Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Metab ; 36(2): 422-437.e8, 2024 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-38325337

RESUMO

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.


Assuntos
Cetonas , Miocárdio , Camundongos , Animais , Cetonas/metabolismo , Miocárdio/metabolismo , Mitocôndrias/metabolismo , Oxirredução , Coração , Músculo Esquelético/metabolismo
2.
Cell Metab ; 35(6): 1038-1056.e8, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37060901

RESUMO

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.


Assuntos
Mitocôndrias , Ácido Pirúvico , Camundongos , Animais , Ácido Pirúvico/metabolismo , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Mitocôndrias Musculares/metabolismo , Oxirredução , Lipídeos , Ácidos Graxos/metabolismo
3.
iScience ; 25(10): 105189, 2022 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-36274957

RESUMO

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.

4.
iScience ; 25(1): 103635, 2022 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-35028529

RESUMO

Nicotinamide riboside supplements (NRS) have been touted as a nutraceutical that promotes cardiometabolic and musculoskeletal health by enhancing nicotinamide adenine dinucleotide (NAD+) biosynthesis, mitochondrial function, and/or the activities of NAD-dependent sirtuin deacetylase enzymes. This investigation examined the impact of NRS on whole body energy homeostasis, skeletal muscle mitochondrial function, and corresponding shifts in the acetyl-lysine proteome, in the context of diet-induced obesity using C57BL/6NJ mice. The study also included a genetically modified mouse model that imposes greater demand on sirtuin flux and associated NAD+ consumption, specifically within muscle tissues. In general, whole body glucose control was marginally improved by NRS when administered at the midpoint of a chronic high-fat diet, but not when given as a preventative therapy upon initiation of the diet. Contrary to anticipated outcomes, the study produced little evidence that NRS increases tissue NAD+ levels, augments mitochondrial function, and/or mitigates diet-induced hyperacetylation of the skeletal muscle proteome.

5.
Cell Metab ; 31(1): 131-147.e11, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31813822

RESUMO

This study sought to examine the functional significance of mitochondrial protein acetylation using a double knockout (DKO) mouse model harboring muscle-specific deficits in acetyl-CoA buffering and lysine deacetylation, due to genetic ablation of carnitine acetyltransferase and Sirtuin 3, respectively. DKO mice are highly susceptible to extreme hyperacetylation of the mitochondrial proteome and develop a more severe form of diet-induced insulin resistance than either single KO mouse line. However, the functional phenotype of hyperacetylated DKO mitochondria is largely normal. Of the >120 measures of respiratory function assayed, the most consistently observed traits of a markedly heightened acetyl-lysine landscape are enhanced oxygen flux in the context of fatty acid fuel and elevated rates of electron leak. In sum, the findings challenge the notion that lysine acetylation causes broad-ranging damage to mitochondrial quality and performance and raise the possibility that acetyl-lysine turnover, rather than acetyl-lysine stoichiometry, modulates redox balance and carbon flux.


Assuntos
Carnitina O-Acetiltransferase/genética , Resistência à Insulina/genética , Lisina/metabolismo , Mitocôndrias Musculares/metabolismo , Proteínas Mitocondriais/metabolismo , Estresse Oxidativo/genética , Sirtuína 3/genética , Acetilcoenzima A/metabolismo , Acetilação , Animais , Carnitina O-Acetiltransferase/metabolismo , Creatina Quinase/metabolismo , Dieta Hiperlipídica , Metabolismo Energético/genética , Homeostase , Peróxido de Hidrogênio/metabolismo , Insulina/sangue , Lisina/análogos & derivados , Masculino , Potencial da Membrana Mitocondrial/genética , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Camundongos Knockout , Mitocôndrias Musculares/genética , Proteínas Mitocondriais/genética , Oxirredução , Proteoma/genética , Proteoma/metabolismo , Sirtuína 3/metabolismo , Termodinâmica
6.
Cell Rep ; 26(6): 1557-1572.e8, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30726738

RESUMO

Acyl CoA metabolites derived from the catabolism of carbon fuels can react with lysine residues of mitochondrial proteins, giving rise to a large family of post-translational modifications (PTMs). Mass spectrometry-based detection of thousands of acyl-PTMs scattered throughout the proteome has established a strong link between mitochondrial hyperacylation and cardiometabolic diseases; however, the functional consequences of these modifications remain uncertain. Here, we use a comprehensive respiratory diagnostics platform to evaluate three disparate models of mitochondrial hyperacylation in the mouse heart caused by genetic deletion of malonyl CoA decarboxylase (MCD), SIRT5 demalonylase and desuccinylase, or SIRT3 deacetylase. In each case, elevated acylation is accompanied by marginal respiratory phenotypes. Of the >60 mitochondrial energy fluxes evaluated, the only outcome consistently observed across models is a ∼15% decrease in ATP synthase activity. In sum, the findings suggest that the vast majority of mitochondrial acyl PTMs occur as stochastic events that minimally affect mitochondrial bioenergetics.


Assuntos
Metabolismo Energético , Mitocôndrias Cardíacas/metabolismo , Processamento de Proteína Pós-Traducional , Acetilação , Animais , Carboxiliases/metabolismo , Respiração Celular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sirtuína 3/metabolismo , Sirtuínas/metabolismo
7.
Nucleic Acids Res ; 47(4): e23, 2019 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-30590691

RESUMO

Genetic manipulation via transgene overexpression, RNAi, or Cas9-based methods is central to biomedical research. Unfortunately, use of these tools is often limited by vector options. We have created a modular platform (pMVP) that allows a gene of interest to be studied in the context of an array of promoters, epitope tags, conditional expression modalities, and fluorescent reporters, packaged in 35 custom destination vectors, including adenovirus, lentivirus, PiggyBac transposon, and Sleeping Beauty transposon, in aggregate >108,000 vector permutations. We also used pMVP to build an epigenetic engineering platform, pMAGIC, that packages multiple gRNAs and either Sa-dCas9 or x-dCas9(3.7) fused to one of five epigenetic modifiers. Importantly, via its compatibility with adenoviral vectors, pMAGIC uniquely enables use of dCas9/LSD1 fusions to interrogate enhancers within primary cells. To demonstrate this, we used pMAGIC to target Sa-dCas9/LSD1 and modify the epigenetic status of a conserved enhancer, resulting in altered expression of the homeobox transcription factor PDX1 and its target genes in pancreatic islets and insulinoma cells. In sum, the pMVP and pMAGIC systems empower researchers to rapidly generate purpose-built, customized vectors for manipulation of gene expression, including via targeted epigenetic modification of regulatory elements in a broad range of disease-relevant cell types.


Assuntos
Sistemas CRISPR-Cas/genética , Engenharia Genética/métodos , Vetores Genéticos/genética , Proteínas de Homeodomínio/genética , Transativadores/genética , Transgenes/genética , Adenoviridae/genética , Animais , Elementos de DNA Transponíveis/genética , Elementos Facilitadores Genéticos/genética , Epigenômica/métodos , Edição de Genes/métodos , Regulação da Expressão Gênica/genética , Células HEK293 , Histona Desmetilases/genética , Humanos , Insulinoma/metabolismo , Ilhotas Pancreáticas/metabolismo , Lentivirus/genética , Camundongos , Regiões Promotoras Genéticas/genética , RNA Guia de Cinetoplastídeos/genética , Ratos
8.
PLoS Genet ; 11(11): e1005553, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26540294

RESUMO

Levels of certain circulating short-chain dicarboxylacylcarnitine (SCDA), long-chain dicarboxylacylcarnitine (LCDA) and medium chain acylcarnitine (MCA) metabolites are heritable and predict cardiovascular disease (CVD) events. Little is known about the biological pathways that influence levels of most of these metabolites. Here, we analyzed genetics, epigenetics, and transcriptomics with metabolomics in samples from a large CVD cohort to identify novel genetic markers for CVD and to better understand the role of metabolites in CVD pathogenesis. Using genomewide association in the CATHGEN cohort (N = 1490), we observed associations of several metabolites with genetic loci. Our strongest findings were for SCDA metabolite levels with variants in genes that regulate components of endoplasmic reticulum (ER) stress (USP3, HERC1, STIM1, SEL1L, FBXO25, SUGT1) These findings were validated in a second cohort of CATHGEN subjects (N = 2022, combined p = 8.4x10-6-2.3x10-10). Importantly, variants in these genes independently predicted CVD events. Association of genomewide methylation profiles with SCDA metabolites identified two ER stress genes as differentially methylated (BRSK2 and HOOK2). Expression quantitative trait loci (eQTL) pathway analyses driven by gene variants and SCDA metabolites corroborated perturbations in ER stress and highlighted the ubiquitin proteasome system (UPS) arm. Moreover, culture of human kidney cells in the presence of levels of fatty acids found in individuals with cardiometabolic disease, induced accumulation of SCDA metabolites in parallel with increases in the ER stress marker BiP. Thus, our integrative strategy implicates the UPS arm of the ER stress pathway in CVD pathogenesis, and identifies novel genetic loci associated with CVD event risk.


Assuntos
Doenças Cardiovasculares/genética , Metabolômica , Complexo de Endopeptidases do Proteassoma/genética , Locos de Características Quantitativas , Ubiquitina/genética , Doenças Cardiovasculares/patologia , Carnitina/análogos & derivados , Carnitina/metabolismo , Metilação de DNA , Estresse do Retículo Endoplasmático/genética , Humanos , Polimorfismo de Nucleotídeo Único , Fatores de Risco
9.
Diabetes ; 64(5): 1532-43, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25422105

RESUMO

This study used mice with muscle-specific overexpression of PGC-1α, a transcriptional coactivator that promotes mitochondrial biogenesis, to determine whether increased oxidative potential facilitates metabolic improvements in response to lifestyle modification. MCK-PGC1α mice and nontransgenic (NT) littermates were fed a high-fat diet (HFD) for 10 weeks, followed by stepwise exposures to voluntary wheel running (HFD+Ex) and then 25% caloric restriction with exercise (Ex/CR), each for an additional 10 weeks with continued HFD. Running and CR improved weight and glucose control similarly in MCK-PGC1α and NT mice. Sedentary MCK-PGC1α mice were more susceptible to diet-induced glucose intolerance, and insulin action measured in isolated skeletal muscles remained lower in the transgenic compared with the NT group, even after Ex/CR. Comprehensive profiling of >200 metabolites and lipid intermediates revealed dramatic group-specific responses to the intervention but did not produce a lead candidate that tracked with changes in glucose tolerance irrespective of genotype. Instead, principal components analysis identified a chemically diverse metabolite cluster that correlated with multiple measures of insulin responsiveness. These findings challenge the notion that increased oxidative capacity defends whole-body energy homeostasis and suggest that the interplay between mitochondrial performance, lipotoxicity, and insulin action is more complex than previously proposed.


Assuntos
Restrição Calórica , Músculo Esquelético/metabolismo , Condicionamento Físico Animal , Fatores de Transcrição/metabolismo , Animais , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/efeitos adversos , Metabolismo Energético , Regulação da Expressão Gênica , Masculino , Camundongos , Mitocôndrias Musculares/metabolismo , Atividade Motora , Oxirredução , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Proteômica , Fatores de Transcrição/genética
10.
J Biol Chem ; 289(12): 8106-20, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24482226

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Metabolismo Energético , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Oxirredutases/metabolismo , Tiorredoxinas/metabolismo , Animais , Proteínas de Transporte/genética , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/genética , Músculo Esquelético/enzimologia , Oxirredução , Tiorredoxinas/genética
11.
J Lipid Res ; 55(4): 635-44, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24395925

RESUMO

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.


Assuntos
Carnitina O-Acetiltransferase/metabolismo , Obesidade/enzimologia , Acetilcoenzima A/metabolismo , Animais , Carnitina/análogos & derivados , Carnitina/metabolismo , Carnitina O-Palmitoiltransferase/metabolismo , Células Cultivadas , Diabetes Mellitus/enzimologia , Humanos , Metabolismo dos Lipídeos , Masculino , Fibras Musculares Esqueléticas/enzimologia , Complexo Piruvato Desidrogenase/metabolismo , Ratos Wistar , Ratos Zucker
12.
Muscle Nerve ; 36(1): 71-80, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17455272

RESUMO

Because previous studies of three-dimensional skeletal muscle cultures have shown limited differentiation, the goal of this study was to establish conditions that would produce mature sarcomeres in a mammalian-derived skeletal muscle construct. We evaluated the differentiation of bioartificial muscles generated from C(2)C(12) myoblasts in a collagen gel cultured under steady, passive tension for up to 36 days. Staining for alpha-actinin, myosin, and F-actin indicated the presence of striated fibers as early as 6 days post-differentiation. Electron microscopy at 16 days post-differentiation revealed multinucleated myotubes with ordered, striated myofibers. At 33 days, the cultures contained collagen fibers and showed localization of paxillin at the fiber termini, suggesting that myotendinous junctions were forming. The present study demonstrates mature muscle synthesis in a three-dimensional system using a pure mammalian myoblast cell line. Our results suggest that this culture model can be used to evaluate the effects of various mechanical and biochemical cues on muscle development under normal and pathological conditions.


Assuntos
Diferenciação Celular/fisiologia , Colágeno/fisiologia , Músculo Esquelético/citologia , Sarcômeros/ultraestrutura , Animais , Técnicas de Cultura de Células , Células Cultivadas , Imageamento Tridimensional , Camundongos , Camundongos Endogâmicos C3H , Microscopia Eletrônica de Transmissão/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...