Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Lipid Res ; 60(3): 528-538, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30610082

RESUMO

During prolonged fasting, the liver plays a central role in maintaining systemic energy homeostasis by producing glucose and ketones in processes fueled by oxidation of fatty acids liberated from adipose tissue. In mice, this is accompanied by transient hepatic accumulation of glycerolipids. We found that the hepatic expression of monoacylglycerol acyltransferase 1 (Mogat1), an enzyme with monoacylglycerol acyltransferase (MGAT) activity that produces diacyl-glycerol from monoacylglycerol, was significantly increased in the liver of fasted mice compared with mice given ad libitum access to food. Basal and fasting-induced expression of Mogat1 was markedly diminished in the liver of mice lacking the transcription factor PPARα. Suppressing Mogat1 expression in liver and adipose tissue with antisense oligonucleotides (ASOs) reduced hepatic MGAT activity and triglyceride content compared with fasted controls. Surprisingly, the expression of many other PPARα target genes and PPARα activity was also decreased in mice given Mogat1 ASOs. When mice treated with control or Mogat1 ASOs were gavaged with the PPARα ligand, WY-14643, and then fasted for 18 h, WY-14643 administration reversed the effects of Mogat1 ASOs on PPARα target gene expression and liver triglyceride content. In conclusion, Mogat1 is a fasting-induced PPARα target gene that may feed forward to regulate liver PPARα activity during food deprivation.


Assuntos
Jejum , Privação de Alimentos , Fígado/enzimologia , N-Acetilglucosaminiltransferases/metabolismo , Tecido Adiposo/metabolismo , Animais , Regulação Enzimológica da Expressão Gênica , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , N-Acetilglucosaminiltransferases/deficiência , N-Acetilglucosaminiltransferases/genética , PPAR alfa/genética , Fatores de Tempo , Triglicerídeos/metabolismo
2.
Liver Transpl ; 24(7): 908-921, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29729104

RESUMO

The prevalence of obesity-associated nonalcoholic fatty liver disease has significantly increased over the past decade, and end-stage liver disease secondary to nonalcoholic steatohepatitis has become 1 of the most common indications for liver transplantation. This both increases the demand for organs and decreases the availability of donor livers deemed suitable for transplantation. Although in the past many steatotic livers were discarded due to concerns over enhanced susceptibility to ischemia/reperfusion injury (IRI) and organ failure, the discrepancy between supply and demand has resulted in increasing use of expanded criteria donor organs including steatotic livers. However, it remains controversial whether steatotic livers can be safely used for transplantation and how best to improve the performance of steatotic grafts. We aimed to evaluate the impact of diet-induced hepatic steatosis in a murine model of IRI. Using a diet of high trans-fat, fructose, and cholesterol (HTF-C) and a diet high in saturated fats, sucrose, and cholesterol (Western diet), we were able to establish models of mixed macrovesicular and microvesicular steatosis (HTF-C) and microvesicular steatosis (Western). We found that the presence of hepatic steatosis, whether it is predominantly macrovesicular or microvesicular, significantly worsens IRI as measured by plasma alanine aminotransferase levels and inflammatory cytokine concentration, and histological evaluation for necrosis. Additionally, we report on a novel finding in which hepatic IRI in the setting of steatosis results in the induction of the necroptosis factors, receptor interacting protein kinase (RIPK) 3, RIPK1, and mixed-lineage kinase domain-like. These data lay the groundwork for additional experimentation to test potential therapeutic approaches to limit IRI in steatotic livers by using a genetically tractable system. Liver Transplantation 24 908-921 2018 AASLD.


Assuntos
Transplante de Fígado/efeitos adversos , Fígado/patologia , Hepatopatia Gordurosa não Alcoólica/patologia , Obesidade/patologia , Traumatismo por Reperfusão/patologia , Animais , Dieta Ocidental/efeitos adversos , Modelos Animais de Doenças , Humanos , Fígado/irrigação sanguínea , Fígado/cirurgia , Testes de Função Hepática , Transplante de Fígado/normas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/etiologia , Obesidade/etiologia , Traumatismo por Reperfusão/etiologia , Coleta de Tecidos e Órgãos/normas
3.
Proc Natl Acad Sci U S A ; 110(2): 642-7, 2013 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-23267081

RESUMO

Lipin 1 is a coregulator of DNA-bound transcription factors and a phosphatidic acid (PA) phosphatase (PAP) enzyme that catalyzes a critical step in the synthesis of glycerophospholipids. Lipin 1 is highly expressed in adipocytes, and constitutive loss of lipin 1 blocks adipocyte differentiation; however, the effects of Lpin1 deficiency in differentiated adipocytes are unknown. Here we report that adipocyte-specific Lpin1 gene recombination unexpectedly resulted in expression of a truncated lipin 1 protein lacking PAP activity but retaining transcriptional regulatory function. Loss of lipin 1-mediated PAP activity in adipocytes led to reduced glyceride synthesis and increased PA content. Characterization of the deficient mice also revealed that lipin 1 normally modulates cAMP-dependent signaling through protein kinase A to control lipolysis by metabolizing PA, which is an allosteric activator of phosphodiesterase 4 and the molecular target of rapamycin. Consistent with these findings, lipin 1 expression was significantly related to adipose tissue lipolytic rates and protein kinase A signaling in adipose tissue of obese human subjects. Taken together, our findings identify lipin 1 as a reciprocal regulator of triglyceride synthesis and hydrolysis in adipocytes, and suggest that regulation of lipolysis by lipin 1 is mediated by PA-dependent modulation of phosphodiesterase 4.


Assuntos
Adipócitos/enzimologia , Redes e Vias Metabólicas/fisiologia , Proteínas Nucleares/genética , Obesidade/fisiopatologia , Fosfatidato Fosfatase/genética , Ácidos Fosfatídicos/metabolismo , Células 3T3-L1 , Alelos , Animais , Western Blotting , Clonagem Molecular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Primers do DNA/genética , Feminino , Cromatografia Gasosa-Espectrometria de Massas , Glicerídeos/biossíntese , Humanos , Lipólise/genética , Lipólise/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Nucleares/deficiência , Proteínas Nucleares/metabolismo , Obesidade/enzimologia , Fosfatidato Fosfatase/deficiência , Fosfatidato Fosfatase/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
4.
J Biol Chem ; 287(28): 23537-48, 2012 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-22621923

RESUMO

Currently approved thiazolidinediones (TZDs) are effective insulin-sensitizing drugs that may have efficacy for treatment of a variety of metabolic and inflammatory diseases, but their use is limited by side effects that are mediated through ectopic activation of the peroxisome proliferator-activated receptor γ (PPARγ). Emerging evidence suggests that the potent anti-diabetic efficacy of TZDs can be separated from the ability to serve as ligands for PPARγ. A novel TZD analog (MSDC-0602) with very low affinity for binding and activation of PPARγ was evaluated for its effects on insulin resistance in obese mice. MSDC-0602 treatment markedly improved several measures of multiorgan insulin sensitivity, adipose tissue inflammation, and hepatic metabolic derangements, including suppressing hepatic lipogenesis and gluconeogenesis. These beneficial effects were mediated at least in part via direct actions on hepatocytes and were preserved in hepatocytes from liver-specific PPARγ(-/-) mice, indicating that PPARγ was not required to suppress these pathways. In conclusion, the beneficial pharmacology exhibited by MSDC-0602 on insulin sensitivity suggests that PPARγ-sparing TZDs are effective for treatment of type 2 diabetes with reduced risk of PPARγ-mediated side effects.


Assuntos
Resistência à Insulina , Obesidade/prevenção & controle , PPAR gama/metabolismo , Tiazolidinedionas/farmacologia , Células 3T3-L1 , Animais , Ligação Competitiva , Células Cultivadas , Feminino , Expressão Gênica/efeitos dos fármacos , Glicólise/genética , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Hipoglicemiantes/química , Hipoglicemiantes/metabolismo , Hipoglicemiantes/farmacologia , Lipogênese/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Estrutura Molecular , Obesidade/genética , Obesidade/metabolismo , PPAR gama/genética , Pioglitazona , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rosiglitazona , Tiazolidinedionas/química , Tiazolidinedionas/metabolismo
5.
J Biol Chem ; 286(13): 11155-62, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21321124

RESUMO

Diabetic cardiac dysfunction is associated with decreased rates of myocardial glucose oxidation (GO) and increased fatty acid oxidation (FAO), a fuel shift that has been shown to sensitize the heart to ischemic insult and ventricular dysfunction. We sought to evaluate the metabolic and functional consequences of chronic suppression of GO in heart as modeled by transgenic mice with cardiac-specific overexpression of pyruvate dehydrogenase kinase 4 (myosin heavy chain (MHC)-PDK4 mice), an inhibitor of pyruvate dehydrogenase. Hearts of MHC-PDK4 mice were shown to exhibit an insulin-resistant substrate utilization profile, characterized by low GO rates and high FAO flux. Surprisingly, MHC-PDK4 mice were not sensitized to cardiac ischemia-reperfusion injury despite a fuel utilization pattern that phenocopied the diabetic heart. In addition, MHC-PDK4 mice were protected against high fat diet-induced myocyte lipid accumulation, likely related to increased capacity for FAO. The high rates of mitochondrial FAO in the MHC-PDK4 heart were related to heightened activity of the AMP-activated protein kinase, reduced levels of malonyl-CoA, and increased capacity for mitochondrial uncoupled respiration. The expression of the known AMP-activated protein kinase target, peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), a master regulator of mitochondrial function and biogenesis, was also activated in the MHC-PDK4 heart. These results demonstrate that chronic activation of PDK4 triggers transcriptional and post-transcriptional mechanisms that re-program the heart for chronic high rates of FAO without the expected deleterious functional or metabolic consequences.


Assuntos
Cardiomiopatias Diabéticas/enzimologia , Glucose/metabolismo , Mitocôndrias Cardíacas/enzimologia , Miocárdio/enzimologia , Proteínas Serina-Treonina Quinases/biossíntese , Animais , Cardiomiopatias Diabéticas/genética , Modelos Animais de Doenças , Glucose/genética , Frequência Cardíaca/genética , Resistência à Insulina/genética , Camundongos , Camundongos Transgênicos , Mitocôndrias Cardíacas/genética , Isquemia Miocárdica/enzimologia , Isquemia Miocárdica/genética , Miocárdio/patologia , Cadeias Pesadas de Miosina/genética , Oxirredução , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Proteínas Serina-Treonina Quinases/genética , Piruvato Desidrogenase Quinase de Transferência de Acetil , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição , Disfunção Ventricular/enzimologia , Disfunção Ventricular/genética
6.
JCI Insight ; 6(9)2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-33986192

RESUMO

Lipin 1 is a bifunctional protein that is a transcriptional regulator and has phosphatidic acid (PA) phosphohydrolase activity, which dephosphorylates PA to generate diacylglycerol. Human lipin 1 mutations lead to episodic rhabdomyolysis, and some affected patients exhibit cardiac abnormalities, including exercise-induced cardiac dysfunction and cardiac triglyceride accumulation. Furthermore, lipin 1 expression is deactivated in failing heart, but the effects of lipin 1 deactivation in myocardium are incompletely understood. We generated mice with cardiac-specific lipin 1 KO (cs-Lpin1-/-) to examine the intrinsic effects of lipin 1 in the myocardium. Cs-Lpin1-/- mice had normal systolic cardiac function but mild cardiac hypertrophy. Compared with littermate control mice, PA content was higher in cs-Lpin1-/- hearts, which also had an unexpected increase in diacylglycerol and triglyceride content. Cs-Lpin1-/- mice exhibited diminished cardiac cardiolipin content and impaired mitochondrial respiration rates when provided with pyruvate or succinate as metabolic substrates. After transverse aortic constriction-induced pressure overload, loss of lipin 1 did not exacerbate cardiac hypertrophy or dysfunction. However, loss of lipin 1 dampened the cardiac ionotropic response to dobutamine and exercise endurance in association with reduced protein kinase A signaling. These data suggest that loss of lipin 1 impairs cardiac functional reserve, likely due to effects on glycerolipid homeostasis, mitochondrial function, and protein kinase A signaling.


Assuntos
Cardiomegalia/genética , Modelos Animais de Doenças , Tolerância ao Exercício/genética , Camundongos , Mitocôndrias Cardíacas/metabolismo , Contração Miocárdica/genética , Miocárdio/metabolismo , Fosfatidato Fosfatase/genética , Animais , Cardiolipinas/metabolismo , Cardiomegalia/metabolismo , Cardiotônicos/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Diglicerídeos/metabolismo , Dobutamina/farmacologia , Tolerância ao Exercício/efeitos dos fármacos , Camundongos Knockout , Contração Miocárdica/efeitos dos fármacos , Ácidos Fosfatídicos/metabolismo , Ácido Pirúvico/metabolismo , Ácido Succínico/metabolismo , Triglicerídeos/metabolismo
7.
J Biol Chem ; 284(40): 27402-8, 2009 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-19648647

RESUMO

Proinflammatory cytokines induce nitric oxide-dependent DNA damage and ultimately beta-cell death. Not only does nitric oxide cause beta-cell damage, it also activates a functional repair process. In this study, the mechanisms activated by nitric oxide that facilitate the repair of damaged beta-cell DNA are examined. JNK plays a central regulatory role because inhibition of this kinase attenuates the repair of nitric oxide-induced DNA damage. p53 is a logical target of JNK-dependent DNA repair; however, nitric oxide does not stimulate p53 activation or accumulation in beta-cells. Further, knockdown of basal p53 levels does not affect DNA repair. In contrast, expression of growth arrest and DNA damage (GADD) 45alpha, a DNA repair gene that can be regulated by p53-dependent and p53-independent pathways, is stimulated by nitric oxide in a JNK-dependent manner, and knockdown of GADD45alpha expression attenuates the repair of nitric oxide-induced beta-cell DNA damage. These findings show that beta-cells have the ability to repair nitric oxide-damaged DNA and that JNK and GADD45alpha mediate the p53-independent repair of this DNA damage.


Assuntos
Proteínas de Ciclo Celular/genética , Dano ao DNA/efeitos dos fármacos , Reparo do DNA , Regulação da Expressão Gênica , Células Secretoras de Insulina/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Óxido Nítrico/farmacologia , Proteínas Nucleares/genética , Animais , Células Cultivadas , Ativação Enzimática , Feminino , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/enzimologia , Ratos , Ratos Sprague-Dawley , Proteína Supressora de Tumor p53/metabolismo
8.
Am J Physiol Endocrinol Metab ; 297(5): E1187-96, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19738038

RESUMO

For many cell types, including pancreatic ß-cells, nitric oxide is a mediator of cell death; however, it is paradoxical that for a given cell type nitric oxide can induce both necrosis and apoptosis. This report tests the hypothesis that cell death mediated by nitric oxide shifts from an early necrotic to a late apoptotic event. Central to this transition is the ability of ß-cells to respond and repair nitric oxide-mediated damage. ß-Cells have the ability to repair DNA that is damaged following 24-h incubation with IL-1; however, cytokine-induced DNA damage becomes irreversible following 36-h incubation. This irreversible DNA damage following 36-h incubation with IL-1 correlates with the activation of caspase-3 (cleavage and activity). The increase in caspase activity correlates with reductions in endogenous nitric oxide production, as nitric oxide is an inhibitor of caspase activity. In contrast, caspase cleavage or activation is not observed under conditions in which ß-cells are capable of repairing damaged DNA (24-h incubation with cytokines). These findings provide evidence that ß-cell death in response to cytokines shifts from an early necrotic process to apoptosis and that this shift is associated with irreversible DNA damage and caspase-3 activation.


Assuntos
Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Citocinas/farmacologia , Dano ao DNA/fisiologia , Células Secretoras de Insulina/efeitos dos fármacos , Óxido Nítrico/farmacologia , Animais , Caspase 3/metabolismo , Separação Celular , Ensaio Cometa , Reparo do DNA/efeitos dos fármacos , Metabolismo Energético/fisiologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Interleucina-1/antagonistas & inibidores , Interleucina-1/farmacologia , Masculino , Necrose , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real
9.
Diabetes ; 68(5): 918-931, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30833469

RESUMO

Low 25-hydroxyvitamin D levels correlate with the prevalence of diabetes; however, the mechanisms remain uncertain. Here, we show that nutritional deprivation-responsive mechanisms regulate vitamin D metabolism. Both fasting and diabetes suppressed hepatic cytochrome P450 (CYP) 2R1, the main vitamin D 25-hydroxylase responsible for the first bioactivation step. Overexpression of coactivator peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α), induced physiologically by fasting and pathologically in diabetes, resulted in dramatic downregulation of CYP2R1 in mouse hepatocytes in an estrogen-related receptor α (ERRα)-dependent manner. However, PGC-1α knockout did not prevent fasting-induced suppression of CYP2R1 in the liver, indicating that additional factors contribute to the CYP2R1 repression. Furthermore, glucocorticoid receptor (GR) activation repressed the liver CYP2R1, suggesting GR involvement in the regulation of CYP2R1. GR antagonist mifepristone partially prevented CYP2R1 repression during fasting, suggesting that glucocorticoids and GR contribute to the CYP2R1 repression during fasting. Moreover, fasting upregulated the vitamin D catabolizing CYP24A1 in the kidney through the PGC-1α-ERRα pathway. Our study uncovers a molecular mechanism for vitamin D deficiency in diabetes and reveals a novel negative feedback mechanism that controls crosstalk between energy homeostasis and the vitamin D pathway.


Assuntos
Diabetes Mellitus/metabolismo , Jejum/sangue , Fatores de Transcrição/sangue , Fatores de Transcrição/metabolismo , Deficiência de Vitamina D/metabolismo , Vitamina D/sangue , Vitamina D/metabolismo , Animais , Colestanotriol 26-Mono-Oxigenase/metabolismo , Diabetes Mellitus/sangue , Jejum/fisiologia , Fígado/metabolismo , Camundongos , Mifepristona/farmacologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Glucocorticoides/antagonistas & inibidores , Receptores de Glucocorticoides/metabolismo , Deficiência de Vitamina D/sangue , Receptor ERRalfa Relacionado ao Estrogênio
10.
J Neurochem ; 107(6): 1753-65, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19014370

RESUMO

GTP binding regulatory protein (G protein)-coupled receptors can activate MAPK pathways via G protein-dependent and -independent mechanisms. However, the physiological outcomes correlated with the cellular signaling events are not as well characterized. In this study, we examine the involvement of G protein and beta-arrestin 2 pathways in kappa opioid receptor-induced, extracellular signal-regulated kinase 1/2 (ERK1/2)-mediated proliferation of both immortalized and primary astrocyte cultures. As different agonists induce different cellular signaling pathways, we tested the prototypic kappa agonist, U69593 as well as the structurally distinct, non-nitrogenous agonist, C(2)-methoxymethyl salvinorin B (MOM-Sal-B). In immortalized astrocytes, U69593, activated ERK1/2 by a rapid (min) initial stimulation that was sustained over 2 h and increased proliferation. Sequestration of activated Gbetagamma subunits attenuated U69593 stimulation of ERK1/2 and suppressed proliferation in these cells. Furthermore, small interfering RNA silencing of beta-arrestin 2 diminished sustained ERK activation induced by U69593. In contrast, MOM-Sal-B induced only the early phase of ERK1/2 phosphorylation and did not affect proliferation of immortalized astrocytes. In primary astrocytes, U69593 produced the same effects as seen in immortalized astrocytes. MOM-Sal-B elicited sustained ERK1/2 activation which was correlated with increased primary astrocyte proliferation. Proliferative actions of both agonists were abolished by either inhibition of ERK1/2, Gbetagamma subunits or beta-arrestin 2, suggesting that both G protein-dependent and -independent ERK pathways are required for this outcome.


Assuntos
Arrestinas/metabolismo , Astrócitos/fisiologia , Proliferação de Células , Proteínas de Ligação ao GTP/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Receptores Opioides kappa/fisiologia , Analgésicos/farmacologia , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Benzenoacetamidas/farmacologia , Bromodesoxiuridina/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Inibidores Enzimáticos/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Toxina Pertussis/farmacologia , Pirrolidinas/farmacologia , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inibidores , Fatores de Tempo , Transfecção/métodos , beta-Arrestina 2 , beta-Arrestinas
11.
PLoS Med ; 3(2): e17, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16354107

RESUMO

BACKGROUND: There are at least two phases of beta-cell death during the development of autoimmune diabetes: an initiation event that results in the release of beta-cell-specific antigens, and a second, antigen-driven event in which beta-cell death is mediated by the actions of T lymphocytes. In this report, the mechanisms by which the macrophage-derived cytokine interleukin (IL)-1 induces beta-cell death are examined. IL-1, known to inhibit glucose-induced insulin secretion by stimulating inducible nitric oxide synthase expression and increased production of nitric oxide by beta-cells, also induces beta-cell death. METHODS AND FINDINGS: To ascertain the mechanisms of cell death, the effects of IL-1 and known activators of apoptosis on beta-cell viability were examined. While IL-1 stimulates beta-cell DNA damage, this cytokine fails to activate caspase-3 or to induce phosphatidylserine (PS) externalization; however, apoptosis inducers activate caspase-3 and the externalization of PS on beta-cells. In contrast, IL-1 stimulates the release of the immunological adjuvant high mobility group box 1 protein (HMGB1; a biochemical maker of necrosis) in a nitric oxide-dependent manner, while apoptosis inducers fail to stimulate HMGB1 release. The release of HMGB1 by beta-cells treated with IL-1 is not sensitive to caspase-3 inhibition, while inhibition of this caspase attenuates beta-cell death in response to known inducers of apoptosis. CONCLUSIONS: These findings indicate that IL-1 induces beta-cell necrosis and support the hypothesis that macrophage-derived cytokines may participate in the initial stages of diabetes development by inducing beta-cell death by a mechanism that promotes antigen release (necrosis) and islet inflammation (HMGB1 release).


Assuntos
Apoptose , Diabetes Mellitus Tipo 1/fisiopatologia , Proteína HMGB1/metabolismo , Células Secretoras de Insulina/patologia , Interleucina-1/fisiologia , Óxido Nítrico/fisiologia , Animais , Caspase 3 , Caspases/metabolismo , Técnicas de Cultura de Células , Dano ao DNA , Diabetes Mellitus Tipo 1/imunologia , Ativação Enzimática , Humanos , Inflamação , Insulinoma/patologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/patologia , Macrófagos/imunologia , Necrose , Ratos , Ratos Sprague-Dawley
12.
JCI Insight ; 1(4): e86038, 2016 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-27158672

RESUMO

To elucidate the mechanisms for reverse LV remodeling, we generated a conditional (doxycycline [dox] off) transgenic mouse tetracycline transactivating factor-TRAF2 (tTA-TRAF2) that develops a dilated heart failure (HF) phenotype upon expression of a proinflammatory transgene, TNF receptor-associated factor 2 (TRAF2), and complete normalization of LV structure and function when the transgene is suppressed. tTA-TRAF2 mice developed a significant increase in LV dimension with decreased contractile function, which was completely normalized in the tTA-TRAF2 mice fed dox for 4 weeks (tTA-TRAF2dox4W). Normalization of LV structure and function was accompanied by partial normalization (~60%) of gene expression associated with incident HF. Similar findings were observed in patients with dilated cardiomyopathy who underwent reverse LV remodeling following mechanical circulatory support. Persistence of the HF gene program was associated with an exaggerated hypertrophic response and increased mortality in tTA-TRAF2dox4W mice following transaortic constriction (TAC). These effects were no longer observed following TAC in tTA-TRAF2dox8W, wherein there was a more complete (88%) reversal of the incident HF genes. These results demonstrate that reverse LV remodeling is associated with improvements in cardiac myocyte biology; however, the persistence of the abnormal HF gene program may be maladaptive following perturbations in hemodynamic loading conditions.

13.
Diabetes ; 63(7): 2284-96, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24595352

RESUMO

Monoacylglycerol acyltransferase (MGAT) enzymes convert monoacylglycerol to diacylglycerol (DAG), a lipid that has been linked to the development of hepatic insulin resistance through activation of protein kinase C (PKC). The expression of genes that encode MGAT enzymes is induced in the livers of insulin-resistant human subjects with nonalcoholic fatty liver disease, but whether MGAT activation is causal of hepatic steatosis or insulin resistance is unknown. We show that the expression of Mogat1, which encodes MGAT1, and MGAT activity are also increased in diet-induced obese (DIO) and ob/obmice. To probe the metabolic effects of MGAT1 in the livers of obese mice, we administered antisense oligonucleotides (ASOs) against Mogat1 to DIO and ob/ob mice for 3 weeks. Knockdown of Mogat1 in liver, which reduced hepatic MGAT activity, did not affect hepatic triacylglycerol content and unexpectedly increased total DAG content. Mogat1 inhibition also increased both membrane and cytosolic compartment DAG levels. However, Mogat1 ASO treatment significantly improved glucose tolerance and hepatic insulin signaling in obese mice. In summary, inactivation of hepatic MGAT activity, which is markedly increased in obese mice, improved glucose tolerance and hepatic insulin signaling independent of changes in body weight, intrahepatic DAG and TAG content, and PKC signaling.


Assuntos
Aciltransferases/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Fígado/efeitos dos fármacos , Obesidade/metabolismo , Aciltransferases/antagonistas & inibidores , Aciltransferases/genética , Animais , Dieta Hiperlipídica , Diglicerídeos/metabolismo , Expressão Gênica/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Camundongos Transgênicos , Obesidade/etiologia , Obesidade/genética , Oligonucleotídeos Antissenso/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Triglicerídeos/metabolismo
14.
Cell Rep ; 7(6): 2042-2053, 2014 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-24910426

RESUMO

Carrier-facilitated pyruvate transport across the inner mitochondrial membrane plays an essential role in anabolic and catabolic intermediary metabolism. Mitochondrial pyruvate carrier 2 (Mpc2) is believed to be a component of the complex that facilitates mitochondrial pyruvate import. Complete MPC2 deficiency resulted in embryonic lethality in mice. However, a second mouse line expressing an N-terminal truncated MPC2 protein (Mpc2(Δ16)) was viable but exhibited a reduced capacity for mitochondrial pyruvate oxidation. Metabolic studies demonstrated exaggerated blood lactate concentrations after pyruvate, glucose, or insulin challenge in Mpc2(Δ16) mice. Additionally, compared with wild-type controls, Mpc2(Δ16) mice exhibited normal insulin sensitivity but elevated blood glucose after bolus pyruvate or glucose injection. This was attributable to reduced glucose-stimulated insulin secretion and was corrected by sulfonylurea KATP channel inhibitor administration. Collectively, these data are consistent with a role for MPC2 in mitochondrial pyruvate import and suggest that Mpc2 deficiency results in defective pancreatic ß cell glucose sensing.


Assuntos
Glucose/farmacologia , Insulina/metabolismo , Proteínas de Membrana Transportadoras/deficiência , Animais , Proteínas de Transporte de Ânions , Feminino , Glucose/metabolismo , Secreção de Insulina , Ácido Láctico/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas de Transporte da Membrana Mitocondrial , Transportadores de Ácidos Monocarboxílicos , Taxa Secretória/efeitos dos fármacos
15.
Mol Metab ; 2(3): 194-204, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24049734

RESUMO

Peroxisome proliferator-activated receptorγ coactivators (PGC-1α and PGC-1ß) play important roles in the transcriptional regulation of intermediary metabolism. To evaluate the effects of overexpressing PGC-1α or PGC-1ß at physiologic levels in liver, we generated transgenic mice with inducible overexpression of PGC-1α or PGC-1ß. Gene expression array profiling revealed that whereas both PGC-1 family proteins induced mitochondrial oxidative enzymes, the expression of several genes involved in converting glucose to fatty acid was induced by PGC-1ß, but not PGC-1α. The increased expression of enzymes involved in carbohydrate utilization and de novo lipogenesis by PGC-1ß required carbohydrate response element binding protein (ChREBP). The interaction between PGC-1ß and ChREBP, as well as PGC-1ß occupancy of the liver-type pyruvate kinase promoter, was influenced by glucose concentration and liver-specific PGC-1ß(-/-) hepatocytes were refractory to the lipogenic response to high glucose conditions. These data suggest that PGC-1ß-mediated coactivation of ChREBP is involved in the lipogenic response to hyperglycemia.

16.
Biol Sex Differ ; 3(1): 11, 2012 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-22620287

RESUMO

BACKGROUND: Although there appear to be no differences in muscle protein turnover in young and middle aged men and women, we have reported significant differences in the rate of muscle protein synthesis between older adult men and women. This suggests that aging may affect muscle protein turnover differently in men and women. METHODS: We measured the skeletal muscle protein fractional synthesis rate (FSR) by using stable isotope-labeled tracer methods during basal postabsorptive conditions and during a hyperaminoacidemic-hyperinsulinemic-euglycemic clamp in eight young men (25-45 y), ten young women (25-45 y), ten old men (65-85 y) and ten old women (65-85 y). RESULTS: The basal muscle protein FSR was not different in young and old men (0.040 ± 0.004 and 0.043 ± 0.005%·h-1, respectively) and combined insulin, glucose and amino acid infusion significantly increased the muscle protein FSR both in young (to 0.063 ± 0.006%·h-1) and old (to 0.051 ± 0.008%·h-1) men but the increase (0.023 ± 0.004 vs. 0.009 ± 0.004%·h-1, respectively) was ~60% less in the old men (P = 0.03). In contrast, the basal muscle protein FSR was ~30% greater in old than young women (0.060 ± 0.003 vs. 0.046 ± 0.004%·h-1, respectively; P < 0.05) and combined insulin, glucose and amino acid infusion significantly increased the muscle protein FSR in young (P < 0.01) but not in old women (P = 0.10) so that the FSR was not different between young and old women during the clamp (0.074 ± 0.006%·h-1 vs. 0.072 ± 0.006%·h-1, respectively). CONCLUSIONS: There is sexual dimorphism in the age-related changes in muscle protein synthesis and thus the metabolic processes responsible for the age-related decline in muscle mass.

17.
J Clin Invest ; 122(12): 4667-74, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23187122

RESUMO

Bariatric surgery in obese patients is a highly effective method of preventing or resolving type 2 diabetes mellitus (T2DM); however, the remission rate is not the same among different surgical procedures. We compared the effects of 20% weight loss induced by laparoscopic adjustable gastric banding (LAGB) or Roux-en-Y gastric bypass (RYGB) surgery on the metabolic response to a mixed meal, insulin sensitivity, and ß cell function in nondiabetic obese adults. The metabolic response to meal ingestion was markedly different after RYGB than after LAGB surgery, manifested by rapid delivery of ingested glucose into the systemic circulation, by an increase in the dynamic insulin secretion rate, and by large, early postprandial increases in plasma glucose, insulin, and glucagon-like peptide-1 concentrations in the RYGB group. However, the improvement in oral glucose tolerance, insulin sensitivity, and overall ß cell function after weight loss were not different between surgical groups. Additionally, both surgical procedures resulted in a similar decrease in adipose tissue markers of inflammation. We conclude that marked weight loss itself is primarily responsible for the therapeutic effects of RYGB and LAGB on insulin sensitivity, ß cell function, and oral glucose tolerance in nondiabetic obese adults.


Assuntos
Derivação Gástrica , Gastroplastia , Resistência à Insulina , Células Secretoras de Insulina/fisiologia , Obesidade/cirurgia , Adulto , Área Sob a Curva , Composição Corporal , Antígeno CD11b/metabolismo , Proteínas de Ligação ao Cálcio , Ceramidas/metabolismo , Citocinas/metabolismo , Diglicerídeos/metabolismo , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia , Masculino , Glicoproteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Mucinas/metabolismo , Músculo Esquelético/metabolismo , Obesidade/metabolismo , Obesidade/patologia , Tamanho do Órgão , Período Pós-Prandial , Receptores Acoplados a Proteínas G/metabolismo , Redução de Peso
18.
PLoS One ; 7(12): e52645, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23285128

RESUMO

PGC-1ß plays pleiotropic roles in regulating intermediary metabolism and has been shown to regulate both catabolic and anabolic processes in liver. We sought to evaluate the effects of PGC-1ß on liver energy metabolism by generating mice with postnatal, liver-specific deletion of PGC-1ß (LS-PGC-1ß(-/-) mice). LS-PGC-1ß(-/-) mice were outwardly normal, but exhibited a significant increase in hepatic triglyceride content at 6 weeks of age. Hepatic steatosis was due, at least in part, to impaired capacity for fatty acid oxidation and marked mitochondrial dysfunction. Mitochondrial DNA content and the expression of genes encoding multiple steps in mitochondrial fatty acid oxidation and oxidative phosphorylation pathways were significantly diminished in LS-PGC-1ß(-/-) mice. Liquid chromatography mass spectrometry-based analyses also revealed that acetylcarnitine and butyrylcarnitine levels were depleted whereas palmitoylcarnitine content was increased in LS-PGC-1ß(-/-) liver, which is consistent with attenuated rates of fatty acid oxidation. Interestingly, loss of PGC-1ß also significantly impaired inducible expression of glycolytic and lipogenic enzymes that occurs with high carbohydrate diet refeeding after a prolonged fast. These results suggest that PGC-1ß plays dual roles in regulating hepatic fatty acid metabolism by controlling the expression of programs of genes involved in both fatty acid oxidation and de novo fatty acid synthesis.


Assuntos
Jejum/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Mitocôndrias Hepáticas/metabolismo , Transativadores/deficiência , Animais , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Metabolismo Energético , Feminino , Deleção de Genes , Expressão Gênica , Glicólise , Camundongos , Camundongos Knockout , Mitocôndrias Hepáticas/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Transativadores/genética , Fatores de Transcrição , Triglicerídeos/metabolismo
19.
Am J Physiol Endocrinol Metab ; 296(4): E672-80, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19158322

RESUMO

To test the hypothesis that somatic mitochondrial (mt)DNA mutation accumulation predisposes mice to beta-cell loss and diabetes development, transgenic mice expressing a proofreading-deficient mtDNA polymerase-gamma under the control of the rat insulin-1 promoter were generated. At 6 wk of age, mtDNA mutations reached 0.01% (1.05 mutations/10,000 bp) in islets isolated from transgenic mice. This mutational burden is associated with impaired glucose tolerance and a diabetes prevalence of 52% in male transgenic mice. Female transgenic mice maintain slightly elevated fasting glucose levels, mild glucose intolerance, and a diabetes prevalence of 14%. Diabetes in transgenic animals is associated with insulin insufficiency that results from a significant reduction in beta-cell mass. Importantly, apoptosis of beta-cells is increased 7-fold in female and 11-fold in male transgenic mice compared with littermate controls. These results are consistent with a causative role of somatic mtDNA mutation accumulation in the loss of beta-cell mass and diabetes development.


Assuntos
Apoptose/genética , DNA Mitocondrial/genética , Diabetes Mellitus/genética , Células Secretoras de Insulina/fisiologia , Mutação/fisiologia , Animais , DNA Polimerase gama , DNA Mitocondrial/metabolismo , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , DNA Polimerase Dirigida por DNA/fisiologia , Diabetes Mellitus/fisiopatologia , Feminino , Glucose/metabolismo , Homeostase/genética , Insulina/genética , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Tamanho do Órgão , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/fisiologia , Transgenes/fisiologia
20.
Diabetes ; 57(1): 124-32, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17928398

RESUMO

OBJECTIVE: The unfolded protein response (UPR) is a conserved cellular response designed to alleviate damage and promote survival of cells experiencing stress; however, prolonged UPR activation can result in apoptotic cell death. The UPR, activated by cytokine-induced nitric oxide (NO) production, has been proposed to mediate beta-cell death in response to cytokines. In this study, the role of UPR activation in cytokine-induced beta-cell death was examined. RESEARCH DESIGN AND METHODS: The effects of cytokine treatment of rat and human islets and RINm5F cells on UPR activation, NO production, and cell viability were examined using molecular and biochemical methodologies. RESULTS: UPR activation correlates with beta-cell death in interleukin (IL)-1-treated rat islets. NO mediates both cytokine-induced UPR activation and beta-cell death as NO synthase inhibitors attenuate each of these IL-1-stimulated events. Importantly, cytokines and tunicamycin, a classical UPR activator, induce beta-cell death by different mechanisms. Cell death in response to the classical UPR activator is associated with a 2.5-fold increase in caspase-3 activity, while IL-1 fails to stimulate caspase-3 activity. In addition, cell death is enhanced by approximately 35% in tunicamycin-treated cells expressing an S51A eIF2 alpha mutant that cannot be phosphorylated or in cells lacking PERK (protein kinase regulated by RNA/endoplasmic reticulum-like kinase). In contrast, neither the absence of PERK nor the expression of the S51A eIF2 alpha mutant affects the levels of cytokine-induced death. CONCLUSIONS: While cytokine-induced beta-cell death temporally correlates with UPR activation, the lack of caspase activity and the ability of NO to attenuate caspase activity suggest that prolonged UPR activation does not mediate cytokine-induced beta-cell death.


Assuntos
Morte Celular/efeitos dos fármacos , Citocinas/farmacologia , Células Secretoras de Insulina/fisiologia , Óxido Nítrico/fisiologia , Desnaturação Proteica , Processamento Alternativo , Animais , Caspase 3/metabolismo , Técnicas de Cultura de Células , Morte Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Perfilação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Insulinoma , Masculino , Nitritos/metabolismo , Proteínas Nucleares/genética , Neoplasias Pancreáticas , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Fatores de Transcrição de Fator Regulador X , Fatores de Transcrição , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA