Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
PLoS Biol ; 19(11): e3001447, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34758018

RESUMO

During the first weeks of postnatal heart development, cardiomyocytes undergo a major adaptive metabolic shift from glycolytic energy production to fatty acid oxidation. This metabolic change is contemporaneous to the up-regulation and activation of the p38γ and p38δ stress-activated protein kinases in the heart. We demonstrate that p38γ/δ contribute to the early postnatal cardiac metabolic switch through inhibitory phosphorylation of glycogen synthase 1 (GYS1) and glycogen metabolism inactivation. Premature induction of p38γ/δ activation in cardiomyocytes of newborn mice results in an early GYS1 phosphorylation and inhibition of cardiac glycogen production, triggering an early metabolic shift that induces a deficit in cardiomyocyte fuel supply, leading to whole-body metabolic deregulation and maladaptive cardiac pathogenesis. Notably, the adverse effects of forced premature cardiac p38γ/δ activation in neonate mice are prevented by maternal diet supplementation of fatty acids during pregnancy and lactation. These results suggest that diet interventions have a potential for treating human cardiac genetic diseases that affect heart metabolism.


Assuntos
Glicogênio Sintase/metabolismo , Proteína Quinase 12 Ativada por Mitógeno/metabolismo , Proteína Quinase 13 Ativada por Mitógeno/metabolismo , Miocárdio/enzimologia , Animais , Animais Recém-Nascidos , Cardiomegalia/enzimologia , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Dieta Hiperlipídica , Ativação Enzimática , Comportamento Alimentar , Feminino , Deleção de Genes , Intolerância à Glucose/enzimologia , Glicogênio/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Resistência à Insulina , Metabolismo dos Lipídeos , Sistema de Sinalização das MAP Quinases , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/enzimologia , Especificidade de Órgãos , Fosforilação
2.
Diabetologia ; 64(11): 2550-2561, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34448880

RESUMO

AIMS/HYPOTHESIS: Nicotinamide nucleotide transhydrogenase (NNT) is involved in mitochondrial NADPH production and its spontaneous inactivating mutation (NntTr [Tr, truncated]) is usually considered to be the main cause of the lower glucose tolerance of C57BL/6J vs C57BL/6N mice. However, the impact of this mutation on glucose tolerance remains disputed. Here, we singled out the impact of NntTr from that of other genetic variants between C57BL/6J and C57BL/6N mice on mitochondrial glutathione redox state (EGSH), glucose-stimulated insulin secretion (GSIS) and glucose tolerance. METHODS: Male and female N5BL/6J mice that express wild-type Nnt (NntWT) or NntTr (N5-WT and N5-Tr mice) on the C57BL/6J genetic background were obtained by crossing N5BL/6J NntWT/Tr heterozygous mice. C57BL/6J and C57BL/6N mice were from Janvier Labs. The Nnt genotype was confirmed by PCR and the genetic background by whole genome sequencing of one mouse of each type. Glucose tolerance was assessed by IPGTT, ITT and fasting/refeeding tests. Stimulus-secretion coupling events and GSIS were measured in isolated pancreatic islets. Cytosolic and mitochondrial EGSH were measured using the fluorescent redox probe GRX1-roGFP2 (glutaredoxin 1 fused to redox-sensitive enhanced GFP). RESULTS: The Nnt genotype and genetic background of each type of mouse were confirmed. As reported previously in C57BL/6N vs C57BL/6J islets, the glucose regulation of mitochondrial (but not cytosolic) EGSH and of NAD(P)H autofluorescence was markedly improved in N5-WT vs N5-Tr islets, confirming the role of NNT in mitochondrial redox regulation. However, ex vivo GSIS was only 1.2-1.4-times higher in N5-WT vs N5-Tr islets, while it was 2.4-times larger in C57BL/6N vs N5-WT islets, questioning the role of NNT in GSIS. In vivo, the ITT results did not differ between N5-WT and N5-Tr or C57BL/6N mice. However, the glucose excursion during an IPGTT was only 15-20% lower in female N5-WT mice than in N5-Tr and C57BL/6J mice and remained 3.5-times larger than in female C57BL/6N mice. Similar observations were made during a fasting/refeeding test. A slightly larger (~30%) impact of NNT on glucose tolerance was found in males. CONCLUSIONS/INTERPRETATION: Although our results confirm the importance of NNT in the regulation of mitochondrial redox state by glucose, they markedly downsize the role of NNT in the alteration of GSIS and glucose tolerance in C57BL/6J vs C57BL/6N mice. Therefore, documenting an NntWT genotype in C57BL/6 mice does not provide proof that their glucose tolerance is as good as in C57BL/6N mice.


Assuntos
Intolerância à Glucose/enzimologia , Glucose/farmacologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , NADP Trans-Hidrogenases/fisiologia , Animais , Cálcio/metabolismo , Células Cultivadas , Feminino , Glutarredoxinas , Glutationa/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , NADP/metabolismo , Oxirredução , Reação em Cadeia da Polimerase , Sequenciamento Completo do Genoma
3.
Mol Cell Endocrinol ; 537: 111425, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34391847

RESUMO

BACKGROUND: Previously, we reported that Mof was highly expressed in α-cells, and its knockdown led to ameliorated fasting blood glucose (FBG) and glucose tolerance in non-diabetic mice, attributed by reduced total α-cell but enhanced prohormone convertase (PC)1/3-positive α-cell mass. However, how Mof and histone 4 lysine 16 acetylation (H4K16ac) control α-cell and whether Mof inhibition improves glucose handling in type 2 diabetes (T2DM) mice remain unknown. METHODS: Mof overexpression and chromatin immunoprecipitation sequence (ChIP-seq) based on H4K16ac were applied to determine the effect of Mof on α-cell transcriptional factors and underlying mechanism. Then we administrated mg149 to α-TC1-6 cell line, wild type, db/db and diet-induced obesity (DIO) mice to observe the impact of Mof inhibition in vitro and in vivo. In vitro, western blotting and TUNEL staining were used to examine α-cell apoptosis and function. In vivo, glucose tolerance, hormone levels, islet population, α-cell ratio and the co-staining of glucagon and PC1/3 or PC2 were examined. RESULTS: Mof activated α-cell-specific transcriptional network. ChIP-seq results indicated that H4K16ac targeted essential genes regulating α-cell differentiation and function. Mof activity inhibition in vitro caused impaired α-cell function and enhanced apoptosis. In vivo, it contributed to ameliorated glucose intolerance and islet dysfunction, characterized by decreased fasting glucagon and elevated post-challenge insulin levels in T2DM mice. CONCLUSION: Mof regulates α-cell differentiation and function via acetylating H4K16ac and H4K16ac binding to Pax6 and Foxa2 promoters. Mof inhibition may be a potential interventional target for T2DM, which led to decreased α-cell ratio but increased PC1/3-positive α-cells.


Assuntos
Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/fisiopatologia , Células Secretoras de Glucagon/enzimologia , Células Secretoras de Glucagon/patologia , Intolerância à Glucose/enzimologia , Intolerância à Glucose/fisiopatologia , Histona Acetiltransferases/antagonistas & inibidores , Acetilação/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Tipo 2/complicações , Dieta , Redes Reguladoras de Genes/efeitos dos fármacos , Células Secretoras de Glucagon/efeitos dos fármacos , Histona Acetiltransferases/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Pró-Proteína Convertase 1/metabolismo , Salicilatos/farmacologia
4.
Eur J Pharmacol ; 899: 173978, 2021 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-33691164

RESUMO

Metabolic syndrome is linked to an increased risk of cardiovascular complications by a mechanism involving mainly decreased nitric oxide (NO) bioavailability and impaired NO-soluble guanylate cyclase (sGC)- cyclic guanosine monophosphate (cGMP) signalling (NO-sGC-cGMP). To further develop this scientific point, this study aimed to investigate the effects of long-term treatment with BAY 41-2272 (a sGC stimulator) on cardiovascular reactivity of spontaneously hypertensive rats (SHR) as a model of metabolic syndrome. SHR were randomly divided into 3 groups: control group, cafeteria diet (CD)-fed group and CD-fed group treated daily with BAY 41-2272 (5 mg/kg) by gastric gavage for 12 weeks. In vivo measurements of body weight, abdominal circumference, blood pressure and glucose tolerance test were performed. At the end of the feeding period, ex vivo cumulative concentration-response curves were performed on isolated perfused heart (isoproterenol (0.1 nM - 1 µM)) and thoracic aorta (phenylephrine (1 nM-10 µM), acetylcholine (1 nM-10 µM), and sodium nitroprusside (SNP) (0.1 nM-0.1 µM)). We showed that chronic CD feeding induced abdominal obesity, hypertriglyceridemia, glucose intolerance and exacerbated arterial hypertension in SHR. Compared to control group, CD-fed group showed a decrease in ß-adrenoceptor-induced cardiac inotropy, in coronary perfusion pressure and in aortic contraction to phenylephrine. While relaxing effects of acetylcholine and SNP were unchanged. BAY 41-2272 long-term treatment markedly prevented arterial hypertension development and glucose intolerance, enhanced the α1-adrenoceptor-induced vasoconstriction, and restored cardiac inotropy and coronary vasodilation. These findings suggest that BAY 41-2272 may be a potential novel drug for preventing metabolic and cardiovascular complications of metabolic syndrome.


Assuntos
Doenças Cardiovasculares/prevenção & controle , Ativadores de Enzimas/farmacologia , Síndrome Metabólica/prevenção & controle , Pirazóis/farmacologia , Piridinas/farmacologia , Guanilil Ciclase Solúvel/metabolismo , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/enzimologia , Aorta Torácica/fisiopatologia , Doenças Cardiovasculares/enzimologia , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/fisiopatologia , Circulação Coronária/efeitos dos fármacos , GMP Cíclico/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Intolerância à Glucose/enzimologia , Intolerância à Glucose/etiologia , Intolerância à Glucose/fisiopatologia , Intolerância à Glucose/prevenção & controle , Hipertensão/enzimologia , Hipertensão/etiologia , Hipertensão/fisiopatologia , Hipertensão/prevenção & controle , Hipertrigliceridemia/enzimologia , Hipertrigliceridemia/etiologia , Hipertrigliceridemia/fisiopatologia , Hipertrigliceridemia/prevenção & controle , Preparação de Coração Isolado , Masculino , Síndrome Metabólica/enzimologia , Síndrome Metabólica/etiologia , Síndrome Metabólica/fisiopatologia , Óxido Nítrico Sintase Tipo II/metabolismo , Obesidade Abdominal/enzimologia , Obesidade Abdominal/etiologia , Obesidade Abdominal/fisiopatologia , Obesidade Abdominal/prevenção & controle , Ratos Endogâmicos SHR , Vasoconstrição/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Função Ventricular Esquerda/efeitos dos fármacos , Pressão Ventricular/efeitos dos fármacos
5.
Clin Nutr ; 40(3): 1420-1429, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32943240

RESUMO

BACKGROUND & AIMS: Several proteins of the innate immune system are known to be deregulated with insulin resistance. We here aimed to investigate the relationship among circulating lysozyme (both plasma concentration and activity) and obesity-associated metabolic disturbances. METHODS: Plasma lysozyme concentration was determined cross-sectionally in a discovery (Cohort 1, n = 137) and in a replication cohort (Cohort 2, n = 181), in which plasma lysozyme activity was also analyzed. Plasma lysozyme was also evaluated longitudinally in participants from the replication cohort (n = 93). Leukocyte lysozyme expression (LYZ mRNA) were also investigated in an independent cohort (Cohort 3, n = 76), and adipose tissue (AT) LYZ mRNA (n = 25) and plasma peptidoglycan levels (n = 61) in subcohorts from discovery cohort. RESULTS: Translocation of peptidoglycan (as inferred from its increased circulating levels) was linked to plasma lysozyme, hyperinsulinemia and dyslipidemia in obese subjects. In both discovery and replication cohorts, plasma lysozyme levels and activity were significantly increased in obesity in direct association with obesity-associated metabolic disturbances and inflammatory parameters, being circulating lysozyme negatively correlated with fasting glucose, HbA1c and insulin resistance (HOMA-IR) in obese subjects. Of note, total cholesterol (p < 0.0001) and LDL cholesterol (p = 0.003) contributed independently to age-, gender- and BMI adjusted plasma lysozyme activity. Longitudinally, changes in HbA1c levels and serum LDL cholesterol were negatively associated with circulating lysozyme antimicrobial activity. On the contrary, the change in glucose infusion rate during the clamp (insulin sensitivity) was positively associated with lysozyme concentration. CONCLUSIONS: Increased plasma lysozyme levels and activity are found in obese subjects. The longitudinal findings suggest that plasma lysozyme might be protective on the development of obesity-associated metabolic disturbances.


Assuntos
Intolerância à Glucose/enzimologia , Sistema Imunitário/enzimologia , Inflamação/enzimologia , Muramidase/sangue , Obesidade/enzimologia , Tecido Adiposo/enzimologia , Adulto , Glicemia/análise , Estudos de Coortes , Dislipidemias/enzimologia , Feminino , Humanos , Resistência à Insulina , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Obesidade/metabolismo , Peptidoglicano/sangue
6.
Molecules ; 25(19)2020 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-32987643

RESUMO

Diabetes mellitus (DM) is one of the major causes of death in the world. There are two types of DM-type 1 DM and type 2 DM. Type 1 DM can only be treated by insulin injection whereas type 2 DM is commonly treated using anti-hyperglycemic agents. Despite its effectiveness in controlling blood glucose level, this therapeutic approach is not able to reduce the decline in the number of functional pancreatic ß cells. MST1 is a strong pro-apoptotic kinase that is expressed in pancreatic ß cells. It induces ß cell death and impairs insulin secretion. Recently, a potent and specific inhibitor for MST1, called XMU-MP-1, was identified and characterized. We hypothesized that treatment with XMU-MP-1 would produce beneficial effects by improving the survival and function of the pancreatic ß cells. We used INS-1 cells and STZ-induced diabetic mice as in vitro and in vivo models to test the effect of XMU-MP-1 treatment. We found that XMU-MP-1 inhibited MST1/2 activity in INS-1 cells. Moreover, treatment with XMU-MP-1 produced a beneficial effect in improving glucose tolerance in the STZ-induced diabetic mouse model. Histological analysis indicated that XMU-MP-1 increased the number of pancreatic ß cells and enhanced Langerhans islet area in the severe diabetic mice. Overall, this study showed that MST1 could become a promising therapeutic target for diabetes mellitus.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Intolerância à Glucose/tratamento farmacológico , Células Secretoras de Insulina/enzimologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Sulfonamidas/farmacologia , Animais , Linhagem Celular , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Experimental/patologia , Intolerância à Glucose/induzido quimicamente , Intolerância à Glucose/enzimologia , Intolerância à Glucose/patologia , Células Secretoras de Insulina/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Serina-Treonina Quinases/metabolismo , Serina-Treonina Quinase 3
7.
Biosci Rep ; 39(7)2019 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-31270248

RESUMO

Cardiometabolic syndrome (CMS) describes the cluster of metabolic and cardiovascular diseases that are generally characterized by impaired glucose tolerance, intra-abdominal adiposity, dyslipidemia, and hypertension. CMS currently affects more than 25% of the world's population and the rates of diseases are rapidly rising. These CMS conditions represent critical risk factors for cardiovascular diseases including atherosclerosis, heart failure, myocardial infarction, and peripheral artery disease (PAD). Therefore, it is imperative to elucidate the underlying signaling involved in disease onset and progression. The c-Jun N-terminal Kinases (JNKs) are a family of stress signaling kinases that have been recently indicated in CMS. The purpose of this review is to examine the in vivo implications of JNK as a potential therapeutic target for CMS. As the constellation of diseases associated with CMS are complex and involve multiple tissues and environmental triggers, carefully examining what is known about the JNK pathway will be important for specificity in treatment strategies.


Assuntos
Dislipidemias/genética , Intolerância à Glucose/genética , Hipertensão/genética , Insulina/genética , MAP Quinase Quinase 4/genética , Tecido Adiposo/enzimologia , Tecido Adiposo/patologia , Adiposidade/genética , Animais , Citocinas/genética , Citocinas/metabolismo , Dislipidemias/enzimologia , Dislipidemias/patologia , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica , Intolerância à Glucose/enzimologia , Intolerância à Glucose/patologia , Humanos , Hipertensão/enzimologia , Hipertensão/patologia , Insulina/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Fígado/enzimologia , Fígado/patologia , MAP Quinase Quinase 4/deficiência , Camundongos , Camundongos Knockout , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Transdução de Sinais , Síndrome
8.
Endocrine ; 65(2): 295-303, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31115769

RESUMO

PURPOSE: Emerging clinical evidence has implied that alkaline phosphatase (ALP) may contribute to gestational diabetes mellitus (GDM). However, there were no studies to reveal the independent and prospective associations between ALP and GDM. Our aim was to explore the independent and prospective associations between early maternal ALP level and GDM risk and glucose regulation. METHODS: In a prospective cohort study with 2073 singleton mothers at four maternity units in China, maternal serum ALP levels were measured before 20 gestational weeks. Using logistic regression, we analyzed the relationship between maternal ALP level and risk of GDM. We further explored the relationships of ALP level to fasting blood glucose (FBG), 1-h and 2-h post-load blood glucose (1-h, 2-h PBG) with multiple linear regression. Finally, we analyzed the association between maternal ALP level and isolated impaired fasting glucose (i-IFG) and isolated impaired glucose tolerance (i-IGT) risk. RESULTS: The maximum value of maternal ALP level was 90 U/L, within the normal range. After adjustment for confounding factors, the odds ratio (ORs) of GDM increased linearly with ALP level (p for overall association = 0.002, p for nonlinear association = 0.799), with the OR comparing the highest versus lowest quartile of 2.47 (95% CI 1.47, 4.15). Moreover, each additional of 10 U/L ALP level was associated with a 2% higher FBG (p = 0.043) and a 12% higher 1-h PBG (p = 0.004). Higher ALP level also increased the risk of i-IFG (OR 3.73, 95% CI 1.17-11.86) and i-IGT (OR 2.03, 95% CI 1.07-3.84). CONCLUSIONS: Even within the upper limit of normal, higher early maternal ALP level could increase the risk of GDM. Moreover, both FBG and PBG were increased with early maternal ALP.


Assuntos
Fosfatase Alcalina/sangue , Diabetes Gestacional/enzimologia , Intolerância à Glucose/enzimologia , Adolescente , Adulto , Glicemia , China , Diabetes Gestacional/sangue , Diabetes Gestacional/etiologia , Feminino , Intolerância à Glucose/sangue , Intolerância à Glucose/etiologia , Humanos , Gravidez , Estudos Prospectivos , Adulto Jovem
9.
Cardiovasc Diabetol ; 17(1): 103, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-30016962

RESUMO

BACKGROUND: The cardiac hormones atrial (ANP) and B-type natriuretic peptides (BNP) moderate arterial blood pressure and improve energy metabolism as well as insulin sensitivity via their shared cGMP-producing guanylyl cyclase-A (GC-A) receptor. Obesity is associated with impaired NP/GC-A/cGMP signaling, which possibly contributes to the development of type 2 diabetes and its cardiometabolic complications. In vitro, synthetic ANP, via GC-A, stimulates glucose-dependent insulin release from cultured pancreatic islets and ß-cell proliferation. However, the relevance for systemic glucose homeostasis in vivo is not known. To dissect whether the endogenous cardiac hormones modulate the secretory function and/or proliferation of ß-cells under (patho)physiological conditions in vivo, here we generated a novel genetic mouse model with selective disruption of the GC-A receptor in ß-cells. METHODS: Mice with a floxed GC-A gene were bred to Rip-CreTG mice, thereby deleting GC-A selectively in ß-cells (ß GC-A KO). Weight gain, glucose tolerance, insulin sensitivity, and glucose-stimulated insulin secretion were monitored in normal diet (ND)- and high-fat diet (HFD)-fed mice. ß-cell size and number were measured by immunofluorescence-based islet morphometry. RESULTS: In vitro, the insulinotropic and proliferative actions of ANP were abolished in islets isolated from ß GC-A KO mice. Concordantly, in vivo, infusion of BNP mildly enhanced baseline plasma insulin levels and glucose-induced insulin secretion in control mice. This effect of exogenous BNP was abolished in ß GC-A KO mice, corroborating the efficient inactivation of the GC-A receptor in ß-cells. Despite this under physiological, ND conditions, fasted and fed insulin levels, glucose-induced insulin secretion, glucose tolerance and ß-cell morphology were similar in ß GC-A KO mice and control littermates. However, HFD-fed ß GC-A KO animals had accelerated glucose intolerance and diminished adaptative ß-cell proliferation. CONCLUSIONS: Our studies of ß GC-A KO mice demonstrate that the cardiac hormones ANP and BNP do not modulate ß-cell's growth and secretory functions under physiological, normal dietary conditions. However, endogenous NP/GC-A signaling improves the initial adaptative response of ß-cells to HFD-induced obesity. Impaired ß-cell NP/GC-A signaling in obese individuals might contribute to the development of type 2 diabetes.


Assuntos
Fator Natriurético Atrial/metabolismo , Glicemia/metabolismo , Deleção de Genes , Intolerância à Glucose/etiologia , Células Secretoras de Insulina/enzimologia , Obesidade/complicações , Receptores do Fator Natriurético Atrial/deficiência , Animais , Proliferação de Células , Modelos Animais de Doenças , Progressão da Doença , Predisposição Genética para Doença , Intolerância à Glucose/enzimologia , Intolerância à Glucose/genética , Intolerância à Glucose/patologia , Insulina/sangue , Células Secretoras de Insulina/patologia , Camundongos Knockout , Peptídeo Natriurético Encefálico/metabolismo , Obesidade/enzimologia , Obesidade/genética , Fenótipo , Receptores do Fator Natriurético Atrial/genética , Transdução de Sinais , Técnicas de Cultura de Tecidos
10.
Nutrients ; 10(2)2018 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-29385050

RESUMO

Selenium (Se) is an essential micronutrient known for its antioxidant properties and health benefits, attributed to its presence in selenoproteins as the amino acid, selenocysteine. Selenocysteine lyase (Scly) catalyzes hydrolysis of selenocysteine to selenide and alanine, facilitating re-utilization of Se for de novo selenoprotein synthesis. Previously, it was reported that male Scly-/- mice develop increased body weight and body fat composition, and altered lipid and carbohydrate metabolism, compared to wild type mice. Strikingly, females appeared to present with a less severe phenotype, suggesting the relationship between Scly and energy metabolism may be regulated in a sex-specific manner. Here, we report that while body weight and body fat gain occur in both male and female Scly-/- mice, strikingly, males are susceptible to developing glucose intolerance, whereas female Scly-/- mice are protected. Because Se is critical for male reproduction, we hypothesized that castration would attenuate the metabolic dysfunction observed in male Scly-/- mice by eliminating sequestration of Se in testes. We report that fasting serum insulin levels were significantly reduced in castrated males compared to controls, but islet area was unchanged between groups. Finally, both male and female Scly-/- mice exhibit reduced hypothalamic expression of selenoproteins S, M, and glutathione peroxidase 1.


Assuntos
Metabolismo Energético , Liases/deficiência , Caracteres Sexuais , Adiposidade , Fatores Etários , Animais , Glicemia/metabolismo , Metabolismo Energético/genética , Feminino , Genótipo , Intolerância à Glucose/sangue , Intolerância à Glucose/enzimologia , Intolerância à Glucose/genética , Glutationa Peroxidase/metabolismo , Hipotálamo/enzimologia , Insulina/sangue , Liases/genética , Masculino , Camundongos Knockout , Orquiectomia , Fenótipo , Selenoproteínas/metabolismo , Fatores de Tempo , Aumento de Peso , Glutationa Peroxidase GPX1
11.
Am J Physiol Regul Integr Comp Physiol ; 314(3): R478-R488, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351427

RESUMO

Protein tyrosine phosphatase 1B (PTP1B) is a negative regulator of leptin receptor signaling and may contribute to leptin resistance in diet-induced obesity. Although PTP1B inhibition has been suggested as a potential weight loss therapy, the role of specific neuronal PTP1B signaling in cardiovascular and metabolic regulation and the importance of sex differences in this regulation are still unclear. In this study, we investigated the impact of proopiomelanocortin (POMC) neuronal PTP1B deficiency in cardiometabolic regulation in male and female mice fed a high-fat diet (HFD). When compared with control mice (PTP1B flox/flox), male and female mice deficient in POMC neuronal PTP1B (PTP1B flox/flox/POMC-Cre) had attenuated body weight gain (males: -18%; females: -16%) and fat mass (males: -33%; female: -29%) in response to HFD. Glucose tolerance was improved by 40%, and liver lipid accumulation was reduced by 40% in PTP1B/POMC-Cre males but not in females. When compared with control mice, deficiency of POMC neuronal PTP1B did not alter mean arterial pressure (MAP) in male or female mice (males: 112 ± 1 vs. 112 ± 1 mmHg in controls; females: 106 ± 3 vs. 109 ± 3 mmHg in controls). Deficiency of POMC neuronal PTP1B also did not alter MAP response to acute stress in males or females compared with control mice (males: Δ32 ± 0 vs. Δ29 ± 4 mmHg; females: Δ22 ± 2 vs. Δ27 ± 4 mmHg). These data demonstrate that POMC-specific PTP1B deficiency improved glucose tolerance and attenuated diet-induced fatty liver only in male mice and attenuated weight gain in males and females but did not enhance the MAP and HR responses to a HFD or to acute stress.


Assuntos
Núcleo Arqueado do Hipotálamo/enzimologia , Glicemia/metabolismo , Intolerância à Glucose/enzimologia , Metabolismo dos Lipídeos , Fígado/metabolismo , Neurônios/enzimologia , Hepatopatia Gordurosa não Alcoólica/enzimologia , Obesidade/enzimologia , Pró-Opiomelanocortina/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Núcleo Solitário/enzimologia , Animais , Núcleo Arqueado do Hipotálamo/fisiopatologia , Biomarcadores/sangue , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Intolerância à Glucose/sangue , Intolerância à Glucose/fisiopatologia , Intolerância à Glucose/prevenção & controle , Fígado/patologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/sangue , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Obesidade/etiologia , Obesidade/fisiopatologia , Obesidade/prevenção & controle , Proteína Tirosina Fosfatase não Receptora Tipo 1/deficiência , Proteína Tirosina Fosfatase não Receptora Tipo 1/genética , Fatores Sexuais , Núcleo Solitário/fisiopatologia , Aumento de Peso
12.
Toxicol Sci ; 160(1): 4-14, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29036520

RESUMO

The lung is constantly exposed to ambient pollutants such as ambient fine particulate matter (PM2.5), making it one of the most frequent locations of inflammation in the body. Given the establishment of crucial role of inflammation in the pathogenesis of cardiometabolic diseases, pulmonary inflammation is thus widely believed to be an important risk factor for cardiometabolic diseases. However, the causality between them has not yet been well established. To determine if pulmonary inflammation is sufficient to cause adverse cardiometabolic effects, SFTPC-rtTA+/-tetO-cre+/-pROSA-inhibitor κB kinase 2(IKK2)ca+/- (LungIKK2ca) and littermate SFTPC-rtTA+/-tetO-cre-/-pROSA-IKK2ca+/- wildtype (WT) mice were fed with doxycycline diet to induce constitutively active Ikk2 (Ikk2ca) overexpression in the lung and their pulmonary, systemic, adipose, and hypothalamic inflammations, vascular function, and glucose homeostasis were assessed. Feeding with doxycycline diet resulted in IKK2ca overexpression in the lungs of LungIKK2ca but not WT mice. This induction of IKK2ca was accompanied by marked pulmonary inflammation as evidenced by significant increases in bronchoalveolar lavage fluid leukocytes, pulmonary macrophage infiltration, and pulmonary mRNA expression of tumor necrosis factor α (Tnfα) and interleukin-6 (Il-6). This pulmonary inflammation due to lung-specific overexpression of IKK2ca was sufficient to increase circulating TNFα and IL-6 levels, adipose expression of Tnfα and Il-6 mRNA, aortic endothelial dysfunction, and systemic insulin resistance. Unexpectedly, no significant alteration in hypothalamic expression of Tnfα and Il-6 mRNA and glucose intolerance were observed in these mice. Pulmonary inflammation is sufficient to induce systemic inflammation, endothelial dysfunction, and insulin resistance, but not hypothalamic inflammation and glucose intolerance.


Assuntos
Tecido Adiposo/enzimologia , Encefalite/enzimologia , Intolerância à Glucose/enzimologia , Hipotálamo/enzimologia , Quinase I-kappa B/metabolismo , Pulmão/enzimologia , Paniculite/enzimologia , Pneumonia/enzimologia , Animais , Aorta/enzimologia , Aorta/fisiopatologia , Glicemia/metabolismo , Encefalite/genética , Endotélio Vascular/enzimologia , Endotélio Vascular/fisiopatologia , Ativação Enzimática , Predisposição Genética para Doença , Intolerância à Glucose/sangue , Intolerância à Glucose/genética , Quinase I-kappa B/genética , Resistência à Insulina , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmão/patologia , Camundongos Transgênicos , Paniculite/genética , Fenótipo , Pneumonia/genética , Pneumonia/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
13.
PLoS One ; 12(9): e0183488, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28873424

RESUMO

Here, we aimed to investigate the potential role of DUSP6, a dual specificity phosphatase, that specifically inactivates extracellular signal-regulated kinase (ERK), for the regulation of body weight and glucose homeostasis. We further assessed whether metabolic challenges affect Dusp6 expression in selected brain areas or white adipose tissue. Hypothalamic Dusp6 mRNA levels remained unchanged in chow-fed lean vs. high fat diet (HFD) fed obese C57Bl/6J mice, and in C57Bl/6J mice undergoing prolonged fasting or refeeding with fat free diet (FFD) or HFD. Similarly, Dusp6 expression levels were unchanged in selected brain regions of Lepob mice treated with 1 mg/kg of leptin for 6 days, compared to pair-fed or saline-treated Lepob controls. Dusp6 expression levels remained unaltered in vitro in primary adipocytes undergoing differentiation, but were increased in eWAT of HFD-fed obese C57Bl/6J mice, compared to chow-fed lean controls. Global chow-fed DUSP6 KO mice displayed reduced body weight and lean mass and slightly increased fat mass at a young age, which is indicative for early-age weight retardation. Subsequent exposure to HFD led to a significant increase in lean mass and body weight in DUSP6 deficient mice, compared to WT controls. Nevertheless, after 26 weeks of high-fat diet exposure, we observed comparable body weight, fat and lean mass in DUSP6 WT and KO mice, suggesting overall normal susceptibility to develop obesity. In line with the increased weight gain to compensate for early-age weight retardation, HFD-fed DUSP6 KO displayed increased expression levels of anabolic genes involved in lipid and cholesterol metabolism in the epididymal white adipose tissue (eWAT), compared to WT controls. Glucose tolerance was perturbed in both chow-fed lean or HFD-fed obese DUSP6 KO, compared to their respective WT controls. Overall, our data indicate that DUSP6 deficiency has limited impact on the regulation of energy metabolism, but impairs systemic glucose tolerance. Our data are in conflict to earlier reports that propose protection from diet-induced obesity and glucose intolerance in DUSP6 deficient mice. Reasons for the discrepancies remain elusive, but may entail differential genetic backgrounds, environmental factors such as the type and source of HFD, or alterations in the gut microbiome between facilities.


Assuntos
Peso Corporal , Fosfatase 6 de Especificidade Dupla/deficiência , Intolerância à Glucose/enzimologia , Tecido Adiposo Branco/metabolismo , Animais , Composição Corporal , Dieta Hiperlipídica , Fosfatase 6 de Especificidade Dupla/genética , Fosfatase 6 de Especificidade Dupla/metabolismo , Epididimo/metabolismo , Jejum , Perfilação da Expressão Gênica , Intolerância à Glucose/complicações , Intolerância à Glucose/genética , Intolerância à Glucose/patologia , Homeostase , Leptina/metabolismo , Metabolismo dos Lipídeos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenômenos Fisiológicos da Nutrição , Obesidade/complicações , Obesidade/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
14.
Cell Rep ; 20(3): 655-667, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28723568

RESUMO

The liver has a large capacity for mitochondrial fatty acid ß-oxidation, which is critical for systemic metabolic adaptations such as gluconeogenesis and ketogenesis. To understand the role of hepatic fatty acid oxidation in response to a chronic high-fat diet (HFD), we generated mice with a liver-specific deficiency of mitochondrial long-chain fatty acid ß-oxidation (Cpt2L-/- mice). Paradoxically, Cpt2L-/- mice were resistant to HFD-induced obesity and glucose intolerance with an absence of liver damage, although they exhibited serum dyslipidemia, hepatic oxidative stress, and systemic carnitine deficiency. Feeding an HFD induced hepatokines in mice, with a loss of hepatic fatty acid oxidation that enhanced systemic energy expenditure and suppressed adiposity. Additionally, the suppression in hepatic gluconeogenesis was sufficient to improve HFD-induced glucose intolerance. These data show that inhibiting hepatic fatty acid oxidation results in a systemic hormetic response that protects mice from HFD-induced obesity and glucose intolerance.


Assuntos
Carnitina O-Palmitoiltransferase/metabolismo , Ácidos Graxos/metabolismo , Intolerância à Glucose/enzimologia , Fígado/enzimologia , Mitocôndrias Hepáticas/enzimologia , Obesidade/enzimologia , Animais , Carnitina O-Palmitoiltransferase/deficiência , Carnitina O-Palmitoiltransferase/genética , Ácidos Graxos/genética , Intolerância à Glucose/genética , Intolerância à Glucose/patologia , Fígado/patologia , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/metabolismo , Erros Inatos do Metabolismo/patologia , Camundongos , Camundongos Knockout , Mitocôndrias Hepáticas/genética , Mitocôndrias Hepáticas/patologia , Obesidade/genética , Obesidade/patologia , Oxirredução
15.
Oncotarget ; 8(9): 15205-15212, 2017 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-28148899

RESUMO

AIM: Gestational diabetes mellitus (GDM) has been shown to be associated with a high risk of diabetes in offspring. In mitochondria, the inhibition of pyruvate dehydrogenase (PDH) activity by PDH phosphorylation is involved in the development of diabetes. We aimed to determine the role of PDH phosphorylation in the liver in GDM-induced offspring glucose intolerance. RESULTS: PDH phosphorylation was increased in lymphocytes from the umbilical cord blood of the GDM patients and in high glucose-treated hepatic cells. Both the male and female offspring from GDM mice had elevated liver weights and glucose intolerance. Further, PDH phosphorylation was increased in the livers of both the male and female offspring from GDM mice, and elevated acetylation may have contributed to this increased phosphorylation. MATERIALS AND METHODS: We obtained lymphocytes from umbilical cord blood collected from both normal and GDM pregnant women. In addition, we obtained the offspring of streptozotocin-induced GDM female pregnant mice. The glucose tolerance test was performed to assess glucose tolerance in the offspring. Further, Western blotting was conducted to detect changes in protein levels. CONCLUSIONS: Intrauterine hyperglycemia induced offspring glucose intolerance by inhibiting PDH activity, along with increased PDH phosphorylation in the liver, and this effect might be mediated by enhanced mitochondrial protein acetylation.


Assuntos
Diabetes Gestacional/fisiopatologia , Regulação Enzimológica da Expressão Gênica , Intolerância à Glucose/etiologia , Hiperglicemia/complicações , Cetona Oxirredutases/metabolismo , Animais , Animais Recém-Nascidos , Glicemia/metabolismo , Feminino , Intolerância à Glucose/enzimologia , Intolerância à Glucose/patologia , Teste de Tolerância a Glucose , Humanos , Masculino , Camundongos , Fosforilação , Gravidez , Piruvatos/metabolismo
16.
Surg Obes Relat Dis ; 12(8): 1523-1531, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27425837

RESUMO

BACKGROUND: Vertical sleeve gastrectomy (VSG) ameliorates metabolic complications in obese and diabetic patients through unknown mechanisms. OBJECTIVE: The objective of this study was to investigate the role of lipocalin-type prostaglandin D2 synthase (L-PGDS) in glucose regulation in response to VSG using L-PGDS knock-out (KO), knock-in (KI), and C57BL/6 (wild type) mice. SETTING: Winthrop University Hospital Research Institute. METHODS: Animals were divided into 6 groups: L-PGDS KO sham/VSG (n = 5), L-PGDS KI sham/VSG (n = 5), and C57BL/6 (wild type) sham/VSG (n = 5). Related parameters were measured in fasting animals after 10 weeks. RESULTS: Our intraperitoneal glucose tolerance tests and homeostatic model assessment insulin resistance results showed significant glycemic improvement 10 weeks post-VSG in both C57BL/6 and KI groups compared with the sham group. In contrast, the KO group developed glucose intolerance and insulin resistance similar to or greater than the sham group 10 weeks post-VSG. Interestingly, weight gain was insignificant 10 weeks post-VSG in all the groups and even trended higher in the KO group compared with sham. Peptide YY levels in the KO group post-VSG were slightly increased but significantly less than other groups. Similarly, the KO group showed significantly less leptin sensitivity in response to VSG compared with the KI group. Total cholesterol level remained unchanged in all groups irrespective of sham or surgery but interestingly, the KO group had significantly higher cholesterol levels. In parallel, adipocyte size was also found to be significantly increased in the KO group post-VSG compared with the sham group. CONCLUSION: Our findings propose that L-PGDS plays an important role in the beneficial metabolic effects observed after VSG.


Assuntos
Gastrectomia/métodos , Intolerância à Glucose/enzimologia , Oxirredutases Intramoleculares/fisiologia , Lipocalinas/fisiologia , Adipócitos/patologia , Análise de Variância , Animais , Cirurgia Bariátrica/métodos , Glicemia/metabolismo , Colesterol/metabolismo , Dieta Hiperlipídica , Jejum/sangue , Intolerância à Glucose/patologia , Intolerância à Glucose/cirurgia , Homeostase , Resistência à Insulina/fisiologia , Leptina/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Obesidade/enzimologia , Obesidade/patologia , Obesidade/cirurgia , Redução de Peso
17.
Nat Commun ; 7: 11869, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27323669

RESUMO

Receptor-interacting protein kinase 3 (RIPK3) mediates necroptosis, a form of programmed cell death that promotes inflammation in various pathological conditions, suggesting that it might be a privileged pharmacological target. However, its function in glucose homeostasis and obesity has been unknown. Here we show that RIPK3 is over expressed in the white adipose tissue (WAT) of obese mice fed with a choline-deficient high-fat diet. Genetic inactivation of Ripk3 promotes increased Caspase-8-dependent adipocyte apoptosis and WAT inflammation, associated with impaired insulin signalling in WAT as the basis for glucose intolerance. Similarly to mice, in visceral WAT of obese humans, RIPK3 is overexpressed and correlates with the body mass index and metabolic serum markers. Together, these findings provide evidence that RIPK3 in WAT maintains tissue homeostasis and suppresses inflammation and adipocyte apoptosis, suggesting that systemic targeting of necroptosis might be associated with the risk of promoting insulin resistance in obese patients.


Assuntos
Tecido Adiposo Branco/enzimologia , Deficiência de Colina/genética , Intolerância à Glucose/genética , Gordura Intra-Abdominal/enzimologia , Necrose/enzimologia , Obesidade/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Adipócitos/enzimologia , Adipócitos/patologia , Tecido Adiposo Branco/patologia , Animais , Apoptose/genética , Índice de Massa Corporal , Caspase 8/genética , Caspase 8/metabolismo , Colina/metabolismo , Deficiência de Colina/enzimologia , Deficiência de Colina/etiologia , Deficiência de Colina/patologia , Dieta Hiperlipídica , Regulação da Expressão Gênica , Intolerância à Glucose/enzimologia , Intolerância à Glucose/etiologia , Intolerância à Glucose/patologia , Homeostase , Humanos , Inflamação , Insulina/metabolismo , Resistência à Insulina , Gordura Intra-Abdominal/patologia , Masculino , Camundongos , Necrose/genética , Necrose/patologia , Obesidade/enzimologia , Obesidade/etiologia , Obesidade/patologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo
18.
Proc Natl Acad Sci U S A ; 113(20): 5754-9, 2016 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-27140617

RESUMO

Metabolic syndrome is characterized by insulin resistance, obesity, and dyslipidemia. It is the consequence of an imbalance between caloric intake and energy consumption. Adiponectin protects against metabolic syndrome. Insulin-induced signaling includes activation of PI3 kinase and protein kinase B (PKB)/Akt. PKB/Akt in turn inactivates glycogen synthase kinase (GSK) 3, a major regulator of metabolism. Here, we studied the significance of PI3K-dependent GSK3 inactivation for adiponectin formation in diet-induced metabolic syndrome. Mice expressing PI3K-insensitive GSK3 (gsk3(KI)) and wild-type mice (gsk3(WT)) were fed a high-fat diet. Compared with gsk3(WT) mice, gsk3(KI) mice were protected against the development of metabolic syndrome as evident from a markedly lower weight gain, lower total body and liver fat accumulation, better glucose tolerance, stronger hepatic insulin-dependent PKB/Akt phosphorylation, lower serum insulin, cholesterol, and triglyceride levels, as well as higher energy expenditure. Serum adiponectin concentration and the activity of transcription factor C/EBPα controlling the expression of adiponectin in adipose tissue was significantly higher in gsk3(KI) mice than in gsk3(WT) mice. Treatment with GSK3 inhibitor lithium significantly decreased the serum adiponectin concentration of gsk3(KI) mice and abrogated the difference in C/EBPα activity between the genotypes. Taken together, our data demonstrate that the expression of PI3K-insensitive GSK3 stimulates the production of adiponectin and protects from diet-induced metabolic syndrome.


Assuntos
Adiponectina/biossíntese , Quinase 3 da Glicogênio Sintase/fisiologia , Síndrome Metabólica/enzimologia , Tecido Adiposo/metabolismo , Animais , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Dieta Hiperlipídica/efeitos adversos , Intolerância à Glucose/enzimologia , Resistência à Insulina , Fígado/enzimologia , Masculino , Síndrome Metabólica/etiologia , Camundongos Transgênicos , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia
19.
Diabetes ; 65(5): 1268-82, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26861785

RESUMO

Encoding acyl-CoA thioesterase-7 (Acot7) is one of ∼60 genes expressed ubiquitously across tissues but relatively silenced, or disallowed, in pancreatic ß-cells. The capacity of ACOT7 to hydrolyze long-chain acyl-CoA esters suggests potential roles in ß-oxidation, lipid biosynthesis, signal transduction, or insulin exocytosis. We explored the physiological relevance of ß-cell-specific Acot7 silencing by re-expressing ACOT7 in these cells. ACOT7 overexpression in clonal MIN6 and INS1(832/13) ß-cells impaired insulin secretion in response to glucose plus fatty acids. Furthermore, in a panel of transgenic mouse lines, we demonstrate that overexpression of mitochondrial ACOT7 selectively in the adult ß-cell reduces glucose tolerance dose dependently and impairs glucose-stimulated insulin secretion. By contrast, depolarization-induced secretion was unaffected, arguing against a direct action on the exocytotic machinery. Acyl-CoA levels, ATP/ADP increases, membrane depolarization, and Ca(2+) fluxes were all markedly reduced in transgenic mouse islets, whereas glucose-induced oxygen consumption was unchanged. Although glucose-induced increases in ATP/ADP ratio were similarly lowered after ACOT7 overexpression in INS1(832/13) cells, changes in mitochondrial membrane potential were unaffected, consistent with an action of Acot7 to increase cellular ATP consumption. Because Acot7 mRNA levels are increased in human islets in type 2 diabetes, inhibition of the enzyme might provide a novel therapeutic strategy.


Assuntos
Regulação para Baixo , Ácidos Graxos não Esterificados/metabolismo , Regulação Enzimológica da Expressão Gênica , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Palmitoil-CoA Hidrolase/metabolismo , Animais , Sinalização do Cálcio , Linhagem Celular Tumoral , Células Clonais , Feminino , Intolerância à Glucose/enzimologia , Intolerância à Glucose/metabolismo , Intolerância à Glucose/patologia , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Especificidade de Órgãos , Palmitoil-CoA Hidrolase/genética , Ratos , Proteínas Recombinantes/metabolismo , Caracteres Sexuais , Técnicas de Cultura de Tecidos , Regulação para Cima
20.
Am J Physiol Heart Circ Physiol ; 308(12): H1530-9, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25888508

RESUMO

Diet-induced obesity deteriorates the recovery of cardiac function after ischemia-reperfusion (I/R) injury. While mechanistic target of rapamycin (mTOR) is a key mediator of energy metabolism, the effects of cardiac mTOR in ischemic injury under metabolic syndrome remains undefined. Using cardiac-specific transgenic mice overexpressing mTOR (mTOR-Tg mice), we studied the effect of mTOR on cardiac function in both ex vivo and in vivo models of I/R injury in high-fat diet (HFD)-induced obese mice. mTOR-Tg and wild-type (WT) mice were fed a HFD (60% fat by calories) for 12 wk. Glucose intolerance and insulin resistance induced by the HFD were comparable between WT HFD-fed and mTOR-Tg HFD-fed mice. Functional recovery after I/R in the ex vivo Langendorff perfusion model was significantly lower in HFD-fed mice than normal chow diet-fed mice. mTOR-Tg mice demonstrated better cardiac function recovery and had less of the necrotic markers creatine kinase and lactate dehydrogenase in both feeding conditions. Additionally, mTOR overexpression suppressed expression of proinflammatory cytokines, including IL-6 and TNF-α, in both feeding conditions after I/R injury. In vivo I/R models showed that at 1 wk after I/R, HFD-fed mice exhibited worse cardiac function and larger myocardial scarring along myofibers compared with normal chow diet-fed mice. In both feeding conditions, mTOR overexpression preserved cardiac function and prevented myocardial scarring. These findings suggest that cardiac mTOR overexpression is sufficient to prevent the detrimental effects of diet-induced obesity on the heart after I/R, by reducing cardiac dysfunction and myocardial scarring.


Assuntos
Dieta Hiperlipídica , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/enzimologia , Obesidade/complicações , Serina-Treonina Quinases TOR/metabolismo , Animais , Modelos Animais de Doenças , Intolerância à Glucose/enzimologia , Intolerância à Glucose/etiologia , Mediadores da Inflamação/metabolismo , Resistência à Insulina , Masculino , Camundongos Transgênicos , Contração Miocárdica , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/etiologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio/patologia , Necrose , Obesidade/sangue , Obesidade/fisiopatologia , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Fatores de Tempo , Função Ventricular Esquerda , Pressão Ventricular , Remodelação Ventricular , Aumento de Peso
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...