Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Commun Biol ; 3(1): 389, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32680996

ABSTRACT

Mitochondrial dysfunction contributes to cardiac pathologies. Barriers to new therapies include an incomplete understanding of underlying molecular culprits and a lack of effective mitochondria-targeted medicines. Here, we test the hypothesis that the cardiolipin-binding peptide elamipretide, a clinical-stage compound under investigation for diseases of mitochondrial dysfunction, mitigates impairments in mitochondrial structure-function observed after rat cardiac ischemia-reperfusion. Respirometry with permeabilized ventricular fibers indicates that ischemia-reperfusion induced decrements in the activity of complexes I, II, and IV are alleviated with elamipretide. Serial block face scanning electron microscopy used to create 3D reconstructions of cristae ultrastructure reveals that disease-induced fragmentation of cristae networks are improved with elamipretide. Mass spectrometry shows elamipretide did not protect against the reduction of cardiolipin concentration after ischemia-reperfusion. Finally, elamipretide improves biophysical properties of biomimetic membranes by aggregating cardiolipin. The data suggest mitochondrial structure-function are interdependent and demonstrate elamipretide targets mitochondrial membranes to sustain cristae networks and improve bioenergetic function.


Subject(s)
Cardiolipins/metabolism , Cardiotonic Agents/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Oligopeptides/therapeutic use , Animals , Hydrogen Peroxide/metabolism , Male , Mass Spectrometry , Microscopy, Electron, Transmission , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/ultrastructure , Rats , Rats, Sprague-Dawley
2.
J Cardiovasc Pharmacol Ther ; 19(1): 121-32, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24288396

ABSTRACT

We recently showed that Bendavia, a novel mitochondria-targeting peptide, reduced infarction and no-reflow across several experimental models. The purpose of this study was to determine the therapeutic timing and mechanism of action that underlie Bendavia's cytoprotective property. In rabbits exposed to in vivo ischemia/reperfusion (30/180 min), Bendavia administered 20 minutes prior to reperfusion (0.05 mg/kg/h, intravenously) reduced myocardial infarct size by ∼50% when administered for either 1 or 3 hours of reperfusion. However, when Bendavia perfusion began just 10 minutes after the onset of reperfusion, the protection against infarction and no-reflow was completely lost, indicating that the mechanism of protection is occurring early in reperfusion. Experiments in isolated mouse liver mitochondria found no discernible effect of Bendavia on blocking the permeability transition pore, and studies in isolated heart mitochondria showed no effect of Bendavia on respiratory rates. As Bendavia significantly lowered reactive oxygen species (ROS) levels in isolated heart mitochondria, the ROS-scavenging capacity of Bendavia was compared to well-known ROS scavengers using in vitro (cell-free) systems that enzymatically generate ROS. Across doses ranging from 1 nmol/L to 1 mmol/L, Bendavia showed no discernible ROS-scavenging properties, clearly differentiating itself from prototypical scavengers. In conclusion, Bendavia is a promising candidate to reduce cardiac injury when present at the onset of reperfusion but not after reperfusion has already commenced. Given that both infarction and no-reflow are related to increased cellular ROS, Bendavia's protective mechanism of action likely involves reduced ROS generation (as opposed to augmented scavenging) by endothelial and myocyte mitochondria.


Subject(s)
Mitochondria, Liver/drug effects , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/drug therapy , Oligopeptides/pharmacology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Free Radical Scavengers/administration & dosage , Free Radical Scavengers/pharmacology , Guinea Pigs , Male , Mice , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Liver/metabolism , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Permeability Transition Pore , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/physiopathology , Oligopeptides/administration & dosage , Rabbits , Reactive Oxygen Species/metabolism , Time Factors
3.
Am J Physiol Heart Circ Physiol ; 305(4): H477-83, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23771689

ABSTRACT

We have previously shown that myocardial infarct size in nonreperfused hearts of mice with a functional deletion of the circadian rhythm gene mPer2 (mPer2-M) was reduced by 43%. We hypothesized that acute ischemia-reperfusion injury (I/R = 30 min I/2 h R) would also be reduced in these mice and that ischemic preconditioning (IPC) (3 × 5 min cycles) before I/R, which enhances protection in wild-type (WT) hearts, would provide further protection in mPer2-M hearts. We observed a 69 and 75% decrease in infarct size in mPer2-M mouse hearts compared with WT following I/R and IPC, respectively. This was coincident with 67% less neutrophil infiltration and 57% less apoptotic cardiomyocytes. IPC in mPer2-M mice before I/R had 48% less neutrophil density and 46% less apoptosis than their WT counterparts. Macrophage density was not different between WT and mPer2-M I/R, but it was 45% higher in mPer2-M IPC mouse hearts compared with WT IPC. There were no baseline differences in cardiac mitochondrial function between WT and mPer2-M mice, but, following I/R, WT exhibited a marked decrease in maximal O2 consumption supported by complex I-mediated substrates, whereas mPer2-M did not, despite no difference in complex I content. Moreover, cardiac mitochondria from WT mice exhibited a very robust increase in ADP-stimulated O2 consumption in response to exogenously added cytochrome c, along with a high rate of reactive oxygen species production, none of which was exhibited by cardiac mitochondria from mPer2-M following I/R. Taken together, these findings suggest that mPer2 deletion preserves mitochondrial membrane structure and functional integrity in heart following I/R injury, the consequence of which is preservation of myocardial viability. Understanding the mechanisms connecting cardiac events, mitochondrial function, and mPer2 could lead to preventative and therapeutic strategies for at risk populations.


Subject(s)
Mitochondria, Heart/metabolism , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Period Circadian Proteins/metabolism , Adenosine Diphosphate/metabolism , Animals , Apoptosis , Biomarkers/metabolism , Blotting, Western , Disease Models, Animal , Female , Immunohistochemistry , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mitochondria, Heart/pathology , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/pathology , Mutation , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Neutrophil Infiltration , Oxidative Stress , Oxygen Consumption , Period Circadian Proteins/genetics , Reactive Oxygen Species/metabolism
5.
Cardiovasc Res ; 98(1): 47-55, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23341578

ABSTRACT

AIMS: We have previously shown that exercise leads to sustainable cardioprotection through a mechanism involving improved glutathione replenishment. This study was conducted to determine if redox-dependent modifications in glutathione reductase (GR) were involved in exercise cardioprotection. Furthermore, we sought to determine if reactive oxygen species generated by NADPH oxidase and/or mitochondria during exercise were triggering events for GR modulations. METHODS AND RESULTS: Rats were exercised for 10 consecutive days, after which isolated hearts were exposed to ischaemia/reperfusion (25 min/120 min). Exercise protected against infarction and arrhythmia, and preserved coronary flow. The GR inhibitor BCNU abolished the beneficial effects. GR activity was increased following exercise in a redox-dependent manner, with no change in GR protein levels. Because fluorescent labelling of GR protein thiols showed lower amounts of reduced thiols after exercise, we sought to determine the source of intracellular reactive oxygen species that may be activating GR. Subsets of animals were exercised immediately after treatment with either NADPH-oxidase inhibitors apocynin or Vas2870, or with mitoTEMPO or Bendavia, which reduce mitochondrial reactive oxygen species levels. The cardioprotective effects of exercise were abolished if animals exercised in the presence of NADPH oxidase inhibitors, in clear contrast to the mitochondrial reagents. These changes correlated with thiol-dependent modifications of GR. CONCLUSION: Adaptive cardioprotective signalling is triggered by reactive oxygen species from NADPH oxidase, and leads to improved glutathione replenishment through redox-dependent modifications in GR.


Subject(s)
Glutathione Reductase/physiology , Mitochondria/physiology , NADPH Oxidases/physiology , Physical Conditioning, Animal , Animals , Female , Glutathione/metabolism , Myocardial Reperfusion Injury/etiology , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
6.
J Mol Cell Cardiol ; 52(5): 1009-18, 2012 May.
Article in English | MEDLINE | ID: mdl-22406429

ABSTRACT

Mitochondria from diabetic hearts are sensitized to mitochondrial permeability transition pore (PTP) opening, which may be responsible for the increased propensity for cardiac injury in diabetic hearts. The purpose of this study was to determine if redox-dependent PTP opening contributes to augmented injury in diabetic hearts, and if compounds targeted at mitochondrial PTP, ROS, and calcium influx protected diabetic hearts from injury. Hearts from control or streptozotocin-induced diabetic rats were excised for either whole-heart or isolated mitochondria experiments. Myocardial glutathione content was oxidized in diabetic hearts when compared to control, and this translated to increased oxidation of the adenine nucleotide translocase in diabetic hearts. Diabetic mitochondria displayed significantly greater sensitivity to PTP opening than non-diabetic counterparts, which was reversed with the thiol-reducing agent dithiothreitol. The thiol-oxidant diamide increased calcium sensitivity in control, but not diabetic mitochondria. Diabetic animals treated with the mitochondria-targeted ROS suppressing peptide MTP-131 also showed improved resistance to PTP opening. In separate experiments hearts underwent ex vivo ischemia/reperfusion (IR). Diabetic hearts were more susceptible to IR injury, with infarct sizes of 60 ± 4% of the area-at-risk (vs. 46 ± 2% in non-diabetics; P<0.05). Administration of the PTP blocker NIM811 (5 µM), MTP-131 (1 nM) or the mitochondrial calcium uniporter blocker minocycline (1 µM) at the onset of reperfusion reduced infarct sizes in both control and diabetic hearts. These findings suggest that augmented susceptibility to injury in the diabetic heart is mediated by redox-dependent shifts in PTP opening, and that three novel mitochondria-targeted agents administered at reperfusion may be suitable adjuvant reperfusion therapies to attenuate injury in diabetic patients.


Subject(s)
Calcium/metabolism , Diabetes Mellitus, Experimental/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Myocardial Reperfusion Injury/metabolism , Oligopeptides/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Signaling , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Cyclosporine/pharmacology , Cyclosporine/therapeutic use , Diabetes Mellitus, Experimental/complications , Glutathione/metabolism , Glutathione Disulfide/metabolism , Heart/drug effects , Heart/physiopathology , Hemodynamics , In Vitro Techniques , Male , Minocycline/pharmacology , Minocycline/therapeutic use , Mitochondria, Heart/drug effects , Mitochondrial ADP, ATP Translocases/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Permeability Transition Pore , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Oligopeptides/therapeutic use , Oxidation-Reduction , Permeability , Rats , Rats, Sprague-Dawley
7.
J Clin Endocrinol Metab ; 97(3): E329-40, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22238402

ABSTRACT

CONTEXT: Indications of adipose tissue dysfunction correlate with systemic insulin resistance and type 2 diabetes. It has been suggested that a defect in adipose tissue turnover may be involved in the development of these disorders. Whether this dysfunction causes or exacerbates systemic insulin resistance is not fully understood. OBJECTIVES, PARTICIPANTS, AND MEASURES: We tested whether the expression of members of the mitogenic ErbB family was reduced in adipose tissue of insulin-resistant individuals and whether ErbB1 and ErbB2 were involved in adipogenesis. Thirty-two women covering a wide range of body mass index values and insulin sensitivity participated in the cross-sectional portion of this study. We also studied preadipocytes isolated from 12 insulin-sensitive individuals to evaluate the impact of ErbB1 or ErbB2 inhibition on adipogenesis in vitro. For this purpose, we measured phospho-ErbB1 and phospho-ErbB2 levels using ELISA and the expression of peroxisome proliferator-activated receptor γ (PPARγ) and PPARγ-regulated genes by real-time PCR. RESULTS: Among the ErbB family members, only ErbB1 expression was correlated with insulin sensitivity. Additionally, ErbB1 levels correlated positively with PPARγ and several PPARγ-regulated genes including acyl-coenzyme A synthetase long-chain family member 1 (ACSL1), adiponectin, adipose tissue triacylglycerol lipase (ATGL), diacylglycerol acyl transferase 1 (DGAT1), glycerol-3-phosphate dehydrogenase 1 (GPD1), and lipoprotein lipase (LPL), but negatively with CD36 and fatty acid-binding protein 4 (FABP4). In preadipocyte culture, ErbB1, but not ErbB2, inhibition was associated with a reduction in the expression of all the above-mentioned genes. CONCLUSIONS: These findings demonstrate a key role for ErbB1 in adipogenesis and suggest that lower ErbB1 protein abundance may lead to adipose tissue dysfunction.


Subject(s)
Adipose Tissue/metabolism , Diabetes Mellitus, Type 2/metabolism , ErbB Receptors/metabolism , Insulin Resistance/physiology , Adipocytes/cytology , Adipocytes/metabolism , Adult , Body Mass Index , Cells, Cultured , Cross-Sectional Studies , Diabetes Mellitus, Type 2/genetics , ErbB Receptors/genetics , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Female , Humans , Insulin/metabolism , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Middle Aged , PPAR gamma/genetics , PPAR gamma/metabolism , Phosphorylation
8.
J Sex Med ; 6 Suppl 3: 269-78, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19267849

ABSTRACT

INTRODUCTION: The combination of independent risk factors for erectile dysfunction, obesity, hypertension, and diabetes are collectively manifested in a condition known as metabolic syndrome X (MSX). However, the regulatory mechanisms responsible for the erectile dysfunction (ED) are not fully understood. Clinical studies suggest that a pleiotropic effect of statin's ability to enhance vascular relaxation might be through an impact on nitric oxide signaling or through a regulation of RhoA activation. AIM: We hypothesized that regulatory aspects of short-term statin therapy involve the alteration of the RhoA/Rho-kinase signaling cascade and will reverse the ED seen in a rat model of MSX. MAIN OUTCOME MEASURES: The magnitude and sensitivity of the voltage-dependent maintenance of intracavernosal blood pressure and mean arterial blood pressure. These responses were correlated with tissue protein and mRNA expression levels of RhoA and Rho kinases. METHODS: Erectile function was evaluated by assessing voltage-dependent stimulation of the cavernosal nerve in 16-20 weeks old lean and obese-diabetic Zucker rats treated with 5 mg/kg/day of rosuvastatin intraperitoneally for 3 days. Cavernosal tissue RhoA and Rho-kinases expression levels were evaluated by real-time reverse transcriptase-polymerase chain reaction, Western blot. RESULTS: The voltage-dependent erectile responses were suppressed by >30% in the obese-diabetic Zucker rat. The 3-day treatment with rosuvastatin partially restored the erectile response. The Rho-kinase inhibitor, H-1152, dose dependently increased the erectile responses and shifted the voltage sensitivity with statin treatment. Analysis of protein expression levels suggested elevation of RhoA and Rho kinases in obese-diabetics and statin treatment lowering Rho-kinase II. The RhoA and Rho-kinase II mRNA levels were significantly reduced in the rosuvastatin-treated obese-diabetic animals. CONCLUSIONS: These results support a hypothesis that short-term statin therapy may lower RhoA/Rho-kinase expression levels and improve cavernosal blood pressure response to Rho-kinase inhibition and voltage-stimulation, and reversing an augmented vasoconstricted state associated with diabetes and/or hypertension in MSX.


Subject(s)
Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Diabetes Mellitus, Experimental/epidemiology , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , Erectile Dysfunction , Fluorobenzenes/pharmacology , Fluorobenzenes/therapeutic use , Impotence, Vasculogenic/etiology , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Metabolic Syndrome/epidemiology , Nitric Oxide/metabolism , Obesity/epidemiology , Phosphodiesterase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , rho-Associated Kinases/metabolism , Animals , Anticholesteremic Agents/administration & dosage , Blotting, Western , Enzyme Inhibitors/administration & dosage , Erectile Dysfunction/drug therapy , Erectile Dysfunction/epidemiology , Erectile Dysfunction/physiopathology , Fluorobenzenes/administration & dosage , Impotence, Vasculogenic/enzymology , Male , Phosphodiesterase Inhibitors/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Rats , Rats, Zucker , Rosuvastatin Calcium , Sulfonamides/administration & dosage
9.
J Lipid Res ; 50(1): 59-70, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18698091

ABSTRACT

Vascular dysfunction in response to reactive oxygen species (ROS) plays an important role in the development and progression of atherosclerotic lesions. In most cells, mitochondria are the major source of cellular ROS during aerobic respiration. Under most conditions the rates of ROS formation and elimination are balanced through mechanisms that sense relative ROS levels. However, a chronic imbalance in redox homeostasis is believed to contribute to various chronic diseases, including atherosclerosis. Uncoupling protein-2 (UCP2) is a mitochondrial inner membrane protein shown to be a negative regulator of macrophage ROS production. In response to a cholesterol-containing atherogenic diet, C57BL/6J mice significantly increased expression of UCP2 in the aorta, while mice lacking UCP2, in the absence of any other genetic modification, displayed significant endothelial dysfunction following the atherogenic diet. Compared with wild-type mice, Ucp2(-/-) mice had decreased endothelial nitric oxide synthase, an increase in vascular cell adhesion molecule-1 expression, increased ROS production, and an impaired ability to increase total antioxidant capacity. These changes in Ucp2(-/-) mice were associated with increased aortic macrophage infiltration and more numerous and larger atherosclerotic lesions. These data establish that in the vasculature UCP2 functions as an adaptive antioxidant defense to protect against the development of atherosclerosis in response to a fat and cholesterol diet.


Subject(s)
Antioxidants/metabolism , Atherosclerosis/metabolism , Ion Channels/genetics , Ion Channels/physiology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , Animals , Aorta/metabolism , Diet , Female , Homeostasis , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Oxidation-Reduction , Reactive Oxygen Species , Uncoupling Protein 2
10.
J Biol Chem ; 283(18): 12056-63, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18347011

ABSTRACT

In this study, we tested the hypothesis that human neutrophil alpha-defensins (HNPs) inhibit hepatic glucose production through a signaling pathway distinct from insulin. The effect of HNP-1 on fasting blood glucose levels and the expression of hepatic gluconeogenic genes was first examined. Using hyperinsulinemic-euglycemic clamps, we determined the effect of HNP-1 on endogenous glucose production, hepatic expression of key gluconeogenic genes and glucose uptake in skeletal muscle in Zucker diabetic fatty rats. In isolated primary hepatocytes, we studied the effect of HNP-1 and -2 on glucose production, expression of gluconeogenic genes, and phosphorylation of Akt, c-Src, and FoxO1. Our results show that HNP-1 reduced blood glucose levels of both normal mice and Zucker diabetic fatty rats predominantly through suppression of hepatic glucose production. HNPs inhibited glycogenolysis and gluconeogenesis in isolated hepatocytes. HNPs also suppressed expression of key gluconeogenic genes including phosphoenoylpyruvate carboxyl kinase and glucose-6-phosphatase. To investigate the mechanism, we found that HNPs stimulated phosphorylation of Akt and FoxO1 without activating IRS1. Nevertheless, HNPs activated c-Src. Blockade of c-Src activity with either a chemical inhibitor PP2 or an alternative inhibitor CSK prevented the inhibitory effect of HNPs on gluconeogenesis. Together, our results support the hypothesis that HNPs can suppress hepatic glucose production through an intracellular mechanism distinct from the classical insulin signaling pathway.


Subject(s)
Glucose/biosynthesis , Insulin/metabolism , Liver/drug effects , Liver/metabolism , Signal Transduction/drug effects , alpha-Defensins/pharmacology , Animals , Blood Glucose/metabolism , Cell Line, Tumor , Cell Separation , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gluconeogenesis/drug effects , Gluconeogenesis/genetics , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Mice , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Rats , Rats, Zucker , Transcription, Genetic/drug effects
11.
J Biol Chem ; 282(19): 14205-12, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17384440

ABSTRACT

Free fatty acid (FFA) is believed to be a major environmental factor linking obesity to Type II diabetes. We have recently reported that FFA can induce gluconeogenesis in hepatocytes through p38 mitogen-activated protein kinase (p38). In this study, we have investigated the role of p38 in oleate-induced hepatic insulin resistance. Our results show that a prolonged treatment of primary hepatocytes with oleate blunted insulin suppression of hepatic gluconeogenesis, and decreased insulin-induced phosphorylation of Akt in a p38-dependent manner. Reduction of the insulin-induced Akt phosphorylation by oleate correlated with activation of p38. In the presence of p38 inhibition, prolonged exposure of hepatocytes to oleate failed to reduce insulin-stimulated phosphorylation of Akt. An siRNA against p38alpha prevented oleate suppression of the insulin-induced phosphorylation of Akt. Furthermore, a prolonged exposure of hepatocytes to oleate decreased insulin-induced tyrosine phosphorylation of IRS1/2, while slightly increasing serine phosphorylation of IRS. The decrease of insulin-stimulated tyrosine phosphorylation of IRS1/2 in hepatocytes by oleate was reversed by the inhibition of p38. We further show that a prolonged exposure of primary hepatocytes to oleate elevated the protein level of the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) gene in a p38-dependent manner, but had no effect on the mRNA level of PTEN. Knocking down the PTEN gene prevented oleate to inhibit insulin activation of Akt and insulin suppression of gluconeogenesis. Together, results from this study demonstrate a critical role for p38 in oleate-induced hepatic insulin resistance.


Subject(s)
Hepatocytes/drug effects , Hypoglycemic Agents/pharmacology , Insulin Resistance , Insulin/pharmacology , Oleic Acid/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis , Blotting, Western , Caspase 3/metabolism , Gluconeogenesis , Hepatocytes/metabolism , Immunoprecipitation , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tyrosine/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
12.
J Biol Chem ; 281(49): 37794-802, 2006 Dec 08.
Article in English | MEDLINE | ID: mdl-17032647

ABSTRACT

Catecholamine-stimulated lipolysis is primarily a beta-adrenergic and cAMP-dependent event. In previous studies we established that the beta(3)-adrenergic receptor (beta(3)AR) in adipocytes utilizes a unique mechanism to stimulate extracellular signal-regulated kinases 1 and 2 (ERK) by direct recruitment and activation of Src kinase. Therefore, we investigated the role of the ERK pathway in adipocyte metabolism and found that the beta(3)AR agonist CL316,243 regulates lipolysis through both cAMP-dependent protein kinase (PKA) and ERK. Inhibition of PKA activity completely eliminated lipolysis at low (subnanomolar) CL316,243 concentrations and by 75-80% at higher nanomolar concentrations. The remaining 20-25% of PKA-independent lipolysis, as well as ERK activation, was abolished by inhibiting the activity of either Src (PP2 or small interfering RNA), epidermal growth factor receptor (EGFR with AG1478 or small interfering RNA), or mitogen-activated protein kinase kinase 1 or 2 (MKK1/2 with PD098059). PD098059 inhibited lipolysis by 53% in mice as well. Finally, the effect of estradiol, a reported acute activator of ERK and lipolysis, was also totally prevented by PP2, AG1478, and PD098059. These results suggest that ERK activation by beta(3)AR depends upon Src and epidermal growth factor receptor kinase activities and is responsible for the PKA-independent portion of the lipolytic response. Together these results illustrate the distinct and complementary roles for PKA and ERK in catecholamine-stimulated lipolysis.


Subject(s)
ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Lipolysis/physiology , Receptors, Adrenergic, beta-3/metabolism , src-Family Kinases/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adrenergic beta-3 Receptor Agonists , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Dioxoles/pharmacology , Enzyme Activation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Estradiol/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Isoquinolines/pharmacology , Lipolysis/drug effects , Mice , Quinazolines , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Sulfonamides/pharmacology , Tyrphostins/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
13.
J Virol ; 80(14): 6745-56, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16809280

ABSTRACT

The genetic diversity among globally circulating human immunodeficiency virus type 1 (HIV-1) strains is a serious challenge for HIV-1 vaccine design. We have generated a synthetic group M consensus env gene (CON6) for induction of cross-subtype immune responses and report here a comparative study of T-cell responses to this and natural strain env immunogens in a murine model. Three different strains of mice were immunized with CON6 as well as subtype A, B, or C env immunogens, using a DNA prime-recombinant vaccinia virus boost strategy. T-cell epitopes were mapped by gamma interferon enzyme-linked immunospot analysis using five overlapping Env peptide sets from heterologous subtype A, B, and C viruses. The CON6-derived vaccine was immunogenic and induced a greater number of T-cell epitope responses than any single wild-type subtype A, B, and C env immunogen and similar T-cell responses to a polyvalent vaccine. The responses were comparable to within-clade responses but significantly more than between-clade responses. The magnitude of the T-cell responses induced by CON6 (measured by individual epitope peptides) was also greater than the magnitude of responses induced by individual wild-type env immunogens. Though the limited major histocompatibility complex repertoire in inbred mice does not necessarily predict responses in nonhuman primates and humans, these results suggest that synthetic centralized env immunogens represent a promising approach for HIV-1 vaccine design that merits further characterization.


Subject(s)
AIDS Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , Gene Products, env/immunology , HIV Antigens/immunology , HIV Infections/immunology , HIV-1/immunology , T-Lymphocytes/immunology , Vaccines, DNA/immunology , AIDS Vaccines/genetics , AIDS Vaccines/pharmacology , Animals , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/pharmacology , Gene Products, env/genetics , Gene Products, env/pharmacology , Genetic Variation , HIV Antigens/genetics , HIV Antigens/pharmacology , HIV Infections/genetics , HIV Infections/prevention & control , HIV-1/genetics , Histocompatibility Antigens/immunology , Humans , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , Peptides/genetics , Peptides/immunology , Peptides/pharmacology , Species Specificity , Vaccines, DNA/genetics , Vaccines, DNA/pharmacology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/pharmacology
14.
Mol Cell Biol ; 25(13): 5466-79, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15964803

ABSTRACT

The sympathetic nervous system regulates the activity and expression of uncoupling protein 1 (UCP1) through the three beta-adrenergic receptor subtypes and their ability to raise intracellular cyclic AMP (cAMP) levels. Unexpectedly, we recently discovered that the cAMP-dependent regulation of multiple genes in brown adipocytes, including Ucp1, occurred through the p38 mitogen-activated protein kinases (MAPK) (W. Cao, K. W. Daniel, J. Robidoux, P. Puigserver, A. V. Medvedev, X. Bai, L. M. Floering, B. M. Spiegelman, and S. Collins, Mol. Cell. Biol. 24:3057-3067, 2004). However, no well-defined pathway linking cAMP accumulation or cAMP-dependent protein kinase (PKA) to p38 MAPK has been described. Therefore, in the present study using both in vivo and in vitro models, we have initiated a retrograde approach to define the required components, beginning with the p38 MAPK isoforms themselves and the MAP kinase kinase(s) that regulates them. Our strategy included ectopic expression of wild-type and mutant kinases as well as targeted inhibition of gene expression using small interfering RNA. The results indicate that the beta-adrenergic receptors and PKA lead to a highly selective activation of the p38alpha isoform of MAPK, which in turn promotes Ucp1 gene transcription. In addition, this specific activation of p38alpha relies solely on the presence of MAP kinase kinase 3, despite the expression in brown fat of MKK3, -4, and -6. Finally, of the three scaffold proteins of the JIP family expressed in brown adipocytes, only JIP2 co-immunoprecipitates p38alpha MAPK and MKK3. Therefore, in the brown adipocyte the recently described scaffold protein JIP2 assembles the required factors MKK3 and p38alpha MAPK linking PKA to the control of thermogenic gene expression.


Subject(s)
Adipocytes/metabolism , Carrier Proteins/metabolism , Cyclic AMP/metabolism , Gene Expression Regulation , MAP Kinase Kinase 3/metabolism , Membrane Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adipocytes/cytology , Animals , Blotting, Western , Cell Line , Enzyme Activation , Genes, Reporter , Ion Channels , Isoenzymes/metabolism , Luciferases/metabolism , MAP Kinase Kinase 3/genetics , Mice , Mice, Inbred Strains , Mitochondrial Proteins , Mutation , Phosphorylation , Precipitin Tests , Thermogenesis/physiology , Uncoupling Protein 1 , p38 Mitogen-Activated Protein Kinases/analysis , p38 Mitogen-Activated Protein Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL