Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 165(3): 566-79, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27087445

RESUMEN

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.


Asunto(s)
Ayuno/metabolismo , Proteínas de Microfilamentos/metabolismo , Fragmentos de Péptidos/metabolismo , Hormonas Peptídicas/metabolismo , Tejido Adiposo Blanco/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos/administración & dosificación , Ritmo Circadiano , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ayuno/sangre , Femenino , Retardo del Crecimiento Fetal/metabolismo , Fibrilina-1 , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Proteínas de Microfilamentos/sangre , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Datos de Secuencia Molecular , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Hormonas Peptídicas/sangre , Hormonas Peptídicas/química , Hormonas Peptídicas/genética , Progeria/metabolismo , Proteínas Recombinantes/administración & dosificación , Alineación de Secuencia
2.
J Biol Chem ; 299(9): 105185, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37611830

RESUMEN

A substantial body of evidence has established the contributions of both mitochondrial dynamics and lipid metabolism to the pathogenesis of diabetic kidney disease (DKD). However, the precise interplay between these two key metabolic regulators of DKD is not fully understood. Here, we uncover a link between mitochondrial dynamics and lipid metabolism by investigating the role of carbohydrate-response element-binding protein (ChREBP), a glucose-responsive transcription factor and a master regulator of lipogenesis, in kidney podocytes. We find that inducible podocyte-specific knockdown of ChREBP in diabetic db/db mice improves key biochemical and histological features of DKD in addition to significantly reducing mitochondrial fragmentation. Because of the critical role of ChREBP in lipid metabolism, we interrogated whether and how mitochondrial lipidomes play a role in ChREBP-mediated mitochondrial fission. Our findings suggest a key role for a family of ether phospholipids in ChREBP-induced mitochondrial remodeling. We find that overexpression of glyceronephosphate O-acyltransferase, a critical enzyme in the biosynthesis of plasmalogens, reverses the protective phenotype of ChREBP deficiency on mitochondrial fragmentation. Finally, our data also points to Gnpat as a direct transcriptional target of ChREBP. Taken together, our results uncover a distinct mitochondrial lipid signature as the link between ChREBP-induced mitochondrial dynamics and progression of DKD.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Animales , Ratones , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Diabetes Mellitus/metabolismo , Nefropatías Diabéticas/metabolismo , Regulación de la Expresión Génica , Riñón/metabolismo , Lipidómica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Am J Physiol Endocrinol Metab ; 319(4): E667-E677, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32799658

RESUMEN

MicroRNA-30a (miR-30a) impacts adipocyte function, and its expression in white adipose tissue (WAT) correlates with insulin sensitivity in obesity. Bioinformatic analysis demonstrates that miR-30a expression contributes to 2% of all miRNA expression in human tissues. However, molecular mechanisms of miR-30a function in fat cells remain unclear. Here, we expanded our understanding of how miR-30a expression contributes to antidiabetic peroxisome proliferator-activated receptor-γ (PPARγ) agonist activity and metabolic functions in adipocytes. We found that WAT isolated from diabetic patients shows reduced miR-30a levels and diminished expression of the canonical PPARγ target genes ADIPOQ and FABP4 relative to lean counterparts. In human adipocytes, miR-30a required PPARγ for maximal expression, and the PPARγ agonist rosiglitazone robustly induced miR-30a but not other miR-30 family members. Transcriptional activity studies in human adipocytes also revealed that ectopic expression of miR-30a enhanced the activity of rosiglitazone coupled with higher expression of fatty acid and glucose metabolism markers. Diabetic mice that overexpress ectopic miR-30a in subcutaneous WAT display durable reductions in serum glucose and insulin levels for more than 30 days. In agreement with our in vitro findings, RNA-seq coupled with Gene Set Enrichment Analysis (GSEA) suggested that miR-30a enabled activation of the beige fat program in vivo, as evidenced by enhanced mitochondrial biogenesis and induction of UCP1 expression. Metabolomic and gene expression profiling established that the long-term effects of ectopic miR-30a expression enable accelerated glucose metabolism coupled with subcutaneous WAT hyperplasia. Together, we establish a putative role of miR-30a in mediating PPARγ activity and advancing metabolic programs of white to beige fat conversion.


Asunto(s)
Adipocitos Marrones/fisiología , Redes Reguladoras de Genes/genética , MicroARNs/fisiología , Adipocitos Blancos/metabolismo , Animales , Glucemia/metabolismo , Células Cultivadas , Proteínas de Unión a Ácidos Grasos/metabolismo , Humanos , Hipoglucemiantes/farmacología , Resistencia a la Insulina/genética , Metabolómica , Ratones , MicroARNs/genética , Oligopéptidos/metabolismo , Biogénesis de Organelos , PPAR gamma/agonistas , Rosiglitazona/farmacología
4.
Hum Mol Genet ; 25(14): 3029-3041, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27288453

RESUMEN

Rett syndrome (RTT; OMIM 312750), a progressive neurological disorder, is caused by mutations in methyl-CpG-binding protein 2 (MECP2; OMIM 300005), a ubiquitously expressed factor. A genetic suppressor screen designed to identify therapeutic targets surprisingly revealed that downregulation of the cholesterol biosynthesis pathway improves neurological phenotypes in Mecp2 mutant mice. Here, we show that MeCP2 plays a direct role in regulating lipid metabolism. Mecp2 deletion in mice results in a host of severe metabolic defects caused by lipid accumulation, including insulin resistance, fatty liver, perturbed energy utilization, and adipose inflammation by macrophage infiltration. We show that MeCP2 regulates lipid homeostasis by anchoring the repressor complex containing NCoR1 and HDAC3 to its lipogenesis targets in hepatocytes. Consistently, we find that liver targeted deletion of Mecp2 causes fatty liver disease and dyslipidemia similar to HDAC3 liver-specific deletion. These findings position MeCP2 as a novel component in metabolic homeostasis. Rett syndrome patients also show signs of peripheral dyslipidemia; thus, together these data suggest that RTT should be classified as a neurological disorder with systemic metabolic components. We previously showed that treatment of Mecp2 mice with statin drugs alleviated motor symptoms and improved health and longevity. Lipid metabolism is a highly treatable target; therefore, our results shed light on new metabolic pathways for treatment of Rett syndrome.


Asunto(s)
Metabolismo de los Lípidos/genética , Proteína 2 de Unión a Metil-CpG/genética , Síndrome de Rett/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Histona Desacetilasas/genética , Resistencia a la Insulina/genética , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Mutación , Co-Represor 1 de Receptor Nuclear/genética , Síndrome de Rett/tratamiento farmacológico , Síndrome de Rett/metabolismo , Síndrome de Rett/patología , Eliminación de Secuencia
5.
Hepatology ; 66(2): 498-509, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28378930

RESUMEN

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).


Asunto(s)
Ácidos Grasos/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica , Homeostasis/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Receptores Citoplasmáticos y Nucleares/genética , Envejecimiento/genética , Análisis de Varianza , Animales , Autofagia/genética , Células Cultivadas , Modelos Animales de Enfermedad , Glucosa/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Aleatoria
6.
Nat Commun ; 15(1): 6540, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39095402

RESUMEN

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.


Asunto(s)
Aterosclerosis , Gotas Lipídicas , Receptores X del Hígado , Macrófagos , Ratones Noqueados , Animales , Aterosclerosis/metabolismo , Aterosclerosis/genética , Aterosclerosis/prevención & control , Aterosclerosis/patología , Receptores X del Hígado/metabolismo , Receptores X del Hígado/genética , Ratones , Masculino , Ligandos , Femenino , Gotas Lipídicas/metabolismo , Macrófagos/metabolismo , Esteroles/metabolismo , Células Espumosas/metabolismo , Ratones Transgénicos , Ratones Endogámicos C57BL , Humanos , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Activación de Macrófagos , Esterol Esterasa
7.
Res Sq ; 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38260478

RESUMEN

N-acetylaspartate (NAA), the brain's second most abundant metabolite, provides essential substrates for myelination through its hydrolysis. However, activities and physiological roles of NAA in other tissues remain unknown. Here, we show aspartoacylase (ASPA) expression in white adipose tissue (WAT) governs systemic NAA levels for postprandial body temperature regulation. Proteomics and mass spectrometry revealed NAA accumulation in WAT of Aspa knockout mice stimulated the pentose phosphate pathway and pyrimidine production. Stable isotope tracing confirmed higher incorporation of glucose-derived carbon into pyrimidine metabolites in Aspa knockout cells. Additionally, serum NAA positively correlates with the pyrimidine intermediate orotidine and this relationship predicted lower body mass index in humans. Using whole-body and tissue-specific knockout mouse models, we demonstrate that fat cells provided plasma NAA and suppressed postprandial body temperature elevation. Furthermore, exogenous NAA supplementation reduced body temperature. Our study unveils WAT-derived NAA as an endocrine regulator of postprandial body temperature and physiological homeostasis.

8.
Cytokine ; 63(1): 43-51, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23643185

RESUMEN

Selective recruitment of IFN-γ biased Th1 effector cells at the pathologic site(s) determines the local immunity of tuberculosis (TB). We observed the enrichment of CXCR3, CCR5 and CD11a(high) T cells in the peripheral blood, pleural fluid and bronchoalveolar lavage of TB pleural effusion (TB-PE) and miliary tuberculosis (MTB) patients respectively. CXCR3(+)CCR5(+) T cells were significantly high at the local disease site(s) in both the forms of TB and their frequency was highest among activated lymphocytes in TB-PE. Interestingly, all CCR5(+) cells were invariably positive for CXCR3 but all CXCR3(+) cells did not co-express CCR5 in pleural fluid whereas the situation was reverse in bronchoalveolar lavage. These CXCR3(+)CCR5(+) cells dominantly produced IFN-γ in response to Mycobacterium tuberculosis antigen. In vitro chemotaxis assay indicates dominant role of RANTES and IP-10 in the selective recruitment of CXCR3(+)CCR5(+)cells at the tubercular pathologic sites.


Asunto(s)
Receptores CCR3/metabolismo , Receptores CCR5/metabolismo , Células TH1/metabolismo , Tuberculosis/inmunología , Adulto , Antígeno CD11a/metabolismo , Complejo CD3/metabolismo , Movimiento Celular/efectos de los fármacos , Quimiocinas/farmacología , Demografía , Femenino , Humanos , Interferón gamma/metabolismo , Interleucina-4/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Ligandos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Masculino , Derrame Pleural/inmunología , Derrame Pleural/patología , Tuberculosis/patología , Tuberculosis Miliar/inmunología , Tuberculosis Miliar/patología
9.
Proc Natl Acad Sci U S A ; 106(44): 18831-6, 2009 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-19850873

RESUMEN

Constitutive androstane receptor CAR (NR1I3) has been identified as a central mediator of coordinate responses to xenobiotic and endobiotic stress. Here we use leptin-deficient mice (ob/ob) and ob/ob, CAR(-/-) double mutant mice to identify a metabolic role of CAR in type 2 diabetes. Activation of CAR significantly reduces serum glucose levels and improves glucose tolerance and insulin sensitivity. Gene expression analyses and hyperinsulinemic euglycemic clamp results suggest that CAR activation ameliorates hyperglycemia by suppressing glucose production and stimulating glucose uptake and usage in the liver. In addition, CAR activation dramatically improves fatty liver by both inhibition of hepatic lipogenesis and induction of beta-oxidation. We conclude that CAR activation improves type 2 diabetes, and that these actions of CAR suggest therapeutic approaches to the disease.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/prevención & control , Hígado Graso/complicaciones , Hígado Graso/prevención & control , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Receptor de Androstano Constitutivo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/patología , Hígado Graso/sangre , Hígado Graso/patología , Regulación de la Expresión Génica/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Insulina/farmacología , Lipogénesis/efectos de los fármacos , Lipogénesis/genética , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Ratones , Ratones Obesos , Oxidación-Reducción/efectos de los fármacos , Sulfotransferasas/metabolismo
10.
J Health Popul Nutr ; 30(4): 404-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23304906

RESUMEN

Lead poisoning is a major public-health problem in Bangladesh. A cross-sectional study was conducted to determine the extent of and risk factors for elevated blood lead levels (BLLs) in children in Bangladesh during September 2007-July 2009. The study included 919 children aged less than 16 years. The children were recruited from six urban locations in Dhaka and one rural area in Chirirbandar, Dinajpur. In total, 495 (54%) children had high BLLs (> 10 microg/dL), with higher BLLs observed among children aged 5-9 years compared to children of other ages (p < 0.001). The BLLs among children in urban Dhaka were significantly higher than those in rural areas (13.45 +/- 8.21 microg/dL vs 7.29 +/- 6.25 microg/dL, p < 0.001). The high BLLs correlated with low body mass index (r = -0.23, p < 0.001) and low haemoglobin status (r = -0.10, p = 0.02). On bivariate analysis, proximity to industry (p < 0.001), drinking-water from municipal supply or tubewell (p < 0.001), brass or lead water-taps (p < 0.001), use of melamine plate (p = 0.001), and indigenous medicinal (kabiraji) treatments (p = 0.004) significantly correlated with higher BLLs. Proximity to industry and the use of indigenous medicines remained significant predictors of high BLLs after controlling for the confounders. Several risk factors appropriate for future educational interventions to prevent exposure to lead poisoning were identified.


Asunto(s)
Intoxicación por Plomo/epidemiología , Adolescente , Bangladesh/epidemiología , Niño , Preescolar , Estudios Transversales , Humanos , Lactante , Intoxicación por Plomo/sangre , Intoxicación por Plomo/prevención & control , Análisis Multivariante , Prevalencia , Factores de Riesgo , Población Rural , Población Urbana
11.
PLoS One ; 17(12): e0279041, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36520818

RESUMEN

Gestational diabetes mellitus (GDM) affects 7-18% of all pregnancies. Despite its high prevalence, there is no widely accepted animal model. To address this, we recently developed a mouse model of GDM. The goal of this work was to further characterize this animal model by assessing insulin resistance and beta cell function. Mice were randomly assigned to either control (CD) or high fat, high sugar (HFHS) diet and mated 1 week later. At day 0 (day of mating) mice were fasted and intraperitoneal insulin tolerance tests (ipITT) were performed. Mice were then euthanized and pancreata were collected for histological analysis. Euglycemic hyperinsulinemic clamp experiments were performed on day 13.5 of pregnancy to assess insulin resistance. Beta cell function was assessed by glucose stimulated insulin secretion (GSIS) assay performed on day 0, 13.5 and 17.5 of pregnancy. At day 0, insulin tolerance and beta cell numbers were not different. At day 13.5, glucose infusion and disposal rates were significantly decreased (p<0.05) in Pregnant (P) HFHS animals (p<0.05) suggesting development of insulin resistance in P HFHS dams. Placental and fetal glucose uptake was significantly increased (p<0.01) in P HFHS dams at day 13.5 of pregnancy and by day 17.5 of pregnancy fetal weights were increased (p<0.05) in P HFHS dams compared to P CD dams. Basal and secreted insulin levels were increased in HFHS fed females at day 0, however at day 13.5 and 17.5 GSIS was decreased (p<0.05) in P HFHS dams. In conclusion, this animal model results in insulin resistance and beta cell dysfunction by mid-pregnancy further validating its relevance in studying the pathophysiology GDM.


Asunto(s)
Diabetes Gestacional , Resistencia a la Insulina , Insulinas , Ratones , Femenino , Embarazo , Animales , Humanos , Azúcares , Ratones Endogámicos C57BL , Placenta , Dieta Alta en Grasa/efectos adversos , Glucosa , Insulina
12.
Cell Metab ; 34(12): 1932-1946.e7, 2022 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-36243005

RESUMEN

Low-grade, sustained inflammation in white adipose tissue (WAT) characterizes obesity and coincides with type 2 diabetes mellitus (T2DM). However, pharmacological targeting of inflammation lacks durable therapeutic effects in insulin-resistant conditions. Through a computational screen, we discovered that the FDA-approved rheumatoid arthritis drug auranofin improved insulin sensitivity and normalized obesity-associated abnormalities, including hepatic steatosis and hyperinsulinemia in mouse models of T2DM. We also discovered that auranofin accumulation in WAT depleted inflammatory responses to a high-fat diet without altering body composition in obese wild-type mice. Surprisingly, elevated leptin levels and blunted beta-adrenergic receptor activity achieved by leptin receptor deletion abolished the antidiabetic effects of auranofin. These experiments also revealed that the metabolic benefits of leptin reduction were superior to immune impacts of auranofin in WAT. Our studies uncover important metabolic properties of anti-inflammatory treatments and contribute to the notion that leptin reduction in the periphery can be accomplished to treat obesity and T2DM.


Asunto(s)
Artritis Reumatoide , Diabetes Mellitus Tipo 2 , Animales , Ratones , Ratones Obesos , Hipoglucemiantes , Auranofina/farmacología , Auranofina/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Obesidad/tratamiento farmacológico
13.
Cell Metab ; 3(5): 379-86, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16679295

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Leptina/fisiología , Obesidad/metabolismo , Hormonas Peptídicas/fisiología , Animales , Glucemia/metabolismo , Regulación de la Temperatura Corporal , Peso Corporal , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/fisiopatología , Regulación hacia Abajo , Ghrelina , Insulina/sangre , Canales Iónicos/genética , Canales Iónicos/metabolismo , Leptina/genética , Leptina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Obesidad/sangre , Obesidad/fisiopatología , Páncreas/metabolismo , Hormonas Peptídicas/genética , Hormonas Peptídicas/metabolismo , Fenotipo , ARN Mensajero/metabolismo , Proteína Desacopladora 2
14.
J Biol Chem ; 285(52): 40581-92, 2010 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-20956520

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Dieta/efectos adversos , Hipoglucemiantes/farmacología , Ácido Oleanólico/análogos & derivados , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA Carboxilasa/genética , Acetil-CoA Carboxilasa/metabolismo , Tejido Adiposo/metabolismo , Animales , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/genética , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Ácidos Grasos no Esterificados/sangre , Glucosa/metabolismo , Hipoglucemiantes/uso terapéutico , Insulina/sangre , Ratones , Ratones Mutantes , Músculo Esquelético/metabolismo , Ácido Oleanólico/farmacología , Ácido Oleanólico/uso terapéutico , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Triglicéridos/sangre
15.
Nat Med ; 10(6): 625-32, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15133506

RESUMEN

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.


Asunto(s)
Tejido Adiposo/irrigación sanguínea , Apoptosis/fisiología , Vasos Sanguíneos/metabolismo , Obesidad/terapia , Péptidos/uso terapéutico , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Secuencia de Aminoácidos , Animales , Dieta , Metabolismo Energético , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Biblioteca de Péptidos , Péptidos/metabolismo , Prohibitinas , Proteínas Represoras/metabolismo
16.
Proc Natl Acad Sci U S A ; 105(49): 19229-34, 2008 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-19047629

RESUMEN

Activating signal cointegrator-2 (ASC-2), a transcriptional coactivator of multiple transcription factors that include the adipogenic factors peroxisome proliferator-activated receptor gamma (PPARgamma) and C/EBPalpha, is associated with histone H3-Lys-4-methyltransferase (H3K4MT) MLL3 or its paralogue MLL4 in a complex named ASCOM (ASC-2 complex). Indeed, ASC-2-null mouse embryonic fibroblasts (MEFs) have been demonstrated to be refractory to PPARgamma-stimulated adipogenesis and fail to express the PPARgamma-responsive adipogenic marker gene aP2. However, the specific roles for MLL3 and MLL4 in adipogenesis remain undefined. Here, we provide evidence that MLL3 plays crucial roles in adipogenesis. First, MLL3(Delta/Delta) mice expressing a H3K4MT-inactivated mutant of MLL3 have significantly less white fat. Second, MLL3(Delta/Delta) MEFs are mildly but consistently less responsive to inducers of adipogenesis than WT MEFs. Third, ASC-2, MLL3, and MLL4 are recruited to the PPARgamma-activated aP2 gene during adipogenesis, and PPARgamma is shown to interact directly with the purified ASCOM. Moreover, although H3K4 methylation of aP2 is readily induced in WT MEFs, it is not induced in ASC-2(-/-) MEFs and only partially induced in MLL3(Delta/Delta) MEFs. These results suggest that ASCOM-MLL3 and ASCOM-MLL4 likely function as crucial but redundant H3K4MT complexes for PPARgamma-dependent adipogenesis.


Asunto(s)
Adipogénesis/fisiología , N-Metiltransferasa de Histona-Lisina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Obesidad/fisiopatología , Adipocitos Blancos/enzimología , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/metabolismo , Animales , Peso al Nacer , Grasas de la Dieta/farmacología , Femenino , N-Metiltransferasa de Histona-Lisina/genética , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Células 3T3 NIH , Coactivadores de Receptor Nuclear , Obesidad/genética , PPAR gamma/metabolismo , Fenotipo
17.
J Family Med Prim Care ; 10(5): 2038-2040, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-34195146

RESUMEN

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.

18.
Cell Rep ; 37(1): 109767, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34610308

RESUMEN

Cardiac metabolism is a high-oxygen-consuming process, showing a preference for long-chain fatty acid (LCFA) as the fuel source under physiological conditions. However, a metabolic switch (favoring glucose instead of LCFA) is commonly reported in ischemic or late-stage failing hearts. The mechanism regulating this metabolic switch remains poorly understood. Here, we report that loss of PHD2/3, the cellular oxygen sensors, blocks LCFA mitochondria uptake and ß-oxidation in cardiomyocytes. In high-fat-fed mice, PHD2/3 deficiency improves glucose metabolism but exacerbates the cardiac defects. Mechanistically, we find that PHD2/3 bind to CPT1B, a key enzyme of mitochondrial LCFA uptake, promoting CPT1B-P295 hydroxylation. Further, we show that CPT1B-P295 hydroxylation is indispensable for its interaction with VDAC1 and LCFA ß-oxidation. Finally, we demonstrate that a CPT1B-P295A mutant constitutively binds to VDAC1 and rescues LCFA metabolism in PHD2/3-deficient cardiomyocytes. Together, our data identify an oxygen-sensitive regulatory axis involved in cardiac metabolism.


Asunto(s)
Carnitina O-Palmitoiltransferasa/metabolismo , Ácidos Grasos/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/metabolismo , Animales , Carnitina/metabolismo , Carnitina O-Palmitoiltransferasa/deficiencia , Carnitina O-Palmitoiltransferasa/genética , Dieta Alta en Grasa , Ácidos Grasos/química , Glucosa/metabolismo , Hidroxilación , Prolina Dioxigenasas del Factor Inducible por Hipoxia/deficiencia , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Peroxidación de Lípido , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mutagénesis Sitio-Dirigida , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Procolágeno-Prolina Dioxigenasa/deficiencia , Procolágeno-Prolina Dioxigenasa/genética , Unión Proteica , Canal Aniónico 1 Dependiente del Voltaje/genética
19.
Nat Commun ; 12(1): 1927, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33772019

RESUMEN

Accumulating evidence suggests that chronic inflammation of metabolic tissues plays a causal role in obesity-induced insulin resistance. Yet, how specific endothelial factors impact metabolic tissues remains undefined. Bone morphogenetic protein (BMP)-binding endothelial regulator (BMPER) adapts endothelial cells to inflammatory stress in diverse organ microenvironments. Here, we demonstrate that BMPER is a driver of insulin sensitivity. Both global and endothelial cell-specific inducible knockout of BMPER cause hyperinsulinemia, glucose intolerance and insulin resistance without increasing inflammation in metabolic tissues in mice. BMPER can directly activate insulin signaling, which requires its internalization and interaction with Niemann-Pick C1 (NPC1), an integral membrane protein that transports intracellular cholesterol. These results suggest that the endocrine function of the vascular endothelium maintains glucose homeostasis. Of potential translational significance, the delivery of BMPER recombinant protein or its overexpression alleviates insulin resistance and hyperglycemia in high-fat diet-fed mice and Leprdb/db (db/db) diabetic mice. We conclude that BMPER exhibits therapeutic potential for the treatment of diabetes.


Asunto(s)
Proteínas Portadoras/genética , Endotelio Vascular/metabolismo , Resistencia a la Insulina/genética , Transducción de Señal/genética , Animales , Glucemia/metabolismo , Proteínas Portadoras/metabolismo , Células Cultivadas , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Intolerancia a la Glucosa/genética , Células HEK293 , Humanos , Hiperinsulinismo/genética , Hiperinsulinismo/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Niemann-Pick C1/genética , Proteína Niemann-Pick C1/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo
20.
JCI Insight ; 6(11)2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-33974562

RESUMEN

The hypothalamus is a critical regulator of glucose metabolism and is capable of correcting diabetes conditions independently of an effect on energy balance. The small GTPase Rap1 in the forebrain is implicated in high-fat diet-induced (HFD-induced) obesity and glucose imbalance. Here, we report that increasing Rap1 activity selectively in the medial hypothalamus elevated blood glucose without increasing the body weight of HFD-fed mice. In contrast, decreasing hypothalamic Rap1 activity protected mice from diet-induced hyperglycemia but did not prevent weight gain. The remarkable glycemic effect of Rap1 was reproduced when Rap1 was specifically deleted in steroidogenic factor-1-positive (SF-1-positive) neurons in the ventromedial hypothalamic nucleus (VMH) known to regulate glucose metabolism. While having no effect on body weight regardless of sex, diet, and age, Rap1 deficiency in the VMH SF1 neurons markedly lowered blood glucose and insulin levels, improved glucose and insulin tolerance, and protected mice against HFD-induced neural leptin resistance and peripheral insulin resistance at the cellular and whole-body levels. Last, acute pharmacological inhibition of brain exchange protein directly activated by cAMP 2, a direct activator of Rap1, corrected glucose imbalance in obese mouse models. Our findings uncover the primary role of VMH Rap1 in glycemic control and implicate Rap1 signaling as a potential target for therapeutic intervention in diabetes.


Asunto(s)
Glucemia/metabolismo , Hiperglucemia/metabolismo , Insulina/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Animales , Dieta Alta en Grasa , Técnicas de Silenciamiento del Gen , Homeostasis , Hipotálamo/metabolismo , Resistencia a la Insulina , Leptina/metabolismo , Ratones , Factor Esteroidogénico 1/metabolismo , Proteínas de Unión al GTP rap1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA