Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
BMC Biol ; 18(1): 149, 2020 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-33092598

RESUMO

BACKGROUND: The classical functions of the skeleton encompass locomotion, protection and mineral homeostasis. However, cell-specific gene deletions in the mouse and human genetic studies have identified the skeleton as a key endocrine regulator of metabolism. The bone-specific phosphatase, Phosphatase, Orphan 1 (PHOSPHO1), which is indispensable for bone mineralisation, has been recently implicated in the regulation of energy metabolism in humans, but its role in systemic metabolism remains unclear. Here, we probe the mechanism underlying metabolic regulation by analysing Phospho1 mutant mice. RESULTS: Phospho1-/- mice exhibited improved basal glucose homeostasis and resisted high-fat-diet-induced weight gain and diabetes. The metabolic protection in Phospho1-/- mice was manifested in the absence of altered levels of osteocalcin. Osteoblasts isolated from Phospho1-/- mice were enriched for genes associated with energy metabolism and diabetes; Phospho1 both directly and indirectly interacted with genes associated with glucose transport and insulin receptor signalling. Canonical thermogenesis via brown adipose tissue did not underlie the metabolic protection observed in adult Phospho1-/- mice. However, the decreased serum choline levels in Phospho1-/- mice were normalised by feeding a 2% choline rich diet resulting in a normalisation in insulin sensitivity and fat mass. CONCLUSION: We show that mice lacking the bone mineralisation enzyme PHOSPHO1 exhibit improved basal glucose homeostasis and resist high-fat-diet-induced weight gain and diabetes. This study identifies PHOSPHO1 as a potential bone-derived therapeutic target for the treatment of obesity and diabetes.


Assuntos
Metabolismo Energético , Resistência à Insulina/genética , Obesidade/genética , Monoéster Fosfórico Hidrolases/genética , Animais , Colina/metabolismo , Glucose/metabolismo , Homeostase , Masculino , Camundongos , Monoéster Fosfórico Hidrolases/metabolismo
2.
J Biol Chem ; 287(6): 4188-97, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22158867

RESUMO

In obesity, rapidly expanding adipose tissue becomes hypoxic, precipitating inflammation, fibrosis, and insulin resistance. Compensatory angiogenesis may prevent these events. Mice lacking the intracellular glucocorticoid-amplifying enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ßHSD1(-/-)) have "healthier" adipose tissue distribution and resist metabolic disease with diet-induced obesity. Here we show that adipose tissues of 11ßHSD1(-/-) mice exhibit attenuated hypoxia, induction of hypoxia-inducible factor (HIF-1α) activation of the TGF-ß/Smad3/α-smooth muscle actin (α-SMA) signaling pathway, and fibrogenesis despite similar fat accretion with diet-induced obesity. Moreover, augmented 11ßHSD1(-/-) adipose tissue angiogenesis is associated with enhanced peroxisome proliferator-activated receptor γ (PPARγ)-inducible expression of the potent angiogenic factors VEGF-A, apelin, and angiopoietin-like protein 4. Improved adipose angiogenesis and reduced fibrosis provide a novel mechanism whereby suppression of intracellular glucocorticoid regeneration promotes safer fat expansion with weight gain.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Tecido Adiposo/enzimologia , Hipóxia/enzimologia , Neovascularização Fisiológica , Obesidade/enzimologia , Transdução de Sinais , Actinas/genética , Actinas/metabolismo , Adipocinas , Tecido Adiposo/irrigação sanguínea , Tecido Adiposo/patologia , Proteína 4 Semelhante a Angiopoietina , Angiopoietinas/genética , Angiopoietinas/metabolismo , Animais , Apelina , Fibrose/enzimologia , Fibrose/genética , Fibrose/fisiopatologia , Hipóxia/patologia , Hipóxia/fisiopatologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Resistência à Insulina/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos , Camundongos Knockout , Obesidade/patologia , Obesidade/fisiopatologia , PPAR gama/genética , PPAR gama/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Aumento de Peso/genética
3.
J Biol Chem ; 287(24): 20088-99, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22511782

RESUMO

The importance of AMP-activated protein kinase (AMPK) and protein kinase C (PKC) as effectors of metformin (Met) action on glucose uptake (GU) in skeletal muscle cells was investigated. GU in L6 myotubes was stimulated 2-fold following 16 h of Met treatment and acutely enhanced by insulin in an additive fashion. Insulin-stimulated GU was sensitive to PI3K inhibition, whereas that induced by Met was not. Met and its related biguanide, phenformin, stimulated AMPK activation/phosphorylation to a level comparable with that induced by the AMPK activator, 5-amino-1-ß-d-ribofuranosyl-imidazole-4-carboxamide (AICAR). However, the increase in GU elicited by AICAR was significantly lower than that induced by either biguanide. Expression of a constitutively active AMPK mimicked the effects of AICAR on GU, whereas a dominant interfering AMPK or shRNA silencing of AMPK prevented AICAR-stimulated GU and Met-induced AMPK signaling but only repressed biguanide-stimulated GU by ∼20%. Consistent with this, analysis of GU in muscle cells from α1(-/-)/α2(-/-) AMPK-deficient mice revealed a significant retention of Met-stimulated GU, being reduced by ∼35% compared with that of wild type cells. Atypical PKCs (aPKCs) have been implicated in Met-stimulated GU, and in line with this, Met and phenformin induced activation/phosphorylation of aPKC in L6 myotubes. However, although cellular depletion of aPKC (>90%) led to loss in biguanide-induced aPKC phosphorylation, it had no effect on Met-stimulated GU, whereas inhibitors targeting novel/conventional PKCs caused a significant reduction in biguanide-induced GU. Our findings indicate that although Met activates AMPK, a significant component of Met-stimulated GU in muscle cells is mediated via an AMPK-independent mechanism that involves novel/conventional PKCs.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Glucose/metabolismo , Hipoglicemiantes/farmacologia , Metformina/farmacologia , Fibras Musculares Esqueléticas/enzimologia , Proteína Quinase C/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/genética , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativadores de Enzimas/farmacologia , Glucose/genética , Camundongos , Camundongos Knockout , Fibras Musculares Esqueléticas/citologia , Fenformin/farmacologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteína Quinase C/genética , Ribonucleotídeos/farmacologia , Fatores de Tempo
4.
PLoS One ; 16(2): e0245179, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33566837

RESUMO

In type 2 diabetes (T2D), both muscle and liver are severely resistant to insulin action. Muscle insulin resistance accounts for more than 80% of the impairment in total body glucose disposal in T2D patients and is often characterized by an impaired insulin signaling. Mitsugumin 53 (MG53), a muscle-specific TRIM family protein initially identified as a key regulator of cell membrane repair machinery has been suggested to be a critical regulator of muscle insulin signaling pathway by acting as ubiquitin E3 ligase targeting both the insulin receptor and insulin receptor substrate 1 (IRS1). Here, we show using in vitro and in vivo approaches that MG53 is not a critical regulator of insulin signaling and glucose homeostasis. First, MG53 expression is not consistently regulated in skeletal muscle from various preclinical models of insulin resistance. Second, MG53 gene knock-down in muscle cells does not lead to impaired insulin response as measured by Akt phosphorylation on Serine 473 and glucose uptake. Third, recombinant human MG53 does not alter insulin response in both differentiated C2C12 and human skeletal muscle cells. Fourth, ectopic expression of MG53 in HEK293 cells lacking endogenous MG53 expression fails to alter insulin response as measured by Akt phosphorylation. Finally, both male and female mg53 -/- mice were not resistant to high fat induced obesity and glucose intolerance compared to wild-type mice. Taken together, these results strongly suggest that MG53 is not a critical regulator of insulin signaling pathway in skeletal muscle.


Assuntos
Insulina/metabolismo , Músculo Esquelético/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Animais , Antígenos CD/metabolismo , Proteínas de Transporte/metabolismo , Feminino , Células HEK293 , Humanos , Proteínas Substratos do Receptor de Insulina/metabolismo , Resistência à Insulina/fisiologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fibras Musculares Esqueléticas/metabolismo , Receptor de Insulina/metabolismo , Transdução de Sinais , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
5.
Biochem J ; 410(2): 369-79, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17983354

RESUMO

Elevated ceramide concentrations in adipocytes and skeletal muscle impair PKB (protein kinase B; also known as Akt)-directed insulin signalling to key hormonal end points. An important feature of this inhibition involves the ceramide-induced activation of atypical PKCzeta (protein kinase C-zeta), which associates with and negatively regulates PKB. In the present study, we demonstrate that this inhibition is critically dependent on the targeting and subsequent retention of PKCzeta-PKB within CEM (caveolin-enriched microdomains), which is facilitated by kinase interactions with caveolin. Ceramide also recruits PTEN (phosphatase and tensin homologue detected on chromosome 10), a 3'-phosphoinositide phosphatase, thereby creating a repressive membrane microenvironment from which PKB cannot signal. Disrupting the structural integrity of caveolae by cholesterol depletion prevented caveolar targeting of PKCzeta and PKB and suppressed kinase-caveolin association, but, importantly, also ameliorated ceramide-induced inhibition of PKB. Consistent with this, adipocytes from caveolin-1-/- mice, which lack functional caveolae, exhibit greater resistance to ceramide compared with caveolin-1+/+ adipocytes. We conclude that the recruitment and retention of PKB within CEM contribute significantly to ceramide-induced inhibition of PKB-directed signalling.


Assuntos
Ceramidas/farmacologia , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células 3T3 , Adipócitos/enzimologia , Animais , Caveolina 1/deficiência , Colesterol/metabolismo , Ativação Enzimática , Humanos , Insulina/fisiologia , Camundongos , Camundongos Knockout , Músculo Esquelético/enzimologia , Transdução de Sinais
6.
Diabetes ; 54(11): 3161-8, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16249440

RESUMO

It has been proposed that p38 mitogen-activated protein kinase (MAPK) isoforms sensitive to the pyridinylimidazole compounds SB 203580 and SB 202190 may participate in the acute insulin-dependent activation of glucose transporters recruited to the plasma membrane of adipocytes and skeletal muscle. Here, we explore whether these kinases support the insulin stimulation of glucose uptake in these tissues by investigating the effects of a genetic loss in p38beta and that of the p38 MAPK inhibitor SB 203580. Glucose uptake in adipocytes and soleus muscle was stimulated by insulin by up to fourfold irrespective of whether tissues were isolated from wild-type or p38beta-null mice. Consistent with this finding, mice lacking p38beta exhibited normal glucose tolerance, insulinemia, and glycemia compared with their wild-type counterparts. Insulin-stimulated glucose uptake was not inhibited by SB 203580 when adipocytes were preincubated with the drug at a cytocrit of 50%, but intriguingly, uptake was suppressed (by 35%) when the cytocrit was reduced by one-half. Despite the activation of glucose uptake at the higher cytocrit, insulin failed to induce any detectable activation of p38 MAPK, whereas p38 signaling was robustly activated by anisomycin in a SB 203580-sensitive manner. Although insulin also failed to induce any detectable activation of p38 MAPK in muscle, insulin-dependent glucose uptake was reduced by SB 203580 (approximately 44%) in muscle of both wild-type and p38beta-null mice. Our results indicate that p38beta is not required for insulin-stimulated glucose uptake in adipocytes or muscle. Moreover, given that insulin fails to promote any significant activation of p38 MAPK in these tissues and the finding that sensitivity of glucose uptake, but not that of the kinase, to SB 203580 can be influenced by cytocrit, we suggest that p38 signaling is unlikely to participate in any putative activation of transporters recruited to the cell surface by insulin and that SB 203580 suppresses insulin-stimulated glucose transport by a mechanism unrelated to its inhibitory effect on p38 MAPK.


Assuntos
Tecido Adiposo/enzimologia , Glucose/metabolismo , Insulina/farmacologia , Músculo Esquelético/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Anisomicina/farmacologia , Transporte Biológico/efeitos dos fármacos , Glicemia/metabolismo , Peso Corporal , Deleção de Genes , Imidazóis/farmacologia , Insulina/sangue , Insulina/metabolismo , Camundongos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Piridinas/farmacologia , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/genética
7.
Diabetes ; 52(2): 277-82, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12540597

RESUMO

It has been established that leptin exerts a negative control on food intake, allowing one to maintain stable caloric intake over time. The aim of the present study was to investigate whether leptin regulates food intake when a supply of calories is provided by the systemic route. Experiments were carried out in leptin receptor-deficient obese fa/fa rats and lean Fa/fa controls. In both groups, 48 h of glucose infusion reduced food intake in proportion to caloric supply, resulting in virtually no change in total caloric intake as compared to before the infusion. This hypophagic response was reproduced without adding systemic calories, but by increasing glucose and insulin concentrations specifically in the brain through carotid artery infusion. Concomitant intracerebroventricular administration of 5-(tetradecyloxy)-2-furoic acid, an acetyl CoA carboxylase inhibitor that precludes malonyl-CoA synthesis, abolished the restriction of feeding in carotid-infused lean and obese rats. These data indicate that a supply of calories via glucose infusion induces a hypophagic response independent of leptin signaling in the rat, and support the hypothesis that a rise in central malonyl-CoA, triggered by increased glucose and insulin concentrations, participates in this adaptation. This process could contribute to the limiting of hyperphagia, primarily when leptin signaling is altered, as in the obese state.


Assuntos
Glicemia/metabolismo , Dieta Redutora , Carboidratos da Dieta , Insulina/sangue , Leptina/sangue , Obesidade/fisiopatologia , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/fisiologia , Animais , Ventrículos Cerebrais/efeitos dos fármacos , Ventrículos Cerebrais/fisiologia , Ingestão de Energia , Furanos/administração & dosagem , Furanos/farmacologia , Técnica Clamp de Glucose , Injeções Intraventriculares , Obesidade/genética , Ratos , Ratos Zucker , Receptores de Superfície Celular/genética , Receptores para Leptina , Magreza
8.
Biochem J ; 382(Pt 2): 619-29, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15193147

RESUMO

Non-esterified fatty acids (NEFAs) have been implicated in the pathogenesis of skeletal muscle insulin resistance that may develop, in part, as a consequence of a direct inhibitory effect on early insulin signalling events. Here we report work investigating the mechanism by which palmitate (a saturated free fatty acid) inhibits insulin action in rat L6 myotubes. Palmitate suppressed the insulin-induced plasma membrane recruitment and phosphorylation of protein kinase B (PKB) and this was associated with a loss in insulin-stimulated glucose transport. The inhibition in PKB was not due to a loss in insulin receptor substrate (IRS)1 tyrosine phosphorylation, IRS-1/p85 (phosphoinositide 3-kinase) association or suppression in phosphatidyl 3,4,5 triphosphate synthesis, but was attributable to an elevated intracellular synthesis of ceramide (6-fold) from palmitate and a concomitant activation of protein kinase PKCzeta (5-fold). Inhibitors of serine palmitoyl transferase suppressed the intracellular synthesis of ceramide from palmitate, prevented PKCzeta activation, and antagonized the inhibition in PKB recruitment/phosphorylation and the loss in insulin-stimulated glucose transport elicited by the NEFA. Inhibiting the palmitate-induced activation of PKCzeta with Ro 31.8220, also prevented the loss in the insulin-dependent phosphorylation of PKB caused by palmitate. These findings indicate that intracellular ceramide synthesis and PKCzeta activation are important aspects of the mechanism by which palmitate desensitizes L6 muscle cells to insulin.


Assuntos
Ceramidas/biossíntese , Resistência à Insulina/fisiologia , Espaço Intracelular/enzimologia , Espaço Intracelular/metabolismo , Fibras Musculares Esqueléticas/enzimologia , Fibras Musculares Esqueléticas/metabolismo , Palmitatos/metabolismo , Proteína Quinase C/fisiologia , Células 3T3-L1 , Adipócitos , Animais , Linhagem Celular , Ativação Enzimática/fisiologia , Camundongos , Músculo Esquelético/enzimologia , Músculo Esquelético/metabolismo , Ratos
9.
FEBS Lett ; 549(1-3): 77-82, 2003 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-12914929

RESUMO

Skeletal muscle, a primary insulin target tissue, expresses the GLUT5 fructose transporter. Although insulin has no acute effect on GLUT5 expression and function in muscle, we show here that long-term (24 h) insulin treatment of L6 muscle cells induces a dose-dependent increase in GLUT5 protein (by up to two-fold), leading to a concomitant increase in fructose uptake. The increase in GLUT5 expression and function was suppressed by inhibitors of gene transcription and protein synthesis, suggesting that insulin promotes de novo carrier synthesis. Transfection of the GLUT5 gene promoter fused to luciferase into L6 cells revealed that insulin induced a 1.8-fold increase in GLUT5 promoter activity. Our findings indicate that insulin is capable of increasing the abundance and functional activity of GLUT5 in skeletal muscle cells and that this is most likely mediated via activation of the GLUT5 promoter.


Assuntos
Insulina/farmacologia , Proteínas de Transporte de Monossacarídeos/biossíntese , Proteínas de Transporte de Monossacarídeos/efeitos dos fármacos , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Frutose/farmacocinética , Transportador de Glucose Tipo 5 , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Ratos , Transcrição Gênica/efeitos dos fármacos
10.
C R Biol ; 325(2): 77-87, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11980179

RESUMO

Leptin acts on the hypothalamus to reduce food intake and on a number of non-neuronal tissues via specific receptors (Lepr). The use of in situ hybridisation to map the Lepr gene in pre-natal mice revealed transcripts in the yolk sac in various structures of the central nervous system and in mesoderm-derived tissues, such as cartilage/bone primordia and musculoaponeurotic laminae. At later stages, significant amounts of Lepr were expressed in the region surrounding the developing eye of the embryo. Lepr was also found to be expressed in the choroid, sclera and connective tissues of the limbus in the adult eye. In conclusion, we have identified new targets for leptin action during embryogenesis and adulthood.


Assuntos
Proteínas de Transporte/biossíntese , Proteínas Fetais/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Leptina/fisiologia , Mesoderma/metabolismo , Receptores de Superfície Celular , Animais , Proteínas de Transporte/genética , Cartilagem/embriologia , Cartilagem/metabolismo , Tecido Conjuntivo/embriologia , Tecido Conjuntivo/metabolismo , Olho/crescimento & desenvolvimento , Olho/metabolismo , Proteínas do Olho/biossíntese , Proteínas do Olho/genética , Proteínas Fetais/genética , Idade Gestacional , Sistema Hematopoético/embriologia , Sistema Hematopoético/metabolismo , Hibridização In Situ , Camundongos , Proteínas Musculares/biossíntese , Proteínas Musculares/genética , Músculo Liso/embriologia , Músculo Liso/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Especificidade de Órgãos , Receptores para Leptina
11.
Artigo em Inglês | MEDLINE | ID: mdl-25352830

RESUMO

Progression and severity of type 1 diabetes is dependent upon inflammatory induction of nitric oxide production and consequent pancreatic ß-cell damage. Glucocorticoids (GCs) are highly effective anti-inflammatory agents but have been precluded in type 1 diabetes and in islet transplantation protocols because they exacerbated insulin resistance and suppressed ß-cell insulin secretion at the high-doses employed clinically. In contrast, physiological-range elevation of GC action within ß-cells ameliorated lipotoxic ß-cell failure in transgenic mice overexpressing the intracellular enzyme 11ß-hydroxysteroid dehydrogenase type 1 (MIP-HSD1(tg/+) mice). Here, we tested the hypothesis that elevated ß-cell 11beta-HSD1 protects against the ß-cell destruction elicited by streptozotocin (STZ), a toxin that dose-dependently mimics aspects of inflammatory and autoimmune ß-cell destruction. MIP-HSD1(tg/+) mice exhibited an episodic protection from the severe hyperglycemia caused by a single high dose of STZ associated with higher and sustained ß-cell survival, maintained ß-cell replicative potential, higher plasma and islet insulin levels, reduced inflammatory macrophage infiltration and increased anti-inflammatory T regulatory cell content. MIP-HSD1(tg/+) mice also completely resisted mild hyperglycemia and insulitis induced by multiple low-dose STZ administration. In vitro, MIP-HSD1(tg/+) islets exhibited attenuated STZ-induced nitric oxide production, an effect reversed with a specific 11beta-HSD1 inhibitor. GC regeneration selectively within ß-cells protects against inflammatory ß-cell destruction, suggesting therapeutic targeting of 11beta-HSD1 may ameliorate processes that exacerbate type 1 diabetes and that hinder islet transplantation.

12.
PLoS One ; 9(4): e94680, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24732937

RESUMO

Maternal obesity is linked with increased adverse pregnancy outcomes for both mother and child. The metabolic impact of excessive fat within the context of pregnancy is not fully understood. We used a mouse model of high fat (HF) feeding to induce maternal obesity to identify adipose tissue-mediated mechanisms driving metabolic dysfunction in pregnant and non-pregnant obese mice. As expected, chronic HF-feeding for 12 weeks preceding pregnancy increased peripheral (subcutaneous) and visceral (mesenteric) fat mass. However, unexpectedly at late gestation (E18.5) HF-fed mice exhibited a remarkable normalization of visceral but not peripheral adiposity, with a 53% reduction in non-pregnant visceral fat mass expressed as a proportion of body weight (P<0.001). In contrast, in control animals, pregnancy had no effect on visceral fat mass proportion. Obesity exaggerated glucose intolerance at mid-pregnancy (E14.5). However by E18.5, there were no differences, in glucose tolerance between obese and control mice. Transcriptomic analysis of visceral fat from HF-fed dams at E18.5 revealed reduced expression of genes involved in de novo lipogenesis (diacylglycerol O-acyltransferase 2--Dgat2) and inflammation (chemokine C-C motif ligand 20--Ccl2) and upregulation of estrogen receptor α (ERα) compared to HF non pregnant. Attenuation of adipose inflammation was functionally confirmed by a 45% reduction of CD11b+CD11c+ adipose tissue macrophages (expressed as a proportion of all stromal vascular fraction cells) in HF pregnant compared to HF non pregnant animals (P<0.001). An ERα selective agonist suppressed both de novo lipogenesis and expression of lipogenic genes in adipocytes in vitro. These data show that, in a HF model of maternal obesity, late gestation is associated with amelioration of visceral fat hypertrophy, inflammation and glucose intolerance, and suggest that these effects are mediated in part by elevated visceral adipocyte ERα signaling.


Assuntos
Adipócitos/citologia , Adiposidade , Estrogênios/metabolismo , Gordura Intra-Abdominal/metabolismo , Camundongos Obesos , Adipócitos/metabolismo , Animais , Feminino , Glucose/metabolismo , Teste de Tolerância a Glucose , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Prenhez , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Fatores de Tempo , Transcriptoma
13.
PLoS One ; 9(7): e101865, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25058613

RESUMO

Ceramides are known to promote insulin resistance in a number of metabolically important tissues including skeletal muscle, the predominant site of insulin-stimulated glucose disposal. Depending on cell type, these lipid intermediates have been shown to inhibit protein kinase B (PKB/Akt), a key mediator of the metabolic actions of insulin, via two distinct pathways: one involving the action of atypical protein kinase C (aPKC) isoforms, and the second dependent on protein phosphatase-2A (PP2A). The main aim of this study was to explore the mechanisms by which ceramide inhibits PKB/Akt in three different skeletal muscle-derived cell culture models; rat L6 myotubes, mouse C2C12 myotubes and primary human skeletal muscle cells. Our findings indicate that the mechanism by which ceramide acts to repress PKB/Akt is related to the myocellular abundance of caveolin-enriched domains (CEM) present at the plasma membrane. Here, we show that ceramide-enriched-CEMs are markedly more abundant in L6 myotubes compared to C2C12 myotubes, consistent with their previously reported role in coordinating aPKC-directed repression of PKB/Akt in L6 muscle cells. In contrast, a PP2A-dependent pathway predominantly mediates ceramide-induced inhibition of PKB/Akt in C2C12 myotubes. In addition, we demonstrate for the first time that ceramide engages an aPKC-dependent pathway to suppress insulin-induced PKB/Akt activation in palmitate-treated cultured human muscle cells as well as in muscle cells from diabetic patients. Collectively, this work identifies key mechanistic differences, which may be linked to variations in plasma membrane composition, underlying the insulin-desensitising effects of ceramide in different skeletal muscle cell models that are extensively used in signal transduction and metabolic studies.


Assuntos
Ceramidas/farmacologia , Insulina/farmacologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Caveolinas/genética , Caveolinas/metabolismo , Regulação da Expressão Gênica , Humanos , Insulina/metabolismo , Camundongos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Cultura Primária de Células , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos
14.
Diabetes ; 61(3): 642-52, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22315313

RESUMO

Type 2 diabetes ultimately results from pancreatic ß-cell failure. Abnormally elevated intracellular regeneration of glucocorticoids by the enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) in fat or liver may underlie pathophysiological aspects of the metabolic syndrome. Elevated 11ß-HSD1 is also found in pancreatic islets of obese/diabetic rodents and is hypothesized to suppress insulin secretion and promote diabetes. To define the direct impact of elevated pancreatic ß-cell 11ß-HSD1 on insulin secretion, we generated ß-cell-specific, 11ß-HSD1-overexpressing (MIP-HSD1) mice on a strain background prone to ß-cell failure. Unexpectedly, MIP-HSD1(tg/+) mice exhibited a reversal of high fat-induced ß-cell failure through augmentation of the number and intrinsic function of small islets in association with induction of heat shock, protein kinase A, and extracellular signal-related kinase and p21 signaling pathways. 11ß-HSD1(-/-) mice showed mild ß-cell impairment that was offset by improved glucose tolerance. The benefit of higher ß-cell 11ß-HSD1 exhibited a threshold because homozygous MIP-HSD1(tg/tg) mice and diabetic Lep(db/db) mice with markedly elevated ß-cell 11ß-HSD1 levels had impaired basal ß-cell function. Optimal elevation of ß-cell 11ß-HSD1 represents a novel biological mechanism supporting compensatory insulin hypersecretion rather than exacerbating metabolic disease. These findings have immediate significance for current therapeutic strategies for type 2 diabetes.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/fisiologia , Diabetes Mellitus Tipo 2/prevenção & controle , Dieta Hiperlipídica/efeitos adversos , Células Secretoras de Insulina/fisiologia , Animais , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Diabetes Mellitus/etiologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Teste de Tolerância a Glucose , Insulina/metabolismo , Secreção de Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ácido Palmítico/farmacologia
15.
FEBS Lett ; 585(2): 269-74, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21176778

RESUMO

The role of protein kinase C (PKCs) isoforms in the regulation of glucose metabolism by insulin is complex, partly due to the large PKC family consisting of three sub-groups: conventional, novel and atypical. Activation of some conventional and novel PKCs in response to increased levels of diacylglycerol (DAG) have been shown to counteract insulin signalling. However, roles of atypical PKCs (aPKCs) remain poorly understood. aPKCs act as molecular switches by promoting or suppressing signalling pathways, in response to insulin or ceramides respectively. Understanding how DAG- and ceramide-activated PKCs impair insulin signalling would help to develop treatments to fight insulin resistance.


Assuntos
Resistência à Insulina , Proteína Quinase C/metabolismo , Ceramidas , Diglicerídeos , Humanos , Metabolismo dos Lipídeos , Isoformas de Proteínas
16.
Diabetes ; 60(4): 1158-67, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21350084

RESUMO

OBJECTIVE: The study objective was to determine the key early mechanisms underlying the beneficial redistribution, function, and inflammatory profile of adipose tissue in 11ß-hydroxysteroid dehydrogenase type 1 knockout (11ß-HSD1(-/-)) mice fed a high-fat (HF) diet. RESEARCH DESIGN AND METHODS: By focusing on the earliest divergence in visceral adiposity, subcutaneous and visceral fat depots from 11ß-HSD1(-/-) and C57Bl/6J control mice fed an HF diet for 4 weeks were used for comparative microarray analysis of gene expression, and differences were validated with real-time PCR. Key changes in metabolic signaling pathways were confirmed using Western blotting/immunoprecipitation, and fat cell size was compared with the respective chow-fed control groups. Altered adipose inflammatory cell content and function after 4 weeks (early) and 18 weeks (chronic) of HF feeding was investigated using fluorescence (and magnetic)-activated cell sorting analysis, immunohistochemistry, and in situ hybridization. RESULTS: In subcutaneous fat, HF-fed 11ß-HSD1(-/-) mice showed evidence of enhanced insulin and ß-adrenergic signaling associated with accretion of smaller metabolically active adipocytes. In contrast, reduced 11ß-HSD1(-/-) visceral fat accumulation was characterized by maintained AMP kinase activation, not insulin sensitization, and higher adipocyte interleukin-6 release. Intracellular glucocorticoid deficiency was unexpectedly associated with suppressed inflammatory signaling and lower adipocyte monocyte chemoattractant protein-1 secretion with strikingly reduced cytotoxic T-cell and macrophage infiltration, predominantly in visceral fat. CONCLUSIONS: Our data define for the first time the novel and distinct depot-specific mechanisms driving healthier fat patterning and function as a result of reduced intra-adipose glucocorticoid levels.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Inflamação/metabolismo , Obesidade Abdominal/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , Células 3T3-L1 , Proteínas Quinases Ativadas por AMP/metabolismo , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Western Blotting , Gorduras na Dieta/efeitos adversos , Ativação Enzimática/efeitos dos fármacos , Citometria de Fluxo , Glucocorticoides/farmacologia , Imuno-Histoquímica , Hibridização In Situ , Inflamação/genética , Interleucina-6/farmacologia , Gordura Intra-Abdominal/metabolismo , Masculino , Camundongos , Camundongos Mutantes , Obesidade Abdominal/induzido quimicamente , Obesidade Abdominal/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Am J Physiol Endocrinol Metab ; 282(1): E59-66, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11739084

RESUMO

We investigated angiotensinogen (AGT) expression in adipose tissue and liver of Zucker rats during the onset of obesity. The developmental pattern of AGT expression (protein and mRNA) in liver was similar in both genotypes. In inguinal adipose tissue, AGT cell content was similar in suckling and weaned pups in lean rats, whereas it continuously increased with age in obese rats. AGT amount in adipocytes was unaffected by the genotype until weaning. Thereafter, adipocytes from obese rats displayed a significant increase in AGT content that was strengthened with age. Compared with the cell content, the amount of secreted AGT over 24 h was higher, and a genotype effect was observed as early as 14 days of age. Using fat cell populations differing by size, we showed that this AGT oversecretion was not solely related to adipocyte hypertrophy. Our results demonstrate that the fa genotype exerts a control on the production of AGT in a tissue-specific manner, suggesting a local role of AGT in the overdevelopment of adipose tissue.


Assuntos
Tecido Adiposo/metabolismo , Angiotensinogênio/metabolismo , Obesidade/metabolismo , Ratos Zucker/metabolismo , Adipócitos/metabolismo , Tecido Adiposo/citologia , Angiotensinogênio/genética , Animais , Tamanho Celular , Células Cultivadas , Genótipo , Fígado/metabolismo , Obesidade/genética , RNA Mensageiro/metabolismo , Ratos , Valores de Referência
18.
Biochem J ; 362(Pt 1): 113-8, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11829746

RESUMO

In the obese state, enlarged adipose cells display an altered gene-expression profile and metabolic capacity. The aim of this study was to gain insight into their secretory function, by assessing two secreted proteins, leptin and angiotensinogen, in adipose cells of obese (fa/fa) Zucker rats. A marked and co-ordinate increase in leptin mRNA, gene transcription and promoter activity was observed in obese compared with lean (Fa/fa) rat adipose cells, and this resulted in increased leptin release in culture. Two sets of observations suggest that this effect is due to the fa mutation. First, adipose-cell leptin release was higher in heterozygous (Fa/fa) than in homozygous (Fa/Fa) lean rats. Second, leptin release was not enhanced in enlarged adipose cells of FalFa rats fed a high-fat diet for 15 days. At variance with leptin, angiotensinogen production was not significantly increased in the obese cells. Dexamethasone stimulated both leptin and angiotensinogen release in lean and obese rat adipose cells. The magnitude of leptin stimulation was higher in fa/fa than in Fa/fa rats, whereas angiotensinogen release was increased to the same extent in both genotypes. These observations suggest that leptin production is specifically enhanced in enlarged adipose cells of obese Zucker rats and that cell hypertrophy is not the sole determinant of this feature. Increased leptin production might be related to disruption of leptin signalling by the fa mutation.


Assuntos
Tecido Adiposo/metabolismo , Leptina/biossíntese , Obesidade/metabolismo , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/patologia , Angiotensinogênio/genética , Animais , Dexametasona/farmacologia , Dieta , Dosagem de Genes , Expressão Gênica , Masculino , Mutação , Ratos , Ratos Zucker , Transcrição Gênica , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA