Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(7)2022 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-35408841

RESUMEN

Post-translational modifications of proteins ensure optimized cellular processes, including proteostasis, regulated signaling, cell survival, and stress adaptation to maintain a balanced homeostatic state. Abnormal post-translational modifications are associated with cellular dysfunction and the occurrence of life-threatening diseases, such as cancer and neurodegenerative diseases. Therefore, some of the frequently seen protein modifications have been used as disease markers, while others are targeted for developing specific therapies. The ubiquitin and ubiquitin-like post-translational modifiers, namely, small ubiquitin-like modifier (SUMO) and neuronal precursor cell-expressed developmentally down-regulated protein 8 (NEDD8), share several features, such as protein structures, enzymatic cascades mediating the conjugation process, and targeted amino acid residues. Alterations in the regulatory mechanisms lead to aberrations in biological processes during tumorigenesis, including the regulation of tumor metabolism, immunological modulation of the tumor microenvironment, and cancer stem cell stemness, besides many more. Novel insights into ubiquitin and ubiquitin-like pathways involved in cancer biology reveal a potential interplay between ubiquitination, SUMOylation, and NEDDylation. This review outlines the current understandings of the regulatory mechanisms and assay capabilities of ubiquitination, SUMOylation, and NEDDylation. It will further highlight the role of ubiquitination, SUMOylation, and NEDDylation in tumorigenesis.


Asunto(s)
Neoplasias , Sumoilación , Carcinogénesis , Humanos , Neoplasias/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas/metabolismo , Microambiente Tumoral , Ubiquitina/metabolismo , Ubiquitinación
2.
G3 (Bethesda) ; 11(12)2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34586382

RESUMEN

The spindle assembly checkpoint protects the integrity of the genome by ensuring that chromosomes are properly attached to the mitotic spindle before they are segregated during anaphase. Activation of the spindle checkpoint results in inhibition of the Anaphase-Promoting Complex (APC), an E3 ubiquitin ligase that triggers the metaphase-anaphase transition. Here, we show that levels of Ubc1, an E2 enzyme that functions in complex with the APC, modulate the response to spindle checkpoint activation in Saccharomyces cerevisiae. Overexpression of Ubc1 increased resistance to microtubule poisons, whereas Ubc1 shut-off sensitized cells. We also found that Ubc1 levels are regulated by the spindle checkpoint. Checkpoint activation or direct APC inhibition led to a decrease in Ubc1 levels, charging, and half-life. Additionally, stabilization of Ubc1 prevented its down-regulation by the spindle checkpoint and increased resistance to checkpoint-activating drugs. These results suggest that down-regulation of Ubc1 in response to spindle checkpoint signaling is necessary for a robust cell cycle arrest.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Anafase , Ciclosoma-Complejo Promotor de la Anafase/genética , Proteínas Cdc20 , Proteínas de Ciclo Celular/genética , Puntos de Control de la Fase M del Ciclo Celular , Mitosis , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Huso Acromático , Enzimas Ubiquitina-Conjugadoras/genética
3.
Sci Rep ; 11(1): 5749, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33707480

RESUMEN

Reactive oxygen species (ROS) are implicated in triggering cell signalling events and pathways to promote and maintain tumorigenicity. Chemotherapy and radiation can induce ROS to elicit cell death allows for targeting ROS pathways for effective anti-cancer therapeutics. Coenzyme Q10 is a critical cofactor in the electron transport chain with complex biological functions that extend beyond mitochondrial respiration. This study demonstrates that delivery of oxidized Coenzyme Q10 (ubidecarenone) to increase mitochondrial Q-pool is associated with an increase in ROS generation, effectuating anti-cancer effects in a pancreatic cancer model. Consequent activation of cell death was observed in vitro in pancreatic cancer cells, and both human patient-derived organoids and tumour xenografts. The study is a first to demonstrate the effectiveness of oxidized ubidecarenone in targeting mitochondrial function resulting in an anti-cancer effect. Furthermore, these findings support the clinical development of proprietary formulation, BPM31510, for treatment of cancers with high ROS burden with potential sensitivity to ubidecarenone.


Asunto(s)
Apoptosis , Mitocondrias/metabolismo , Neoplasias Pancreáticas/patología , Especies Reactivas de Oxígeno/metabolismo , Ubiquinona/análogos & derivados , Animales , Línea Celular Tumoral , Proliferación Celular , Respiración de la Célula , Supervivencia Celular , Complejo II de Transporte de Electrones/metabolismo , Glicerol-3-Fosfato Deshidrogenasa (NAD+) , Humanos , Potencial de la Membrana Mitocondrial , Ratones Desnudos , Organoides/patología , Estrés Oxidativo , Consumo de Oxígeno , Neoplasias Pancreáticas/metabolismo , Especificidad por Sustrato , Ubiquinona/metabolismo
4.
Sci Rep ; 10(1): 13899, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32807842

RESUMEN

Metabolic reprogramming in cancer cells, vs. non-cancer cells, elevates levels of reactive oxygen species (ROS) leading to higher oxidative stress. The elevated ROS levels suggest a vulnerability to excess prooxidant loads leading to selective cell death, a therapeutically exploitable difference. Co-enzyme Q10 (CoQ10) an endogenous mitochondrial resident molecule, plays an important role in mitochondrial redox homeostasis, membrane integrity, and energy production. BPM31510 is a lipid-drug conjugate nanodispersion specifically formulated for delivery of supraphysiological concentrations of ubidecarenone (oxidized CoQ10) to the cell and mitochondria, in both in vitro and in vivo model systems. In this study, we sought to investigate the therapeutic potential of ubidecarenone in the highly treatment-refractory glioblastoma. Rodent (C6) and human (U251) glioma cell lines, and non-tumor human astrocytes (HA) and rodent NIH3T3 fibroblast cell lines were utilized for experiments. Tumor cell lines exhibited a marked increase in sensitivity to ubidecarenone vs. non-tumor cell lines. Further, elevated mitochondrial superoxide production was noted in tumor cells vs. non-tumor cells hours before any changes in proliferation or the cell cycle could be detected. In vitro co-culture experiments show ubidecarenone differentially affecting tumor cells vs. non-tumor cells, resulting in an equilibrated culture. In vivo activity in a highly aggressive orthotopic C6 glioma model demonstrated a greater than 25% long-term survival rate. Based on these findings we conclude that high levels of ubidecarenone delivered using BPM31510 provide an effective therapeutic modality targeting cancer-specific modulation of redox mechanisms for anti-cancer effects.


Asunto(s)
Sistemas de Liberación de Medicamentos , Glioma/patología , Lípidos/química , Nanopartículas/química , Preparaciones Farmacéuticas/química , Ubiquinona/análogos & derivados , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Glioma/tratamiento farmacológico , Humanos , Ratones , Células 3T3 NIH , Oxidación-Reducción , Ratas Wistar , Superóxidos/metabolismo , Ubiquinona/administración & dosificación , Ubiquinona/farmacología , Ubiquinona/uso terapéutico
5.
J Pharm Biomed Anal ; 173: 40-46, 2019 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-31108422

RESUMEN

Ubiquitin plays an essential role in modulating protein function, and deregulation of the ubiquitin system leads to the development of a variety of human diseases. E3 Ubiquitin ligases that mediate ubiquitination and degradation of caspases prevent apoptosis, and as such belong to the family of inhibitors of apoptosis proteins (IAPs). Diablo is a substrate of IAPs but also a negative regulator of IAPs in apoptotic pathway as it blocks the interaction between IAPs and caspases. In efforts to identify IAP inhibitors, we developed sandwich immunoassays in conjunction with an electrochemical luminescence (ECL) platform for quantitation of total Diablo, ubiquitinated Diablo, and ubiquitinated Diablo with K48-specific linkage. The assay panel detects Diablo ubiquitination level changes in the presence of IAP inhibitor or proteasome inhibitor, demonstrating its potential as a cost-efficient high-throughput method for drug discovery involving IAP ubiquitination cascade. The ECL based sandwich assay panel performance was subsequently evaluated for precision, linearity, and limit of quantification.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/aislamiento & purificación , Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Proteínas Mitocondriales/aislamiento & purificación , Proteína Inhibidora de la Apoptosis Ligada a X/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Humanos , Inmunoensayo/métodos , Mediciones Luminiscentes/métodos , Proteínas Mitocondriales/metabolismo , Inhibidores de Proteasoma/farmacología , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Ubiquitinación/efectos de los fármacos , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
6.
Cell Biosci ; 8: 45, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30140426

RESUMEN

BACKGROUND: Diabetes mellitus is associated with an increased risk in diabetic cardiomyopathy (DCM) that is distinctly not attributed to co-morbidities with other vasculature diseases. To date, while dysregulation of calcium handling is a key hallmark in cardiomyopathy, studies have been inconsistent in the types of alterations involved. In this study human cardiomyocytes were exposed to an environmental nutritional perturbation of high glucose, fatty acids, and l-carnitine to model DCM and iTRAQ-coupled LC-MS/MS proteomic analysis was used to capture proteins affected by the perturbation. The proteins captured were then compared to proteins currently annotated in the cardiovascular disease (CVD) gene ontology (GO) database to identify proteins not previously described as being related to CVD. Subsequently, GO analysis for calcium regulating proteins and endoplasmic/sarcoplasmic reticulum (ER/SR) associated proteins was carried out. RESULTS: Here, we identified CCDC47 (calumin) as a unique calcium regulating protein altered in our in vitro nutritional perturbation model. The cellular and functional role of CCDC47 was then assessed in rat cardiomyocytes. In rat H9C2 myocytes, overexpression of CCDC47 resulted in increase in ionomycin-induced calcium release and reuptake. Of interest, in a diet-induced obese (DIO) rat model of DCM, CCDC47 mRNA expression was increased in the atrium and ventricle of the heart, but CCDC47 protein expression was significantly increased only in the atrium of DIO rats compared to lean control rats. Notably, no changes in ANP, BNP, or ß-MHC were observed between DIO rats and lean control rats. CONCLUSIONS: Together, our in vitro and in vivo studies demonstrate that CCDC47 is a unique calcium regulating protein that is associated with early onset hypertrophic cardiomyopathy.

7.
Nutr Metab (Lond) ; 15: 11, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29434648

RESUMEN

BACKGROUND: Inhibition of Hsp90 has been shown to improve glucose tolerance and insulin sensitivity in mouse models of diabetes. In the present report, the specific isoform Hsp90ab1, was identified as playing a major role in regulating insulin signaling and glucose metabolism. METHODS: In a diet-induced obese (DIO) mouse model of diabetes, expression of various Hsp90 isoforms in skeletal tissue was examined. Subsequent experiments characterized the role of Hsp90ab1 isoform in glucose metabolism and insulin signaling in primary human skeletal muscle myoblasts (HSMM) and a DIO mouse model. RESULTS: In DIO mice Hsp90ab1 mRNA was upregulated in skeletal muscle compared to lean mice and knockdown using anti-sense oligonucleotide (ASO) resulted in reduced expression in skeletal muscle that was associated with improved glucose tolerance, reduced fed glucose and fed insulin levels compared to DIO mice that were treated with a negative control oligonucleotide. In addition, knockdown of HSP90ab1 in DIO mice was associated with reduced pyruvate dehydrogenase kinase-4 mRNA and phosphorylation of the muscle pyruvate dehydrogenase complex (at serine 232, 293 and 300), but increased phosphofructokinase 1, glycogen synthase 1 and long-chain specific acyl-CoA dehydrogenase mRNA. In HSMM, siRNA knockdown of Hsp90ab1 induced an increase in substrate metabolism, mitochondrial respiration capacity, and insulin sensitivity, providing further evidence for the role of Hsp90ab1 in metabolism. CONCLUSIONS: The data support a novel role for Hsp90ab1 in arbitrating skeletal muscle plasticity via modulation of substrate utilization including glucose and fatty acids in normal and disease conditions. Hsp90ab1 represents a novel target for potential treatment of metabolic disease including diabetes.

8.
BMC Genomics ; 18(1): 987, 2017 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-29273013

RESUMEN

BACKGROUND: Exosomes and other extracellular vesicles (EVs) have emerged as an important mechanism of cell-to-cell communication. However, previous studies either did not fully resolve what genetic materials were shuttled by exosomes or only focused on a specific set of miRNAs and mRNAs. A more systematic method is required to identify the genetic materials that are potentially transferred during cell-to-cell communication through EVs in an unbiased manner. RESULTS: In this work, we present a novel next generation of sequencing (NGS) based approach to identify EV mediated mRNA exchanges between co-cultured adipocyte and macrophage cells. We performed molecular and genomic profiling and jointly considered data from RNA sequencing (RNA-seq) and genotyping to track the "sequence varying mRNAs" transferred between cells. We identified 8 mRNAs being transferred from macrophages to adipocytes and 21 mRNAs being transferred in the opposite direction. These mRNAs represented biological functions including extracellular matrix, cell adhesion, glycoprotein, and signal peptides. CONCLUSIONS: Our study sheds new light on EV mediated RNA communications between adipocyte and macrophage cells, which may play a significant role in developing insulin resistance in diabetic patients. This work establishes a new method that is applicable to examining genetic material exchanges in many cellular systems and has the potential to be extended to in vivo studies as well.


Asunto(s)
Comunicación Celular , Vesículas Extracelulares/metabolismo , ARN Mensajero/metabolismo , Adipocitos/metabolismo , Línea Celular , Técnicas de Cocultivo , Expresión Génica , Técnicas de Genotipaje , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Macrófagos/metabolismo , Transporte de ARN , Análisis de Secuencia de ARN
9.
Mediators Inflamm ; 2017: 9067049, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28458470

RESUMEN

Obesity is marked by chronic, low-grade inflammation. Here, we examined whether intrinsic differences between white and brown adipocytes influence the inflammatory status of macrophages. White and brown adipocytes were characterized by transcriptional regulation of UCP-1, PGC1α, PGC1ß, and CIDEA and their level of IL-6 secretion. The inflammatory profile of PMA-differentiated U937 and THP-1 macrophages, in resting state and after stimulation with LPS/IFN-gamma and IL-4, was assessed by measuring IL-6 secretion and transcriptional regulation of a panel of inflammatory genes after mono- or indirect coculture with white and brown adipocytes. White adipocyte monocultures show increased IL-6 secretion compared to brown adipocytes. White adipocytes cocultured with U937 and THP-1 macrophages induced a greater increase in IL-6 secretion compared to brown adipocytes cocultured with both macrophages. White adipocytes cocultured with macrophages increased inflammatory gene expression in both types. In contrast, macrophages cocultured with brown adipocytes induced downregulation or no alterations in inflammatory gene expression. The effects of adipocytes on macrophages appear to be independent of stimulation state. Brown adipocytes exhibit an intrinsic ability to dampen inflammatory profile of macrophages, while white adipocytes enhance it. These data suggest that brown adipocytes may be less prone to adipose tissue inflammation that is associated with obesity.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Adipocitos Marrones/efectos de los fármacos , Adipocitos Marrones/inmunología , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/inmunología , Adulto , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Perfilación de la Expresión Génica , Humanos , Inflamación/inmunología , Interleucina-4/farmacología , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Persona de Mediana Edad
10.
Future Sci OA ; 3(1): FSO161, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28344825

RESUMEN

AIM: A novel strategy for prostate cancer (PrCa) biomarker discovery is described. MATERIALS & METHODS: In vitro perturbation biology, proteomics and Bayesian causal analysis identified biomarkers that were validated in in vitro models and clinical specimens. RESULTS: Filamin-B (FLNB) and Keratin-19 were identified as biomarkers. Filamin-A (FLNA) was found to be causally linked to FLNB. Characterization of the biomarkers in a panel of cells revealed differential mRNA expression and regulation. Moreover, FLNA and FLNB were detected in the conditioned media of cells. Last, in patients without PrCa, FLNA and FLNB blood levels were positively correlated, while in patients with adenocarcinoma the relationship is dysregulated. CONCLUSION: These data support the strategy and the potential use of the biomarkers for PrCa.

11.
Obesity (Silver Spring) ; 24(8): 1695-703, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27312141

RESUMEN

OBJECTIVE: Collagen VI alpha 3 (COL6A3) is associated with insulin resistance and adipose tissue inflammation. In this study, the role of COL6A3 in human adipocyte function was characterized. METHODS: Immortalized human preadipocyte cell lines stably expressing control or COL6A3 shRNA were used to study adipocyte function and inflammation. RESULTS: COL6A3 knockdown increased triglyceride content, lipolysis, insulin-induced Akt phosphorylation, and mRNA expression of key adipogenic genes (peroxisome proliferator-activated receptor-γ, glucose transporter, adiponectin, and fatty acid binding protein), indicating increased adipocyte function and insulin sensitivity. However, COL6A3 knockdown decreased basal adipocyte chemokine (C-C motif) ligand 2 [CCL2, monocyte chemoattractant protein (MCP1)] mRNA expression, reduced secreted protein levels, and abrogated tumor necrosis factor-α- and lipopolysaccharide-induced MCP1 mRNA expression. In addition, while control adipocytes co-cultured with THP1 macrophages showed a threefold increase in adipocyte MCP1 mRNA expression, in COL6A3 knockdown adipocytes MCP1 mRNA expression was unaltered by co-culturing. Lastly, in normal differentiated adipocytes, matrix metalloproteinase-11 treatment reduced expression of COL6A3 protein, MCP1 mRNA, MCP1 secretion, and abrogated tumor necrosis factor-α- and lipopolysaccharide-induced MCP1 mRNA expression and protein secretion. CONCLUSIONS: COL6A3 knockdown in adipocytes leads to the development of a unique state of inflammatory resistance via suppression of MCP1 induction.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Colágeno Tipo VI/metabolismo , ARN Mensajero/metabolismo , Adiponectina , Animales , Diferenciación Celular/efectos de los fármacos , Quimiocina CCL2/metabolismo , Colágeno/metabolismo , Proteínas de Unión a Ácidos Grasos/metabolismo , Humanos , Inflamación/metabolismo , Insulina/metabolismo , Resistencia a la Insulina , Macrófagos/metabolismo , Obesidad/metabolismo , PPAR gamma/metabolismo
12.
Proc Natl Acad Sci U S A ; 110(28): 11409-14, 2013 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-23798383

RESUMEN

Visceral and s.c. fat exhibit different intrinsic properties, including rates of lipolysis, and are associated with differential risk for the development of type 2 diabetes. These effects are in part related to cell autonomous differences in gene expression. In the present study, we show that expression of Shox2 (Short stature homeobox 2) is higher in s.c. than visceral fat in both rodents and humans and that levels are further increased in humans with visceral obesity. Fat-specific disruption of Shox2 in male mice results in protection from high fat diet-induced obesity, with a preferential loss of s.c. fat. The reduced adipocyte size is secondary to a twofold increase in the expression of ß3 adrenergic receptor (Adrb3) at both the mRNA and protein level and a parallel increase in lipolytic rate. These effects are mimicked by knockdown of Shox2 in C3H10T1/2 cells. Conversely, overexpression of Shox2 leads to a repression of Adrb3 expression and decrease lipolytic rate. Shox2 does not affect differentiation but directly interacts with CCAAT/enhancer binding protein alpha and attenuates its transcriptional activity of the Adrb3 promoter. Thus, Shox2 can regulate the expression of Adrb3 and control the rate of lipolysis and, in this way, exerts control of the phenotypic differences between visceral and s.c. adipocytes.


Asunto(s)
Adipocitos/citología , Proteínas de Homeodominio/fisiología , Animales , Dieta , Proteínas de Homeodominio/genética , Resistencia a la Insulina , Lipólisis , Ratones , Ratones Noqueados , Obesidad/etiología , Obesidad/genética
13.
Diabetes ; 61(7): 1691-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22596050

RESUMEN

Obesity and body fat distribution are important risk factors for the development of type 2 diabetes and metabolic syndrome. Evidence has accumulated that this risk is related to intrinsic differences in behavior of adipocytes in different fat depots. In the current study, we demonstrate that adipocyte precursor cells (APCs) isolated from visceral and subcutaneous white adipose depots of mice have distinct patterns of gene expression, differentiation potential, and response to environmental and genetic influences. APCs derived from subcutaneous fat differentiate well in the presence of classical induction cocktail, whereas those from visceral fat differentiate poorly but can be induced to differentiate by addition of bone morphogenetic protein (BMP)-2 or BMP-4. This difference correlates with major differences in gene expression signature between subcutaneous and visceral APCs. The number of APCs is higher in obesity-prone C57BL/6 mice than obesity-resistant 129 mice, and the number in both depots is increased by up to 270% by exposure of mice to high-fat diet. Thus, APCs from visceral and subcutaneous depots are dynamic populations, which have intrinsic differences in gene expression, differentiation properties, and responses to environmental/genetic factors. Regulation of these populations may provide a new target for the treatment and prevention of obesity and its metabolic complications.


Asunto(s)
Adipocitos Blancos/metabolismo , Grasa Intraabdominal/metabolismo , Células Madre/metabolismo , Grasa Subcutánea Abdominal/metabolismo , Animales , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular , Dieta Alta en Grasa , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Obesidad/metabolismo
14.
Cell Metab ; 14(4): 491-503, 2011 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-21982709

RESUMEN

In obesity, adipocytes distant from vasculature become hypoxic and dysfunctional. This hypoxic response is mediated by hypoxia-inducible factors (Hif1α, Hif2α, and Hif3α) and their obligate partner, Hif1ß (Arnt). We show that mice lacking Hif1ß in fat (FH1ßKO) are lean, exhibit reduced adipocyte size, and are protected from age- and diet-induced glucose intolerance. There is also reduced Vegf and vascular permeability in FH1ßKO fat, but diet-induced inflammation and fibrosis is unchanged. Adipocytes from FH1ßKO mice have reduced glucose uptake due to decreased Glut1 and Glut4, which is mirrored in 3T3-L1 adipocytes with Hif1ß knockdown. Hif1ß knockdown cells also fail to respond appropriately to hypoxia with reduced cellular respiration and reduced mitochondrial gene expression. Some, but not all, of these effects are reproduced by Hif1α knockdown. Thus, Hif1ß/Arnt regulates glucose uptake, mitochondrial gene expression, and vascular permeability to control adipose mass and function, providing a target for obesity therapy.


Asunto(s)
Adipocitos/metabolismo , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Fibrosis/fisiopatología , Inflamación/fisiopatología , Células 3T3-L1 , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/antagonistas & inhibidores , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Hipoxia de la Célula , Femenino , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Obesidad/metabolismo
15.
Proc Natl Acad Sci U S A ; 108(7): 2771-6, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21282637

RESUMEN

Increased intraabdominal (visceral) fat is associated with a high risk of diabetes and metabolic syndrome. We have previously shown that the mesodermal developmental transcription factor Tbx15 is highly differentially expressed between visceral and subcutaneous (s.c.) fat in both humans and rodents, and in humans visceral fat Tbx15 expression is decreased in obesity. Here we show that, in mice, Tbx15 is 260-fold more highly expressed in s.c. preadipocytes than in epididymal preadipocytes. Overexpression of Tbx15 in 3T3-L1 preadipocytes impairs adipocyte differentiation and decreases triglyceride content. This defect in differentiation can be corrected by stimulating cells with the PPARγ agonist rosiglitazone (Rosi). However, triglyceride accumulation remains decreased by ∼50%, due to a decrease in basal lipogenic rate and increase in basal lipolytic rate. 3T3-L1 preadipocytes overexpressing Tbx15 also have a 15% reduction in mitochondrial mass and a 28% reduction in basal mitochondrial respiration (P = 0.004) and ATP turnover (P = 0.02), and a 45% (P = 0.003) reduction in mitochondrial respiratory capacity. Thus, differential expression of Tbx15 between fat depots plays an important role in the interdepot differences in adipocyte differentiation, triglyceride accumulation, and mitochondrial function that may contribute to the risk of diabetes and metabolic disease.


Asunto(s)
Adipocitos/fisiología , Diferenciación Celular/genética , Respiración de la Célula/genética , Mitocondrias/fisiología , Grasa Subcutánea/metabolismo , Proteínas de Dominio T Box/metabolismo , Células 3T3-L1 , Adenosina Trifosfato/metabolismo , Animales , Compuestos Azo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Respiración de la Célula/fisiología , Clonación Molecular , Cartilla de ADN/genética , Metabolismo Energético/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Consumo de Oxígeno/fisiología , PPAR gamma/agonistas , Reacción en Cadena de la Polimerasa , Rosiglitazona , Proteínas de Dominio T Box/genética , Tiazolidinedionas/farmacología
16.
Obesity (Silver Spring) ; 18(5): 872-878, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20111017

RESUMEN

We have previously demonstrated that subcutaneous and intra-abdominal adipose tissue show different patterns of expression for developmental genes (Shox2, En1, Tbx15 Hoxa5, Hoxc8, and Hoxc9), and that the expression level of Tbx15 and Hoxa5 in humans correlated with the level of obesity and fat distribution. To further explore the role of these developmental genes in adipose tissue, we have characterized their expression in different adipose depots in mice, and studied their regulation in obesity and by fasting. Developmental and adipogenic gene expression was compared in two subcutaneous and three intra-abdominal white adipose tissue (WAT) depots as well as brown adipose tissue (BAT) from lean or obese mice in a fed or fasting state. Each of these six adipose depots display a unique pattern of developmental gene expression, whereas expression of adipogenic transcription factors PPARgamma2 C/EBPalpha, beta, and Delta showed constant expression levels in all depots. Expression levels of developmental genes were similar in obese (ob/ob and high-fat diet (HFD)) and lean mice in most depots. Fasting systematically decreased expression of Hoxc8, PPARgamma2, and increased C/EBPDelta in both lean and ob/ob mice, but produced only variable changes in the expression of other developmental and adipogenic genes. These data indicate that each fat depot has a unique developmental gene expression signature, which is largely independent of nutritional state. This finding further supports a fundamental role of developmental genes in fat distribution and the development and/or function of specific adipose tissue depots.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Grasa Intraabdominal/metabolismo , Obesidad/genética , Adiposidad/genética , Animales , Ayuno/fisiología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Obesos , Obesidad/metabolismo , Distribución Aleatoria , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Diabetes ; 58(11): 2607-15, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19720802

RESUMEN

OBJECTIVE: The G-protein-coupled receptor GPR40 mediates fatty acid potentiation of glucose-stimulated insulin secretion, but its contribution to insulin secretion in vivo and mechanisms of action remain uncertain. This study was aimed to ascertain whether GPR40 controls insulin secretion in vivo and modulates intracellular fuel metabolism in islets. RESEARCH DESIGN AND METHODS: Insulin secretion and sensitivity were assessed in GPR40 knockout mice and their wild-type littermates by hyperglycemic clamps and hyperinsulinemic euglycemic clamps, respectively. Transcriptomic analysis, metabolic studies, and lipid profiling were used to ascertain whether GPR40 modulates intracellular fuel metabolism in islets. RESULTS: Both glucose- and arginine-stimulated insulin secretion in vivo were decreased by approximately 60% in GPR40 knockout fasted and fed mice, without changes in insulin sensitivity. Neither gene expression profiles nor intracellular metabolism of glucose and palmitate in isolated islets were affected by GPR40 deletion. Lipid profiling of isolated islets revealed that the increase in triglyceride and decrease in lyso-phosphatidylethanolamine species in response to palmitate in vitro was similar in wild-type and knockout islets. In contrast, the increase in intracellular inositol phosphate levels observed in wild-type islets in response to fatty acids in vitro was absent in knockout islets. CONCLUSIONS: These results indicate that deletion of GPR40 impairs insulin secretion in vivo not only in response to fatty acids but also to glucose and arginine, without altering intracellular fuel metabolism in islets, via a mechanism that may involve the generation of inositol phosphates downstream of GPR40 activation.


Asunto(s)
Glucosa/farmacología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Tejido Adiposo/anatomía & histología , Animales , Ácido Araquidónico/metabolismo , Arginina/farmacología , Ácidos Grasos/metabolismo , Ácidos Grasos/farmacología , Perfilación de la Expresión Génica , Fosfatos de Inositol/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Lípidos/aislamiento & purificación , Lípidos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
18.
Diabetes ; 58(2): 460-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19017762

RESUMEN

OBJECTIVE: Low birth weight (LBW) is associated with increased risk of obesity, diabetes, and cardiovascular disease during adult life. Moreover, this programmed disease risk can progress to subsequent generations. We previously described a mouse model of LBW, produced by maternal caloric undernutrition (UN) during late gestation. LBW offspring (F(1)-UN generation) develop progressive obesity and impaired glucose tolerance (IGT) with aging. We aimed to determine whether such metabolic phenotypes can be transmitted to subsequent generations in an experimental model, even in the absence of altered nutrition during the second pregnancy. RESEARCH DESIGN AND METHODS: We intercrossed female and male F(1) adult control (C) and UN mice and characterized metabolic phenotypes in F(2) offspring. RESULTS: We demonstrate that 1) reduced birth weight progresses to F(2) offspring through the paternal line (Cfemale -Cmale = 1.64 g; Cfemale -UNmale = 1.57 g, P < 0.05; UNfemale -Cmale = 1.64 g; UNfemale -UNmale = 1.60 g, P < 0.05), 2) obesity progresses through the maternal line (percent body fat: Cfemale -Cmale = 22.4%; Cfemale -UNmale = 22.9%; UNfemale -Cmale = 25.9%, P < 0.05; UNfemale -UNmale = 27.5%, P < 0.05), and 3) IGT progresses through both parental lineages (glucose tolerance test area under curve Cfemale -Cmale = 100; Cfemale -UNmale = 122, P < 0.05; UNfemale -Cmale = 131, P < 0.05; UNfemale -UNmale = 151, P < 0.05). Mechanistically, IGT in both F(1) and F(2) generations is linked to impaired beta-cell function, explained, in part, by dysregulation of Sur1 expression. CONCLUSIONS: Maternal undernutrition during pregnancy (F(0)) programs reduced birth weight, IGT, and obesity in both first- and second-generation offspring. Sex-specific transmission of phenotypes implicates complex mechanisms including alterations in the maternal metabolic environment (transmaternal inheritance of obesity), gene expression mediated by developmental and epigenetic pathways (transpaternal inheritance of LBW), or both (IGT).


Asunto(s)
Intolerancia a la Glucosa/fisiopatología , Desnutrición/fisiopatología , Obesidad/fisiopatología , Animales , Animales Recién Nacidos , Peso al Nacer/fisiología , Femenino , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/metabolismo , Masculino , Desnutrición/complicaciones , Desnutrición/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Intercambio Materno-Fetal/fisiología , Ratones , Obesidad/etiología , Obesidad/metabolismo , Embarazo , Factores de Tiempo
19.
Cell Metab ; 7(5): 410-20, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18460332

RESUMEN

Subcutaneous (SC) and visceral (VIS) obesity are associated with different risks of diabetes and the metabolic syndrome. To elucidate whether these differences are due to anatomic location or intrinsic differences in adipose depots, we characterized mice after transplantation of SC or VIS fat from donor mice into either SC or VIS regions of recipient mice. The group with SC fat transplanted into the VIS cavity exhibited decreased body weight, total fat mass, and glucose and insulin levels. These mice also exhibited improved insulin sensitivity during hyperinsulinemic-euglycemic clamps with increased whole-body glucose uptake, glucose uptake into endogenous fat, and insulin suppression of hepatic glucose production. These effects were observed to a lesser extent with SC fat transplanted to the SC area, whereas VIS fat transplanted to the VIS area was without effect. These data suggest that SC fat is intrinsically different from VIS fat and produces substances that can act systemically to improve glucose metabolism.


Asunto(s)
Tejido Adiposo/metabolismo , Grasa Intraabdominal/trasplante , Redes y Vías Metabólicas/fisiología , Obesidad/complicaciones , Grasa Subcutánea/trasplante , Abdomen , Animales , Peso Corporal , Técnica de Clampeo de la Glucosa , Prueba de Tolerancia a la Glucosa , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/fisiopatología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Aging Cell ; 6(6): 827-39, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18001293

RESUMEN

Caloric restriction, leanness and decreased activity of insulin/insulin-like growth factor 1 (IGF-1) receptor signaling are associated with increased longevity in a wide range of organisms from Caenorhabditis elegans to humans. Fat-specific insulin receptor knock-out (FIRKO) mice represent an interesting dichotomy, with leanness and increased lifespan, despite normal or increased food intake. To determine the mechanisms by which a lack of insulin signaling in adipose tissue might exert this effect, we performed physiological and gene expression studies in FIRKO and control mice as they aged. At the whole body level, FIRKO mice demonstrated an increase in basal metabolic rate and respiratory exchange ratio. Analysis of gene expression in white adipose tissue (WAT) of FIRKO mice from 6 to 36 months of age revealed persistently high expression of the nuclear-encoded mitochondrial genes involved in glycolysis, tricarboxylic acid cycle, beta-oxidation and oxidative phosphorylation as compared to expression of the same genes in WAT from controls that showed a tendency to decline in expression with age. These changes in gene expression were correlated with increased cytochrome c and cytochrome c oxidase subunit IV at the protein level, increased citrate synthase activity, increased expression of peroxisome proliferator-activated receptor gamma coactivator 1alpha (PGC-1alpha) and PGC-1beta, and an increase in mitochondrial DNA in WAT of FIRKO mice. Together, these data suggest that maintenance of mitochondrial activity and metabolic rates in adipose tissue may be important contributors to the increased lifespan of the FIRKO mouse.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Expresión Génica , Genes Mitocondriales/genética , Longevidad/fisiología , Mitocondrias/metabolismo , Delgadez/metabolismo , Tejido Adiposo Blanco/química , Animales , Citrato (si)-Sintasa/metabolismo , Ciclo del Ácido Cítrico/genética , Citocromos c/metabolismo , ADN Mitocondrial/análisis , Transporte de Electrón/genética , Complejo IV de Transporte de Electrones/metabolismo , Glucólisis/genética , Longevidad/genética , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/ultraestructura , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxidación-Reducción , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Receptor de Insulina/genética , Delgadez/genética , Transactivadores/metabolismo , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...