Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Lipid Res ; 58(12): 2324-2333, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29066466

RESUMO

Diacylglycerol kinases (DGKs) regulate the balance between diacylglycerol (DAG) and phosphatidic acid. DGKζ is highly abundant in skeletal muscle and induces fiber hypertrophy. We hypothesized that DGKζ influences functional and metabolic adaptations in skeletal muscle and whole-body fuel utilization. DAG content was increased in skeletal muscle and adipose tissue, but unaltered in liver of DGKζ KO mice. Linear growth, body weight, fat mass, and lean mass were reduced in DGKζ KO versus wild-type mice. Conversely, male DGKζ KO and wild-type mice displayed a similar robust increase in plantaris weight after functional overload, suggesting that DGKζ is dispensable for muscle hypertrophy. Although glucose tolerance was similar, insulin levels were reduced in high-fat diet (HFD)-fed DGKζ KO versus wild-type mice. Submaximal insulin-stimulated glucose transport and p-Akt Ser473 were increased, suggesting enhanced skeletal muscle insulin sensitivity. Energy homeostasis was altered in DGKζ KO mice, as evidenced by an elevated respiratory exchange ratio, independent of altered physical activity or food intake. In conclusion, DGKζ deficiency increases tissue DAG content and leads to modest growth retardation, reduced adiposity, and protection against insulin resistance. DGKζ plays a role in the control of growth and metabolic processes, further highlighting specialized functions of DGK isoforms in type 2 diabetes pathophysiology.


Assuntos
Diacilglicerol Quinase/genética , Metabolismo Energético/genética , Glucose/metabolismo , Resistência à Insulina/genética , Insulina/metabolismo , Animais , Transporte Biológico , Diacilglicerol Quinase/deficiência , Dieta Hiperlipídica , Diglicerídeos/metabolismo , Expressão Gênica , Homeostase/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Músculo Esquelético , Obesidade/etiologia , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia
2.
Am J Physiol Endocrinol Metab ; 313(4): E483-E491, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28720584

RESUMO

Insulin resistance is central to the development of type 2 diabetes and related metabolic disorders. Because skeletal muscle is responsible for the majority of whole body insulin-stimulated glucose uptake, regulation of glucose metabolism in this tissue is of particular importance. Although Rho GTPases and many of their affecters influence skeletal muscle metabolism, there is a paucity of information on the protein kinase N (PKN) family of serine/threonine protein kinases. We investigated the impact of PKN2 on insulin signaling and glucose metabolism in primary human skeletal muscle cells in vitro and mouse tibialis anterior muscle in vivo. PKN2 knockdown in vitro decreased insulin-stimulated glucose uptake, incorporation into glycogen, and oxidation. PKN2 siRNA increased 5'-adenosine monophosphate-activated protein kinase (AMPK) signaling while stimulating fatty acid oxidation and incorporation into triglycerides and decreasing protein synthesis. At the transcriptional level, PKN2 knockdown increased expression of PGC-1α and SREBP-1c and their target genes. In mature skeletal muscle, in vivo PKN2 knockdown decreased glucose uptake and increased AMPK phosphorylation. Thus, PKN2 alters key signaling pathways and transcriptional networks to regulate glucose and lipid metabolism. Identification of PKN2 as a novel regulator of insulin and AMPK signaling may provide an avenue for manipulation of skeletal muscle metabolism.


Assuntos
Adenilato Quinase/metabolismo , Glucose/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Proteína Quinase C/genética , Animais , Ácidos Graxos/metabolismo , Técnicas de Silenciamento de Genes , Glicogênio/metabolismo , Humanos , Técnicas In Vitro , Resistência à Insulina/genética , Metabolismo dos Lipídeos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Fosforilação , Biossíntese de Proteínas/genética , Proteína Quinase C/metabolismo , Músculo Quadríceps/citologia , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Triglicerídeos/metabolismo
3.
Am J Physiol Endocrinol Metab ; 310(6): E461-72, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26758685

RESUMO

Mechanisms regulating skeletal muscle growth involve a balance between the activity of serine/threonine protein kinases, including the mammalian target of rapamycin (mTOR) and 5'-AMP-activated protein kinase (AMPK). The contribution of different AMPK subunits to the regulation of cell growth size remains inadequately characterized. Using AMPKγ3 mutant-overexpressing transgenic Tg-Prkag3(225Q) and AMPKγ3-knockout (Prkag3(-/-)) mice, we investigated the requirement for the AMPKγ3 isoform in functional overload-induced muscle hypertrophy. Although the genetic disruption of the γ3 isoform did not impair muscle growth, control sham-operated AMPKγ3-transgenic mice displayed heavier plantaris muscles in response to overload hypertrophy and underwent smaller mass gain and lower Igf1 expression compared with wild-type littermates. The mTOR signaling pathway was upregulated with functional overload but unchanged between genetically modified animals and wild-type littermates. Differences in AMPK-related signaling pathways between transgenic, knockout, and wild-type mice did not impact muscle hypertrophy. Glycogen content was increased following overload in wild-type mice. In conclusion, our functional, transcriptional, and signaling data provide evidence against the involvement of the AMPKγ3 isoform in the regulation of skeletal muscle hypertrophy. Thus, the AMPKγ3 isoform is dispensable for functional overload-induced muscle growth. Mechanical loading can override signaling pathways that act as negative effectors of mTOR signaling and consequently promote skeletal muscle hypertrophy.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Fator de Crescimento Insulin-Like I/metabolismo , Músculo Esquelético/crescimento & desenvolvimento , Serina-Treonina Quinases TOR/metabolismo , Animais , Hipertrofia/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Tamanho do Órgão , Transdução de Sinais
4.
Surg Obes Relat Dis ; 11(3): 592-601, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25862179

RESUMO

BACKGROUND: Roux-en-Y gastric bypass (RYGB) surgery rapidly increases whole body insulin sensitivity, with changes in several organs including skeletal muscle. Objectives were to determine whether improvements in insulin action in skeletal muscle may occur directly at the level of the myocyte or secondarily from changes in systemic factors associated with weight loss. Myotubes were derived before and after RYGB surgery. The setting was Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden. METHODS: Eight patients (body mass index (BMI) 41.8 kg/m(2); age 41 yr) underwent RYGB surgery. Before and 6 months after RYGB surgery, skeletal muscle biopsies were collected from vastus lateralis muscle. Satellite cells derived from skeletal muscle biopsies were propagated in vitro as myoblasts and differentiated into myotubes. RESULTS: Expression of myogenic markers is increased in myoblasts derived from biopsies taken 6 months after bypass surgery, compared with their respective presurgery condition. Furthermore, glycogen synthesis, tyrosine phosphorylation of insulin receptor (IRS)-1-Tyr612 and Interleukin (IL)-8 secretion were increased, while fatty acid oxidation and circulating IL8 levels remain unaltered. Myotubes derived from muscle biopsies obtained after RYGB surgery displayed increased insulin-stimulated phosphorylation of protein kinase B (PKB)-Thr308 and proline-rich Akt substrate of 40 kDa (PRAS40)-Thr246. CONCLUSIONS: RYGB surgery is accompanied by enhanced glucose metabolism and insulin signaling, altered IL8 secretion and changes in mRNA levels and myogenic markers in cultured skeletal muscle cells. Thus, RYGB surgery involves intrinsic reprogramming of skeletal muscle to increase peripheral insulin sensitivity and glucose metabolism.


Assuntos
Derivação Gástrica/métodos , Glucose/metabolismo , Resistência à Insulina/fisiologia , Laparoscopia , Metabolismo dos Lipídeos , Músculo Esquelético/metabolismo , Obesidade Mórbida/cirurgia , Adulto , Biópsia , Células Cultivadas , Feminino , Humanos , Masculino , Músculo Esquelético/patologia , Obesidade Mórbida/metabolismo , Obesidade Mórbida/patologia
5.
Physiol Rep ; 3(3)2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25780092

RESUMO

A common polymorphism (R577X) in the α-actinin (ACTN) 3 gene, which leads to complete deficiency of a functional protein in skeletal muscle, could directly influence metabolism in the context of health and disease. Therefore, we tested the hypothesis that states of glucose tolerance are associated with the ACTN3 R577X genotype. We analyzed the prevalence of the ACTN3 R577X polymorphism in people with normal glucose tolerance (NGT) and type 2 diabetes (T2D) and measured muscle-specific α-actinin 2 and 3 mRNA and protein abundance in skeletal muscle biopsies. Furthermore, we investigated the protein abundance of the myosin heavy chain isoforms and the components of the mitochondrial electron transport chain in skeletal muscle from people with NGT or T2D. mRNA of selected sarcomeric z-disk proteins was also assessed. Although the prevalence of the ACTN3 577XX genotype was higher in T2D patients, genotype distribution was unrelated to metabolic control or obesity. ACTN2 and ACTN3 mRNA expression and protein abundance was unchanged between NGT and T2D participants. Protein abundance of mitochondrial complexes II and IV was related to genotype and glucose tolerance status. Gene expression of sarcomeric z-disk proteins was increased in skeletal muscle from NGT participants with the ACTN3 577XX genotype. While genetic variation in ACTN3 does not influence metabolic control, genotype does appear to influence gene expression of other sarcomeric proteins, which could contribute to the functional properties of skeletal muscle and the fatigue-resistant phenotype associated with the R577X polymorphism.

6.
Obesity (Silver Spring) ; 21(7): 1389-95, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23754443

RESUMO

INTRODUCTION: Trefoil factor family member 2 (Tff2) is a small gut peptide, mainly known for its protective and healing functions. As previously demonstrated, high-fat (HF) feeding can rapidly and specifically modulate Tff2 transcription in key tissues of mice, including the duodenum and mesenteric adipose tissue, therefore suggesting a novel role for this gene in energy balance. DESIGN AND METHODS: To explore whether and how Tff2 can influence feeding behavior and energy metabolism, Tff2 knock-out (KO) mice were challenged with HF diet for 12 weeks, hence food and energy intakes, body composition, as well as energy excretion and serum lipid and hormonal levels were analyzed. Finally, energy efficiency was estimated. RESULTS: Tff2 KO mice showed a greater appetite and higher energy intake compared to wild-type (WT). Consistently, they presented lower levels of serum leptin, and increased transcription of agouti-related protein (Agrp) in the hypothalamus. Though energy and triglyceride fecal excretion were augmented in Tff2 KO mice, digestible energy intake was superior. However, KO mice were finally protected from HF diet-induced obesity, and accumulated less weight and fat depots than WT animals, while keeping a normal lean mass. Energy efficiency was lower in HF-KO mice, while energy expenditure and locomotor activity were globally increased. CONCLUSIONS: The present work demonstrates previously unsuspected roles for Tff2 and suggests it to be a mastermind in the control of energy balance and a promising therapeutic target for obesity.


Assuntos
Dieta Hiperlipídica , Mucinas/genética , Proteínas Musculares/genética , Obesidade/genética , Peptídeos/genética , Tecido Adiposo/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Apetite , Composição Corporal , Ingestão de Energia , Metabolismo Energético , Regulação da Expressão Gênica , Hipotálamo/metabolismo , Leptina/sangue , Lipídeos/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucinas/metabolismo , Proteínas Musculares/metabolismo , Obesidade/sangue , Peptídeos/metabolismo , Saciação , Fator Trefoil-2
7.
Exp Gerontol ; 45(11): 896-903, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20813182

RESUMO

A compromised muscle function due to aging, sarcopenia and reduced level of physical activity can lead to metabolic complications and chronic diseases. Endurance exercise counters these diseases by inducing beneficial adaptations whose molecular mechanisms remain unclear. We have investigated the transcriptomic changes following mild-intensity endurance training in skeletal muscle of elderly men. Seven healthy subjects followed an exercise program of cycle ergometer training at lactate threshold (LT) level for 60 min/day, five times/week during six weeks. Physiological and transcriptomic changes were analyzed before and after training. LT training decreased percentage body fat and fasting levels of plasma glucose, while increasing high-density lipoprotein cholesterol and lecithin-cholesterol acyltransferase levels. Transcriptomic analysis revealed fast-to-slow fiber type transition, increased amount of mtDNA encoded transcripts and modulation of 12 transcripts notably related to extracellular matrix (ECM), oxidative phosphorylation (OXPHOS), as well as partially characterized and novel transcripts. The training simultaneously induced the expression of genes related to slow fiber type transition, OXPHOS and ECM, which might contribute to the improvement of glucose and lipid metabolisms and whole body aerobic capacity.


Assuntos
Perfilação da Expressão Gênica , Ácido Láctico/sangue , Músculo Esquelético/metabolismo , Educação Física e Treinamento , Resistência Física , Tecido Adiposo/anatomia & histologia , Idoso , Glicemia/análise , HDL-Colesterol/sangue , DNA Mitocondrial/genética , Limiar Diferencial , Matriz Extracelular/metabolismo , Jejum/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Fosforilação Oxidativa , Fosfatidilcolina-Esterol O-Aciltransferase/sangue , RNA/metabolismo , Fatores de Tempo
8.
J Steroid Biochem Mol Biol ; 122(1-3): 91-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20036330

RESUMO

In postmenopausal women, prevalence of metabolic syndrome (MS) is 40%. Aging is associated with a decline in basal metabolic rate and an alteration in tissue metabolism, leading to MS. Hormonal therapy has been shown to be effective against some of the MS-related features but its effects on sarcopenia and skeletal muscle metabolism remain unclear. We have analyzed the effects of estradiol (E(2)) on global gene expression in skeletal muscle of ovariectomized (OVX) female C57BL6 mice using the serial analysis of gene expression method. Animals were randomly assigned to six groups of each 14 mice: the vehicle group (OVX), and five groups in which E(2) was injected 1h, 3h, 6h, 18 h or 24h prior to sacrifice. E(2) modulated 177 transcripts, including 11 partially characterized transcripts and 52 potentially novel transcripts. Most of the differentially expressed transcripts were up-regulated at E(2)3h and E(2)18 h, while down-regulated transcripts were observed at E(2)6h and E(2)24h, illustrating two cycles of up and down E(2)-responsive genes. Modulated transcripts were involved in skeletal muscle structure/growth, fiber type distribution and energy metabolism. These results suggest that a single physiological dose of E(2) can concomitantly modulate transcripts determining skeletal muscle type and energy metabolism, which may in turn affect sarcopenia and MS.


Assuntos
Estradiol/farmacologia , Síndrome Metabólica/metabolismo , Fibras Musculares Esqueléticas/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Animais , Biologia Computacional , Metabolismo Energético/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Ovariectomia , RNA/química , RNA/genética , Distribuição Aleatória , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcopenia/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...