Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Diabetes Obes Metab ; 25(7): 1985-1994, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36999233

RESUMO

AIM: To determine the effects of astaxanthin treatment on lipids, cardiovascular disease (CVD) markers, glucose tolerance, insulin action and inflammation in individuals with prediabetes and dyslipidaemia. MATERIALS AND METHODS: Adult participants with dyslipidaemia and prediabetes (n = 34) underwent baseline blood draw, an oral glucose tolerance test and a one-step hyperinsulinaemic-euglycaemic clamp. They were then randomized (n = 22 treated, 12 placebo) to receive astaxanthin 12 mg daily or placebo for 24 weeks. Baseline studies were repeated after 12 and 24 weeks of therapy. RESULTS: After 24 weeks, astaxanthin treatment significantly decreased low-density lipoprotein (-0.33 ± 0.11 mM) and total cholesterol (-0.30 ± 0.14 mM) (both P < .05). Astaxanthin also reduced levels of the CVD risk markers fibrinogen (-473 ± 210 ng/mL), L-selectin (-0.08 ± 0.03 ng/mL) and fetuin-A (-10.3 ± 3.6 ng/mL) (all P < .05). While the effects of astaxanthin treatment did not reach statistical significance, there were trends toward improvements in the primary outcome measure, insulin-stimulated, whole-body glucose disposal (+0.52 ± 0.37 mg/m2 /min, P = .078), as well as fasting [insulin] (-5.6 ± 8.4 pM, P = .097) and HOMA2-IR (-0.31 ± 0.16, P = .060), suggesting improved insulin action. No consistent significant differences from baseline were observed for any of these outcomes in the placebo group. Astaxanthin was safe and well tolerated with no clinically significant adverse events. CONCLUSIONS: Although the primary endpoint did not meet the prespecified significance level, these data suggest that astaxanthin is a safe over-the-counter supplement that improves lipid profiles and markers of CVD risk in individuals with prediabetes and dyslipidaemia.


Assuntos
Doenças Cardiovasculares , Dislipidemias , Estado Pré-Diabético , Adulto , Humanos , Estado Pré-Diabético/complicações , Estado Pré-Diabético/tratamento farmacológico , Antioxidantes/uso terapêutico , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/prevenção & controle , Glicemia , Fatores de Risco , Insulina/uso terapêutico , Glucose/uso terapêutico , Colesterol , Fatores de Risco de Doenças Cardíacas , Dislipidemias/tratamento farmacológico
2.
Mol Cell ; 56(3): 425-435, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25458843

RESUMO

Pyruvate lies at a central biochemical node connecting carbohydrate, amino acid, and fatty acid metabolism, and the regulation of pyruvate flux into mitochondria represents a critical step in intermediary metabolism impacting numerous diseases. To characterize changes in mitochondrial substrate utilization in the context of compromised mitochondrial pyruvate transport, we applied (13)C metabolic flux analysis (MFA) to cells after transcriptional or pharmacological inhibition of the mitochondrial pyruvate carrier (MPC). Despite profound suppression of both glucose and pyruvate oxidation, cell growth, oxygen consumption, and tricarboxylic acid (TCA) metabolism were surprisingly maintained. Oxidative TCA flux was achieved through enhanced reliance on glutaminolysis through malic enzyme and pyruvate dehydrogenase (PDH) as well as fatty acid and branched-chain amino acid oxidation. Thus, in contrast to inhibition of complex I or PDH, suppression of pyruvate transport induces a form of metabolic flexibility associated with the use of lipids and amino acids as catabolic and anabolic fuels.


Assuntos
Pró-Proteína Convertase 1/metabolismo , Pró-Proteína Convertase 2/metabolismo , Ácido Pirúvico/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Ciclo do Ácido Cítrico , Ácidos Graxos/metabolismo , Glutamina/metabolismo , Humanos , Lipogênese , Análise do Fluxo Metabólico , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Oxirredução
3.
Mol Biol Rep ; 48(1): 601-610, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33411234

RESUMO

Angiogenesis is a multistep process requiring endothelial cell activation, migration, proliferation and tube formation. We recently reported that elevated secretion of interlukin 8 (IL8) by myotubes (MT) from subjects with Type-2 Diabetes (T2D) reduced angiogenesis by human umbilical vein endothelial cells (HUVEC) and human skeletal muscle explants. This lower vascularization was mediated through impaired activation of the phosphatidylinositol 3-kinase (PI3K)-pathway. We sought to investigate additional signaling elements that might mediate reduced angiogenesis. HUVEC were exposed to levels of IL8 equal to those secreted by MT from non-diabetic (ND) and T2D subjects and the involvement of components in the angiogenic response pathway examined. Cellular content of reactive oxygen species and Nitrate secretion were similar after treatment with [ND-IL8] and [T2D-IL8]. CXCR1 protein was down-regulated after treatment with [T2D-IL8] (p < 0.01 vs [ND-IL8] treatment); CXCR2 expression was unaltered. Addition of neutralizing antibodies against CXCR1 and CXCR2 to HUVEC treated with IL8 confirmed that CXCR1 alone mediated the angiogenic response to IL8. A key modulator of angiogenesis is matrix metalloproteinase-2 (MMP2). MMP2 secretion was higher after treatment with [ND-IL8] vs [T2D-IL8] (p < 0.01). MMP2 inhibition reduced tube formation to greater extent with [ND-IL8] than with [T2D-IL8] (p < 0.005). The PI3K-pathway inhibitor LY294002 reduced IL8-induced MMP2 release. IL8 regulation of MMP2 release was CXCR1 dependent, as anti-CXCR1 significantly reduced MMP2 release (p < 0.05). These results suggest that high levels of IL8 secreted by T2D MT trigger reduced capillarization via lower activation of a CXCR1-PI3K pathway, followed by impaired release and activity of MMP2.


Assuntos
Diabetes Mellitus Tipo 2/genética , Interleucina-8/genética , Metaloproteinase 2 da Matriz/genética , Fibras Musculares Esqueléticas/metabolismo , Receptores de Interleucina-8A/genética , Movimento Celular/genética , Proliferação de Células/genética , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Interleucina-8/farmacologia , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Fosfatidilinositol 3-Quinases/genética , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
Nat Chem Biol ; 14(11): 1021-1031, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30327559

RESUMO

Fatty acid synthase (FASN) predominantly generates straight-chain fatty acids using acetyl-CoA as the initiating substrate. However, monomethyl branched-chain fatty acids (mmBCFAs) are also present in mammals but are thought to be primarily diet derived. Here we demonstrate that mmBCFAs are de novo synthesized via mitochondrial BCAA catabolism, exported to the cytosol by adipose-specific expression of carnitine acetyltransferase (CrAT), and elongated by FASN. Brown fat exhibits the highest BCAA catabolic and mmBCFA synthesis fluxes, whereas these lipids are largely absent from liver and brain. mmBCFA synthesis is also sustained in the absence of microbiota. We identify hypoxia as a potent suppressor of BCAA catabolism that decreases mmBCFA synthesis in obese adipose tissue, such that mmBCFAs are significantly decreased in obese animals. These results identify adipose tissue mmBCFA synthesis as a novel link between BCAA metabolism and lipogenesis, highlighting roles for CrAT and FASN promiscuity influencing acyl-chain diversity in the lipidome.


Assuntos
Tecido Adiposo/enzimologia , Aminoácidos de Cadeia Ramificada/metabolismo , Ácido Graxo Sintases/metabolismo , Ácidos Graxos/biossíntese , Obesidade/enzimologia , Células 3T3 , Adipócitos/citologia , Animais , Sistemas CRISPR-Cas , Carnitina O-Acetiltransferase/metabolismo , Citosol/metabolismo , Feminino , Hipóxia , Lentivirus/genética , Lipogênese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , RNA Interferente Pequeno/metabolismo
5.
Diabetes Obes Metab ; 21(4): 812-821, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30447037

RESUMO

AIM: To evaluate the impact of the sodium glucose co-transporter 2 inhibitor canagliflozin on intrahepatic triglyceride (IHTG) accumulation and its relationship to changes in body weight and glucose metabolism. MATERIALS AND METHODS: In this double-blind, parallel-group, placebo-controlled, 24-week trial subjects with inadequately controlled type 2 diabetes mellitus (T2DM; HbA1c = 7.7% ± 0.7%) from two centres were randomly assigned (1:1) to canagliflozin 300 mg or placebo. We measured IHTG by proton-magnetic resonance spectroscopy (primary outcome), hepatic/muscle/adipose tissue insulin sensitivity during a 2-step euglycaemic insulin clamp, and beta-cell function during a mixed meal tolerance test. Analyses were per protocol. RESULTS: Between 8 September 2014-13 June 2016, 56 patients were enrolled. Canagliflozin reduced HbA1c (placebo-subtracted change: -0.71% [-1.08; -0.33]) and body weight (-3.4% [-5.4; -1.4]; both P ≤ 0.001). A numerically larger absolute decrease in IHTG occurred with canagliflozin (-4.6% [-6.4; -2.7]) versus placebo (-2.4% [-4.2; -0.6]; P = 0.09). In patients with non-alcoholic fatty liver disease (n = 37), the decrease in IHTG was -6.9% (-9.5; -4.2) versus -3.8% (-6.3; -1.3; P = 0.05), and strongly correlated with the magnitude of weight loss (r = 0.69, P < 0.001). Body weight loss ≥5% with a ≥30% relative reduction in IHTG occurred more often with canagliflozin (38% vs. 7%, P = 0.009). Hepatic insulin sensitivity improved with canagliflozin (P < 0.01), but not muscle or adipose tissue insulin sensitivity. Beta-cell glucose sensitivity, insulin clearance, and disposition index improved more with canagliflozin (P < 0.05). CONCLUSIONS: Canagliflozin improves hepatic insulin sensitivity and insulin secretion and clearance in patients with T2DM. IHTG decreases in proportion to the magnitude of body weight loss, which tended to be greater and occur more often with canagliflozin.


Assuntos
Canagliflozina/uso terapêutico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Resistência à Insulina , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Fígado/metabolismo , Inibidores do Transportador 2 de Sódio-Glicose/uso terapêutico , Triglicerídeos/metabolismo , Idoso , Diabetes Mellitus Tipo 2/metabolismo , Método Duplo-Cego , Feminino , Técnica Clamp de Glucose , Hemoglobinas Glicadas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Espectroscopia de Prótons por Ressonância Magnética , Resultado do Tratamento , Redução de Peso
6.
Diabetologia ; 61(12): 2674, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30324489

RESUMO

Owing to an oversight, the authors omitted to note that Dr Taub is a co-founder of and equity holder in Cardero Therapeutics.

7.
J Biol Chem ; 292(29): 12339-12350, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28572512

RESUMO

Chronic inflammation may contribute to insulin resistance via molecular cross-talk between pathways for pro-inflammatory and insulin signaling. Interleukin 1 receptor-associated kinase 1 (IRAK-1) mediates pro-inflammatory signaling via IL-1 receptor/Toll-like receptors, which may contribute to insulin resistance, but this hypothesis is untested. Here, we used male Irak1 null (k/o) mice to investigate the metabolic role of IRAK-1. C57BL/6 wild-type (WT) and k/o mice had comparable body weights on low-fat and high-fat diets (LFD and HFD, respectively). After 12 weeks on LFD (but not HFD), k/o mice (versus WT) had substantially improved glucose tolerance (assessed by the intraperitoneal glucose tolerance test (IPGTT)). As assessed with the hyperinsulinemic euglycemic glucose clamp technique, insulin sensitivity was 30% higher in the Irak1 k/o mice on chow diet, but the Irak1 deletion did not affect IPGTT outcomes in mice on HFD, suggesting that the deletion did not overcome the impact of obesity on glucose tolerance. Moreover, insulin-stimulated glucose-disposal rates were higher in the k/o mice, but we detected no significant difference in hepatic glucose production rates (± insulin infusion). Positron emission/computed tomography scans indicated higher insulin-stimulated glucose uptake in muscle, but not liver, in Irak1 k/o mice in vivo Moreover, insulin-stimulated phosphorylation of Akt was higher in muscle, but not in liver, from Irak1 k/o mice ex vivo In conclusion, Irak1 deletion improved muscle insulin sensitivity, with the effect being most apparent in LFD mice.


Assuntos
Intolerância à Glucose/metabolismo , Resistência à Insulina , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Músculo Esquelético/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Cruzamentos Genéticos , Dieta com Restrição de Gorduras , Dieta Hiperlipídica/efeitos adversos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Técnica Clamp de Glucose , Intolerância à Glucose/etiologia , Intolerância à Glucose/fisiopatologia , Intolerância à Glucose/prevenção & controle , Hemizigoto , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Quinases Associadas a Receptores de Interleucina-1/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/enzimologia , Obesidade/etiologia , Obesidade/fisiopatologia , Especificidade de Órgãos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Gordura Subcutânea Abdominal/efeitos dos fármacos , Gordura Subcutânea Abdominal/enzimologia , Gordura Subcutânea Abdominal/metabolismo
8.
Nat Chem Biol ; 12(1): 15-21, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26571352

RESUMO

Adipose tissue plays important roles in regulating carbohydrate and lipid homeostasis, but less is known about the regulation of amino acid metabolism in adipocytes. Here we applied isotope tracing to pre-adipocytes and differentiated adipocytes to quantify the contributions of different substrates to tricarboxylic acid (TCA) metabolism and lipogenesis. In contrast to proliferating cells, which use glucose and glutamine for acetyl-coenzyme A (AcCoA) generation, differentiated adipocytes showed increased branched-chain amino acid (BCAA) catabolic flux such that leucine and isoleucine from medium and/or from protein catabolism accounted for as much as 30% of lipogenic AcCoA pools. Medium cobalamin deficiency caused methylmalonic acid accumulation and odd-chain fatty acid synthesis. Vitamin B12 supplementation reduced these metabolites and altered the balance of substrates entering mitochondria. Finally, inhibition of BCAA catabolism compromised adipogenesis. These results quantitatively highlight the contribution of BCAAs to adipocyte metabolism and suggest that BCAA catabolism has a functional role in adipocyte differentiation.


Assuntos
Adipócitos/citologia , Adipócitos/metabolismo , Aminoácidos de Cadeia Ramificada/metabolismo , Lipogênese , Obesidade/metabolismo , 3-Metil-2-Oxobutanoato Desidrogenase (Lipoamida)/genética , 3-Metil-2-Oxobutanoato Desidrogenase (Lipoamida)/metabolismo , Células 3T3-L1/efeitos dos fármacos , Acetilcoenzima A/metabolismo , Adipócitos/efeitos dos fármacos , Adipogenia/fisiologia , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Humanos , Camundongos , Dados de Sequência Molecular , Obesidade/cirurgia , Ácidos Tricarboxílicos/metabolismo , Vitamina B 12/farmacologia
9.
Diabetologia ; 60(10): 2052-2065, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28770317

RESUMO

AIMS/HYPOTHESIS: Mitochondria are important regulators of the metabolic phenotype in type 2 diabetes. A key factor in mitochondrial physiology is the H+-ATP synthase. The expression and activity of its physiological inhibitor, ATPase inhibitory factor 1 (IF1), controls tissue homeostasis, metabolic reprogramming and signalling. We aimed to characterise the putative role of IF1 in mediating skeletal muscle metabolism in obesity and diabetes. METHODS: We examined the 'mitochondrial signature' of obesity and type 2 diabetes in a cohort of 100 metabolically characterised human skeletal muscle biopsy samples. The expression and activity of H+-ATP synthase, IF1 and key mitochondrial proteins were characterised, including their association with BMI, fasting plasma insulin, fasting plasma glucose and HOMA-IR. IF1 was also overexpressed in primary cultures of human myotubes derived from the same biopsies to unveil the possible role played by the pathological inhibition of the H+-ATP synthase in skeletal muscle. RESULTS: The results indicate that type 2 diabetes and obesity act via different mechanisms to impair H+-ATP synthase activity in human skeletal muscle (76% reduction in its catalytic subunit vs 280% increase in IF1 expression, respectively) and unveil a new pathway by which IF1 influences lipid metabolism. Mechanistically, IF1 altered cellular levels of α-ketoglutarate and L-carnitine metabolism in the myotubes of obese (84% of control) and diabetic (76% of control) individuals, leading to limited ß-oxidation of fatty acids (60% of control) and their cytosolic accumulation (164% of control). These events led to enhanced release of TNF-α (10 ± 2 pg/ml, 27 ± 5 pg/ml and 35 ± 4 pg/ml in control, obese and type 2 diabetic participants, respectively), which probably contributes to an insulin resistant phenotype. CONCLUSIONS/INTERPRETATION: Overall, our data highlight IF1 as a novel regulator of lipid metabolism and metabolic disorders, and a possible target for therapeutic intervention.


Assuntos
Dislipidemias/metabolismo , Resistência à Insulina/fisiologia , Mitocôndrias Musculares/metabolismo , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Músculo Esquelético/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Humanos , Masculino , Obesidade/metabolismo , Proteômica
10.
Proc Natl Acad Sci U S A ; 110(14): 5422-7, 2013 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-23513224

RESUMO

Facilitated pyruvate transport across the mitochondrial inner membrane is a critical step in carbohydrate, amino acid, and lipid metabolism. We report that clinically relevant concentrations of thiazolidinediones (TZDs), a widely used class of insulin sensitizers, acutely and specifically inhibit mitochondrial pyruvate carrier (MPC) activity in a variety of cell types. Respiratory inhibition was overcome with methyl pyruvate, localizing the effect to facilitated pyruvate transport, and knockdown of either paralog, MPC1 or MPC2, decreased the EC50 for respiratory inhibition by TZDs. Acute MPC inhibition significantly enhanced glucose uptake in human skeletal muscle myocytes after 2 h. These data (i) report that clinically used TZDs inhibit the MPC, (ii) validate that MPC1 and MPC2 are obligatory components of facilitated pyruvate transport in mammalian cells, (iii) indicate that the acute effect of TZDs may be related to insulin sensitization, and (iv) establish mitochondrial pyruvate uptake as a potential therapeutic target for diseases rooted in metabolic dysfunction.


Assuntos
Respiração Celular/efeitos dos fármacos , Proteínas de Membrana Transportadoras/metabolismo , Redes e Vias Metabólicas/fisiologia , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/antagonistas & inibidores , Tiazolidinedionas/farmacologia , Acrilatos/farmacologia , Análise de Variância , Animais , Proteínas de Transporte de Ânions , Western Blotting , Linhagem Celular , Citocromos c/metabolismo , Glucose/metabolismo , Humanos , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Proteínas de Transporte da Membrana Mitocondrial , Proteínas Mitocondriais/metabolismo , Transportadores de Ácidos Monocarboxílicos , Músculo Esquelético/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Carreadoras de Solutos , Tiazolidinedionas/metabolismo
11.
Am J Physiol Endocrinol Metab ; 309(1): E22-34, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25944879

RESUMO

Reduced capillary density is a feature of skeletal muscle (SkM) in type 2 diabetes (T2D), which is associated with multiple metabolic and functional abnormalities. SkM has been identified as a secretory tissue, releasing myokines that regulate multiple processes, including vascularization. We sought to determine how myokines secreted from T2D myotubes might influence SkM angiogenesis. Conditioned media (CM) were generated by myotubes from T2D and nondiabetic (ND) subjects. Primary human endothelial cells (HUVEC) and SkM explants were exposed to CM or recombinant myokines, and tube number or capillary outgrowth was determined as well as measurement of protein expression and phosphorylation. CM from ND myotubes stimulated tube formation of HUVEC to a greater extent than T2D myotubes (T2D-CM = 100%, ND-CM = 288 ± 90% after 48 h, P < 0.05). The effects of T2D myotube CM were mediated by IL-8, not IL-15 or GROα, and were due not to cell damage but rather through regulating tube production and maintenance (response to T2D-IL-8 = 100%, response to ND-IL-8 = 263 ± 46% after 48 h, P < 0.05). A similar effect was seen in SkM explants with exposure to IL-8. The dose-dependent effect of IL-8 on tube formation was also observable in the PI3K and FAK signaling pathways and mediated at least in part by PI3K, leading to regulation of Tie2 expression. These results suggest that elevated levels of IL-8 secreted from T2D myotubes create a muscle microenvironment that supports reduced capillarization in T2D. Impaired vascularization of SkM limits the availability of substrates, including glucose and contributes to the T2D phenotype.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Interleucina-8/metabolismo , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Receptor TIE-2/fisiologia , Estudos de Casos e Controles , Proliferação de Células , Células Cultivadas , Diabetes Mellitus Tipo 2/patologia , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Interleucina-8/sangue , Masculino , Pessoa de Meia-Idade , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/patologia , Transdução de Sinais/fisiologia
12.
Am J Physiol Endocrinol Metab ; 306(7): E756-68, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24473437

RESUMO

Adipose dysfunction resulting from chronic inflammation and impaired adipogenesis has increasingly been recognized as a major contributor to obesity-mediated insulin resistance, but the molecular mechanisms that maintain healthy adipocytes and limit adipose inflammation remain unclear. Here, we used genetic and pharmacological approaches to delineate a novel role for sphingosine kinase 1 (SK1) in metabolic disorders associated with obesity. SK1 phosphorylates sphingosine to form sphingosine 1 phosphate (S1P), a bioactive sphingolipid with numerous roles in inflammation. SK1 mRNA expression was increased in adipose tissue of diet-induced obese (DIO) mice and obese type 2 diabetic humans. In DIO mice, SK1 deficiency increased markers of adipogenesis and adipose gene expression of the anti-inflammatory molecules IL-10 and adiponectin and reduced adipose tissue macrophage (ATM) recruitment and proinflammatory molecules TNFα and IL-6. These changes were associated with enhanced insulin signaling in adipose and muscle and improved systemic insulin sensitivity and glucose tolerance in SK1(-/-) mice. Specific pharmacological inhibition of SK1 in WT DIO mice also reduced adipocyte and ATM inflammation and improved overall glucose homeostasis. These data suggest that the SK1-S1P axis could be an attractive target for the development of treatments to ameliorate adipose inflammation and insulin resistance associated with obesity and type 2 diabetes.


Assuntos
Tecido Adiposo/metabolismo , Mediadores da Inflamação/metabolismo , Resistência à Insulina/genética , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Adipócitos/imunologia , Adipócitos/metabolismo , Tecido Adiposo/imunologia , Adulto , Idoso , Animais , Estudos de Casos e Controles , Células Cultivadas , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Obesidade/complicações , Obesidade/metabolismo , Paniculite/complicações , Paniculite/genética , Paniculite/metabolismo
13.
Clin Sci (Lond) ; 125(8): 383-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23642227

RESUMO

HF (heart failure) and T2D (Type 2 diabetes) associate with detrimental alterations in SkM (skeletal muscle) structure/function. We have demonstrated recently that (-)-ERC (epicatechin-rich cocoa) improves SkM mitochondrial structure [Taub, Ramirez-Sanchez, Ciaraldi, Perkins, Murphy, Naviaux, Hogan, Ceballos, Maisel, Henry et al. (2012) Clin. Trans. Sci. 5, 43-47]. We hypothesized that an improved mitochondrial structure may facilitate the reversal of detrimental alterations in sarcomeric microstructure. In a pilot study, five patients with HF and T2D consumed ERC for 3 months; treadmill testing [VO2max (maximum oxygen consumption)] and SkM biopsies were performed. Western blot analysis, immunohistochemistry and electron microscopy were used. We report severe perturbations in components of the DAPC (dystrophin-associated protein complex) as well as sarcomeric microstructure at baseline. ERC induced recovery/enhancement of DAPC protein levels, sarcomeric microstructure and, in a co-ordinated fashion, alterations in markers of SkM growth/differentiation consistent with myofibre regeneration. VO2max increased (~24%) but did not reach statistical significance. These initial results warrant further rigorous investigation, since the use of ERC (or pure epicatechin) may represent a safe and novel means of improving muscle function.


Assuntos
Cacau/química , Catequina/farmacologia , Diabetes Mellitus Tipo 2/fisiopatologia , Insuficiência Cardíaca/fisiopatologia , Músculo Esquelético/efeitos dos fármacos , Sarcômeros/efeitos dos fármacos , Idoso , Western Blotting , Catequina/administração & dosagem , Catequina/uso terapêutico , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/prevenção & controle , Disferlina , Distrofina/metabolismo , Complexo de Proteínas Associadas Distrofina/metabolismo , Teste de Esforço , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/prevenção & controle , Humanos , Imuno-Histoquímica , Masculino , Proteínas de Membrana/metabolismo , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Projetos Piloto , Sarcoglicanas/metabolismo , Sarcômeros/metabolismo , Sarcômeros/ultraestrutura , Resultado do Tratamento , Utrofina/metabolismo
14.
Adipocyte ; 11(1): 69-83, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35094654

RESUMO

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that interacts via 5 G-protein coupled receptors, S1PR1-5, to regulate signalling pathways critical to biological processes including cell growth, immune cell trafficking, and inflammation.We demonstrate that in Type 2 diabetic (T2D) subjects, plasma S1P levels significantly increased in response to the anti-diabetic drug, rosiglitazone, and, S1P levels correlated positively with measures of improved glucose homeostasis. In HFD-induced obese C57BL/6 J mice S1PR3 gene expression was increased in adipose tissues (AT) and liver compared with low fat diet (LFD)-fed counterparts. On a HFD, weight gain was similar in both S1PR3-/- mice and WT littermates; however, HFD-fed S1PR3-/- mice exhibited a phenotype of partial lipodystrophy, exacerbated insulin resistance and glucose intolerance. This worsened metabolic phenotype of HFD-fed S1PR3-/- mice was mechanistically linked with increased adipose inflammation, adipose macrophage and T-cell accumulation, hepatic inflammation and hepatic steatosis. In 3T3-L1 preadipocytes S1P increased adipogenesis and S1P-S1PR3 signalling regulated the expression of PPARγ, suggesting a novel role for this signalling pathway in the adipogenic program. These results reveal an anti-diabetic role for S1P, and, that S1P-S1PR3 signalling in the adipose and liver defends against excessive inflammation and steatosis to maintain metabolic homeostasis at key regulatory pathways.


Assuntos
Fenômenos Biológicos , Fígado Gorduroso , Animais , Dieta Hiperlipídica/efeitos adversos , Humanos , Inflamação/metabolismo , Lisofosfolipídeos , Camundongos , Camundongos Endogâmicos C57BL , Obesidade , Esfingosina/análogos & derivados , Receptores de Esfingosina-1-Fosfato
15.
JCI Insight ; 7(1)2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-34813504

RESUMO

While current thinking posits that insulin signaling to glucose transporter 4 (GLUT4) exocytic translocation and glucose uptake in skeletal muscle and adipocytes is controlled by phosphorylation-based signaling, many proteins in this pathway are acetylated on lysine residues. However, the importance of acetylation and lysine acetyltransferases to insulin-stimulated glucose uptake is incompletely defined. Here, we demonstrate that combined loss of the acetyltransferases E1A binding protein p300 (p300) and cAMP response element binding protein binding protein (CBP) in mouse skeletal muscle caused a complete loss of insulin-stimulated glucose uptake. Similarly, brief (i.e., 1 hour) pharmacological inhibition of p300/CBP acetyltransferase activity recapitulated this phenotype in human and rodent myotubes, 3T3-L1 adipocytes, and mouse muscle. Mechanistically, these effects were due to p300/CBP-mediated regulation of GLUT4 exocytic translocation and occurred downstream of Akt signaling. Taken together, we highlight a fundamental role for acetylation and p300/CBP in the direct regulation of insulin-stimulated glucose transport in skeletal muscle and adipocytes.


Assuntos
Adipócitos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteína p300 Associada a E1A/metabolismo , Glucose/metabolismo , Músculo Esquelético , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Feminino , Insulina/metabolismo , Masculino , Camundongos , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo
16.
Am J Physiol Endocrinol Metab ; 300(3): E536-42, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21189360

RESUMO

To determine whether serine/threonine ROCK1 is activated by insulin in vivo in humans and whether impaired activation of ROCK1 could play a role in the pathogenesis of insulin resistance, we measured the activity of ROCK1 and the protein content of the Rho family in vastus lateralis muscle of lean, obese nondiabetic, and obese type 2 diabetic subjects. Biopsies were taken after an overnight fast and after a 3-h hyperinsulinemic euglycemic clamp. Insulin-stimulated GDR was reduced 38% in obese nondiabetic subjects compared with lean, 62% in obese diabetic subjects compared with lean, and 39% in obese diabetic compared with obese nondiabetic subjects (all comparisons P < 0.001). Insulin-stimulated IRS-1 tyrosine phosphorylation is impaired 41-48% in diabetic subjects compared with lean or obese subjects. Basal activity of ROCK1 was similar in all groups. Insulin increased ROCK1 activity 2.1-fold in lean and 1.7-fold in obese nondiabetic subjects in muscle. However, ROCK1 activity did not increase in response to insulin in muscle of obese type 2 diabetic subjects without change in ROCK1 protein levels. Importantly, insulin-stimulated ROCK1 activity was positively correlated with insulin-mediated GDR in lean subjects (P < 0.01) but not in obese or type 2 diabetic subjects. Moreover, RhoE GTPase that inhibits the catalytic activity of ROCK1 by binding to the kinase domain of the enzyme is notably increased in obese type 2 diabetic subjects, accounting for defective ROCK1 activity. Thus, these data suggest that ROCK1 may play an important role in the pathogenesis of resistance to insulin action on glucose disposal in muscle of obese type 2 diabetic subjects.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Músculo Esquelético/metabolismo , Quinases Associadas a rho/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Adulto , Biotransformação/efeitos dos fármacos , Western Blotting , Índice de Massa Corporal , Proteínas do Citoesqueleto/metabolismo , Feminino , Glucose/metabolismo , Técnica Clamp de Glucose , Humanos , Proteínas Substratos do Receptor de Insulina/biossíntese , Proteínas Substratos do Receptor de Insulina/genética , Resistência à Insulina , Cinética , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/efeitos dos fármacos , Obesidade/metabolismo , Fosforilação , Tirosina/metabolismo , Proteínas rho de Ligação ao GTP/biossíntese , Proteínas rho de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
18.
Nat Commun ; 11(1): 2024, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-32332780

RESUMO

Crosstalk between liver and skeletal muscle is vital for glucose homeostasis. Hepatokines, liver-derived proteins that play an important role in regulating muscle metabolism, are important to this communication. Here we identify apolipoprotein J (ApoJ) as a novel hepatokine targeting muscle glucose metabolism and insulin sensitivity through a low-density lipoprotein receptor-related protein-2 (LRP2)-dependent mechanism, coupled with the insulin receptor (IR) signaling cascade. In muscle, LRP2 is necessary for insulin-dependent IR internalization, an initial trigger for insulin signaling, that is crucial in regulating downstream signaling and glucose uptake. Of physiologic significance, deletion of hepatic ApoJ or muscle LRP2 causes insulin resistance and glucose intolerance. In patients with polycystic ovary syndrome and insulin resistance, pioglitazone-induced improvement of insulin action is associated with an increase in muscle ApoJ and LRP2 expression. Thus, the ApoJ-LRP2 axis is a novel endocrine circuit that is central to the maintenance of normal glucose homeostasis and insulin sensitivity.


Assuntos
Clusterina/metabolismo , Glucose/metabolismo , Resistência à Insulina , Músculo Esquelético/metabolismo , Síndrome do Ovário Policístico/metabolismo , Adulto , Animais , Linhagem Celular , Clusterina/sangue , Clusterina/genética , Modelos Animais de Doenças , Feminino , Técnica Clamp de Glucose , Humanos , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Insulina/metabolismo , Fígado/metabolismo , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Masculino , Camundongos , Camundongos Knockout , Pioglitazona/farmacologia , Pioglitazona/uso terapêutico , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/tratamento farmacológico , Receptor de Insulina/metabolismo , Transdução de Sinais/efeitos dos fármacos
19.
J Clin Endocrinol Metab ; 94(1): 157-63, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18854391

RESUMO

OBJECTIVE: The potential differential contributions of skeletal muscle and adipose tissue to whole body insulin resistance were evaluated in subjects with polycystic ovary syndrome (PCOS). RESEARCH DESIGN AND METHODS: Forty-two PCOS subjects and 15 body mass index-matched control subjects were studied. Insulin action was evaluated by the hyperinsulinemic/euglycemic clamp procedure. Isolated adipocytes and cultured muscle cells were analyzed for glucose transport activity; adipocytes, muscle tissue, and myotubes were analyzed for the expression and phosphorylation of insulin-signaling proteins. RESULTS: Fifty-seven per cent of the PCOS subjects had impaired glucose tolerance and the lowest rate of maximal insulin-stimulated whole body glucose disposal compared to controls (P < 0.01). PCOS subjects with normal glucose tolerance had intermediate reduction in glucose disposal rate (P < 0.05 vs. both control and impaired glucose tolerance subjects). However, rates of maximal insulin-stimulated glucose transport (insulin responsiveness) into isolated adipocytes were comparable between all three groups, whereas PCOS subjects displayed impaired insulin sensitivity. In contrast, myotubes from PCOS subjects displayed reduced insulin responsiveness for glucose uptake and normal sensitivity. There were no differences between groups in the expression of glucose transporter 4 or insulin-signaling proteins or maximal insulin stimulation of phosphorylation of Akt in skeletal muscle, myotubes, or adipocytes. CONCLUSIONS: Individuals with PCOS display impaired insulin responsiveness in skeletal muscle and myotubes, whereas isolated adipocytes display impaired insulin sensitivity but normal responsiveness. Skeletal muscle and adipose tissue contribute differently to insulin resistance in PCOS. Insulin resistance in PCOS cannot be accounted for by differences in the expression of selected signaling molecules or maximal phosphorylation of Akt.


Assuntos
Resistência à Insulina , Síndrome do Ovário Policístico/metabolismo , Adipócitos/metabolismo , Adulto , Feminino , Glucose/metabolismo , Transportador de Glucose Tipo 4/análise , Humanos , Insulina/farmacologia , Músculo Esquelético/metabolismo , Especificidade de Órgãos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo
20.
J Clin Endocrinol Metab ; 94(2): 469-76, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18984667

RESUMO

CONTEXT: Polycystic ovary syndrome (PCOS) is characterized by insulin resistance, compensatory hyperinsulinemia, increased prevalence of impaired glucose tolerance, and increased ovarian androgen biosynthesis. OBJECTIVE: The aim of the study was to evaluate effects of pioglitazone on whole body insulin action and ovarian androgen biosynthesis in PCOS. DESIGN: We performed a randomized placebo-controlled trial. SETTING: The study was conducted at the Special Diagnostic and Treatment Unit of the Veterans Affairs Medical Center, San Diego, and the University of California, San Diego, General Clinical Research Center. PATIENTS OR OTHER PARTICIPANTS: A total of 23 subjects with PCOS were evaluated at baseline and end of treatment. Six age- and body mass index-matched women without PCOS were normal controls for baseline evaluation. INTERVENTION: Subjects with PCOS were randomized to oral placebo or pioglitazone 45 mg daily for 6 months. MAIN OUTCOME MEASURE(S): The primary outcome measures were whole body insulin action as measured by hyperinsulinemic euglycemic clamp and ovarian androgen biosynthesis as measured by leuprolide-stimulated production of 17-hydroxyprogesterone (17-OHP). RESULTS: Compared with placebo, pioglitazone treatment significantly improved multiple measures of insulin action, including glucose disposal rate (P < 0.01), 2-h glucose during 75-g oral glucose tolerance test (P < 0.01), area under the curve glucose during oral glucose tolerance test (P < 0.01), serum adiponectin (P < 0.01), and fasting hyperinsulinemia (P < 0.01). Compared to placebo, pioglitazone treatment reduced the increment of leuprolide-stimulated 17-OHP (P < 0.02). Improvements in glucose disposal rate correlated with reductions in 17-OHP stimulation (P < 0.02). CONCLUSIONS: Compared to placebo, pioglitazone treatment in PCOS was associated with improvements in insulin action and glucose homeostasis and ameliorated the hyperandrogenic ovarian response.


Assuntos
Hormônios Esteroides Gonadais/metabolismo , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/metabolismo , Tiazolidinedionas/farmacologia , Tiazolidinedionas/uso terapêutico , Adulto , Glicemia/metabolismo , Feminino , Intolerância à Glucose/metabolismo , Teste de Tolerância a Glucose , Hormônios Esteroides Gonadais/sangue , Humanos , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Insulina/sangue , Insulina/metabolismo , Pioglitazona , Placebos , Síndrome do Ovário Policístico/sangue
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa