Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 18(2): e0281887, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36821579

RESUMEN

Gut microbiota has been established as a main regulator of health. However, how changes in gut microbiota are directly associated with physiological and cellular alterations has been difficult to tackle on a large-scale basis, notably because of the cost and labor-extensive resources required for rigorous experiments in mammals. In the present study, we used the nematode Caenorhabditis elegans as a model organism to elucidate microbiota-host interactions. We developed a method to extract gut microbiota (MCB) from murine feces, and tested its potential as food source for and its impact on C. elegans biology compared to the standard bacterial diet Escherichia coli OP50. Although less preferred than OP50, MCB was not avoided but had a lower energy density (triglycerides and glucose). Consistently, MCB-fed worms exhibited smaller body length and size, lower fertility, and lower fat content than OP50-fed worms, but had a longer mean lifespan, which resembles the effects of calorie restriction in mammals. However, these outcomes were altered when bacteria were inactivated, suggesting an important role of symbiosis of MCB beyond nutrient source. Taken together, our findings support the effectiveness of gut MCB processing to test its effects in C. elegans. More work comparing MCB of differently treated mice or humans is required to further validate relevance to mammals before large-scale screening assays.


Asunto(s)
Caenorhabditis elegans , Microbioma Gastrointestinal , Humanos , Animales , Ratones , Caenorhabditis elegans/fisiología , Escherichia coli/fisiología , Longevidad/fisiología , Mamíferos
2.
Front Physiol ; 13: 949378, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36105289

RESUMEN

Hypoxia is common in lung diseases and a potent stimulator of the long non-coding RNA Metastasis-Associated Lung Adenocarcinoma Transcript 1 (MALAT1). Herein, we investigated the impact of Malat1 on hypoxia-induced lung dysfunction in mice. Malat1-deficient mice and their wild-type littermates were tested after 8 days of normoxia or hypoxia (10% oxygen). Hypoxia decreased elastance of the lung by increasing lung volume and caused in vivo hyperresponsiveness to methacholine without altering the contraction of airway smooth muscle. Malat1 deficiency also modestly decreased lung elastance but only when tested at low lung volumes and without altering lung volume and airway smooth muscle contraction. The in vivo responsiveness to methacholine was also attenuated by Malat1 deficiency, at least when elastance, a readout sensitive to small airway closure, was used to assess the response. More impressively, in vivo hyperresponsiveness to methacholine caused by hypoxia was virtually absent in Malat1-deficient mice, especially when hysteresivity, a readout sensitive to small airway narrowing heterogeneity, was used to assess the response. Malat1 deficiency also increased the coefficient of oxygen extraction and decreased ventilation in conscious mice, suggesting improvements in gas exchange and in clinical signs of respiratory distress during natural breathing. Combined with a lower elastance at low lung volumes at baseline, as well as a decreased propensity for small airway closure and narrowing heterogeneity during a methacholine challenge, these findings represent compelling evidence suggesting that the lack of Malat1 protects the access to alveoli for air entering the lung.

3.
Cell Rep Med ; 2(4): 100248, 2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33948578

RESUMEN

Insulin-like growth factor-binding protein (IGFBP)-2 is a circulating biomarker of cardiometabolic health. Here, we report that circulating IGFBP-2 concentrations robustly increase after different bariatric procedures in humans, reaching higher levels after biliopancreatic diversion with duodenal switch (BPD-DS) than after Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG). This increase is closely associated with insulin sensitization. In mice and rats, BPD-DS and RYGB operations also increase circulating IGFBP-2 levels, which are not affected by SG or caloric restriction. In mice, Igfbp2 deficiency significantly impairs surgery-induced loss in adiposity and early improvement in insulin sensitivity but does not affect long-term enhancement in glucose homeostasis. This study demonstrates that the modulation of circulating IGFBP-2 may play a role in the early improvement of insulin sensitivity and loss of adiposity brought about by bariatric surgery.


Asunto(s)
Cirugía Bariátrica , Fenómenos Bioquímicos/fisiología , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Obesidad Mórbida/cirugía , Animales , Cirugía Bariátrica/métodos , Desviación Biliopancreática/métodos , Gastrectomía/métodos , Derivación Gástrica/métodos , Humanos , Ratones , Obesidad/cirugía , Obesidad Mórbida/metabolismo
4.
Immun Ageing ; 18(1): 8, 2021 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-33622333

RESUMEN

BACKGROUND: The resident immune population of pancreatic islets has roles in islet development, beta cell physiology, and the pathology of diabetes. These roles have largely been attributed to islet macrophages, comprising 90% of islet immune cells (in the absence of islet autoimmunity), and, in the case of type 1 diabetes, to infiltrating autoreactive T cells. In adipose, tissue-resident and recruited T and B cells have been implicated in the development of insulin resistance during diet-induced obesity and ageing, but whether this is paralleled in the pancreatic islets is not known. Here, we investigated the non-macrophage component of resident islet immune cells in islets isolated from C57BL/6 J male mice during ageing (3 to 24 months of age) and following similar weight gain achieved by 12 weeks of 60% high fat diet. Immune cells were also examined by flow cytometry in cadaveric non-diabetic human islets. RESULTS: Immune cells comprised 2.7 ± 1.3% of total islet cells in non-diabetic mouse islets, and 2.3 ± 1.7% of total islet cells in non-diabetic human islets. In 3-month old mice on standard diet, B and T cells each comprised approximately 2-4% of the total islet immune cell compartment, and approximately 0.1% of total islet cells. A similar amount of T cells were present in non-diabetic human islets. The majority of islet T cells expressed the αß T cell receptor, and were comprised of CD8-positive, CD4-positive, and regulatory T cells, with a minor population of γδ T cells. Interestingly, the number of islet T cells increased linearly (R2 = 0.9902) with age from 0.10 ± 0.05% (3 months) to 0.38 ± 0.11% (24 months) of islet cells. This increase was uncoupled from body weight, and was not phenocopied by a degree similar weight gain induced by high fat diet in mice. CONCLUSIONS: This study reveals that T cells are a part of the normal islet immune population in mouse and human islets, and accumulate in islets during ageing in a body weight-independent manner. Though comprising only a small subset of the immune cells within islets, islet T cells may play a role in the physiology of islet ageing.

5.
Cell Metab ; 32(2): 287-300.e7, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32755608

RESUMEN

Stimulation of brown adipose tissue (BAT) thermogenesis in humans has emerged as an attractive target to improve metabolic health. Pharmacological stimulations targeting the ß3-adrenergic receptor (ß3-AR), the adrenergic receptor believed to mediate BAT thermogenesis, have historically performed poorly in human clinical trials. Here we report that, in contrast to rodents, human BAT thermogenesis is not mediated by the stimulation of ß3-AR. Oral administration of the ß3-AR agonist mirabegron only elicited increases in BAT thermogenesis when ingested at the maximal allowable dose. This led to off-target binding to ß1-AR and ß2-AR, thereby increasing cardiovascular responses and white adipose tissue lipolysis, respectively. ADRB2 was co-expressed with UCP1 in human brown adipocytes. Pharmacological stimulation and inhibition of the ß2-AR as well as knockdown of ADRB1, ADRB2, or ADRB3 in human brown adipocytes all confirmed that BAT lipolysis and thermogenesis occur through ß2-AR signaling in humans (ClinicalTrials.govNCT02811289).


Asunto(s)
Adipocitos Marrones/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Termogénesis , Adolescente , Adulto , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Adulto Joven
6.
J Gerontol A Biol Sci Med Sci ; 75(11): 2031-2036, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32298404

RESUMEN

Long-term caloric restriction (CR) has been shown to be beneficial to various tissues and organs. In contrast, CR exerts differential effects on bone, which could be due in part to the nature of the protein regime utilized. Male Sprague Dawley rats (8-month-old) were subjected for 12 months to 40% CR in macronutrients and compared with rats fed ad libitum for the same period. Casein- and soy-fed groups were compared. There was a significant decrease in bone quality in both CR groups, which was independent of the source of protein in the diet. In contrast, the group fed soy protein ad libitum showed better bone quality and higher levels of bone formation compared with casein-fed animals. Notably, bone marrow adipocytes were not mobilized upon CR as demonstrated by an absence of change in adipocyte number and tissue expression of leptin. This study demonstrates that the negative effect of CR on bone quality could not be prevented by the most common protein regimes.


Asunto(s)
Tejido Adiposo/metabolismo , Médula Ósea/metabolismo , Restricción Calórica , Proteínas en la Dieta/farmacología , Osteoporosis/etiología , Osteoporosis/prevención & control , Adipocitos/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Animales , Caseínas/farmacología , Modelos Animales de Enfermedad , Leptina/metabolismo , Masculino , Osteogénesis/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Proteínas de Soja/farmacología
7.
PLoS One ; 13(5): e0196603, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29746487

RESUMEN

Several studies have suggested that signals emerging from white adipose tissue can contribute to the control of longevity. In turn, aging is associated with perturbed regulation and partitioning of fat depots and insulin resistance. However, the exact mechanisms involved in these relationships remain undetermined. Using RAP-PCR on adipose tissue of young and old male mice coupled with qPCR validation, we have uncovered the long non-coding RNA Malat1 as a gene robustly downregulated in visceral white adipose tissue (vWAT) during normal aging in male mice and men. Reductions in Malat1 expression in subcutaneous WAT (scWAT) were also observed in genetic (ob and db) as well as diet-induced models of obesity. Based on these findings, Malat1+/+ and Malat1-/- mouse littermates were thus probed to detect whether loss of Malat1 would impact age or diet-induced gain in fat mass and development of glucose intolerance. Contrary to this hypothesis, male and female Malat1-deficient mice gained as much weight, and developed insulin resistance to a similar extent as their Malat1+/+ littermates when studied up to eight months old on regular chow or a high-fat, high-sucrose diet. Moreover, we observed no marked difference in oxygen consumption, food intake, or lipid profiles between Malat1+/+ and Malat1-/- mice. Therefore, we conclude that the overall metabolic impact of the absence of Malat1 on adipose tissue accretion and glucose intolerance is either physiologically not relevant upon aging and obesity, or that it is masked by as yet unknown compensatory mechanisms.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Envejecimiento/metabolismo , Resistencia a la Insulina/fisiología , Insulina/metabolismo , ARN Largo no Codificante/metabolismo , Tejido Adiposo Blanco/fisiología , Envejecimiento/fisiología , Animales , Peso Corporal/fisiología , Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos/fisiología , Femenino , Glucosa/metabolismo , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/fisiopatología , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Obesidad/fisiopatología , Consumo de Oxígeno/fisiología
8.
Diabetes ; 67(7): 1285-1296, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29496744

RESUMEN

The current demographic shift toward an aging population has led to a robust increase in the prevalence of age-associated metabolic disorders. Recent studies have demonstrated that the etiology of obesity-related insulin resistance that develops with aging differs from that induced by high-calorie diets. Whereas the role of adaptive immunity in changes in energy metabolism driven by nutritional challenges has recently gained attention, its impact on aging remains mostly unknown. Here we found that the number of follicular B2 lymphocytes and expression of the B-cell-specific transcriptional coactivator OcaB increase with age in spleen and in intra-abdominal epididymal white adipose tissue (eWAT), concomitantly with higher circulating levels of IgG and impaired glucose homeostasis. Reduction of B-cell maturation and Ig production-especially that of IgG2c-by ablation of OcaB prevented age-induced glucose intolerance and insulin resistance and promoted energy expenditure by stimulating fatty acid utilization in eWAT and brown adipose tissue. Transfer of wild-type bone marrow in OcaB-/- mice replenished the eWAT B2-cell population and IgG levels, which diminished glucose tolerance, insulin sensitivity, and energy expenditure while increasing body weight gain in aged mice. Thus these findings demonstrate that upon aging, modifications in B-cell-driven adaptive immunity contribute to glucose intolerance and fat accretion.


Asunto(s)
Envejecimiento/metabolismo , Linfocitos B/fisiología , Metabolismo Energético/genética , Resistencia a la Insulina/genética , Metabolismo de los Lípidos/genética , Obesidad , Transactivadores/genética , Adolescente , Adulto , Anciano , Envejecimiento/genética , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Epidídimo , Femenino , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/inmunología , Intolerancia a la Glucosa/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Obesidad/complicaciones , Obesidad/genética , Obesidad/inmunología , Obesidad/metabolismo , Adulto Joven
9.
G3 (Bethesda) ; 7(8): 2739-2747, 2017 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-28620088

RESUMEN

Despite many advances, the molecular links between energy metabolism and longevity are not well understood. Here, we have used the nematode model Caenorhabditis elegans to study the role of the yet-uncharacterized gene R148.3 in fat accumulation and lifespan. In wild-type worms, a R148.3p::GFP reporter showed enhanced expression throughout life in the pharynx, in neurons, and in muscles. Functionally, a protein fusing a predicted 22 amino acid N-terminal signal sequence (SS) of R148.3 to mCherry displayed robust accumulation in coelomyocytes, indicating that R148.3 is a secreted protein. Systematic depletion of R148.3 by RNA interference (RNAi) at L1 but not at young-adult stage enhanced triglyceride accumulation, which was associated with increased food uptake and lower expression of genes involved in lipid oxidation. However, RNAi of R148.3 at both L1 and young-adult stages robustly diminished mean and maximal lifespan of wild-type worms, and also abolished the long-lived phenotypes of eat-2 and daf-2/InsR mutants. Based on these data, we propose that R148.3 is an SS that modulates fat mass and longevity in an independent manner.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiología , Genes de Helminto , Metabolismo de los Lípidos/genética , Longevidad/genética , Secuencia de Aminoácidos , Animales , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/metabolismo , Mutación/genética , Estrés Oxidativo , Fenotipo , Señales de Clasificación de Proteína , Triglicéridos/metabolismo
10.
Oncol Rep ; 37(4): 2153-2160, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28260109

RESUMEN

Long non-coding RNAs (lncRNA) are key regulators of gene expression both at the transcriptional and post-transcriptional levels. The lncRNA metastasis associated lung adenocarcinoma transcript 1 (Malat1) is overexpressed in many types of cancer, including hepatocarcinoma, and induces cell proliferation in several cell lines in vitro. However, the direct causal effects of Malat1 on hepatocyte proliferation and liver carcinogenesis in vivo are not fully understood. To better determine the contribution of Malat1 to hepatocarcinoma oncogenesis, this study was aimed at testing the hypothesis that its absence confers resistance to the development of liver tumors. Male Malat1-/- mice and their wild-type (WT) littermates were studied one year after treatment with the genotoxic agent diethylnitrosamine (DEN), a potent inducer of liver cancer. As expected, in WT mice, Malat1 expression was significantly higher in hepatic tumors than in healthy liver regions. Altered hepatic mRNA levels of Ki67, HDAC3, NFκB and p27 were observed in DEN-treated Malat1-/- mice. Despite this, these mice were characterized by similar liver weight, prevalence of tumors, and histological features compared to those of their WT littermates. In parallel, plasma lipids and glucose homeostasis did not significantly differ between DEN-treated groups. These findings support a role for Malat1 as a marker of liver carcinogenesis, but also suggest that its role in the regulation of hepatocyte hyperproliferation in mice is either minimal or masked by redundant and/or overwhelming mechanisms.


Asunto(s)
Carcinoma Hepatocelular/patología , Dietilnitrosamina/efectos adversos , Neoplasias Hepáticas/patología , ARN Largo no Codificante/genética , Animales , Glucemia/metabolismo , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/genética , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Lípidos/sangre , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/genética , Masculino , Ratones , Tamaño de los Órganos , Regulación hacia Arriba
11.
Mol Pharmacol ; 91(3): 167-177, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28049773

RESUMEN

Alternative splicing at the human glucuronosyltransferase 1 gene locus (UGT1) produces alternate isoforms UGT1A_i2s that control glucuronidation activity through protein-protein interactions. Here, we hypothesized that UGT1A_i2s function as a complex protein network connecting other metabolic pathways with an influence on cancer cell metabolism. This is based on a pathway enrichment analysis of proteomic data that identified several high-confidence candidate interaction proteins of UGT1A_i2 proteins in human tissues-namely, the rate-limiting enzyme of glycolysis pyruvate kinase (PKM), which plays a critical role in cancer cell metabolism and tumor growth. The partnership of UGT1A_i2 and PKM2 was confirmed by coimmunoprecipitation in the HT115 colon cancer cells and was supported by a partial colocalization of these two proteins. In support of a functional role for this partnership, depletion of UGT1A_i2 proteins in HT115 cells enforced the Warburg effect, with a higher glycolytic rate at the expense of mitochondrial respiration, and led to lactate accumulation. Untargeted metabolomics further revealed a significantly altered cellular content of 58 metabolites, including many intermediates derived from the glycolysis and tricarboxylic acid cycle pathways. These metabolic changes were associated with a greater migration potential. The potential relevance of our observations is supported by the down-regulation of UGT1A_i2 mRNA in colon tumors compared with normal tissues. Alternate UGT1A variants may thus be part of the expanding compendium of metabolic pathways involved in cancer biology directly contributing to the oncogenic phenotype of colon cancer cells. Findings uncover new aspects of UGT functions diverging from their transferase activity.


Asunto(s)
Empalme Alternativo/genética , Neoplasias del Colon/enzimología , Neoplasias del Colon/metabolismo , Glucuronosiltransferasa/genética , Proteínas Portadoras/metabolismo , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Respiración de la Célula , Supervivencia Celular , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Metabolismo Energético , Regulación Neoplásica de la Expresión Génica , Glucuronosiltransferasa/metabolismo , Glucólisis , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Ácido Láctico/metabolismo , Proteínas de la Membrana/metabolismo , Metabolómica , Mitocondrias/metabolismo , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Hormonas Tiroideas/metabolismo , Proteínas de Unión a Hormona Tiroide
12.
Int J Oncol ; 49(4): 1731-6, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27499160

RESUMEN

Increased expression levels of the long non-coding RNA metastasis-associated lung adenocarcinoma transcript 1 (Malat1) have been associated with enhanced proliferation and metastasis of several cancer cell types. Hypoxia, a hallmark characteristic of solid tumors, has been linked to an increase in the activity of the ATP-generating AMPK protein. Since Malat1 was recently shown to be upregulated during hypoxia, the objective of this study was to determine the contribution of AMPK in the mechanistic pathways regulating Malat1 expression in low oxygen conditions. Compared to those cultured in 21% O2 conditions, HeLa cells incubated in 1.5% O2 expressed more Malat1 transcripts. This observation was mimicked in HEK293T cells using a synthetic reporter construct containing 5.6 kb of the human Malat1 promoter, suggesting that hypoxia directly impacted Malat1 gene transcription. Interestingly, pharmacological stimulation of AMPK increased Malat1 promoter transactivation in 21% O2 conditions, whereas inhibition of either AMPK or its upstream activator CaMKK completely abolished the augmentation of Malat1 under hypoxia. Pharmacological modulation of LKB1, another major regulator of AMPK, had no impact on Malat1 promoter transactivation, suggesting that calcium inputs are important in the control of Malat1 expression by AMPK. Overexpression of hypoxia-inducible factor-1α (HIF-1α) increased Malat1 expression in 21% O2 conditions, whereas pharmacological inhibition of HIF-1α blocked the impact of hypoxia on the Malat1 promoter. Taken together, these findings strongly suggest that Malat1 expression is regulated in hypoxic conditions by a CaMKK/AMPK/HIF-1α axis. More research is needed in physiological settings to test the clinical relevance of this pathway.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Regulación Neoplásica de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/fisiopatología , Proteínas Quinasas/metabolismo , ARN Largo no Codificante/genética , Quinasas de la Proteína-Quinasa Activada por el AMP , Western Blotting , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Células HEK293 , Células HeLa , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética , Proteínas Quinasas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Activación Transcripcional , Regulación hacia Arriba
13.
PLoS One ; 7(4): e35110, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22493735

RESUMEN

The development of human calcified aortic stenosis (AS) includes age-dependent processes that have been involved in atherosclerosis, such as infiltration of macrophages in aortic valves, which then promote production of many pro-inflammatory cytokines, including resistin. However, the molecular mechanisms contributing to these processes are not established. Since Sirt1 has been shown to modulate macrophage biology and inflammation, we examined its levels in human AS and tested its impact on resistin expression. Sirt1 mRNA (p = 0.01) and protein (p<0.05) levels were reduced in explanted valves from AS patients (n = 51) compared to those from control (n = 11) patients. Sirt1 mRNA levels were negatively associated with resistin mRNA levels quantified in AS valves (p = 0.02). Stimulation of Sirt1 by resveratrol or virus-driven overexpression robustly diminished resistin mRNA and protein expression in macrophages, whereas down-regulation of Sirt1 triggered a large increase in resistin expression. These effects were direct, as chromatin immunoprecipitation assays showed that Sirt1 physically interacted with the resistin promoter region at an AP-1 response element. Moreover, Sirt1 blocked c-jun-induced resistin transactivation in gene reporter assays. These findings demonstrate that, in calcified AS, levels of Sirt1 are reduced whereas those of resistin are increased within aortic valve leaflets. Our results also suggest that this loss of Sirt1 expression alleviates its inhibition of resistin transcription in macrophages. Although the overall contribution of this process to the underlying mechanisms for AS disease development remains unresolved, these observations suggest that modification of Sirt1 expression and/or activity could represent a novel approach against inflammation in AS.


Asunto(s)
Estenosis de la Válvula Aórtica/metabolismo , Válvula Aórtica/metabolismo , Macrófagos/metabolismo , Resistina/genética , Sirtuina 1/genética , Anciano , Animales , Válvula Aórtica/efectos de los fármacos , Válvula Aórtica/inmunología , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/inmunología , Sitios de Unión , Células Cultivadas , Inmunoprecipitación de Cromatina , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Persona de Mediana Edad , Unión Proteica , ARN Mensajero/biosíntesis , Resistina/metabolismo , Elementos de Respuesta , Resveratrol , Sirtuina 1/metabolismo , Estilbenos/farmacología , Transcripción Genética
14.
Mol Cell Endocrinol ; 358(1): 63-8, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22410287

RESUMEN

Insulin-like growth factor binding protein 2 (IGFBP-2) has been implicated in the etiology of several diseases, including the metabolic syndrome. Although IGFBP-2 derives mostly from the liver, recent evidence in mice and humans indicate that aging and obesity are associated with altered IGFBP-2 levels in white adipocytes. The present study was aimed at determining the mechanisms that control IGFBP-2 expression in mature adipocytes. IGFBP-2 mRNA and protein expression in serum-deprived 3T3-L1 adipocytes were twofold increased by acute insulin treatment. Co-treatments with the phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin or the mammalian target of rapamycin (mTOR) inhibitor rapamycin blunted the effects of insulin. Coherently, IGFBP-2 mRNA levels were robustly increased in adipocytes lacking either TSC2 or 4E-BP1. Insulin triggered the recruitment of CAAT/enhancer binding protein α (C/EBPα) to the IGFBP-2 proximal promoter. These findings suggest that insulin upregulates IGFBP-2 expression through a PI3K/mTOR/C/EBPα pathway in white adipocytes.


Asunto(s)
Adipocitos Blancos/metabolismo , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/biosíntesis , Insulina/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Células 3T3-L1 , Proteínas Adaptadoras Transductoras de Señales , Androstadienos/farmacología , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Portadoras , Proteínas de Ciclo Celular , Línea Celular , Factores Eucarióticos de Iniciación , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Ratones , Fosfoproteínas/deficiencia , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , Sirolimus/farmacología , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/deficiencia , Wortmanina
15.
Aging Cell ; 8(4): 449-59, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19485965

RESUMEN

Age is an important risk factor for the development of metabolic diseases (e.g. obesity, diabetes and atherosclerosis). Yet, little is known about the molecular mechanisms occurring upon aging that affect energy metabolism. Although visceral white adipose tissue (vWAT) is known for its key impact on metabolism, recent studies have indicated it could also be a key regulator of lifespan, suggesting that it can serve as a node for age-associated fat accretion. Here we show that aging triggers changes in the transcriptional milieu of the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma) in vWAT, which leads to a modified potential for transactivation of target genes upon ligand treatment. We found that in vWAT of mice, rats and men, aging induced a specific decrease in the expression of steroid receptor coactivator-1 (SRC-1), whose recruitment to PPARgamma is associated with improved insulin sensitivity and low adipogenic activity. In contrast, aging and oxidative stress did not impact on PPARgamma expression and PPARgamma ligand production. Age-induced loss of PPARgamma/SRC-1 interactions increased the binding of PPARgamma to the promoter of the adipogenic gene aP2. These findings suggest that strategies aimed at increasing SRC-1 expression and recruitment to PPARgamma upon aging might help improve age-associated metabolic disorders.


Asunto(s)
Tejido Adiposo/metabolismo , Envejecimiento/fisiología , Histona Acetiltransferasas/metabolismo , PPAR gamma/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Núcleo Celular , Expresión Génica , Histona Acetiltransferasas/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Coactivador 1 de Receptor Nuclear , Estrés Oxidativo , PPAR gamma/genética , Unión Proteica , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Factores de Transcripción/genética , Transcripción Genética
16.
Mol Cell Biol ; 26(20): 7561-74, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17015477

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARgamma) might not be permissive to ligand activation in prostate cancer cells. Association of PPARgamma with repressing factors or posttranslational modifications in PPARgamma protein could explain the lack of effect of PPARgamma ligands in a recent randomized clinical trial. Using cells and prostate cancer xenograft mouse models, we demonstrate in this study that a combination treatment using the PPARgamma agonist pioglitazone and the histone deacetylase inhibitor valproic acid is more efficient at inhibiting prostate tumor growth than each individual therapy. We show that the combination treatment impairs the bone-invasive potential of prostate cancer cells in mice. In addition, we demonstrate that expression of E-cadherin, a protein involved in the control of cell migration and invasion, is highly up-regulated in the presence of valproic acid and pioglitazone. We show that E-cadherin expression responds only to the combination treatment and not to single PPARgamma agonists, defining a new class of PPARgamma target genes. These results open up new therapeutic perspectives in the treatment of prostate cancer.


Asunto(s)
Cadherinas/metabolismo , PPAR gamma/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Animales , Cadherinas/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Quimioterapia Combinada , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/metabolismo , Humanos , Masculino , Ratones , Invasividad Neoplásica/patología , Trasplante de Neoplasias , PPAR gamma/agonistas , PPAR gamma/genética , Fosforilación/efectos de los fármacos , Pioglitazona , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , ARN Mensajero/genética , Proteína de Retinoblastoma/metabolismo , Tiazolidinedionas/uso terapéutico , Ácido Valproico/uso terapéutico
17.
Mol Cell Biol ; 25(22): 9985-95, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16260612

RESUMEN

In addition to their role in cell cycle progression, new data reveal an emerging role of D-type cyclins in transcriptional regulation and cellular differentiation processes. Using 3T3-L1 cell lines to study adipogenesis, we observed an up-regulation of cyclin D3 expression throughout the differentiation process. Surprisingly, cyclin D3 was only minimally expressed during the initial stages of adipogenesis, when mitotic division is prevalent. This seemingly paradoxical expression led us to investigate a potential cell cycle-independent role for cyclin D3 during adipogenesis. We show here a direct interaction between cyclin D3 and the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma). Our experiments reveal cyclin D3 acts as a ligand-dependent PPARgamma coactivator, which, together with its cyclin-dependent kinase partner, phosphorylates the A-B domain of the nuclear receptor. Overexpression and knockdown studies with cyclin D3 had marked effects on PPARgamma activity and subsequently on adipogenesis. Chromatin immunoprecipitation assays confirm the participation of cyclin D3 in the regulation of PPARgamma target genes. We show that cyclin D3 mutant mice are protected from diet-induced obesity, display smaller adipocytes, have reduced adipogenic gene expression, and are insulin sensitive. Our results indicate that cyclin D3 is an important factor governing adipogenesis and obesity.


Asunto(s)
Adipocitos/citología , Ciclinas/fisiología , PPAR gamma/metabolismo , Adipocitos/metabolismo , Animales , Compuestos Azo/farmacología , Northern Blotting , Western Blotting , Células COS , Línea Celular , Chlorocebus aethiops , Inmunoprecipitación de Cromatina , Ciclina D3 , Quinasa 6 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Dieta , Regulación de la Expresión Génica , Inmunoprecipitación , Insulina/metabolismo , Ratones , Ratones Noqueados , Microscopía Fluorescente , Mutación , Células 3T3 NIH , Obesidad/metabolismo , Plásmidos/metabolismo , ARN Interferente Pequeño/metabolismo , Factores de Tiempo , Transcripción Genética , Regulación hacia Arriba
18.
J Clin Invest ; 113(9): 1288-95, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15124020

RESUMEN

We evaluated the effects of E2F1 on glucose homeostasis using E2F1(-/-) mice. E2F1(-/-) mice show an overall reduction in pancreatic size as the result of impaired postnatal pancreatic growth. Furthermore, these animals have dysfunctional beta cells, linked to impaired PDX-1 activity. Because of the disproportionate small pancreas and dysfunctional islets, E2F1(-/-) mice secrete insufficient amounts of insulin in response to a glucose load, resulting in glucose intolerance. Despite this glucose intolerance, E2F1(-/-) mice do not develop overt diabetes mellitus because they have insulin hypersensitivity, which is secondary to a diminished adipose tissue mass and altered adipocytokine levels, which compensates for the defect in insulin secretion. These data demonstrate that factors controlling cell proliferation, such as E2F1, determine pancreatic growth and function, subsequently affecting metabolic homeostasis.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Homeodominio , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/fisiología , Páncreas/crecimiento & desarrollo , Proteínas Adaptadoras Transductoras de Señales , Adipocitos/citología , Adipocitos/metabolismo , Animales , Apoptosis , Glucemia/metabolismo , División Celular , Regulación de la Expresión Génica , Glucosa/metabolismo , Intolerancia a la Glucosa , Homeostasis , Insulina/sangre , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/citología , Leptina/sangre , Masculino , Ratones , Ratones Noqueados , Músculo Liso/citología , Músculo Liso/metabolismo , Páncreas/citología , Proteínas de Unión al ARN , Factores de Tiempo , Transactivadores/metabolismo
19.
J Lipid Res ; 44(9): 1652-9, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12837847

RESUMEN

Selective cyclo-oxygenase-2 (COX-2) inhibitors are nonsteroidal antiinflammatory drugs used in the management of inflammatory diseases. We demonstrate here that inhibition of the COX-2 enzyme impairs adipocyte differentiation. The inhibition of adipogenesis occurs in the early clonal expansion phase. In particular, COX-2 inhibition limits cell cycle reentry required before terminal adipocyte differentiation. This inhibition of adipogenesis is independent of the production of the peroxisome proliferator activated receptor gamma ligand prostaglandin J2, but dependent on the production of proliferative prostaglandins, such as prostaglandin E2. Modulation of the activity of the COX-2 enzyme via COX-2 selective inhibitors might open up new perspectives in the control of obesity and related metabolic diseases.


Asunto(s)
Adipocitos/citología , Adipocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Clonales/citología , Células Clonales/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/farmacología , Isoenzimas/antagonistas & inhibidores , Células 3T3 , Animales , Antiinflamatorios no Esteroideos/farmacología , Western Blotting , Bromodesoxiuridina/análisis , División Celular/efectos de los fármacos , Ciclina E/análisis , Ciclina E/biosíntesis , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Técnica del Anticuerpo Fluorescente , Ratones , Mitógenos/farmacología , Prostaglandina-Endoperóxido Sintasas , Receptores Citoplasmáticos y Nucleares/agonistas , Factores de Transcripción/agonistas
20.
Oncogene ; 22(27): 4186-93, 2003 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-12833141

RESUMEN

The nuclear receptor PPARgamma is implicated in the control of cell proliferation and apoptosis. However, the molecular mechanisms by which it controls these processes remain largely elusive. We show here that PPARgamma activation in the presence of the retinoblastoma protein (RB) results in the arrest of cells at the G1 phase of the cell cycle, whereas in the absence of RB, cells accumulate in G2/M, endoreduplicate, and undergo apoptosis. Through the use of HDAC inhibitors and coimmunoprecipitations, we furthermore demonstrate that the effects of RB on PPARgamma-mediated control of the cell cycle and apoptosis depend on the recruitment of histone deacetylase 3 (HDAC3) to PPARgamma. In combination, these data hence demonstrate that the effects of PPARgamma on cell proliferation and apoptosis are dependent on the presence of an RB-HDAC3 complex.


Asunto(s)
Apoptosis , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteína de Retinoblastoma/metabolismo , Tiazolidinedionas , Factores de Transcripción/metabolismo , Células 3T3 , Adipocitos/metabolismo , Animales , Northern Blotting , Western Blotting , Bromodesoxiuridina/farmacología , Ciclo Celular , División Celular , Núcleo Celular/metabolismo , Citometría de Flujo , Fase G1 , Fase G2 , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Etiquetado Corte-Fin in Situ , Luciferasas/metabolismo , Ratones , Microscopía Fluorescente , Mitosis , Neoplasias/metabolismo , Plásmidos/metabolismo , Pruebas de Precipitina , ARN Interferente Pequeño/metabolismo , Rosiglitazona , Tiazoles/farmacología , Transcripción Genética , Activación Transcripcional , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...