglycemia and showed a significant increase in muscle-specific insulin-stimulated glucose uptake (71% soleus, 58% gastrocnemius) and peripheral glucose clearance as documented by a 48% increase in glucose disposal rate. CDDO-Me activates AMP-activated protein kinase (AMPK) and via LKB1 activation in muscle and liver in vivo. Treatment of isolated hepatocytes with CDDO-Me directly stimulates AMPK activity and LKB1 phosphorylation and decreases acetyl-coA carboxylase activity; it also down-regulates lipogenic gene expression, suppresses gluconeogenesis, and increases glucose uptake. Inhibition of AMPK phosphorylation using compound C and lentiviral-mediated knockdown of AMPK completely blocks the CDDO-Me-induced effect on hepatocytes as well as C(2)C(12) cells. We conclude that the triterpenoid CDDO-Me has potent anti-diabetic action in diabetic mouse models that is mediated at least in part through AMPK activation. The in vivo anti-diabetogenic effects occur at a dose substantially lower than that used for anti-leukemia therapy. We suggest that CDDO-Me holds promise as a potential anti-diabetic agent.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Dieta/efeitos adversos , Hipoglicemiantes/farmacologia , Ácido Oleanólico/análogos & derivados , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Acetil-CoA Carboxilase/genética , Acetil-CoA Carboxilase/metabolismo , Tecido Adiposo/metabolismo , Animais , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/genética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Ácidos Graxos não Esterificados/sangue , Glucose/metabolismo , Hipoglicemiantes/uso terapêutico , Insulina/sangue , Camundongos , Camundongos Mutantes , Músculo Esquelético/metabolismo , Ácido Oleanólico/farmacologia , Ácido Oleanólico/uso terapêutico , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteínas Serina-Treonina Quinases/metabolismo , Triglicerídeos/sangue
16.
Hepatology ; 46(1): 147-57, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17526026

RESUMO

UNLABELLED: The orphan receptor Small Heterodimer Partner (SHP, NROB2) regulates metabolic pathways, including hepatic bile acid, lipid, and glucose homeostasis. We reported that SHP-deletion in leptin-deficient OB(-/-) mice increases insulin sensitivity, and prevents the development of fatty liver. The prevention of steatosis in OB(-/-)/SHP(-/-) double mutants is not due to decreased body weight but is associated with increased hepatic very-low-density lipoprotein (VLDL) secretion and elevated microsomal triglyceride transfer protein (MTP) mRNA and protein levels. SHP represses the transactivation of the MTP promoter and the induction of MTP mRNA by LRH-1 in hepatocytes. Adenoviral overexpression of SHP inhibits MTP activity as well as VLDL-apoB protein secretion, and RNAi knockdown of SHP exhibits opposite effects. The expression of SHP in induced in fatty livers of OB(-/-) mice and other genetic or dietary models of steatosis, and acute overexpression of SHP by adenovirus, result in rapid accumulation of neutral lipids in hepatocytes. In addition, the pathways for hepatic lipid uptake and lipogenic program are also downregulated in OB(-/-)/SHP(-/-) mice, which may contribute to the decreased hepatic lipid content. CONCLUSION: These studies demonstrate that SHP regulates the development of fatty liver by modulating hepatic lipid export, uptake, and synthesis, and that the improved peripheral insulin sensitivity in OB(-/-)/SHP(-/-) mice is associated with decreased hepatic steatosis.


Assuntos
Fígado Gorduroso/genética , Lipídeos/fisiologia , Receptores Citoplasmáticos e Nucleares/genética , Tecido Adiposo Marrom/fisiologia , Animais , Transporte Biológico , Células Cultivadas , Fígado Gorduroso/fisiopatologia , Deleção de Genes , Hepatócitos/fisiologia , Leptina/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , RNA/genética , RNA/isolamento & purificação
17.
Cell Metab ; 3(5): 379-86, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16679295

RESUMO

Ghrelin and leptin are suggested to regulate energy homeostasis as mutual antagonists on hypothalamic neurons that regulate feeding behavior. We employed reverse genetics to investigate the interplay between ghrelin and leptin. Leptin-deficient mice (ob/ob) are hyperphagic, obese, and hyperglycemic. Unexpectedly, ablation of ghrelin in ob/ob mice fails to rescue the obese hyperphagic phenotype, indicating that the ob/ob phenotype is not a consequence of ghrelin unopposed by leptin. Remarkably, deletion of ghrelin augments insulin secretion in response to glucose challenge and increases peripheral insulin sensitivity; indeed, the hyperglycemia exhibited by ob/ob mice is markedly reduced when ob/ob mice are bred onto the ghrelin(-/-) background. We further demonstrate that ablation of ghrelin reduces expression of Ucp2 mRNA in the pancreas, which contributes toward enhanced glucose-induced insulin secretion. Hence, chronically, ghrelin controls glucose homeostasis by regulating pancreatic Ucp2 expression and insulin sensitivity.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Leptina/fisiologia , Obesidade/metabolismo , Hormônios Peptídicos/fisiologia , Animais , Glicemia/metabolismo , Regulação da Temperatura Corporal , Peso Corporal , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/fisiopatologia , Regulação para Baixo , Grelina , Insulina/sangue , Canais Iônicos/genética , Canais Iônicos/metabolismo , Leptina/genética , Leptina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Obesidade/sangue , Obesidade/fisiopatologia , Pâncreas/metabolismo , Hormônios Peptídicos/genética , Hormônios Peptídicos/metabolismo , Fenótipo , RNA Mensageiro/metabolismo , Proteína Desacopladora 2
18.
Nat Med ; 10(6): 625-32, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15133506

RESUMO

Obesity is an increasingly prevalent human condition in developed societies. Despite major progress in the understanding of the molecular mechanisms leading to obesity, no safe and effective treatment has yet been found. Here, we report an antiobesity therapy based on targeted induction of apoptosis in the vasculature of adipose tissue. We used in vivo phage display to isolate a peptide motif (sequence CKGGRAKDC) that homes to white fat vasculature. We show that the CKGGRAKDC peptide associates with prohibitin, a multifunctional membrane protein, and establish prohibitin as a vascular marker of adipose tissue. Targeting a proapoptotic peptide to prohibitin in the adipose vasculature caused ablation of white fat. Resorption of established white adipose tissue and normalization of metabolism resulted in rapid obesity reversal without detectable adverse effects. Because prohibitin is also expressed in blood vessels of human white fat, this work may lead to the development of targeted drugs for treatment of obese patients.


Assuntos
Tecido Adiposo/irrigação sanguínea , Apoptose/fisiologia , Vasos Sanguíneos/metabolismo , Obesidade/terapia , Peptídeos/uso terapêutico , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Sequência de Aminoácidos , Animais , Dieta , Metabolismo Energético , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Biblioteca de Peptídeos , Peptídeos/metabolismo , Proibitinas , Proteínas Repressoras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA