Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
FASEB J ; 28(1): 316-26, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24076965

ABSTRACT

Mitochondrial fission, regulated by dynamin-related protein-1 (Drp1), is a newly recognized determinant of mitochondrial function, but its contribution to left ventricular (LV) impairment following ischemia-reperfusion (IR) injury is unknown. We report that Drp1 activation during IR results in LV dysfunction and that Drp1 inhibition is beneficial. In both isolated neonatal murine cardiomyocytes and adult rat hearts (Langendorff preparation) mitochondrial fragmentation and swelling occurred within 30 min of IR. Drp1-S637 (serine 637) dephosphorylation resulted in Drp1 mitochondrial translocation and increased mitochondrial fission. The Drp1 inhibitor Mdivi-1 preserved mitochondrial morphology, reduced cytosolic calcium, and prevented cell death. Drp1 siRNA similarly preserved mitochondrial morphology. In Langendorff hearts, Mdivi-1 reduced mitochondrial reactive oxygen species, improved LV developed pressure (92±5 vs. 28±10 mmHg, P<0.001), and lowered LV end diastolic pressure (10±1 vs. 86±13 mmHg, P<0.001) following IR. Mdivi-1 was protective if administered prior to or following ischemia. Because Drp1-S637 dephosphorylation is calcineurin sensitive, we assessed the effects of a calcineurin inhibitor, FK506. FK506 treatment prior to IR prevented Drp1-S637 dephosphorylation and preserved cardiac function. Likewise, therapeutic hypothermia (30°C) inhibited Drp1-S637 dephosphorylation and preserved mitochondrial morphology and myocardial function. Drp1 inhibition is a novel strategy to improve myocardial function following IR.


Subject(s)
Dynamins/metabolism , Myocardial Reperfusion Injury/drug therapy , Animals , Calcineurin Inhibitors , Cells, Cultured , Dynamins/genetics , Immunoblotting , Immunosuppressive Agents/pharmacology , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mitochondrial Dynamics/drug effects , Mitochondrial Dynamics/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxygen Consumption/drug effects , Oxygen Consumption/genetics , Quinazolinones/pharmacology , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Tacrolimus/pharmacology
2.
Am J Respir Crit Care Med ; 187(8): 865-78, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23449689

ABSTRACT

RATIONALE: Pulmonary arterial hypertension (PAH) is a lethal, female-predominant, vascular disease. Pathologic changes in PA smooth muscle cells (PASMC) include excessive proliferation, apoptosis-resistance, and mitochondrial fragmentation. Activation of dynamin-related protein increases mitotic fission and promotes this proliferation-apoptosis imbalance. The contribution of decreased fusion and reduced mitofusin-2 (MFN2) expression to PAH is unknown. OBJECTIVES: We hypothesize that decreased MFN2 expression promotes mitochondrial fragmentation, increases proliferation, and impairs apoptosis. The role of MFN2's transcriptional coactivator, peroxisome proliferator-activated receptor γ coactivator 1-α (PGC1α), was assessed. MFN2 therapy was tested in PAH PASMC and in models of PAH. METHODS: Fusion and fission mediators were measured in lungs and PASMC from patients with PAH and female rats with monocrotaline or chronic hypoxia+Sugen-5416 (CH+SU) PAH. The effects of adenoviral mitofusin-2 (Ad-MFN2) overexpression were measured in vitro and in vivo. MEASUREMENTS AND MAIN RESULTS: In normal PASMC, siMFN2 reduced expression of MFN2 and PGC1α; conversely, siPGC1α reduced PGC1α and MFN2 expression. Both interventions caused mitochondrial fragmentation. siMFN2 increased proliferation. In rodent and human PAH PASMC, MFN2 and PGC1α were decreased and mitochondria were fragmented. Ad-MFN2 increased fusion, reduced proliferation, and increased apoptosis in human PAH and CH+SU. In CH+SU, Ad-MFN2 improved walking distance (381 ± 35 vs. 245 ± 39 m; P < 0.05); decreased pulmonary vascular resistance (0.18 ± 0.02 vs. 0.38 ± 0.14 mm Hg/ml/min; P < 0.05); and decreased PA medial thickness (14.5 ± 0.8 vs. 19 ± 1.7%; P < 0.05). Lung vascularity was increased by MFN2. CONCLUSIONS: Decreased expression of MFN2 and PGC1α contribute to mitochondrial fragmentation and a proliferation-apoptosis imbalance in human and experimental PAH. Augmenting MFN2 has therapeutic benefit in human and experimental PAH.


Subject(s)
GTP Phosphohydrolases/deficiency , Heat-Shock Proteins/deficiency , Hypertension, Pulmonary/physiopathology , Mitochondrial Dynamics/physiology , Mitochondrial Proteins/deficiency , Transcription Factors/deficiency , Animals , Apoptosis/physiology , Cell Proliferation/drug effects , Disease Models, Animal , Exercise Tolerance/drug effects , Familial Primary Pulmonary Hypertension , Female , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Lung/cytology , Lung/pathology , Membrane Proteins/administration & dosage , Membrane Proteins/deficiency , Mitochondrial Dynamics/genetics , Mitochondrial Proteins/administration & dosage , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/physiology , Optic Atrophy, Autosomal Dominant/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Rats, Sprague-Dawley
3.
Circ Res ; 112(5): 802-15, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23334860

ABSTRACT

RATIONALE: Closure of the ductus arteriosus (DA) is essential for the transition from fetal to neonatal patterns of circulation. Initial PO2-dependent vasoconstriction causes functional DA closure within minutes. Within days a fibrogenic, proliferative mechanism causes anatomic closure. Though modulated by endothelial-derived vasodilators and constrictors, O2 sensing is intrinsic to ductal smooth muscle cells and oxygen-induced DA constriction persists in the absence of endothelium, endothelin, and cyclooxygenase mediators. O2 increases mitochondrial-derived H2O2, which constricts ductal smooth muscle cells by raising intracellular calcium and activating rho kinase. However, the mechanism by which oxygen changes mitochondrial function is unknown. OBJECTIVE: The purpose of this study was to determine whether mitochondrial fission is crucial for O2-induced DA constriction and closure. METHODS AND RESULTS: Using DA harvested from 30 term infants during correction of congenital heart disease, as well as DA from term rabbits, we demonstrate that mitochondrial fission is crucial for O2-induced constriction and closure. O2 rapidly (<5 minutes) causes mitochondrial fission by a cyclin-dependent kinase- mediated phosphorylation of dynamin-related protein 1 (Drp1) at serine 616. Fission triggers a metabolic shift in the ductal smooth muscle cells that activates pyruvate dehydrogenase and increases mitochondrial H2O2 production. Subsequently, fission increases complex I activity. Mitochondrial-targeted catalase overexpression eliminates PO2-induced increases in mitochondrial-derived H2O2 and cytosolic calcium. The small molecule Drp1 inhibitor, Mdivi-1, and siDRP1 yield concordant results, inhibiting O2-induced constriction (without altering the response to phenylephrine or KCl) and preventing O2-induced increases in oxidative metabolism, cytosolic calcium, and ductal smooth muscle cells proliferation. Prolonged Drp1 inhibition reduces DA closure in a tissue culture model. CONCLUSIONS: Mitochondrial fission is an obligatory, early step in mammalian O2 sensing and offers a promising target for modulating DA patency.


Subject(s)
Ductus Arteriosus/physiology , GTP Phosphohydrolases/physiology , Microtubule-Associated Proteins/physiology , Mitochondrial Dynamics/physiology , Mitochondrial Proteins/physiology , Muscle, Smooth, Vascular/physiology , Oxygen/physiology , Vasoconstriction/physiology , Animals , Animals, Newborn , Calcium/metabolism , Cell Proliferation , Cells, Cultured , Ductus Arteriosus/cytology , Dynamins , Female , Humans , Hydrogen Peroxide/metabolism , Infant, Newborn , Male , Mitochondria/metabolism , Models, Animal , Muscle, Smooth, Vascular/cytology , Oxygen Consumption/physiology , Rabbits , Tissue Culture Techniques , rho-Associated Kinases/metabolism
4.
J Mol Med (Berl) ; 91(3): 333-46, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23247844

ABSTRACT

Pyruvate dehydrogenase kinase (PDK) is activated in right ventricular hypertrophy (RVH), causing an increase in glycolysis relative to glucose oxidation that impairs right ventricular function. The stimulus for PDK upregulation, its isoform specificity, and the long-term effects of PDK inhibition are unknown. We hypothesize that FOXO1-mediated PDK4 upregulation causes bioenergetic impairment and RV dysfunction, which can be reversed by dichloroacetate. Adult male Fawn-Hooded rats (FHR) with pulmonary arterial hypertension (PAH) and right ventricular hypertrophy (RVH; age 6-12 months) were compared to age-matched controls. Glucose oxidation (GO) and fatty acid oxidation (FAO) were measured at baseline and after acute dichloroacetate (1 mM × 40 min) in isolated working hearts and in freshly dispersed RV myocytes. The effects of chronic dichloroacetate (0.75 g/L drinking water for 6 months) on cardiac output (CO) and exercise capacity were measured in vivo. Expression of PDK4 and its regulatory transcription factor, FOXO1, were also measured in FHR and RV specimens from PAH patients (n = 10). Microarray analysis of 168 genes related to glucose or FA metabolism showed >4-fold upregulation of PDK4, aldolase B, and acyl-coenzyme A oxidase. FOXO1 was increased in FHR RV, whereas HIF-1 α was unaltered. PDK4 expression was increased, and the inactivated form of FOXO1 decreased in human PAH RV (P < 0.01). Pyruvate dehydrogenase (PDH) inhibition in RVH increased proton production and reduced GO's contribution to the tricarboxylic acid (TCA) cycle. Acutely, dichloroacetate reduced RV proton production and increased GO's contribution (relative to FAO) to the TCA cycle and ATP production in FHR (P < 0.01). Chronically dichloroacetate decreased PDK4 and FOXO1, thereby activating PDH and increasing GO in FHR. These metabolic changes increased CO (84 ± 14 vs. 69 ± 14 ml/min, P < 0.05) and treadmill-walking distance (239 ± 20 vs. 171 ± 22 m, P < 0.05). Chronic dichloroacetate inhibits FOXO1-induced PDK4 upregulation and restores GO, leading to improved bioenergetics and RV function in RVH.


Subject(s)
Dichloroacetic Acid/administration & dosage , Forkhead Transcription Factors/metabolism , Glucose/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Forkhead Box Protein O1 , Gene Expression Regulation/drug effects , Humans , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Oxidation-Reduction/drug effects , Physical Conditioning, Animal , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Rats , Up-Regulation/drug effects , Ventricular Dysfunction, Right/metabolism , Ventricular Dysfunction, Right/physiopathology
5.
Circ Res ; 110(11): 1484-97, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22511751

ABSTRACT

RATIONALE: Pulmonary arterial hypertension (PAH) is a lethal syndrome characterized by pulmonary vascular obstruction caused, in part, by pulmonary artery smooth muscle cell (PASMC) hyperproliferation. Mitochondrial fragmentation and normoxic activation of hypoxia-inducible factor-1α (HIF-1α) have been observed in PAH PASMCs; however, their relationship and relevance to the development of PAH are unknown. Dynamin-related protein-1 (DRP1) is a GTPase that, when activated by kinases that phosphorylate serine 616, causes mitochondrial fission. It is, however, unknown whether mitochondrial fission is a prerequisite for proliferation. OBJECTIVE: We hypothesize that DRP1 activation is responsible for increased mitochondrial fission in PAH PASMCs and that DRP1 inhibition may slow proliferation and have therapeutic potential. METHODS AND RESULTS: Experiments were conducted using human control and PAH lungs (n=5) and PASMCs in culture. Parallel experiments were performed in rat lung sections and PASMCs and in rodent PAH models induced by the HIF-1α activator, cobalt, chronic hypoxia, and monocrotaline. HIF-1α activation in human PAH leads to mitochondrial fission by cyclin B1/CDK1-dependent phosphorylation of DRP1 at serine 616. In normal PASMCs, HIF-1α activation by CoCl(2) or desferrioxamine causes DRP1-mediated fission. HIF-1α inhibition reduces DRP1 activation, prevents fission, and reduces PASMC proliferation. Both the DRP1 inhibitor Mdivi-1 and siDRP1 prevent mitotic fission and arrest PAH PASMCs at the G2/M interphase. Mdivi-1 is antiproliferative in human PAH PASMCs and in rodent models. Mdivi-1 improves exercise capacity, right ventricular function, and hemodynamics in experimental PAH. CONCLUSIONS: DRP-1-mediated mitotic fission is a cell-cycle checkpoint that can be therapeutically targeted in hyperproliferative disorders such as PAH.


Subject(s)
Cell Proliferation , Dynamins/metabolism , GTP Phosphohydrolases/metabolism , Hypertension, Pulmonary/enzymology , Microtubule-Associated Proteins/metabolism , Mitochondria, Muscle/enzymology , Mitochondrial Proteins/metabolism , Mitosis , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Animals , Antihypertensive Agents/pharmacology , CDC2 Protein Kinase/metabolism , Case-Control Studies , Cell Cycle Checkpoints , Cell Proliferation/drug effects , Cells, Cultured , Cobalt , Cyclin B1/metabolism , Disease Models, Animal , Dynamins/genetics , Enzyme Activation , Familial Primary Pulmonary Hypertension , GTP Phosphohydrolases/genetics , Genetic Therapy/methods , Glycolysis , Humans , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/therapy , Hypoxia/complications , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Microtubule-Associated Proteins/genetics , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/pathology , Mitochondrial Proteins/genetics , Mitosis/drug effects , Monocrotaline , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Phosphorylation , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Quinazolinones/pharmacology , RNA Interference , Rats , Rats, Sprague-Dawley , Serine , Time Factors , Transfection
6.
Am J Respir Crit Care Med ; 185(6): 670-9, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22246173

ABSTRACT

RATIONALE: Pulmonary arterial hypertension (PAH) is a proliferative arteriopathy associated with glucose transporter-1 (Glut1) up-regulation and a glycolytic shift in lung metabolism. Glycolytic metabolism can be detected with the positron emission tomography (PET) tracer (18)F-fluorodeoxyglucose (FDG). OBJECTIVES: The precise cell type in which glycolytic abnormalities occur in PAH is unknown. Moreover, whether FDG-PET is sufficiently sensitive to monitor PAH progression and detect therapeutic regression is untested. We hypothesized that increased lung FDG-PET reflects enhanced glycolysis in vascular cells and is reversible in response to effective therapies. METHODS: PAH was induced in Sprague-Dawley rats by monocrotaline or chronic hypoxia (10% oxygen) in combination with Sugen 5416. Monocrotaline rats were treated with oral dichloroacetate or daily imatinib injections. FDG-PET scans and pulmonary artery acceleration times were obtained weekly. The origin of the PET signal was assessed by laser capture microdissection of airway versus vascular tissue. Metabolism was measured in pulmonary artery smooth muscle cell (PASMC) cultures, using a Seahorse extracellular flux analyzer. MEASUREMENTS AND MAIN RESULTS: Lung FDG increases 1-2 weeks after monocrotaline (when PAH is mild) and is normalized by dichloroacetate and imatinib, which both also regress medial hypertrophy. Glut1 mRNA is up-regulated in both endothelium and PASMCs, but not airway cells or macrophages. PASMCs from monocrotaline rats are hyperproliferative and display normoxic activation of hypoxia-inducible factor-1α (HIF-1α), which underlies their glycolytic phenotype. CONCLUSIONS: HIF-1α-mediated Glut1 up-regulation in proliferating vascular cells in PAH accounts for increased lung FDG-PET uptake. FDG-PET is sensitive to mild PAH and can monitor therapeutic changes in the vasculature.


Subject(s)
Fluorodeoxyglucose F18 , Hypertension, Pulmonary/diagnostic imaging , Monitoring, Physiologic/methods , Positron-Emission Tomography/methods , Pulmonary Wedge Pressure/physiology , Animals , Disease Models, Animal , Disease Progression , Familial Primary Pulmonary Hypertension , Fluorodeoxyglucose F18/pharmacokinetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Oxygen Consumption , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Sprague-Dawley , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...