Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Diabetes ; 72(10): 1350-1363, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36580483

RESUMEN

Increased saturated fatty acid (SFA) levels in membrane phospholipids have been implicated in the development of metabolic disease. Here, we tested the hypothesis that increased SFA content in cell membranes negatively impacts adipocyte insulin signaling. Preadipocyte cell models with elevated SFA levels in phospholipids were generated by disrupting the ADIPOR2 locus, which resulted in a striking twofold increase in SFA-containing phosphatidylcholines and phosphatidylethanolamines, which persisted in differentiated adipocytes. Similar changes in phospholipid composition were observed in white adipose tissues isolated from the ADIPOR2-knockout mice. The SFA levels in phospholipids could be further increased by treating ADIPOR2-deficient cells with palmitic acid and resulted in reduced membrane fluidity and endoplasmic reticulum stress in mouse and human preadipocytes. Strikingly, increased SFA levels in differentiated adipocyte phospholipids had no effect on adipocyte gene expression or insulin signaling in vitro. Similarly, increased adipocyte phospholipid saturation did not impair white adipose tissue function in vivo, even in mice fed a high-saturated fat diet at thermoneutrality. We conclude that increasing SFA levels in adipocyte phospholipids is well tolerated and does not affect adipocyte insulin signaling in vitro and in vivo.


Asunto(s)
Insulina , Fosfolípidos , Ratones , Humanos , Animales , Insulina/metabolismo , Adipocitos/metabolismo , Ácidos Grasos/metabolismo , Membrana Celular/metabolismo , Proteínas Portadoras/metabolismo
2.
Sci Adv ; 8(46): eadd5430, 2022 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-36383675

RESUMEN

We performed collapsing analyses on 454,796 UK Biobank (UKB) exomes to detect gene-level associations with diabetes. Recessive carriers of nonsynonymous variants in MAP3K15 were 30% less likely to develop diabetes (P = 5.7 × 10-10) and had lower glycosylated hemoglobin (ß = -0.14 SD units, P = 1.1 × 10-24). These associations were independent of body mass index, suggesting protection against insulin resistance even in the setting of obesity. We replicated these findings in 96,811 Admixed Americans in the Mexico City Prospective Study (P < 0.05)Moreover, the protective effect of MAP3K15 variants was stronger in individuals who did not carry the Latino-enriched SLC16A11 risk haplotype (P = 6.0 × 10-4). Separately, we identified a Finnish-enriched MAP3K15 protein-truncating variant associated with decreased odds of both type 1 and type 2 diabetes (P < 0.05) in FinnGen. No adverse phenotypes were associated with protein-truncating MAP3K15 variants in the UKB, supporting this gene as a therapeutic target for diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Quinasas Quinasa Quinasa PAM , Humanos , Diabetes Mellitus Tipo 2/genética , Predisposición Genética a la Enfermedad , Transportadores de Ácidos Monocarboxílicos/genética , Obesidad/genética , Estudios Prospectivos , Quinasas Quinasa Quinasa PAM/genética
3.
Nat Commun ; 13(1): 6020, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36241646

RESUMEN

The fatty acid composition of phosphatidylethanolamine (PE) determines cellular metabolism, oxidative stress, and inflammation. However, our understanding of how cells regulate PE composition is limited. Here, we identify a genetic locus on mouse chromosome 11, containing two poorly characterized genes Tlcd1 and Tlcd2, that strongly influences PE composition. We generated Tlcd1/2 double-knockout (DKO) mice and found that they have reduced levels of hepatic monounsaturated fatty acid (MUFA)-containing PE species. Mechanistically, TLCD1/2 proteins act cell intrinsically to promote the incorporation of MUFAs into PEs. Furthermore, TLCD1/2 interact with the mitochondria in an evolutionarily conserved manner and regulate mitochondrial PE composition. Lastly, we demonstrate the biological relevance of our findings in dietary models of metabolic disease, where Tlcd1/2 DKO mice display attenuated development of non-alcoholic steatohepatitis compared to controls. Overall, we identify TLCD1/2 proteins as key regulators of cellular PE composition, with our findings having broad implications in understanding and treating disease.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Fosfatidiletanolaminas , Animales , Ácidos Grasos/metabolismo , Ácidos Grasos Monoinsaturados/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Fosfatidiletanolaminas/metabolismo
4.
Biomedicines ; 10(2)2022 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-35203411

RESUMEN

Dapagliflozin is a sodium-glucose co-transporter 2 (SGLT2) inhibitor used for the treatment of diabetes. This study examines the effects of dapagliflozin on human islets, focusing on alpha and beta cell composition in relation to function in vivo, following treatment of xeno-transplanted diabetic mice. Mouse beta cells were ablated by alloxan, and dapagliflozin was provided in the drinking water while controls received tap water. Body weight, food and water intake, plasma glucose, and human C-peptide levels were monitored, and intravenous arginine/glucose tolerance tests (IVarg GTT) were performed to evaluate islet function. The grafted human islets were isolated at termination and stained for insulin, glucagon, Ki67, caspase 3, and PDX-1 immunoreactivity in dual and triple combinations. In addition, human islets were treated in vitro with dapagliflozin at different glucose concentrations, followed by insulin and glucagon secretion measurements. SGLT2 inhibition increased the animal survival rate and reduced plasma glucose, accompanied by sustained human C-peptide levels and improved islet response to glucose/arginine. SGLT2 inhibition increased both alpha and beta cell proliferation (Ki67+glucagon+ and Ki67+insulin+) while apoptosis was reduced (caspase3+glucagon+ and caspase3+insulin+). Alpha cells were fewer following inhibition of SGLT2 with increased glucagon/PDX-1 double-positive cells, a marker of alpha to beta cell transdifferentiation. In vitro treatment of human islets with dapagliflozin had no apparent impact on islet function. In summary, SGLT2 inhibition supported human islet function in vivo in the hyperglycemic milieu and potentially promoted alpha to beta cell transdifferentiation, most likely through an indirect mechanism.

5.
Mol Metab ; 36: 100964, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32248079

RESUMEN

OBJECTIVE: Peroxisome proliferator-activated receptors (PPARs) are key transcription factors that regulate adipose development and function, and the conversion of white into brown-like adipocytes. Here we investigated whether PPARα and PPARγ activation synergize to induce the browning of white fat. METHODS: A selection of PPAR activators was tested for their ability to induce the browning of both mouse and human white adipocytes in vitro, and in vivo in lean and obese mice. RESULTS: All dual PPARα/γ activators tested robustly increased uncoupling protein 1 (Ucp1) expression in both mouse and human adipocytes in vitro, with tesaglitazar leading to the largest Ucp1 induction. Importantly, dual PPARα/γ activator tesaglitazar strongly induced browning of white fat in vivo in both lean and obese male mice at thermoneutrality, greatly exceeding the increase in Ucp1 observed with the selective PPARγ activator rosiglitazone. While selective PPARγ activation was sufficient for the conversion of white into brown-like adipocytes in vitro, dual PPARα/γ activation was superior to selective PPARγ activation at inducing white fat browning in vivo. Mechanistically, the superiority of dual PPARα/γ activators is mediated at least in part via a PPARα-driven increase in fibroblast growth factor 21 (FGF21). Combined treatment with rosiglitazone and FGF21 resulted in a synergistic increase in Ucp1 mRNA levels both in vitro and in vivo. Tesaglitazar-induced browning was associated with increased energy expenditure, enhanced insulin sensitivity, reduced liver steatosis, and an overall improved metabolic profile compared to rosiglitazone and vehicle control groups. CONCLUSIONS: PPARγ and PPARα synergize to induce robust browning of white fat in vivo, via PPARγ activation in adipose, and PPARα-mediated increase in FGF21.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , PPAR alfa/metabolismo , PPAR gamma/metabolismo , Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Metabolismo Energético , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR alfa/genética , PPAR gamma/genética , Termogénesis/genética , Factores de Transcripción/metabolismo , Proteína Desacopladora 1/metabolismo
6.
Mol Metab ; 22: 49-61, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30772256

RESUMEN

OBJECTIVE: Nonalcoholic fatty liver disease (NAFLD) is becoming a leading cause of advanced chronic liver disease. The progression of NAFLD, including nonalcoholic steatohepatitis (NASH), has a strong genetic component, and the most robust contributor is the patatin-like phospholipase domain-containing 3 (PNPLA3) rs738409 encoding the 148M protein sequence variant. We hypothesized that suppressing the expression of the PNPLA3 148M mutant protein would exert a beneficial effect on the entire spectrum of NAFLD. METHODS: We examined the effects of liver-targeted GalNAc3-conjugated antisense oligonucleotide (ASO)-mediated silencing of Pnpla3 in a knock-in mouse model in which we introduced the human PNPLA3 I148M mutation. RESULTS: ASO-mediated silencing of Pnpla3 reduced liver steatosis (p = 0.038) in homozygous Pnpla3 148M/M knock-in mutant mice but not in wild-type littermates fed a steatogenic high-sucrose diet. In mice fed a NASH-inducing diet, ASO-mediated silencing of Pnpla3 reduced liver steatosis score and NAFLD activity score independent of the Pnpla3 genotype, while reductions in liver inflammation score (p = 0.018) and fibrosis stage (p = 0.031) were observed only in the Pnpla3 knock-in 148M/M mutant mice. These responses were accompanied by reduced liver levels of Mcp1 (p = 0.026) and Timp2 (p = 0.007) specifically in the mutant knock-in mice. This may reduce levels of chemokine attracting inflammatory cells and increase the collagenolytic activity during tissue regeneration. CONCLUSION: This study provides the first evidence that a Pnpla3 ASO therapy can improve all features of NAFLD, including liver fibrosis, and suppress the expression of a strong innate genetic risk factor, Pnpla3 148M, which may open up a precision medicine approach in NASH.


Asunto(s)
Lipasa/genética , Cirrosis Hepática/genética , Proteínas de la Membrana/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Oligonucleótidos Antisentido/genética , Fosfolipasas A2 Calcio-Independiente/genética , Animales , Femenino , Silenciador del Gen , Humanos , Lipasa/metabolismo , Cirrosis Hepática/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Oligonucleótidos Antisentido/metabolismo , Fosfolipasas A2 Calcio-Independiente/metabolismo
7.
Nat Metab ; 1(11): 1089-1100, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-32072135

RESUMEN

Liver X receptors limit cellular lipid uptake by stimulating the transcription of Inducible Degrader of the LDL Receptor (IDOL), an E3 ubiquitin ligase that targets lipoprotein receptors for degradation. The function of IDOL in systemic metabolism is incompletely understood. Here we show that loss of IDOL in mice protects against the development of diet-induced obesity and metabolic dysfunction by altering food intake and thermogenesis. Unexpectedly, analysis of tissue-specific knockout mice revealed that IDOL affects energy balance, not through its actions in peripheral metabolic tissues (liver, adipose, endothelium, intestine, skeletal muscle), but by controlling lipoprotein receptor abundance in neurons. Single-cell RNA sequencing of the hypothalamus demonstrated that IDOL deletion altered gene expression linked to control of metabolism. Finally, we identify VLDLR rather than LDLR as the primary mediator of IDOL effects on energy balance. These studies identify a role for the neuronal IDOL-VLDLR pathway in metabolic homeostasis and diet-induced obesity.


Asunto(s)
Metabolismo Energético/fisiología , Neuronas/metabolismo , Receptores de LDL/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Animales , Glucemia/metabolismo , Dieta , Metabolismo Energético/genética , Hipotálamo/metabolismo , Resistencia a la Insulina , Ratones , Ratones Noqueados , Obesidad/metabolismo , Obesidad/prevención & control , Ubiquitina-Proteína Ligasas/genética
8.
Toxicol Appl Pharmacol ; 355: 147-155, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30008375

RESUMEN

Fibroblast Growth Factors (FGFs) and their receptors (FGFRs) have been proposed as potential drug targets for the treatment of obesity. The aim of this study was to assess the potential toxicity in rats of three anti-FGFR1c mAbs with differential binding activity prior to clinical development. Groups of male rats received weekly injections of either one of two FGFR1c-specific mAbs or an FGFR1c/FGFR4-specific mAb at 10 mg/kg for up to 4 weeks. All three mAbs caused significant reductions in food intake and weight loss leading to some animals being euthanized early for welfare reasons. In all three groups given these mAbs, microscopic changes were seen in the bones and heart valves. In the bones of the femoro-tibial joint, thickening of the diaphyseal cortex of long bones, due to deposition of well organized new lamellar bone, indicated that an osteogenic effect was observed. In the heart, valvulopathy described as an endocardial myxomatous change affecting the mitral, pulmonary, tricuspid and aortic valves was observed in all mAb-treated animals. The presence of FGFR1 mRNA expression in the heart valves was confirmed using in situ hybridization. Targeting the FGF-FGFR1c pathway with anti-FGFR1c mAbs leads to drug induced valvulopathy in rats. In effect, this precluded the development of these mAbs as potential anti-obesity drugs. The valvulopathy observed was similar to that described for fenfluramine and dexafenfluramine. The pathogenesis of the drug-induced valvulopathy is considered FGFR1c-mediated, based on the specificity of the mAbs and FGFR1 mRNA expression in the heart valves.


Asunto(s)
Fármacos Antiobesidad/toxicidad , Anticuerpos Monoclonales/toxicidad , Enfermedades de las Válvulas Cardíacas/inducido químicamente , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/efectos de los fármacos , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/efectos de los fármacos , Animales , Fármacos Antiobesidad/farmacocinética , Anticuerpos Monoclonales/farmacocinética , Huesos/patología , Ingestión de Alimentos/efectos de los fármacos , Enfermedades de las Válvulas Cardíacas/metabolismo , Enfermedades de las Válvulas Cardíacas/patología , Válvulas Cardíacas/metabolismo , Válvulas Cardíacas/patología , Masculino , Osteogénesis/efectos de los fármacos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Wistar , Pérdida de Peso/efectos de los fármacos
9.
PLoS One ; 13(4): e0196601, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29702679

RESUMEN

Beta cell dysfunction accompanies and drives the progression of type 2 diabetes mellitus (T2D), but there are few clinical biomarkers available to assess islet cell stress in humans. Secretagogin, a protein enriched in pancreatic islets, demonstrates protective effects on beta cell function in animals. However, its potential as a circulating biomarker released from human beta cells and islets has not been studied. In this study primary human islets, beta cells and plasma samples were used to explore secretion and expression of secretagogin in relation to the T2D pathology. Secretagogin was abundantly and specifically expressed and secreted from human islets. Furthermore, T2D patients had an elevated plasma level of secretagogin compared with matched healthy controls, which was confirmed in plasma of diabetic mice transplanted with human islets. Additionally, the plasma secretagogin level of the human cohort had an inverse correlation to clinical assessments of beta cell function. To explore the mechanism of secretagogin release in vitro, human beta cells (EndoC-ßH1) were exposed to elevated glucose or cellular stress-inducing agents. Secretagogin was not released in parallel with glucose stimulated insulin release, but was markedly elevated in response to endoplasmic reticulum stressors and cytokines. These findings indicate that secretagogin is a potential novel biomarker, reflecting stress and islet cell dysfunction in T2D patients.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Islotes Pancreáticos/metabolismo , Secretagoginas/sangre , Adulto , Anciano , Animales , Biomarcadores/sangre , Núcleo Celular/metabolismo , Estudios de Cohortes , Citocinas/metabolismo , Citoplasma/metabolismo , Diabetes Mellitus Experimental/sangre , Retículo Endoplásmico/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Glucagón/metabolismo , Glucosa/farmacología , Prueba de Tolerancia a la Glucosa , Humanos , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/fisiopatología , Trasplante de Islotes Pancreáticos , Masculino , Ratones , Persona de Mediana Edad
10.
PLoS One ; 12(12): e0189060, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29206860

RESUMEN

The mechanism behind the glucose lowering effect occurring after specific activation of GPR120 is not completely understood. In this study, a potent and selective GPR120 agonist was developed and its pharmacological properties were compared with the previously described GPR120 agonist Metabolex-36. Effects of both compounds on signaling pathways and GLP-1 secretion were investigated in vitro. The acute glucose lowering effect was studied in lean wild-type and GPR120 null mice following oral or intravenous glucose tolerance tests. In vitro, in GPR120 overexpressing cells, both agonists signaled through Gαq, Gαs and the ß-arrestin pathway. However, in mouse islets the signaling pathway was different since the agonists reduced cAMP production. The GPR120 agonists stimulated GLP-1 secretion both in vitro in STC-1 cells and in vivo following oral administration. In vivo GPR120 activation induced significant glucose lowering and increased insulin secretion after intravenous glucose administration in lean mice, while the agonists had no effect in GPR120 null mice. Exendin 9-39, a GLP-1 receptor antagonist, abolished the GPR120 induced effects on glucose and insulin following an intravenous glucose challenge. In conclusion, GLP-1 secretion is an important mechanism behind the acute glucose lowering effect following specific GPR120 activation.


Asunto(s)
Glucemia/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Receptores Acoplados a Proteínas G/agonistas , Animales , Células CHO , Línea Celular , Cricetulus , AMP Cíclico/biosíntesis , Femenino , Proteínas de Unión al GTP/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , beta-Arrestinas/metabolismo
11.
J Diabetes Res ; 2016: 8630961, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27774459

RESUMEN

Aim. Models combining diabetes and atherosclerosis are important in evaluating the cardiovascular (CV) effects and safety of antidiabetes drugs in the development of treatments targeting CV complications. Our aim was to evaluate if crossing the heterozygous glucokinase knockout mouse (GK+/-) and hyperlipidemic mouse deficient in apolipoprotein E (ApoE-/-) will generate a disease model exhibiting a diabetic and macrovascular phenotype. Methods. The effects of defective glucokinase on the glucose metabolism and on the progression and regression of atherosclerosis on high-fat diets were studied in both genders of GK+/-ApoE-/- and ApoE-/- mice. Coronary vascular function of the female GK+/-ApoE-/- and ApoE-/- mice was also investigated. Results. GK+/-ApoE-/- mice show a stable hyperglycemia which was increased on Western diet. In oral glucose tolerance test, GK+/-ApoE-/- mice showed significant glucose intolerance and impaired glucose-stimulated insulin secretion. Plasma lipids were comparable with ApoE-/- mice; nevertheless the GK+/-ApoE-/- mice showed slightly increased atherosclerosis development. Conclusions. The GK+/-ApoE-/- mice showed a stable and reproducible hyperglycemia, accelerated atherosclerotic lesion progression, and no lesion regression after lipid lowering. This novel model provides a promising tool for drug discovery, enabling the evaluation of compound effects against both diabetic and cardiovascular endpoints simultaneously in one animal model.


Asunto(s)
Apolipoproteínas E/metabolismo , Aterosclerosis/metabolismo , Glucoquinasa/metabolismo , Hiperglucemia/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Dieta Alta en Grasa , Progresión de la Enfermedad , Femenino , Glucoquinasa/genética , Hiperglucemia/genética , Hiperglucemia/patología , Lípidos/sangre , Masculino , Ratones , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología
12.
J Pharmacokinet Pharmacodyn ; 43(2): 207-21, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26932466

RESUMEN

Input estimation is employed in cases where it is desirable to recover the form of an input function which cannot be directly observed and for which there is no model for the generating process. In pharmacokinetic and pharmacodynamic modelling, input estimation in linear systems (deconvolution) is well established, while the nonlinear case is largely unexplored. In this paper, a rigorous definition of the input-estimation problem is given, and the choices involved in terms of modelling assumptions and estimation algorithms are discussed. In particular, the paper covers Maximum a Posteriori estimates using techniques from optimal control theory, and full Bayesian estimation using Markov Chain Monte Carlo (MCMC) approaches. These techniques are implemented using the optimisation software CasADi, and applied to two example problems: one where the oral absorption rate and bioavailability of the drug eflornithine are estimated using pharmacokinetic data from rats, and one where energy intake is estimated from body-mass measurements of mice exposed to monoclonal antibodies targeting the fibroblast growth factor receptor (FGFR) 1c. The results from the analysis are used to highlight the strengths and weaknesses of the methods used when applied to sparsely sampled data. The presented methods for optimal control are fast and robust, and can be recommended for use in drug discovery. The MCMC-based methods can have long running times and require more expertise from the user. The rigorous definition together with the illustrative examples and suggestions for software serve as a highly promising starting point for application of input-estimation methods to problems in drug discovery.


Asunto(s)
Descubrimiento de Drogas/métodos , Eflornitina/farmacocinética , Cadenas de Markov , Método de Montecarlo , Algoritmos , Animales , Teorema de Bayes , Disponibilidad Biológica , Simulación por Computador , Ratones , Modelos Estadísticos , Ratas , Análisis de Regresión , Programas Informáticos
13.
J Pharm Sci ; 104(5): 1825-31, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25631774

RESUMEN

Tissue distribution and pharmacokinetics (PK) of full-length nontargeted antibody and its antigen-binding fragment (FAb) were evaluated for a range of tissues primarily of interest for cardiovascular and metabolic diseases. Mice were intravenously injected with a dose of 10 mg/kg of either human IgG1or its FAb fragment; perfused tissues were collected at a range of time points over 3 weeks for the human IgG1 antibody and 1 week for the human FAb antibody. Tissues were homogenized and antibody concentrations were measured by specific immunoassays on the Gyros system. Exposure in terms of maximum concentration (Cmax ) and area under the curve was assessed for all nine tissues. Tissue exposure of full-length antibody relative to plasma exposure was found to be between 1% and 10%, except for brain (0.2%). Relative concentrations of FAb antibody were the same, except for kidney tissue, where the antibody concentration was found to be ten times higher than in plasma. However, the absolute tissue uptake of full-length IgG was significantly higher than the absolute tissue uptake of the FAb antibody. This study provides a reference PK state for full-length whole and FAb antibodies in tissues related to cardiovascular and metabolic diseases that do not include antigen or antibody binding.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Fragmentos Fab de Inmunoglobulinas/metabolismo , Inmunoglobulina G/metabolismo , Miocardio/metabolismo , Animales , Anticuerpos Monoclonales/administración & dosificación , Femenino , Corazón/efectos de los fármacos , Humanos , Fragmentos Fab de Inmunoglobulinas/administración & dosificación , Inmunoglobulina G/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
14.
PLoS One ; 9(11): e112109, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25427253

RESUMEN

We have generated a novel monoclonal antibody targeting human FGFR1c (R1c mAb) that caused profound body weight and body fat loss in diet-induced obese mice due to decreased food intake (with energy expenditure unaltered), in turn improving glucose control. R1c mAb also caused weight loss in leptin-deficient ob/ob mice, leptin receptor-mutant db/db mice, and in mice lacking either the melanocortin 4 receptor or the melanin-concentrating hormone receptor 1. In addition, R1c mAb did not change hypothalamic mRNA expression levels of Agrp, Cart, Pomc, Npy, Crh, Mch, or Orexin, suggesting that R1c mAb could cause food intake inhibition and body weight loss via other mechanisms in the brain. Interestingly, peripherally administered R1c mAb accumulated in the median eminence, adjacent arcuate nucleus and in the circumventricular organs where it activated the early response gene c-Fos. As a plausible mechanism and coinciding with the initiation of food intake suppression, R1c mAb induced hypothalamic expression levels of the cytokines Monocyte chemoattractant protein 1 and 3 and ERK1/2 and p70 S6 kinase 1 activation.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Órganos Circunventriculares/efectos de los fármacos , Intolerancia a la Glucosa/tratamiento farmacológico , Hipotálamo/efectos de los fármacos , Obesidad/tratamiento farmacológico , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/fisiopatología , Quimiocina CCL2/agonistas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CCL7/agonistas , Quimiocina CCL7/genética , Quimiocina CCL7/metabolismo , Órganos Circunventriculares/metabolismo , Órganos Circunventriculares/fisiopatología , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético , Femenino , Regulación de la Expresión Génica , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/fisiopatología , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiopatología , Leptina/deficiencia , Leptina/genética , Ratones , Ratones Noqueados , Ratones Obesos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Obesidad/genética , Obesidad/metabolismo , Obesidad/fisiopatología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor de Melanocortina Tipo 4/deficiencia , Receptor de Melanocortina Tipo 4/genética , Receptores de Somatostatina/deficiencia , Receptores de Somatostatina/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Factor de Respuesta Sérica/agonistas , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Transducción de Señal
15.
PLoS One ; 8(11): e80330, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24324556

RESUMEN

Adiponectin has been shown to have beneficial cardiovascular effects and to signal through the adiponectin receptors, AdipoR1 and AdipoR2. The original aim of this study was to investigate the effect of combined AdipoR1 and AdipoR2 deficiency (AdipoR1(-/-)AdipoR2(-/-)) on atherosclerosis. However, we made the interesting observation that AdipoR1(-/-) AdipoR2(-/-) leads to embryonic lethality demonstrating the critical importance of the adiponectin signalling system during development. We then investigated the effect of AdipoR2-ablation on the progression of atherosclerosis in apolipoprotein E deficient (ApoE(-/-)) mice. AdipoR2(-/-)ApoE(-/-) mice fed an atherogenic diet had decreased plaque area in the brachiocephalic artery compared with AdipoR2(+/+) ApoE(-/-) littermate controls as visualized in vivo using an ultrasound biomicroscope and confirmed by histological analyses. The decreased plaque area in the brachiocephalic artery could not be explained by plasma cholesterol levels or inflammatory status. However, accumulation of neutral lipids was decreased in peritoneal macrophages from AdipoR2(-/-)ApoE(-/-) mice after incubation with oxidized LDL. This effect was associated with lower CD36 and higher ABCA1 mRNA levels in peritoneal macrophages from AdipoR2(-/-)ApoE(-/-) mice compared with AdipoR2(+/+)ApoE(-/-) controls after incubation with oxidized LDL. In summary, we show that adiponectin receptors are crucial during embryonic development and that AdipoR2-deficiency slows down the progression of atherosclerosis in the brachiocephalic artery of ApoE-deficient mice.


Asunto(s)
Apolipoproteínas E/deficiencia , Apolipoproteínas E/metabolismo , Aterosclerosis/metabolismo , Receptores de Adiponectina/metabolismo , Animales , Apolipoproteínas E/genética , Aterosclerosis/genética , Western Blotting , Peso Corporal/genética , Peso Corporal/fisiología , Femenino , Genotipo , Heterocigoto , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Receptores de Adiponectina/deficiencia , Receptores de Adiponectina/genética
16.
Biochem Biophys Res Commun ; 369(4): 1065-70, 2008 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-18339309

RESUMEN

Glycerol-3-phosphate acyltransferase (GPAT) is involved in triacylglycerol (TAG) and phospholipid synthesis, catalyzing the first committed step. In order to further investigate the in vivo importance of the dominating mitochondrial variant, GPAT1, a novel GPAT1(-/-) mouse model was generated and studied. Female GPAT1(-/-) mice had reduced body weight-gain and adiposity when fed chow diet compared with littermate wild-type controls. Furthermore, GPAT1(-/-) females on chow diet showed decreased liver TAG content, plasma cholesterol and TAG levels and increased ex vivo liver fatty acid oxidation and plasma ketone bodies. However, these beneficial effects were abolished and the glucose tolerance tended to be impaired when GPAT1(-/-) females were fed a long-term high-fat diet (HFD). GPAT1-deficiency was not associated with altered whole body energy expenditure or respiratory exchange ratio. In addition, there were no changes in male GPAT1(-/-) mice fed either diet except for increased plasma ketone bodies on chow diet, indicating a gender-specific phenotype. Thus, GPAT1-deficiency does not protect against HFD-induced obesity, hepatic steatosis or whole body glucose intolerance.


Asunto(s)
Grasas de la Dieta/efectos adversos , Hígado Graso/etiología , Intolerancia a la Glucosa/etiología , Glucosa/metabolismo , Glicerol-3-Fosfato O-Aciltransferasa/fisiología , Obesidad/etiología , Triglicéridos/metabolismo , Animales , Colesterol/sangre , Dieta , Grasas de la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Metabolismo Energético , Hígado Graso/genética , Femenino , Intolerancia a la Glucosa/genética , Glicerol-3-Fosfato O-Aciltransferasa/genética , Homeostasis , Masculino , Ratones , Ratones Mutantes , Mitocondrias/enzimología , Obesidad/genética , Triglicéridos/análisis , Aumento de Peso
17.
Arterioscler Thromb Vasc Biol ; 27(12): 2707-13, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17932310

RESUMEN

OBJECTIVE: Previous studies have indicated that the hyperlipidemia and gene expression changes induced by a short-term high-fat diet (HFD) are mediated through the peroxisome proliferator-activated receptor gamma coactivator (PGC)-1beta, and that in vitro both PGC-1beta and PGC -1alpha increase PPARalpha-mediated transcriptional activities. Here, we examined the in vivo effects of these two coactivators in potentiating the lipid lowering properties of the PPARalpha agonist Wy14,643 (Wy). METHODS AND RESULTS: C57BL/6 mice were fed chow or HFD and transduced with adenoviruses encoding PGC-1alpha or PGC-1beta. On chow, hepatic PGC-1beta overexpression caused severe combined hyperlipidemia including elevated plasma apolipoprotein B levels. Hepatic triglyceride secretion, DGAT1, and FAT/CD36 expression were increased whereas PPARalpha and hepatic lipase mRNA levels were reduced. PGC-1beta overexpression blunted Wy-mediated changes in expression levels of PPARalpha and downstream genes. Furthermore, PGC-1beta did not potentiate Wy-stimulated fatty acid oxidation in primary hepatocytes. PGC-1beta and PGC-1alpha overexpression did not alter SREBP-1c, SREBP-1c target gene expression, nor hepatic triglyceride content. On HFD, PGC-1beta overexpression decreased hepatic SREBP-1c, yet increased FAS and ACCalpha mRNA and plasma triglyceride levels. CONCLUSIONS: Hepatic PGC-1beta overexpression caused combined hyperlipidemia independent of SREBP-1c activation. Hepatic PGC-1beta overexpression reduced the potentially beneficial effects of PPARalpha activation on gene expression. Thus, inhibition of hepatic PGC-1beta may provide a therapy for treating combined hyperlipidemia.


Asunto(s)
Anticolesterolemiantes/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Hiperlipidemia Familiar Combinada/prevención & control , Hígado/efectos de los fármacos , PPAR alfa/agonistas , Pirimidinas/farmacología , Transactivadores/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Adenoviridae/genética , Animales , Anticolesterolemiantes/uso terapéutico , Apolipoproteínas B/sangre , Antígenos CD36/metabolismo , Células Cultivadas , Diacilglicerol O-Acetiltransferasa/metabolismo , Grasas de la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Ácido Graso Sintasas/metabolismo , Ácidos Grasos/metabolismo , Vectores Genéticos , Hiperlipidemia Familiar Combinada/genética , Hiperlipidemia Familiar Combinada/metabolismo , Lipasa/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , PPAR alfa/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Pirimidinas/uso terapéutico , ARN Mensajero/metabolismo , Receptores de Lipoproteína/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Transactivadores/genética , Factores de Transcripción , Transducción Genética , Triglicéridos/metabolismo , Regulación hacia Arriba
18.
Diabetes ; 56(3): 583-93, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17327425

RESUMEN

The adipocyte-derived hormone adiponectin regulates glucose and lipid metabolism and influences the risk for developing obesity, type 2 diabetes, and cardiovascular disease. Adiponectin binds to two different seven-transmembrane domain receptors termed AdipoR1 and AdipoR2. To study the physiological importance of these receptors, AdipoR1 gene knockout mice (AdipoR1(-/-)) and AdipoR2 gene knockout mice (AdipoR2(-/-)) were generated. AdipoR1(-/-) mice showed increased adiposity associated with decreased glucose tolerance, spontaneous locomotor activity, and energy expenditure. However, AdipoR2(-/-) mice were lean and resistant to high-fat diet-induced obesity associated with improved glucose tolerance and higher spontaneous locomotor activity and energy expenditure and reduced plasma cholesterol levels. Thus, AdipoR1 and AdipoR2 are clearly involved in energy metabolism but have opposing effects.


Asunto(s)
Metabolismo Energético/fisiología , Receptores de Superficie Celular/metabolismo , Proteínas Quinasas Activadas por AMP , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Adiposidad/genética , Adiposidad/fisiología , Animales , Peso Corporal/fisiología , Encéfalo/patología , Metabolismo Energético/genética , Conducta Alimentaria , Femenino , Glucosa/metabolismo , Masculino , Ratones , Ratones Noqueados , Actividad Motora/genética , Actividad Motora/fisiología , Complejos Multienzimáticos/metabolismo , PPAR alfa/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Adiponectina , Receptores de Superficie Celular/genética , Transducción de Señal , Testículo/citología , Factores de Tiempo
19.
FASEB J ; 20(3): 434-43, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16507761

RESUMEN

Glycerol-3-phosphate acyltransferase (GPAT) catalyzes the first committed step in triacylglycerol (TAG) and phospholipid biosynthesis. GPAT activity has been identified in both ER and mitochondrial subcellular fractions. The ER activity dominates in most tissues except in liver, where the mitochondrial isoform (mtGPAT) can constitute up to 50% of the total activity. To study the in vivo effects of hepatic mtGPAT overexpression, mice were transduced with adenoviruses expressing either murine mtGPAT or a catalytically inactive variant of the enzyme. Overexpressing mtGPAT resulted in massive 12- and 7-fold accumulation of liver TAG and diacylglycerol, respectively but had no effect on phospholipid or cholesterol ester content. Histological analysis showed extensive lipid accumulation in hepatocytes. Furthermore, mtGPAT transduction markedly increased adipocyte differentiation-related protein and stearoyl-CoA desaturase-1 (SCD-1) in the liver. In line with increased SCD-1 expression, 18:1 and 16:1 in the hepatic TAG fraction increased. In addition, mtGPAT overexpression decreased ex vivo fatty acid oxidation, increased liver TAG secretion rate 2-fold, and increased plasma TAG and cholesterol levels. These results support the hypothesis that increased hepatic mtGPAT activity associated with obesity and insulin resistance contributes to increased TAG biosynthesis and inhibition of fatty acid oxidation, responses that would promote hepatic steatosis and dyslipidemia.


Asunto(s)
Ácidos Grasos/metabolismo , Hígado Graso/enzimología , Glicerol-3-Fosfato O-Aciltransferasa/biosíntesis , Mitocondrias Hepáticas/enzimología , Triglicéridos/metabolismo , Sustitución de Aminoácidos , Animales , Carbohidratos/biosíntesis , Diglicéridos/metabolismo , Inducción Enzimática , Hígado Graso/genética , Glicerol-3-Fosfato O-Aciltransferasa/genética , Resistencia a la Insulina , Lípidos/biosíntesis , Masculino , Malonil Coenzima A/metabolismo , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Oxidación-Reducción , Fosfolípidos/química , ARN Mensajero/biosíntesis , Proteínas Recombinantes de Fusión/fisiología
20.
J Lipid Res ; 47(2): 329-40, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16282640

RESUMEN

Adipose differentiation-related protein (ADRP) is a lipid droplet-associated protein that is expressed in various tissues. In mice treated with the peroxisome proliferator-activated receptor alpha (PPARalpha) agonist Wy14,643 (Wy), hepatic mRNA and protein levels of ADRP as well as hepatic triglyceride content increased. Also in primary mouse hepatocytes, Wy increased ADRP expression and intracellular triglyceride mass. The triglyceride mass increased in spite of unchanged triglyceride biosynthesis and increased palmitic acid oxidation. However, Wy incubation decreased the secretion of newly synthesized triglycerides, whereas apolipoprotein B secretion increased. Thus, decreased availability of triglycerides for VLDL assembly could help to explain the cellular accumulation of triglycerides after Wy treatment. We hypothesized that this effect could be mediated by increased ADRP expression. Similar to PPARalpha activation, adenovirus-mediated ADRP overexpression in mouse hepatocytes enhanced cellular triglyceride mass and decreased the secretion of newly synthesized triglycerides. In ADRP-overexpressing cells, Wy incubation resulted in a further decrease in triglyceride secretion. This effect of Wy was not attributable to decreased cellular triglycerides after increased fatty acid oxidation because the triglyceride mass in Wy-treated ADRP-overexpressing cells was unchanged. In summary, PPARalpha activation prevents the availability of triglycerides for VLDL assembly and increases hepatic triglyceride content in part by increasing the expression of ADRP.


Asunto(s)
Hepatocitos/metabolismo , Proteínas de la Membrana/metabolismo , PPAR alfa/metabolismo , Triglicéridos/metabolismo , Acil-CoA Deshidrogenasa de Cadena Larga/genética , Acil-CoA Oxidasa/genética , Animales , Apolipoproteína B-100 , Apolipoproteína B-48 , Apolipoproteínas B/metabolismo , Carnitina O-Palmitoiltransferasa/genética , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/farmacología , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Hepatocitos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , PPAR alfa/antagonistas & inhibidores , PPAR alfa/genética , Ácido Palmítico/metabolismo , Perilipina-2 , Proliferadores de Peroxisomas/farmacología , Pirimidinas/farmacología , Transfección , Triglicéridos/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...