Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Acta Physiol (Oxf) ; 238(1): e13935, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36650072

RESUMEN

AIM: Valuable studies have tested the role of UCP1 on body temperature maintenance in mice, and we sought to knockout Ucp1 in rats (Ucp1-/- ) to provide insight into thermogenic mechanisms in larger mammals. METHODS: We used CRISPR/Cas9 technology to create Ucp1-/- rats. Body weight and adiposity were measured, and rats were subjected to indirect calorimetry. Rats were maintained at room temperature or exposed to 4°C for either 24 h or 14 days. Analyses of brown and white adipose tissue and skeletal muscle were conducted via histology, western blot comparison of oxidative phosphorylation proteins, and qPCR to compare mitochondrial DNA levels and mRNA expression profiles. RNA-seq was performed in skeletal muscle. RESULTS: Ucp1-/- rats withstood 4°C for 14 days, but core temperature steadily declined. All rats lost body weight after 14 days at 4°C, but controls increased food intake more robustly than Ucp1-/- rats. Brown adipose tissue showed signs of decreased activity in Ucp1-/- rats, while mitochondrial lipid metabolism markers in white adipose tissue and skeletal muscle were increased. Ucp1-/- rats displayed more visible shivering and energy expenditure than controls at 4°C. Skeletal muscle transcriptomics showed more differences between genotypes at 23°C than at 4°C. CONCLUSION: Room temperature presented sufficient cold stress to rats lacking UCP1 to activate compensatory thermogenic mechanisms in skeletal muscle, which were only activated in control rats following exposure to 4°C. These results provide novel insight into thermogenic responses to UCP1 deficiency; and highlight Ucp1-/- rats as an attractive translational model for the study of thermogenesis.


Asunto(s)
Tejido Adiposo Pardo , Frío , Animales , Ratas , Tejido Adiposo Pardo/fisiología , Tejido Adiposo Blanco/fisiología , Peso Corporal , Mamíferos , Proteínas Mitocondriales/metabolismo , Termogénesis , Proteína Desacopladora 1/metabolismo
3.
Elife ; 112022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-35018884

RESUMEN

Agouti-related peptide (AgRP) neurons increase motivation for food, however, whether metabolic sensing of homeostatic state in AgRP neurons potentiates motivation by interacting with dopamine reward systems is unexplored. As a model of impaired metabolic-sensing, we used the AgRP-specific deletion of carnitine acetyltransferase (Crat) in mice. We hypothesised that metabolic sensing in AgRP neurons is required to increase motivation for food reward by modulating accumbal or striatal dopamine release. Studies confirmed that Crat deletion in AgRP neurons (KO) impaired ex vivo glucose-sensing, as well as in vivo responses to peripheral glucose injection or repeated palatable food presentation and consumption. Impaired metabolic-sensing in AgPP neurons reduced acute dopamine release (seconds) to palatable food consumption and during operant responding, as assessed by GRAB-DA photometry in the nucleus accumbens, but not the dorsal striatum. Impaired metabolic-sensing in AgRP neurons suppressed radiolabelled 18F-fDOPA accumulation after ~30 min in the dorsal striatum but not the nucleus accumbens. Impaired metabolic sensing in AgRP neurons suppressed motivated operant responding for sucrose rewards during fasting. Thus, metabolic-sensing in AgRP neurons is required for the appropriate temporal integration and transmission of homeostatic hunger-sensing to dopamine signalling in the striatum.


Asunto(s)
Proteína Relacionada con Agouti/genética , Cuerpo Estriado/fisiología , Dopamina/fisiología , Homeostasis , Neuronas/fisiología , Transducción de Señal , Proteína Relacionada con Agouti/metabolismo , Animales , Ratones , Ratones Noqueados
4.
Cell Rep ; 37(1): 109767, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34610308

RESUMEN

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


Asunto(s)
Carnitina O-Palmitoiltransferasa/metabolismo , Ácidos Grasos/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/metabolismo , Animales , Carnitina/metabolismo , Carnitina O-Palmitoiltransferasa/deficiencia , Carnitina O-Palmitoiltransferasa/genética , Dieta Alta en Grasa , Ácidos Grasos/química , Glucosa/metabolismo , Hidroxilación , Prolina Dioxigenasas del Factor Inducible por Hipoxia/deficiencia , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Peroxidación de Lípido , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mutagénesis Sitio-Dirigida , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Procolágeno-Prolina Dioxigenasa/deficiencia , Procolágeno-Prolina Dioxigenasa/genética , Unión Proteica , Canal Aniónico 1 Dependiente del Voltaje/genética
5.
Biology (Basel) ; 10(6)2021 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-34204316

RESUMEN

Male mice lacking HuR in skeletal muscle (HuRm-/-) have been shown to have decreased gastrocnemius lipid oxidation and increased adiposity and insulin resistance. The same consequences have not been documented in female HuRm-/- mice. Here we examine this sexually dimorphic phenotype. HuRm-/- mice have an increased fat mass to lean mass ratio (FM/LM) relative to controls where food intake is similar. Increased body weight for male mice correlates with increased blood glucose during glucose tolerance tests (GTT), suggesting increased fat mass in male HuRm-/- mice as a driver of decreased glucose clearance. However, HuRm-/- female mice show decreased blood glucose levels during GTT relative to controls. HuRm-/- mice display decreased palmitate oxidation in skeletal muscle relative to controls. This difference is more robust for male HuRm-/- mice and more exaggerated for both sexes at high dietary fat. A high-fat diet stimulates expression of Pgc1α in HuRm-/- male skeletal muscle, but not in females. However, the lipid oxidation Pparα pathway remains decreased in HuRm-/- male mice relative to controls regardless of diet. This pathway is only decreased in female HuRm-/- mice fed high fat diet. A decreased capacity for lipid oxidation in skeletal muscle in the absence of HuR may thus be linked to decreased glucose clearance in male but not female mice.

6.
Med Sci Sports Exerc ; 53(6): 1161-1169, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33315811

RESUMEN

PURPOSE: Toll-like receptor 4 (TLR4) is an inflammatory receptor expressed ubiquitously in immune cells as well as skeletal muscle and other metabolic tissues. Skeletal muscle develops favorable inflammation-mediated metabolic adaptations from exercise training. Multiple inflammatory myokines, downstream from TLR4, are proposed links to the metabolic benefits of exercise. In addition, activation of TLR4 alters skeletal muscle substrate preference. The role of skeletal muscle TLR4 (mTLR4) in exercise metabolism has not previously been investigated. Herein, we aimed to specifically test the significance of mTLR4 to exercise-induced metabolic adaptations. METHODS: We developed a novel muscle-specific TLR4 knockout (mTLR4-/-) mouse model on C57BL/6J background. Male mTLR4-/- mice and wild-type (WT) littermates were compared under sedentary (SED) and voluntary wheel running (WR) conditions for 4 wk. RESULTS: mTLR4 deletion revealed marked reductions in downstream interleukin-1 receptor-associated kinase-4 (IRAK4) phosphorylation. In addition, the disruption of mTLR4 signaling prominently blunted the metabolic adaptations in WR-mTLR4-/- mice as opposed to substantial improvements exhibited by the WT counterparts. Voluntary WR in WT mice, relative to SED, resulted in significant increases in skeletal muscle fatty acid oxidation, glucose oxidation, and associated mitochondrial enzyme activities, all of which were not significantly changed in mTLR4-/- mice. CONCLUSIONS: This study introduces a novel mTLR4-/- mouse model and identifies mTLR4 as an immunomodulatory effector of exercise-induced metabolic adaptations in skeletal muscle.


Asunto(s)
Músculo Esquelético/metabolismo , Condicionamiento Físico Animal/fisiología , Receptor Toll-Like 4/metabolismo , Adaptación Fisiológica , Animales , Composición Corporal , Metabolismo Energético , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Musculares/metabolismo , Modelos Animales , Músculo Esquelético/enzimología , Oxidación-Reducción , Fosforilación , Carrera/fisiología , Transducción de Señal
7.
Mol Metab ; 44: 101140, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33285301

RESUMEN

OBJECTIVE: The expression of the interleukin-1 receptor type I (IL-1R) is enriched in pancreatic islet ß-cells, signifying that ligands activating this pathway are important for the health and function of the insulin-secreting cell. Using isolated mouse, rat, and human islets, we identified the cytokine IL-1α as a highly inducible gene in response to IL-1R activation. In addition, IL-1α is elevated in mouse and rat models of obesity and Type 2 diabetes. Since less is known about the biology of IL-1α relative to IL-1ß in pancreatic tissue, our objective was to investigate the contribution of IL-1α to pancreatic ß-cell function and overall glucose homeostasis in vivo. METHODS: We generated a novel mouse line with conditional IL-1α alleles and subsequently produced mice with either pancreatic- or myeloid lineage-specific deletion of IL-1α. RESULTS: Using this in vivo approach, we discovered that pancreatic (IL-1αPdx1-/-), but not myeloid-cell, expression of IL-1α (IL-1αLysM-/-) was required for the maintenance of whole body glucose homeostasis in both male and female mice. Moreover, pancreatic deletion of IL-1α led to impaired glucose tolerance with no change in insulin sensitivity. This observation was consistent with our finding that glucose-stimulated insulin secretion was reduced in islets isolated from IL-1αPdx1-/- mice. Alternatively, IL-1αLysM-/- mice (male and female) did not have any detectable changes in glucose tolerance, respiratory quotient, physical activity, or food intake when compared with littermate controls. CONCLUSIONS: Taken together, we conclude that there is an important physiological role for pancreatic IL-1α to promote glucose homeostasis by supporting glucose-stimulated insulin secretion and islet ß-cell mass in vivo.


Asunto(s)
Glucosa/metabolismo , Homeostasis , Secreción de Insulina/fisiología , Interleucina-1alfa/metabolismo , Células Mieloides/metabolismo , Páncreas/metabolismo , Animales , Línea Celular , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Intolerancia a la Glucosa/metabolismo , Proteínas de Homeodominio , Inflamación , Insulina/sangre , Insulina/metabolismo , Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratas , Receptores de Citocinas , Receptores Tipo I de Interleucina-1/metabolismo , Transactivadores
8.
Mol Metab ; 37: 100993, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32298772

RESUMEN

OBJECTIVES: Glucose-stimulated insulin secretion is a critical function in the regulation of glucose homeostasis, and its deregulation is associated with the development of type 2 diabetes. Here, we performed a genetic screen using islets isolated from the BXD panel of advanced recombinant inbred (RI) lines of mice to search for novel regulators of insulin production and secretion. METHODS: Pancreatic islets were isolated from 36 RI BXD lines and insulin secretion was measured following exposure to 2.8 or 16.7 mM glucose with or without exendin-4. Islets from the same RI lines were used for RNA extraction and transcript profiling. Quantitative trait loci (QTL) mapping was performed for each secretion condition and combined with transcriptome data to prioritize candidate regulatory genes within the identified QTL regions. Functional studies were performed by mRNA silencing or overexpression in MIN6B1 cells and by studying mice and islets with beta-cell-specific gene inactivation. RESULTS: Insulin secretion under the 16.7 mM glucose plus exendin-4 condition was mapped significantly to a chromosome 2 QTL. Within this QTL, RNA-Seq data prioritized Crat (carnitine O-acetyl transferase) as a strong candidate regulator of the insulin secretion trait. Silencing Crat expression in MIN6B1 cells reduced insulin content and insulin secretion by ∼30%. Conversely, Crat overexpression enhanced insulin content and secretion by ∼30%. When islets from mice with beta-cell-specific Crat inactivation were exposed to high glucose, they displayed a 30% reduction of insulin content as compared to control islets. We further showed that decreased Crat expression in both MIN6B1 cells and pancreatic islets reduced the oxygen consumption rate in a glucose concentration-dependent manner. CONCLUSIONS: We identified Crat as a regulator of insulin secretion whose action is mediated by an effect on total cellular insulin content; this effect also depends on the genetic background of the RI mouse lines. These data also show that in the presence of the stimulatory conditions used the insulin secretion rate is directly related to the insulin content.


Asunto(s)
Carnitina O-Acetiltransferasa/genética , Secreción de Insulina/genética , Células Secretoras de Insulina/metabolismo , Animales , Carnitina O-Acetiltransferasa/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Exenatida/metabolismo , Pruebas Genéticas/métodos , Glucosa/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos , Sitios de Carácter Cuantitativo
9.
Am J Physiol Endocrinol Metab ; 318(1): E11-E21, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31661298

RESUMEN

There are endocrine and immunological changes that occur during onset and progression of the overweight and obese states. The inhibitor of nuclear factor-κB kinase-ε (IKKε) was originally described as an inducible protein kinase; whole body gene deletion or systemic pharmaceutical targeting of this kinase improved insulin sensitivity and glucose tolerance in mice. To investigate the primary sites of action associated with IKKε during weight gain, we describe the first mouse line with conditional elimination of IKKε in the liver (IKKεAlb-/-). IKKεAlb-/- mice and littermate controls gain weight, show similar changes in body composition, and do not display any improvements in insulin sensitivity or whole body glucose tolerance. These studies were conducted using breeder chow diets and matched low- vs. high-fat diets. While glycogen accumulation in the liver is reduced in IKKεAlb-/- mice, lipid storage in liver is similar in IKKεAlb-/- mice and littermate controls. Our results using IKKεAlb-/- mice suggest that the primary action of this kinase to impact insulin sensitivity during weight gain lies predominantly within extrahepatic tissues.


Asunto(s)
Glucemia/metabolismo , Dieta Alta en Grasa , Glicéridos/metabolismo , Glucógeno/metabolismo , Quinasa I-kappa B/genética , Resistencia a la Insulina/genética , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Animales , Dieta con Restricción de Grasas , Prueba de Tolerancia a la Glucosa , Ratones , Ratones Noqueados , Obesidad , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
J Biol Chem ; 294(33): 12313-12327, 2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31097541

RESUMEN

Mitochondrial lipid overload in skeletal muscle contributes to insulin resistance, and strategies limiting this lipid pressure improve glucose homeostasis; however, comprehensive cellular adaptations that occur in response to such an intervention have not been reported. Herein, mice with skeletal muscle-specific deletion of carnitine palmitoyltransferase 1b (Cpt1bM-/-), which limits mitochondrial lipid entry, were fed a moderate fat (25%) diet, and samples were subjected to a multimodal analysis merging transcriptomics, proteomics, and nontargeted metabolomics to characterize the coordinated multilevel cellular responses that occur when mitochondrial lipid burden is mitigated. Limiting mitochondrial fat entry predictably improves glucose homeostasis; however, remodeling of glucose metabolism pathways pales compared with adaptations in amino acid and lipid metabolism pathways, shifts in nucleotide metabolites, and biogenesis of mitochondria and peroxisomes. Despite impaired fat utilization, Cpt1bM-/- mice have increased acetyl-CoA (14-fold) and NADH (2-fold), indicating metabolic shifts yield sufficient precursors to meet energy demand; however, this does not translate to enhance energy status as Cpt1bM-/- mice have low ATP and high AMP levels, signifying energy deficit. Comparative analysis of transcriptomic data with disease-associated gene-sets not only predicted reduced risk of glucose metabolism disorders but was also consistent with lower risk for hepatic steatosis, cardiac hypertrophy, and premature death. Collectively, these results suggest induction of metabolic inefficiency under conditions of energy surfeit likely contributes to improvements in metabolic health when mitochondrial lipid burden is mitigated. Moreover, the breadth of disease states to which mechanisms induced by muscle-specific Cpt1b inhibition may mediate health benefits could be more extensive than previously predicted.


Asunto(s)
Carnitina O-Palmitoiltransferasa/deficiencia , Metabolismo Energético , Metabolismo de los Lípidos , Mitocondrias Musculares/metabolismo , Acetilcoenzima A/genética , Acetilcoenzima A/metabolismo , Adenosina Monofosfato/genética , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Animales , Carnitina O-Palmitoiltransferasa/metabolismo , Ratones , Ratones Noqueados , Mitocondrias Musculares/genética , NAD/genética , NAD/metabolismo
11.
Metabolism ; 97: 40-49, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31129047

RESUMEN

BACKGROUND: Metabolic flexibility can be assessed by changes in respiratory exchange ratio (RER) following feeding. Though metabolic flexibility (difference in RER between fasted and fed state) is often impaired in individuals with obesity or type 2 diabetes, the cellular processes contributing to this impairment are unclear. MATERIALS AND METHODS: From several clinical studies we identified the 16 most and 14 least metabolically flexible male and female subjects out of >100 participants based on differences between 24-hour and sleep RER measured in a whole-room indirect calorimeter. Global skeletal muscle gene expression profiles revealed that, in metabolically flexible subjects, transcripts regulated by the RNA binding protein, HuR, are enriched. We generated and characterized mice with a skeletal muscle-specific knockout of the HuR encoding gene, Elavl1 (HuRm-/-). RESULTS: Male, but not female, HuRm-/- mice exhibit metabolic inflexibility, with mild obesity, impaired glucose tolerance, impaired fat oxidation and decreased in vitro palmitate oxidation compared to HuRfl/fl littermates. Expression levels of genes involved in mitochondrial fatty acid oxidation and oxidative phosphorylation are decreased in both mouse and human muscle when HuR is inhibited. CONCLUSIONS: HuR inhibition results in impaired metabolic flexibility and decreased lipid oxidation, suggesting a role for HuR as an important regulator of skeletal muscle metabolism.


Asunto(s)
Proteína 1 Similar a ELAV/metabolismo , Músculo Esquelético/metabolismo , Proteínas de Unión al ARN/metabolismo , Roedores/metabolismo , Adulto , Animales , Diabetes Mellitus Tipo 2/metabolismo , Ayuno/metabolismo , Ácidos Grasos/metabolismo , Femenino , Intolerancia a la Glucosa/metabolismo , Humanos , Metabolismo de los Lípidos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Obesidad/metabolismo , Oxidación-Reducción , Fosforilación Oxidativa , Intercambio Gaseoso Pulmonar/fisiología
12.
Diabetes ; 68(4): 819-831, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30728184

RESUMEN

Proximal tubular epithelial cells are highly energy demanding. Their energy need is covered mostly from mitochondrial fatty acid oxidation. Whether derailments in fatty acid metabolism and mitochondrial dysfunction are forerunners of tubular damage has been suggested but is not entirely clear. Here we modeled mitochondrial overload by creating mice lacking the enzyme carnitine acetyltransferase (CrAT) in the proximal tubules, thus limiting a primary mechanism to export carbons under conditions of substrate excess. Mice developed tubular disease and, interestingly, secondary glomerulosclerosis. This was accompanied by increased levels of apoptosis regulator and fibrosis markers, increased oxidative stress, and abnormal profiles of acylcarnitines and organic acids suggesting profound impairments in all major forms of nutrient metabolism. When mice with CrAT deletion were fed a high-fat diet, kidney disease was more severe and developed faster. Primary proximal tubular cells isolated from the knockout mice displayed energy deficit and impaired respiration before the onset of pathology, suggesting mitochondrial respiratory abnormalities as a potential underlying mechanism. Our findings support the hypothesis that derailments of mitochondrial energy metabolism may be causative to chronic kidney disease. Our results also suggest that tubular injury may be a primary event followed by secondary glomerulosclerosis, raising the possibility that focusing on normalizing tubular cell mitochondrial function and energy balance could be an important preventative strategy.


Asunto(s)
Carnitina O-Acetiltransferasa/metabolismo , Enfermedades Renales/metabolismo , Túbulos Renales Proximales/metabolismo , Riñón/metabolismo , Animales , Apoptosis/fisiología , Carnitina O-Acetiltransferasa/genética , Dieta Alta en Grasa , Complejo I de Transporte de Electrón/metabolismo , Riñón/patología , Enfermedades Renales/genética , Enfermedades Renales/patología , Túbulos Renales Proximales/patología , Metabolismo de los Lípidos , Masculino , Ratones , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología
13.
Cell ; 175(6): 1459-1460, 2018 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-30500531

RESUMEN

Most theories of meal-induced thermogenesis involve a gut-brain-brown adipose tissue axis driving sympathetic nervous system-mediated thermogenesis. Li et al. demonstrate that secretin released by the gut after a meal binds to abundant receptors in brown adipose tissue to stimulate thermogenesis, inhibiting food intake and thereby suggesting a novel role for secretin regulating satiety.


Asunto(s)
Tejido Adiposo Pardo , Secretina , Ingestión de Alimentos , Saciedad , Termogénesis
14.
Obesity (Silver Spring) ; 26(9): 1439-1447, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30226002

RESUMEN

OBJECTIVE: This study examined the phenotypic effects of adipocyte-specific oncostatin M receptor (OSMR) loss in chow-fed mice. METHODS: Chow-fed adipocyte-specific OSMR knockout (FKO) mice and littermate OSMRfl/fl controls were studied. Tissue weights, insulin sensitivity, adipokine production, and stromal cell immunophenotypes were assessed in epididymal fat (eWAT); serum adipokine production was also assessed. In vitro, adipocytes were treated with oncostatin M, and adipokine gene expression was assessed. RESULTS: Body weights, fasting blood glucose levels, and eWAT weights did not differ between genotypes. However, the eWAT of OSMRFKO mice was modestly less responsive to insulin stimulation than that of OSMRfl/fl mice. Notably, significant increases in adipokines, including C-reactive protein, lipocalin 2, intercellular adhesion molecule-1, and insulinlike growth factor binding protein 6, were observed in the eWAT of OSMRFKO mice. In addition, significant increases in fetuin A and intercellular adhesion molecule-1 were detected in OSMRFKO serum. Flow cytometry revealed a significant increase in leukocyte number and modest, but not statistically significant, increases in B cells and T cells in the eWAT of OSMRFKO mice. CONCLUSIONS: The chow-fed OSMRFKO mice exhibited adipose tissue dysfunction and increased proinflammatory adipokine production. These results suggest that intact adipocyte oncostatin M-OSMR signaling is necessary for adipose tissue immune cell homeostasis.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/fisiopatología , Oncostatina M/metabolismo , Animales , Masculino , Ratones , Ratones Noqueados
15.
J Lipid Res ; 59(9): 1660-1670, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30026188

RESUMEN

Transcriptional coactivator PPAR γ coactivator (PGC)-1α and its splice variant N-terminal (NT)-PGC-1α mediate transcriptional regulation of brown adipose tissue (BAT) thermogenesis in response to changes in ambient temperature. PGC-1α is dispensable for cold-induced BAT thermogenesis as long as NT-PGC-1α is present. However, the functional significance of NT-PGC-1α in BAT has not been determined. In the present study, we generated NT-PGC-1α-/- mice to investigate the effect of NT-PGC-1α deficiency on adaptive BAT thermogenesis. At thermoneutrality, NT-PGC-1α-/- mice exhibited abnormal BAT phenotype with increased accumulation of large lipid droplets concomitant with marked downregulation of FA oxidation (FAO)-related genes. Consistent with transcriptional changes, mitochondrial FAO was significantly diminished in NT-PGC-1α-/- BAT. This alteration, in turn, enhanced glucose utilization within the NT-PGC-1α-/- BAT mitochondria. In line with this, NT-PGC-1α-/- mice had higher reliance on carbohydrates. In response to cold or ß3-adrenergic receptor agonist, NT-PGC-1α-/- mice transiently exhibited lower thermogenesis but reached similar thermogenic capacities as their WT littermates. Collectively, these findings demonstrate that NT-PGC-1α is an important contributor to the maintenance of FAO capacity in BAT at thermoneutrality and provide deeper insights into the relative contributions of PGC-1α and NT-PGC-1α to temperature-regulated BAT remodeling.


Asunto(s)
Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Ácidos Grasos/metabolismo , Mitocondrias/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/química , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/deficiencia , Tejido Adiposo Blanco/metabolismo , Animales , Regulación de la Expresión Génica , Lipólisis , Ratones , Mutación , Oxidación-Reducción , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Temperatura , Termogénesis
16.
FASEB J ; : fj201800634R, 2018 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-29932868

RESUMEN

Hunger-sensing agouti-related peptide (AgRP) neurons ensure survival by adapting metabolism and behavior to low caloric environments. This adaption is accomplished by consolidating food intake, suppressing energy expenditure, and maximizing fat storage (nutrient partitioning) for energy preservation. The intracellular mechanisms responsible are unknown. Here we report that AgRP carnitine acetyltransferase (Crat) knockout (KO) mice exhibited increased fatty acid utilization and greater fat loss after 9 d of calorie restriction (CR). No differences were seen in mice with ad libitum food intake. Eleven days ad libitum feeding after CR resulted in greater food intake, rebound weight gain, and adiposity in AgRP Crat KO mice compared with wild-type controls, as KO mice act to restore pre-CR fat mass. Collectively, this study highlights the importance of Crat in AgRP neurons to regulate nutrient partitioning and fat mass during chronically reduced caloric intake. The increased food intake, body weight gain, and adiposity in KO mice after CR also highlights the detrimental and persistent metabolic consequence of impaired substrate utilization associated with CR. This finding may have significant implications for postdieting weight management in patients with metabolic diseases.-Reichenbach, A., Stark, R., Mequinion, M., Lockie, S. H., Lemus, M. B., Mynatt, R. L., Luquet, S., Andrews, Z. B. Carnitine acetyltransferase (Crat) in hunger-sensing AgRP neurons permits adaptation to calorie restriction.

17.
Mol Metab ; 14: 95-107, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29914854

RESUMEN

OBJECTIVE: Pancreatic tissue, and islets in particular, are enriched in expression of the interleukin-1 receptor type I (IL-1R). Because of this enrichment, islet ß-cells are exquisitely sensitive to the IL-1R ligands IL-1α and IL-1ß, suggesting that signaling through this pathway regulates health and function of islet ß-cells. METHODS: Herein, we report a targeted deletion of IL-1R in pancreatic tissue (IL-1RPdx1-/-) in C57BL/6J mice and in db/db mice on the C57 genetic background. Islet morphology, ß-cell transcription factor abundance, and expression of the de-differentiation marker Aldh1a3 were analyzed by immunofluorescent staining. Glucose and insulin tolerance tests were used to examine metabolic status of these genetic manipulations. Glucose-stimulated insulin secretion was evaluated in vivo and in isolated islets ex vivo by perifusion. RESULTS: Pancreatic deletion of IL-1R leads to impaired glucose tolerance, a phenotype that is exacerbated by age. Crossing the IL-1RPdx1-/- with db/db mice worsened glucose tolerance without altering body weight. There were no detectable alterations in insulin tolerance between IL-1RPdx1-/- mice and littermate controls. However, glucose-stimulated insulin secretion was reduced in islets isolated from IL-1RPdx1-/- relative to control islets. Insulin output in vivo after a glucose challenge was also markedly reduced in IL-1RPdx1-/- mice when compared with littermate controls. Pancreatic islets from IL-1RPdx1-/- mice displayed elevations in Aldh1a3, a marker of de-differentiation, and reduction in nuclear abundance of the ß-cell transcription factor MafA. Nkx6.1 abundance was unaltered. CONCLUSIONS: There is an important physiological role for pancreatic IL-1R to promote glucose homeostasis by suppressing expression of Aldh1a3, sustaining MafA abundance, and supporting glucose-stimulated insulin secretion in vivo.


Asunto(s)
Diferenciación Celular , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Receptores Tipo I de Interleucina-1/genética , Animales , Línea Celular Tumoral , Células Cultivadas , Femenino , Eliminación de Gen , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Homeostasis , Resistencia a la Insulina , Células Secretoras de Insulina/citología , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Retinal-Deshidrogenasa/genética , Retinal-Deshidrogenasa/metabolismo
18.
Endocrinology ; 159(6): 2473-2483, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29697769

RESUMEN

Behavioral adaptation to periods of varying food availability is crucial for survival, and agouti-related protein (AgRP) neurons have been associated with entrainment to temporal restricted feeding. We have shown that carnitine acetyltransferase (Crat) in AgRP neurons enables metabolic flexibility and appropriate nutrient partitioning. In this study, by restricting food availability to 3 h/d during the light phase, we examined whether Crat is a component of a food-entrainable oscillator (FEO) that helps link behavior to food availability. AgRP Crat knockout (KO) mice consumed less food and regained less body weight but maintained blood glucose levels during the 25-day restricted feeding protocol. Importantly, we observed no difference in meal latency, food anticipatory activity (FAA), or brown adipose tissue temperature during the first 13 days of restricted feeding. However, as the restricted feeding paradigm progressed, we noticed an increased FAA in AgRP Crat KO mice. The delayed increase in FAA, which developed during the last 12 days of restricted feeding, corresponded with elevated plasma levels of corticosterone and nonesterified fatty acids, indicating it resulted from greater energy debt incurred by KO mice over the course of the experiment. These experiments highlight the importance of Crat in AgRP neurons in regulating feeding behavior and body weight gain during restricted feeding but not in synchronizing behavior to food availability. Thus, Crat within AgRP neurons forms a component of the homeostatic response to restricted feeding but is not likely to be a molecular component of FEO.


Asunto(s)
Adaptación Fisiológica/genética , Proteína Relacionada con Agouti/metabolismo , Restricción Calórica , Carnitina O-Acetiltransferasa/fisiología , Conducta Alimentaria/fisiología , Homeostasis/genética , Neuronas/metabolismo , Animales , Carnitina O-Acetiltransferasa/genética , Carnitina O-Acetiltransferasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Condicionamiento Físico Animal/fisiología
19.
Cell Rep ; 22(7): 1745-1759, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29444428

RESUMEN

AgRP neurons control peripheral substrate utilization and nutrient partitioning during conditions of energy deficit and nutrient replenishment, although the molecular mechanism is unknown. We examined whether carnitine acetyltransferase (Crat) in AgRP neurons affects peripheral nutrient partitioning. Crat deletion in AgRP neurons reduced food intake and feeding behavior and increased glycerol supply to the liver during fasting, as a gluconeogenic substrate, which was mediated by changes to sympathetic output and peripheral fatty acid metabolism in the liver. Crat deletion in AgRP neurons increased peripheral fatty acid substrate utilization and attenuated the switch to glucose utilization after refeeding, indicating altered nutrient partitioning. Proteomic analysis in AgRP neurons shows that Crat regulates protein acetylation and metabolic processing. Collectively, our studies highlight that AgRP neurons require Crat to provide the metabolic flexibility to optimize nutrient partitioning and regulate peripheral substrate utilization, particularly during fasting and refeeding.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Carnitina O-Acetiltransferasa/metabolismo , Ácidos Grasos/metabolismo , Animales , Colecistoquinina/administración & dosificación , Ingestión de Alimentos , Ayuno , Conducta Alimentaria , Eliminación de Gen , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Inyecciones Intraperitoneales , Inyecciones Intraventriculares , Insulina/administración & dosificación , Integrasas/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones Noqueados , Proteómica , Reproducibilidad de los Resultados
20.
Mol Nutr Food Res ; 62(8): e1700856, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29476602

RESUMEN

SCOPE: The primary disorder underlying metabolic syndrome is insulin resistance due to excess body weight and abdominal visceral fat accumulation. In this study, it is asked if dietary intake of an ethanolic extract from Russian tarragon (Artemisia dracunculus L., termed PMI5011), shown to improve glucose utilization by enhancing insulin signaling in skeletal muscle, could prevent obesity-induced insulin resistance, skeletal muscle metabolic inflexibility, and ectopic lipid accumulation in the skeletal muscle and liver. METHODS AND RESULTS: Male wild-type mice are fed a high-fat diet alone or supplemented with PMI5011 (1% w/w) over 3 months. Dietary intake of PMI5011 improved fatty acid oxidation and metabolic flexibility in the skeletal muscle, reduced insulin levels, and enhanced insulin signaling in the skeletal muscle and liver independent of robust changes in expression of factors that control fatty acid oxidation. This corresponds with significantly reduced lipid accumulation in the skeletal muscle and liver, although body weight gain is comparable to a high-fat diet alone. CONCLUSION: Previous studies showed that PMI5011 enhances insulin sensitivity in the setting of established obesity-induced insulin resistance. The current study demonstrates that dietary intake of PMI5011 prevents high-fat diet-induced insulin resistance, metabolic dysfunction, and ectopic lipid accumulation in the skeletal muscle and liver without reducing body weight.


Asunto(s)
Artemisia/química , Suplementos Dietéticos , Metabolismo de los Lípidos , Lipotrópicos/uso terapéutico , Músculo Esquelético/metabolismo , Obesidad/terapia , Extractos Vegetales/uso terapéutico , Adiposidad , Animales , Fármacos Antiobesidad/uso terapéutico , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético , Regulación de la Expresión Génica , Resistencia a la Insulina , Hígado/metabolismo , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Músculo Esquelético/patología , Obesidad/etiología , Obesidad/patología , Especificidad de Órganos , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/metabolismo , Distribución Aleatoria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...