Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 127(8): 1094-1108, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32660330

RESUMEN

RATIONALE: Circumstantial evidence links the development of heart failure to posttranslational modifications of mitochondrial proteins, including lysine acetylation (Kac). Nonetheless, direct evidence that Kac compromises mitochondrial performance remains sparse. OBJECTIVE: This study sought to explore the premise that mitochondrial Kac contributes to heart failure by disrupting oxidative metabolism. METHODS AND RESULTS: A DKO (dual knockout) mouse line with deficiencies in CrAT (carnitine acetyltransferase) and Sirt3 (sirtuin 3)-enzymes that oppose Kac by buffering the acetyl group pool and catalyzing lysine deacetylation, respectively-was developed to model extreme mitochondrial Kac in cardiac muscle, as confirmed by quantitative acetyl-proteomics. The resulting impact on mitochondrial bioenergetics was evaluated using a respiratory diagnostics platform that permits comprehensive assessment of mitochondrial function and energy transduction. Susceptibility of DKO mice to heart failure was investigated using transaortic constriction as a model of cardiac pressure overload. The mitochondrial acetyl-lysine landscape of DKO hearts was elevated well beyond that observed in response to pressure overload or Sirt3 deficiency alone. Relative changes in the abundance of specific acetylated lysine peptides measured in DKO versus Sirt3 KO hearts were strongly correlated. A proteomics comparison across multiple settings of hyperacetylation revealed ≈86% overlap between the populations of Kac peptides affected by the DKO manipulation as compared with experimental heart failure. Despite the severity of cardiac Kac in DKO mice relative to other conditions, deep phenotyping of mitochondrial function revealed a surprisingly normal bioenergetics profile. Thus, of the >120 mitochondrial energy fluxes evaluated, including substrate-specific dehydrogenase activities, respiratory responses, redox charge, mitochondrial membrane potential, and electron leak, we found minimal evidence of oxidative insufficiencies. Similarly, DKO hearts were not more vulnerable to dysfunction caused by transaortic constriction-induced pressure overload. CONCLUSIONS: The findings challenge the premise that hyperacetylation per se threatens metabolic resilience in the myocardium by causing broad-ranging disruption to mitochondrial oxidative machinery.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Mitocondrias Cardíacas/metabolismo , Proteínas Mitocondriales/metabolismo , Miocitos Cardíacos/metabolismo , Proteoma , Acetilación , Animales , Carnitina O-Acetiltransferasa/deficiencia , Carnitina O-Acetiltransferasa/genética , Modelos Animales de Enfermedad , Metabolismo Energético , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Lisina , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo , Procesamiento Proteico-Postraduccional , Proteómica , Sirtuina 3/deficiencia , Sirtuina 3/genética
2.
Mol Cell ; 50(5): 686-98, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23746352

RESUMEN

Lipid metabolism is tightly controlled by the nutritional state of the organism. Nutrient-rich conditions increase lipogenesis, whereas nutrient deprivation promotes fat oxidation. In this study, we identify the mitochondrial sirtuin, SIRT4, as a regulator of lipid homeostasis. SIRT4 is active in nutrient-replete conditions to repress fatty acid oxidation while promoting lipid anabolism. SIRT4 deacetylates and inhibits malonyl CoA decarboxylase (MCD), an enzyme that produces acetyl CoA from malonyl CoA. Malonyl CoA provides the carbon skeleton for lipogenesis and also inhibits fat oxidation. Mice lacking SIRT4 display elevated MCD activity and decreased malonyl CoA in skeletal muscle and white adipose tissue. Consequently, SIRT4 KO mice display deregulated lipid metabolism, leading to increased exercise tolerance and protection against diet-induced obesity. In sum, this work elucidates SIRT4 as an important regulator of lipid homeostasis, identifies MCD as a SIRT4 target, and deepens our understanding of the malonyl CoA regulatory axis.


Asunto(s)
Carboxiliasas/metabolismo , Metabolismo de los Lípidos , Proteínas Mitocondriales/metabolismo , Sirtuinas/metabolismo , Acetilación , Tejido Adiposo Blanco/metabolismo , Animales , Dieta , Ácidos Grasos/metabolismo , Metabolismo de los Lípidos/genética , Lípidos/biosíntesis , Masculino , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Obesidad/etiología , Obesidad/metabolismo , Oxidación-Reducción , Sirtuinas/genética
3.
J Biol Chem ; 292(11): 4651-4662, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-28154172

RESUMEN

The immunity-related GTPases (IRGs) are a family of proteins that are induced by interferon (IFN)-γ and play pivotal roles in immune and inflammatory responses. IRGs ostensibly function as dynamin-like proteins that bind to intracellular membranes and promote remodeling and trafficking of those membranes. Prior studies have shown that loss of Irgm1 in mice leads to increased lethality to bacterial infections as well as enhanced inflammation to non-infectious stimuli; however, the mechanisms underlying these phenotypes are unclear. In the studies reported here, we found that uninfected Irgm1-deficient mice displayed high levels of serum cytokines typifying profound autoinflammation. Similar increases in cytokine production were also seen in cultured, IFN-γ-primed macrophages that lacked Irgm1. A series of metabolic studies indicated that the enhanced cytokine production was associated with marked metabolic changes in the Irgm1-deficient macrophages, including increased glycolysis and an accumulation of long chain acylcarnitines. Cells were exposed to the glycolytic inhibitor, 2-deoxyglucose, or fatty acid synthase inhibitors to perturb the metabolic alterations, which resulted in dampening of the excessive cytokine production. These results suggest that Irgm1 deficiency drives metabolic dysfunction in macrophages in a manner that is cell-autonomous and independent of infectious triggers. This may be a significant contributor to excessive inflammation seen in Irgm1-deficient mice in different contexts.


Asunto(s)
Citocinas/inmunología , Proteínas de Unión al GTP/genética , Macrófagos/inmunología , Animales , Autofagia , Células Cultivadas , Proteínas de Unión al GTP/inmunología , Eliminación de Gen , Glucólisis , Inflamación/genética , Inflamación/inmunología , Interferón gamma/inmunología , Macrófagos/citología , Macrófagos/metabolismo , Ratones
4.
Biochem Biophys Res Commun ; 479(4): 868-874, 2016 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-27693789

RESUMEN

The purpose of this study was to determine if plasma acylcarnitine (AC) profiling is altered under hyperinsulinemic conditions as part of the aging process. Fifteen young, lean (19-29 years) and fifteen middle-to older-aged (57-82 years) individuals underwent a 2-hr euglycemic-hyperinsulinemic clamp. Plasma samples were obtained at baseline, 20 min, 50 min, and 120 min for analysis of AC species and amino acids. Skeletal muscle biopsies were performed after 60 min of insulin-stimulation for analysis of acetyl-CoA carboxylase (ACC) phosphorylation. Insulin infusion decreased the majority of plasma short-, medium-, and long-chain (SC, MC, and LC, respectively) AC. However, during the initial 50 min, a number of MC and LC AC species (C10, C10:1, C12:1, C14, C16, C16:1, C18) remained elevated in aged individuals compared to their younger counterparts indicating a lag in responsiveness. Additionally, the insulin-induced decline in skeletal muscle ACC phosphorylation was blunted in the aged compared to young individuals (-24% vs. -56%, P < 0.05). These data suggest that a desensitization to insulin during aging, possibly at the level of skeletal muscle ACC phosphorylation, results in a diminished ability to transition to glucose oxidation indicative of metabolic inflexibility.


Asunto(s)
Envejecimiento/sangre , Carnitina/análogos & derivados , Insulina/sangre , Acetil-CoA Carboxilasa/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/metabolismo , Aminoácidos/sangre , Carnitina/sangre , Carnitina/química , Femenino , Glucosa/metabolismo , Técnica de Clampeo de la Glucosa , Humanos , Insulina/administración & dosificación , Resistencia a la Insulina , Masculino , Persona de Mediana Edad , Músculo Esquelético/enzimología , Oxidación-Reducción , Fosforilación , Adulto Joven
5.
J Lipid Res ; 56(9): 1795-807, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26156077

RESUMEN

Palmitic acid (PA) is associated with higher blood concentrations of medium-chain acylcarnitines (MCACs), and we hypothesized that PA may inhibit progression of FA ß-oxidation. Using a cross-over design, 17 adults were fed high PA (HPA) and low PA/high oleic acid (HOA) diets, each for 3 weeks. The [1-(13)C]PA and [13-(13)C]PA tracers were administered with food in random order with each diet, and we assessed PA oxidation (PA OX) and serum AC concentration to determine whether a higher PA intake promoted incomplete PA OX. Dietary PA was completely oxidized during the HOA diet, but only about 40% was oxidized during the HPA diet. The [13-(13)C]PA/[1-(13)C]PA ratio of PA OX had an approximate value of 1.0 for either diet, but the ratio of the serum concentrations of MCACs to long-chain ACs (LCACs) was significantly higher during the HPA diet. Thus, direct measurement of PA OX did not confirm that the HPA diet caused incomplete PA OX, despite the modest, but statistically significant, increase in the ratio of MCACs to LCACs in blood.


Asunto(s)
Carnitina/análogos & derivados , Dieta , Ácidos Grasos/sangre , Palmitatos/administración & dosificación , Adolescente , Adulto , Composición Corporal/efectos de los fármacos , Carnitina/sangre , Citocinas/metabolismo , Grasas de la Dieta/administración & dosificación , Femenino , Humanos , Inmunidad Innata/efectos de los fármacos , Peroxidación de Lípido/genética , Masculino , Ácido Oléico/administración & dosificación , Palmitatos/sangre
6.
J Biol Chem ; 289(12): 8106-20, 2014 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-24482226

RESUMEN

Thioredoxin-interacting protein (TXNIP) is an α-arrestin family member involved in redox sensing and metabolic control. Growing evidence links TXNIP to mitochondrial function, but the molecular nature of this relationship has remained poorly defined. Herein, we employed targeted metabolomics and comprehensive bioenergetic analyses to evaluate oxidative metabolism and respiratory kinetics in mouse models of total body (TKO) and skeletal muscle-specific (TXNIP(SKM-/-)) Txnip deficiency. Compared with littermate controls, both TKO and TXNIP(SKM-/-) mice had reduced exercise tolerance in association with muscle-specific impairments in substrate oxidation. Oxidative insufficiencies in TXNIP null muscles were not due to perturbations in mitochondrial mass, the electron transport chain, or emission of reactive oxygen species. Instead, metabolic profiling analyses led to the discovery that TXNIP deficiency causes marked deficits in enzymes required for catabolism of branched chain amino acids, ketones, and lactate, along with more modest reductions in enzymes of ß-oxidation and the tricarboxylic acid cycle. The decrements in enzyme activity were accompanied by comparable deficits in protein abundance without changes in mRNA expression, implying dysregulation of protein synthesis or stability. Considering that TXNIP expression increases in response to starvation, diabetes, and exercise, these findings point to a novel role for TXNIP in coordinating mitochondrial fuel switching in response to nutrient availability.


Asunto(s)
Proteínas Portadoras/metabolismo , Metabolismo Energético , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Oxidorreductasas/metabolismo , Tiorredoxinas/metabolismo , Animales , Proteínas Portadoras/genética , Metabolómica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/genética , Músculo Esquelético/enzimología , Oxidación-Reducción , Tiorredoxinas/genética
7.
J Lipid Res ; 55(4): 635-44, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24395925

RESUMEN

Carnitine acetyltransferase (CrAT) is a mitochondrial matrix enzyme that catalyzes the interconversion of acetyl-CoA and acetylcarnitine. Emerging evidence suggests that this enzyme functions as a positive regulator of total body glucose tolerance and muscle activity of pyruvate dehydrogenase (PDH), a mitochondrial enzyme complex that promotes glucose oxidation and is feedback inhibited by acetyl-CoA. Here, we used tandem mass spectrometry-based metabolic profiling to identify a negative relationship between CrAT activity and muscle content of lipid intermediates. CrAT specific activity was diminished in muscles from obese and diabetic rodents despite increased protein abundance. This reduction in enzyme activity was accompanied by muscle accumulation of long-chain acylcarnitines (LCACs) and acyl-CoAs and a decline in the acetylcarnitine/acetyl-CoA ratio. In vitro assays demonstrated that palmitoyl-CoA acts as a direct mixed-model inhibitor of CrAT. Similarly, in primary human myocytes grown in culture, nutritional and genetic manipulations that promoted mitochondrial influx of fatty acids resulted in accumulation of LCACs but a pronounced decrease of CrAT-derived short-chain acylcarnitines. These results suggest that lipid-induced antagonism of CrAT might contribute to decreased PDH activity and glucose disposal in the context of obesity and diabetes.


Asunto(s)
Carnitina O-Acetiltransferasa/metabolismo , Obesidad/enzimología , Acetilcoenzima A/metabolismo , Animales , Carnitina/análogos & derivados , Carnitina/metabolismo , Carnitina O-Palmitoiltransferasa/metabolismo , Células Cultivadas , Diabetes Mellitus/enzimología , Humanos , Metabolismo de los Lípidos , Masculino , Fibras Musculares Esqueléticas/enzimología , Complejo Piruvato Deshidrogenasa/metabolismo , Ratas Wistar , Ratas Zucker
8.
Diabetologia ; 57(11): 2282-95, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25091629

RESUMEN

AIMS/HYPOTHESIS: Targeted metabolomic and transcriptomic approaches were used to evaluate the relationship between skeletal muscle metabolite signatures, gene expression profiles and clinical outcomes in response to various exercise training interventions. We hypothesised that changes in mitochondrial metabolic intermediates would predict improvements in clinical risk factors, thereby offering novel insights into potential mechanisms. METHODS: Subjects at risk of metabolic disease were randomised to 6 months of inactivity or one of five aerobic and/or resistance training programmes (n = 112). Pre/post-intervention assessments included cardiorespiratory fitness ([Formula: see text]), serum triacylglycerols (TGs) and insulin sensitivity (SI). In this secondary analysis, muscle biopsy specimens were used for targeted mass spectrometry-based analysis of metabolic intermediates and measurement of mRNA expression of genes involved in metabolism. RESULTS: Exercise regimens with the largest energy expenditure produced robust increases in muscle concentrations of even-chain acylcarnitines (median 37-488%), which correlated positively with increased expression of genes involved in muscle uptake and oxidation of fatty acids. Along with free carnitine, the aforementioned acylcarnitine metabolites were related to improvements in [Formula: see text], TGs and SI (R = 0.20-0.31, p < 0.05). Muscle concentrations of the tricarboxylic acid cycle intermediates succinate and succinylcarnitine (R = 0.39 and 0.24, p < 0.05) emerged as the strongest correlates of SI. CONCLUSIONS/INTERPRETATION: The metabolic signatures of exercise-trained skeletal muscle reflected reprogramming of mitochondrial function and intermediary metabolism and correlated with changes in cardiometabolic fitness. Succinate metabolism and the succinate dehydrogenase complex emerged as a potential regulatory node that intersects with whole-body insulin sensitivity. This study identifies new avenues for mechanistic research aimed at understanding the health benefits of physical activity. Trial registration ClinicalTrials.gov NCT00200993 and NCT00275145 Funding This work was supported by the National Heart, Lung, and Blood Institute (National Institutes of Health), National Institute on Aging (National Institutes of Health) and National Institute of Arthritis and Musculoskeletal and Skin Diseases (National Institutes of Health).


Asunto(s)
Ejercicio Físico/fisiología , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Adolescente , Adulto , Anciano , Aminoácidos de Cadena Ramificada/metabolismo , Carnitina/análogos & derivados , Carnitina/metabolismo , Femenino , Humanos , Masculino , Metabolómica , Persona de Mediana Edad , Ácido Succínico/metabolismo , Adulto Joven
9.
J Pharmacol Exp Ther ; 349(3): 487-96, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24700885

RESUMEN

There is a growing need to understand the underlying mechanisms involved in the progression of cardiovascular disease during obesity and diabetes. Although inhibition of fatty acid oxidation has been proposed as a novel approach to treat ischemic heart disease and heart failure, reduced muscle fatty acid oxidation rates may contribute to the development of obesity-associated insulin resistance. Our aim was to determine whether treatment with the antianginal agent trimetazidine, which inhibits fatty acid oxidation in the heart secondary to inhibition of 3-ketoacyl-CoA thiolase (3-KAT), may have off-target effects on glycemic control in obesity. We fed C57BL/6NCrl mice a high-fat diet (HFD) for 10 weeks before a 22-day treatment with the 3-KAT inhibitor trimetazidine (15 mg/kg per day). Insulin resistance was assessed via glucose/insulin tolerance testing, and lipid metabolite content was assessed in gastrocnemius muscle. Trimetazidine-treatment led to a mild shift in substrate preference toward carbohydrates as an oxidative fuel source in obese mice, evidenced by an increase in the respiratory exchange ratio. This shift in metabolism was accompanied by an accumulation of long-chain acyl-CoA and a trend to an increase in triacylglycerol content in gastrocnemius muscle, but did not exacerbate HFD-induced insulin resistance compared with control-treated mice. It is noteworthy that trimetazidine treatment reduced palmitate oxidation rates in the isolated working mouse heart and neonatal cardiomyocytes but not C2C12 skeletal myotubes. Our findings demonstrate that trimetazidine therapy does not adversely affect HFD-induced insulin resistance, suggesting that treatment with trimetazidine would not worsen glycemic control in obese patients with angina.


Asunto(s)
Acetil-CoA C-Aciltransferasa/antagonistas & inhibidores , Angina de Pecho/metabolismo , Resistencia a la Insulina , Obesidad/metabolismo , Trimetazidina/efectos adversos , Vasodilatadores/efectos adversos , Angina de Pecho/tratamiento farmacológico , Angina de Pecho/enzimología , Angina de Pecho/etiología , Animales , Células Cultivadas , Dieta Alta en Grasa , Ácidos Grasos/metabolismo , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Obesidad/complicaciones , Obesidad/enzimología , Oxidación-Reducción , Ratas , Trimetazidina/administración & dosificación , Trimetazidina/uso terapéutico , Vasodilatadores/administración & dosificación , Vasodilatadores/uso terapéutico
10.
Adv Sci (Weinh) ; : e2400188, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38887849

RESUMEN

Dysferlin is a multi-functional protein that regulates membrane resealing, calcium homeostasis, and lipid metabolism in skeletal muscle. Genetic loss of dysferlin results in limb girdle muscular dystrophy 2B/2R (LGMD2B/2R) and other dysferlinopathies - rare untreatable muscle diseases that lead to permanent loss of ambulation in humans. The mild disease severity in dysferlin-deficient mice and diverse genotype-phenotype relationships in LGMD2B patients have prompted the development of new in vitro models for personalized studies of dysferlinopathy. Here the first 3-D tissue-engineered hiPSC-derived skeletal muscle ("myobundle") model of LGMD2B is described that exhibits compromised contractile function, calcium-handling, and membrane repair, and transcriptomic changes indicative of impaired oxidative metabolism and mitochondrial dysfunction. In response to the fatty acid (FA) challenge, LGMD2B myobundles display mitochondrial deficits and intracellular lipid droplet (LD) accumulation. Treatment with the ryanodine receptor (RyR) inhibitor dantrolene or the dissociative glucocorticoid vamorolone restores LGMD2B contractility, improves membrane repair, and reduces LD accumulation. Lastly, it is demonstrated that chemically induced chronic RyR leak in healthy myobundles phenocopies LGMD2B contractile and metabolic deficit, but not the loss of membrane repair capacity. Together, these results implicate intramyocellular Ca2+ leak as a critical driver of dysferlinopathic phenotype and validate the myobundle system as a platform to study LGMD2B pathogenesis.

11.
Cell Metab ; 36(2): 422-437.e8, 2024 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-38325337

RESUMEN

Time-restricted feeding (TRF) has gained attention as a dietary regimen that promotes metabolic health. This study questioned if the health benefits of an intermittent TRF (iTRF) schedule require ketone flux specifically in skeletal and cardiac muscles. Notably, we found that the ketolytic enzyme beta-hydroxybutyrate dehydrogenase 1 (BDH1) is uniquely enriched in isolated mitochondria derived from heart and red/oxidative skeletal muscles, which also have high capacity for fatty acid oxidation (FAO). Using mice with BDH1 deficiency in striated muscles, we discover that this enzyme optimizes FAO efficiency and exercise tolerance during acute fasting. Additionally, iTRF leads to robust molecular remodeling of muscle tissues, and muscle BDH1 flux does indeed play an essential role in conferring the full adaptive benefits of this regimen, including increased lean mass, mitochondrial hormesis, and metabolic rerouting of pyruvate. In sum, ketone flux enhances mitochondrial bioenergetics and supports iTRF-induced remodeling of skeletal muscle and heart.


Asunto(s)
Cetonas , Miocardio , Ratones , Animales , Cetonas/metabolismo , Miocardio/metabolismo , Mitocondrias/metabolismo , Oxidación-Reducción , Corazón , Músculo Esquelético/metabolismo
12.
Cell Metab ; 7(1): 45-56, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18177724

RESUMEN

Previous studies have suggested that insulin resistance develops secondary to diminished fat oxidation and resultant accumulation of cytosolic lipid molecules that impair insulin signaling. Contrary to this model, the present study used targeted metabolomics to find that obesity-related insulin resistance in skeletal muscle is characterized by excessive beta-oxidation, impaired switching to carbohydrate substrate during the fasted-to-fed transition, and coincident depletion of organic acid intermediates of the tricarboxylic acid cycle. In cultured myotubes, lipid-induced insulin resistance was prevented by manipulations that restrict fatty acid uptake into mitochondria. These results were recapitulated in mice lacking malonyl-CoA decarboxylase (MCD), an enzyme that promotes mitochondrial beta-oxidation by relieving malonyl-CoA-mediated inhibition of carnitine palmitoyltransferase 1. Thus, mcd(-/-) mice exhibit reduced rates of fat catabolism and resist diet-induced glucose intolerance despite high intramuscular levels of long-chain acyl-CoAs. These findings reveal a strong connection between skeletal muscle insulin resistance and lipid-induced mitochondrial stress.


Asunto(s)
Ácidos Grasos/metabolismo , Resistencia a la Insulina , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Animales , Glucemia/metabolismo , Carboxiliasas/genética , Carboxiliasas/metabolismo , Línea Celular , Grasas de la Dieta/administración & dosificación , Prueba de Tolerancia a la Glucosa , Metabolismo de los Lípidos , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/patología , Obesidad/metabolismo , Obesidad/patología , Oxidación-Reducción , Ratas
13.
J Lipid Res ; 54(2): 522-34, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23175776

RESUMEN

Intramuscular accumulation of triacylglycerol, in the form of lipid droplets (LD), has gained widespread attention as a hallmark of metabolic disease and insulin resistance. Paradoxically, LDs also amass in muscles of highly trained endurance athletes who are exquisitely insulin sensitive. Understanding the molecular mechanisms that mediate the expansion and appropriate metabolic control of LDs in the context of habitual physical activity could lead to new therapeutic opportunities. Herein, we show that acute exercise elicits robust upregulation of a broad program of genes involved in regulating LD assembly, morphology, localization, and mobilization. Prominent among these was perilipin-5, a scaffolding protein that affects the spatial and metabolic interactions between LD and their surrounding mitochondrial reticulum. Studies in transgenic mice and primary human skeletal myocytes established a key role for the exercise-responsive transcriptional coactivator PGC-1α in coordinating intramuscular LD programming with mitochondrial remodeling. Moreover, translational studies comparing physically active versus inactive humans identified a remarkably strong association between expression of intramuscular LD genes and enhanced insulin action in exercise-trained subjects. These results reveal an intimate molecular connection between intramuscular LD biology and mitochondrial metabolism that could prove relevant to the etiology and treatment of insulin resistance and other disorders of lipid imbalance.


Asunto(s)
Ejercicio Físico , Proteínas de Choque Térmico/metabolismo , Metabolismo de los Lípidos , Músculo Esquelético/citología , Orgánulos/metabolismo , Condicionamiento Físico Animal , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Femenino , Regulación de la Expresión Génica , Proteínas de Choque Térmico/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Transactivadores/genética , Factores de Transcripción/genética , Triglicéridos/metabolismo , Adulto Joven
14.
Am J Physiol Regul Integr Comp Physiol ; 304(3): R206-17, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23193112

RESUMEN

Disruptions of ovarian function in women are associated with increased risk of metabolic disease due to dysregulation of peripheral glucose homeostasis in skeletal muscle. Our previous evidence suggests that alterations in skeletal muscle lipid metabolism coupled with altered mitochondrial function may also develop. The objective of this study was to use an integrative metabolic approach to identify potential areas of dysfunction that develop in skeletal muscle from ovariectomized (OVX) female mice compared with age-matched ovary-intact adult female mice (sham). The OVX mice exhibited significant increases in body weight, visceral, and inguinal fat mass compared with sham mice. OVX mice also had significant increases in skeletal muscle intramyocellular lipids (IMCL) compared with the sham animals, which corresponded to significant increases in the protein content of the fatty acid transporters CD36/FAT and FABPpm. A targeted metabolic profiling approach identified significantly lower levels of specific acyl carnitine species and various amino acids in skeletal muscle from OVX mice compared with the sham animals, suggesting a potential dysfunction in lipid and amino acid metabolism, respectively. Basal and maximal mitochondrial oxygen consumption rates were significantly impaired in skeletal muscle fibers from OVX mice compared with sham animals. Collectively, these data indicate that loss of ovarian function results in increased IMCL storage that is coupled with alterations in mitochondrial function and changes in the skeletal muscle metabolic profile.


Asunto(s)
Metabolismo Energético/fisiología , Metabolismo de los Lípidos/fisiología , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Ovariectomía , Animales , Femenino , Ratones , Ratones Endogámicos C57BL
15.
Biochem J ; 448(1): 13-20, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22928974

RESUMEN

Published values regarding the sensitivity (IC(50)) of CPT-I (carnitine palmitoyltransferase I) to M-CoA (malonyl-CoA) inhibition in isolated mitochondria are inconsistent with predicted in vivo rates of fatty acid oxidation. Therefore we have re-examined M-CoA inhibition kinetics under various P-CoA (palmitoyl-CoA) concentrations in both isolated mitochondria and PMFs (permeabilized muscle fibres). PMFs have an 18-fold higher IC(50) (0.61 compared with 0.034 µM) in the presence of 25 µM P-CoA and a 13-fold higher IC(50) (6.3 compared with 0.49 µM) in the presence of 150 µM P-CoA compared with isolated mitochondria. M-CoA inhibition kinetics determined in PMFs predicts that CPT-I activity is inhibited by 33% in resting muscle compared with >95% in isolated mitochondria. Additionally, the ability of M-CoA to inhibit CPT-I appears to be dependent on P-CoA concentration, as the relative inhibitory capacity of M-CoA is decreased with increasing P-CoA concentrations. Altogether, the use of PMFs appears to provide an M-CoA IC(50) that better reflects the predicted in vivo rates of fatty acid oxidation. These findings also demonstrate that the ratio of [P-CoA]/[M-CoA] is critical for regulating CPT-I activity and may partially rectify the in vivo disconnect between M-CoA content and CPT-I flux within the context of exercise and Type 2 diabetes.


Asunto(s)
Carnitina O-Palmitoiltransferasa/metabolismo , Ácidos Grasos/metabolismo , Malonil Coenzima A/farmacología , Mitocondrias Musculares/enzimología , Animales , Carnitina/metabolismo , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Permeabilidad de la Membrana Celular , Relación Dosis-Respuesta a Droga , Concentración 50 Inhibidora , Cinética , Malonil Coenzima A/metabolismo , Mitocondrias Musculares/metabolismo , Fibras Musculares de Contracción Lenta/enzimología , Fibras Musculares de Contracción Lenta/metabolismo , Músculo Esquelético/enzimología , Oxidación-Reducción , Consumo de Oxígeno , Palmitoil Coenzima A/metabolismo , Condicionamiento Físico Animal , Ratas , Ratas Sprague-Dawley
16.
Cell Metab ; 35(6): 1038-1056.e8, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37060901

RESUMEN

Even-chain acylcarnitine (AC) metabolites, most of which are generated as byproducts of incomplete fatty acid oxidation (FAO), are viewed as biomarkers of mitochondrial lipid stress attributable to one or more metabolic bottlenecks in the ß-oxidation pathway. The origins and functional implications of FAO bottlenecks remain poorly understood. Here, we combined a sophisticated mitochondrial phenotyping platform with state-of-the-art molecular profiling tools and multiple two-state mouse models of respiratory function to uncover a mechanism that connects AC accumulation to lipid intolerance, metabolic inflexibility, and respiratory inefficiency in skeletal muscle mitochondria. These studies also identified a short-chain carbon circuit at the C4 node of FAO wherein reverse flux of glucose-derived acetyl CoA through medium-chain ketothiolase enhances lipid tolerance and redox stability in heart mitochondria by regenerating free CoA and NAD+. The findings help to explain why diminished FAO capacity, AC accumulation, and metabolic inflexibility are tightly linked to poor health outcomes.


Asunto(s)
Mitocondrias , Ácido Pirúvico , Ratones , Animales , Ácido Pirúvico/metabolismo , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Mitocondrias Musculares/metabolismo , Oxidación-Reducción , Lípidos , Ácidos Grasos/metabolismo
17.
Cell Rep ; 42(5): 112488, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37163372

RESUMEN

Disruption of adipocyte de novo lipogenesis (DNL) by deletion of fatty acid synthase (FASN) in mice induces browning in inguinal white adipose tissue (iWAT). However, adipocyte FASN knockout (KO) increases acetyl-coenzyme A (CoA) and malonyl-CoA in addition to depletion of palmitate. We explore which of these metabolite changes triggers adipose browning by generating eight adipose-selective KO mouse models with loss of ATP-citrate lyase (ACLY), acetyl-CoA carboxylase 1 (ACC1), ACC2, malonyl-CoA decarboxylase (MCD) or FASN, or dual KOs ACLY/FASN, ACC1/FASN, and ACC2/FASN. Preventing elevation of acetyl-CoA and malonyl-CoA by depletion of adipocyte ACLY or ACC1 in combination with FASN KO does not block the browning of iWAT. Conversely, elevating malonyl-CoA levels in MCD KO mice does not induce browning. Strikingly, adipose ACC1 KO induces a strong iWAT thermogenic response similar to FASN KO while also blocking malonyl-CoA and palmitate synthesis. Thus, ACC1 and FASN are strong suppressors of adipocyte thermogenesis through promoting lipid synthesis rather than modulating the DNL intermediates acetyl-CoA or malonyl-CoA.


Asunto(s)
Acetil-CoA Carboxilasa , Adipocitos , Ratones , Animales , Acetil-CoA Carboxilasa/metabolismo , Acetilcoenzima A/metabolismo , Adipocitos/metabolismo , Ratones Noqueados , Ácido Graso Sintasas/metabolismo , Termogénesis , Palmitatos/metabolismo
18.
J Clin Invest ; 133(9)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36927960

RESUMEN

During the development of heart failure (HF), the capacity for cardiomyocyte (CM) fatty acid oxidation (FAO) and ATP production is progressively diminished, contributing to pathologic cardiac hypertrophy and contractile dysfunction. Receptor-interacting protein 140 (RIP140, encoded by Nrip1) has been shown to function as a transcriptional corepressor of oxidative metabolism. We found that mice with striated muscle deficiency of RIP140 (strNrip1-/-) exhibited increased expression of a broad array of genes involved in mitochondrial energy metabolism and contractile function in heart and skeletal muscle. strNrip1-/- mice were resistant to the development of pressure overload-induced cardiac hypertrophy, and CM-specific RIP140-deficient (csNrip1-/-) mice were protected against the development of HF caused by pressure overload combined with myocardial infarction. Genomic enhancers activated by RIP140 deficiency in CMs were enriched in binding motifs for transcriptional regulators of mitochondrial function (estrogen-related receptor) and cardiac contractile proteins (myocyte enhancer factor 2). Consistent with a role in the control of cardiac fatty acid oxidation, loss of RIP140 in heart resulted in augmented triacylglyceride turnover and fatty acid utilization. We conclude that RIP140 functions as a suppressor of a transcriptional regulatory network that controls cardiac fuel metabolism and contractile function, representing a potential therapeutic target for the treatment of HF.


Asunto(s)
Insuficiencia Cardíaca , Proteína de Interacción con Receptores Nucleares 1 , Animales , Ratones , Cardiomegalia/metabolismo , Metabolismo Energético/genética , Ácidos Grasos/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Miocitos Cardíacos/metabolismo , Proteína de Interacción con Receptores Nucleares 1/genética , Proteína de Interacción con Receptores Nucleares 1/metabolismo
19.
J Biol Chem ; 286(49): 42188-42199, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22002063

RESUMEN

Triacylglyceride stored in cytosolic lipid droplets (LDs) constitutes a major energy reservoir in most eukaryotes. The regulated turnover of triacylglyceride in LDs provides fatty acids for mitochondrial ß-oxidation and ATP generation in physiological states of high demand for energy. The mechanisms for the formation of LDs in conditions of energy excess are not entirely understood. Fat storage-inducing transmembrane protein 2 (FIT2/FITM2) is the anciently conserved member of the fat storage-inducing transmembrane family of proteins implicated to be important in the formation of LDs, but its role in energy metabolism has not been tested. Here, we report that expression of FIT2 in mouse skeletal muscle had profound effects on muscle energy metabolism. Mice with skeletal muscle-specific overexpression of FIT2 (CKF2) had significantly increased intramyocellular triacylglyceride and complete protection from high fat diet-induced weight gain due to increased energy expenditure. Mass spectrometry-based metabolite profiling suggested that CKF2 skeletal muscle had increased oxidation of branched chain amino acids but decreased oxidation of fatty acids. Glucose was primarily utilized in CKF2 muscle for synthesis of the glycerol backbone of triacylglyceride and not for glycogen production. CKF2 muscle was ATP-deficient and had activated AMP kinase. Together, these studies indicate that FIT2 expression in skeletal muscle plays an unexpected function in regulating muscle energy metabolism and indicates an important role for lipid droplet formation in this process.


Asunto(s)
Tejido Adiposo/metabolismo , Proteínas de la Membrana/metabolismo , Músculo Esquelético/metabolismo , Adenilato Quinasa/metabolismo , Animales , Cruzamientos Genéticos , Retículo Endoplásmico/metabolismo , Metabolismo Energético , Glucosa/metabolismo , Lípidos/química , Espectrometría de Masas/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo , Músculos/metabolismo , Triglicéridos/química
20.
iScience ; 25(10): 105189, 2022 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-36274957

RESUMEN

Two prominent frontline breast cancer (BC) chemotherapies commonly used in combination, doxorubicin (DOX) and docetaxel (TAX), are associated with long-lasting cardiometabolic and musculoskeletal side effects. Whereas DOX has been linked to mitochondrial dysfunction, mechanisms underlying TAX-induced myotoxicities remain uncertain. Here, the metabolic and functional consequences of TAX ± DOX were investigated using a 3D-bioengineered model of adult human muscle and a drug dosing regimen designed to resemble in vivo pharmacokinetics. DOX potently reduced mitochondrial respiratory capacity, 3D-myobundle size, and contractile force, whereas TAX-induced acetylation and remodeling of the microtubule network led to perturbations in glucose uptake, mitochondrial respiratory sensitivity, and kinetics of fatigue, without compromising tetanic force generation. These findings suggest TAX-induced remodeling of the microtubule network disrupts glucose transport and respiratory control in skeletal muscle and thereby have important clinical implications related to the cardiometabolic health and quality of life of BC patients and survivors.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA