Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Arterioscler Thromb Vasc Biol ; 32(4): 924-33, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22383703

ABSTRACT

OBJECTIVE: We previously reported that cardiac-restricted deletion of focal adhesion kinase (FAK) exacerbated myocyte death following ischemia/reperfusion (I/R). Here, we interrogated whether targeted elevation of myocardial FAK activity could protect the heart from I/R injury. METHODS AND RESULTS: Transgenic mice were generated with myocyte-specific expression of a FAK variant (termed SuperFAK) that conferred elevated allosteric activation. FAK activity in unstressed transgenic hearts was modestly elevated, but this had no discernable effect on anabolic heart growth or cardiac function. Importantly, SuperFAK hearts exhibited a dramatic increase in FAK activity and a reduction in myocyte apoptosis and infarct size 24 to 72 hours following I/R. Moreover, serial echocardiography revealed that the transgenic mice were protected from cardiac decompensation for up to 8 weeks following surgery. Mechanistic studies revealed that elevated FAK activity protected cardiomyocytes from I/R-induced apoptosis by enhancing nuclear factor-κB (NF-κB)-dependent survival signaling during the early period of reperfusion (30 and 60 minutes). Moreover, adenoviral-mediated expression of SuperFAK in cultured cardiomyocytes attenuated H(2)O(2) or hypoxia/reoxygenation-induced apoptosis, whereas blockade of the NF-κB pathway using a pharmacological inhibitor or small interfering RNAs completely abolished the beneficial effect of SuperFAK. CONCLUSIONS: Enhancing cardiac FAK activity attenuates I/R-induced myocyte apoptosis through activation of the prosurvival NF-κB pathway and may represent a novel therapeutic strategy for ischemic heart diseases.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Genetic Therapy , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/enzymology , Animals , Apoptosis , Cells, Cultured , Disease Models, Animal , Enzyme Activation , Focal Adhesion Protein-Tyrosine Kinases/genetics , Hydrogen Peroxide/metabolism , Male , Mice , Mice, Transgenic , Myocardial Infarction/enzymology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , RNA Interference , Rats , Time Factors , Transfection , Ventricular Function, Left , Ventricular Remodeling
2.
Methods Mol Biol ; 843: 111-23, 2012.
Article in English | MEDLINE | ID: mdl-22222526

ABSTRACT

Formation of a fully functional four-chambered heart involves an intricate and complex series of events that includes precise spatial-temporal regulation of cell specification, proliferation, and migration. The formation of the ventricular septum during mid-gestation ensures the unidirectional flow of blood, and is necessary for postnatal viability. Notably, a majority of all congenital malformations of the cardiovascular system in humans involve septal abnormalities which afflict 1 out of 100 newborn children in the United States. Thus, a clear understanding of the precise mechanisms involved in this morphogenetic event will undoubtedly reveal important therapeutic targets. The final step in valvuloseptal morphogenesis occurs, in part, by directed movement of flanking myocytes into the cushion mesenchyme. In order to identify the molecular mechanisms that regulate this critical myocyte function, we have developed two in vitro methodologies; a transwell assay to assess population changes in motility and a single-cell tracking assay to identify signals that drive the coordinated movement of these cells. These methods have proven effective to identify focal adhesion kinase (FAK) as an intracellular component that is critical for myocyte chemotaxis.


Subject(s)
Chemotaxis , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/enzymology , Single-Cell Analysis/methods , Animals , Cell Separation , Mice , Software
3.
Arterioscler Thromb Vasc Biol ; 31(10): 2193-202, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21757658

ABSTRACT

OBJECTIVE: The investment of newly formed endothelial cell tubes with differentiated smooth muscle cells (SMC) is critical for appropriate vessel formation, but the underlying mechanisms remain unknown. We previously showed that depletion of focal adhesion kinase (FAK) in the nkx2.5 expression domain led to aberrant outflow tract (OFT) morphogenesis and strove herein to determine the cell types and mechanisms involved. METHODS AND RESULTS: We crossed fak(loxp) targeted mice with available Cre drivers to deplete FAK in OFT SMC (FAK(wnt) and FAK(nk)) or coronary SMC (FAK(cSMC)). In each case, depletion of FAK led to defective vasculogenesis that was incompatible with postnatal life. Immunohistochemical analysis of the mutant vascular structures revealed that FAK was not required for progenitor cell proliferation, survival, or differentiation into SMC but was necessary for subsequent SMC recruitment to developing vasculature. Using a novel FAK-null SMC culture model, we found that depletion of FAK did not influence SMC growth or survival, but blocked directional SMC motility and invasion toward the potent endothelial-derived chemokine, platelet-derived growth factor PDGFBB. FAK depletion resulted in unstable lamellipodial protrusions due to defective spatial-temporal activation of the small GTPase, Rac-1, and lack of Rac1-dependent recruitment of cortactin (an actin stabilizing protein) to the leading edge. Moreover, FAK null SMC exhibited a significant reduction in stimulated extracellular matrix degradation. CONCLUSIONS: FAK drives PDGFBB-stimulated SMC chemotaxis/invasion and is essential for SMC to appropriately populate the aorticopulmonary septum and the coronary vascular plexus.


Subject(s)
Chemotaxis , Focal Adhesion Kinase 1/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Neovascularization, Physiologic , Animals , Aorta/embryology , Aorta/enzymology , Apoptosis , Becaplermin , Cell Proliferation , Cell Survival , Cells, Cultured , Chemotaxis/genetics , Coronary Vessels/embryology , Coronary Vessels/enzymology , Cortactin/metabolism , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Focal Adhesion Kinase 1/deficiency , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Developmental , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/embryology , Neovascularization, Physiologic/genetics , Neuropeptides/metabolism , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis , Pseudopodia/enzymology , Pulmonary Artery/embryology , Pulmonary Artery/enzymology , Quail/embryology , RNA Interference , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
4.
Circ Res ; 104(10): 1201-8, 2009 May 22.
Article in English | MEDLINE | ID: mdl-19372463

ABSTRACT

Focal adhesion kinase (FAK) is strongly activated by integrins and growth factors and is essential for embryonic development. We previously showed that the C terminus of FAK is expressed as a separate protein termed FAK-related nonkinase (FRNK) in a smooth muscle cell-selective fashion and that FRNK functions to buffer FAK-dependent signals. We now show that FRNK is also transiently expressed in the neonatal myocardium, with peak levels occurring 5 to 7 days postnatal, just before cell cycle withdrawal. Using novel mouse models, we demonstrate that cardiac-selective expression of FRNK (leading to inhibition of FAK) starting at embryonic day 10.5 leads to a severe ventricular noncompaction defect associated with reduced cardiomyocyte proliferation. Remarkably, postnatal expression of nearly identical levels of FRNK is well tolerated and does not affect viability or anabolic cardiac growth. Nonetheless, FRNK expression in the adult heart does attenuate pathological cardiac hypertrophy following aortic banding, confirming and extending our previous data that this compensatory response is blunted in FAK null hearts. Our mechanistic studies in cultured neonatal cardiomyocytes reveal that FRNK expression induces p38/p27(kip)-dependent cell cycle withdrawal and attenuates extracellular signal-regulated kinase-dependent hypertrophic growth. These findings indicate that dynamic expression of FRNK in the neonatal heart may function to promote cardiomyocyte quiescence in an environment that is particularly rich in growth factors and growth promoting extracellular matrices.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Heart/growth & development , Myocytes, Cardiac/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Cell Cycle/physiology , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/cytology , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-myc/metabolism , Rats , p38 Mitogen-Activated Protein Kinases/metabolism
5.
J Mol Cell Cardiol ; 46(2): 241-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19028502

ABSTRACT

Myocyte apoptosis is central to myocardial dysfunction following ischemia/reperfusion (I/R) and during the transition from hypertrophy to heart failure. Focal adhesion kinase (FAK), a non-receptor tyrosine kinase regulates adhesion-dependent survival signals and unopposed FAK activation has been linked to tumor development. We previously showed that conditional myocyte-specific deletion of FAK (MFKO) in the adult heart did not affect basal cardiomyocyte survival or cardiac function but led to dilated cardiomyopathy and heart failure following pressure overload. In the present study, we sought to determine if FAK functions to limit stress-induced cardiomyocyte apoptosis. We reasoned that (I/R), which stimulates robust apoptotic cell death, might uncover an important cardioprotective function for FAK. We found that depletion of FAK markedly exacerbates hypoxia/re-oxygenation-induced cardiomyocyte cell death in vitro. Moreover, deletion of FAK in the adult myocardium resulted in significant increases in I/R-induced infarct size and cardiomyocyte apoptosis with a concomitant reduction in left ventricular function. Finally, our results suggest that NF-kappaB signaling may play a key role in modulating FAK-dependent cardioprotection, since FAK inactivation blunted activation of the NF-kappaB survival signaling pathway and reduced levels of the NF-kappaB target genes, Bcl2 and Bcl-xl. Since the toggling between pro-survival and pro-apoptotic signals remains central to preventing irreversible damage to the heart, we conclude that targeted FAK activation may be beneficial for protecting stress-dependent cardiac remodeling.


Subject(s)
Focal Adhesion Kinase 1/physiology , Focal Adhesion Protein-Tyrosine Kinases/physiology , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Reperfusion Injury/prevention & control , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Focal Adhesion Kinase 1/genetics , Focal Adhesion Protein-Tyrosine Kinases/genetics , Mice , Mice, Mutant Strains , Myocardial Reperfusion Injury/genetics , NF-kappa B/genetics , NF-kappa B/physiology , Reperfusion Injury/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology
6.
Arterioscler Thromb Vasc Biol ; 27(12): 2714-21, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17823367

ABSTRACT

OBJECTIVE: We previously showed that NAD(P)H oxidase deficiency significantly reduces atherosclerosis in apoE(-/-) mice. The present study was designed to determine the relative contribution of monocyte/macrophage versus vascular wall cell NAD(P)H oxidase to atherogenesis in this model. METHODS AND RESULTS: Cell-specific NAD(P)H oxidase inhibition was achieved via allogenic, sex-mismatched bone marrow transplantation. Aortic atherosclerosis and superoxide production in apoE(-/-) mice (Control) with functional NAD(P)H oxidase in both monocytes/macrophages and vascular wall cells was compared with that in apoE(-/-) mice with nonfunctional monocyte/macrophage NAD(P)H oxidase (BMO) or nonfunctional vessel wall NAD(P)H oxidase (VWO). A significant decrease in superoxide production and atherosclerotic lesions was observed in BMO and VWO mice compared with control mice. Interestingly, BMO mice had significantly lower plasma oxidized LDL levels compared with control and VWO mice, whereas aortic sections of VWO mice showed decreased expression of cellular adhesion molecules compared with control and BMO mice. NAD(P)H oxidase deficiency also attenuated neointimal hyperplasia and mitogenic protein activation in apoE(-/-) mice after arterial injury. CONCLUSIONS: We conclude that (1) both monocyte/macrophages and vessel wall cells play critical roles in atherogenesis; (2) decrease in atherosclerosis results from attenuated superoxide generation in monocyte/macrophages or vessel wall cells; and (3) superoxide generation may impact atherosclerosis, in part, by activating smooth muscle cell mitogenic signaling pathways.


Subject(s)
Atherosclerosis/prevention & control , Bone Marrow Transplantation , Endothelium, Vascular/metabolism , Macrophages/metabolism , Muscle, Smooth, Vascular/metabolism , NADPH Oxidases/metabolism , Superoxides/metabolism , Animals , Aorta/enzymology , Aorta/metabolism , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Adhesion Molecules/metabolism , Cell Proliferation , Dietary Fats/administration & dosage , Disease Models, Animal , Endothelium, Vascular/enzymology , Endothelium, Vascular/injuries , Endothelium, Vascular/pathology , Femoral Artery/enzymology , Femoral Artery/injuries , Femoral Artery/metabolism , Hyperplasia , Janus Kinase 2/metabolism , Lipoproteins, LDL/blood , Macrophages/enzymology , Macrophages/pathology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Oxidation-Reduction , Phosphorylation , RNA, Messenger/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Thrombin/metabolism
7.
J Biol Chem ; 282(27): 19808-19, 2007 Jul 06.
Article in English | MEDLINE | ID: mdl-17500057

ABSTRACT

Increase in the expression of leukocyte antigen-related (LAR) protein causes insulin resistance, an important contributor to atherosclerosis. However, the function of LAR in atherosclerosis is not known. To address whether LAR is important in the response of vascular cells to atherogenic stimuli, we investigated cell proliferation, migration, and insulin-like growth factor-1 receptor (IGF-1R) signaling in wild-type and LAR(-/-) mouse vascular smooth muscle cells (VSMC) treated with IGF-1. Absence of LAR significantly enhanced proliferation and migration of VSMC compared with wild-type cells after IGF-1 treatment. U0126 and LY249002, specific inhibitors of MAPK/ERK kinase (MEK) and phosphoinositide 3-kinase, respectively, inhibited IGF-1-induced DNA synthesis and migration in both wild-type and LAR(-/-) VSMC. IGF-1 markedly enhanced IGF-1R phosphorylation in both wild-type and LAR(-/-) VSMC, but the phosphorylation was 90% higher in knock-out cells compared with wild-type cells. Absence of LAR enhanced phosphorylation of insulin receptor substrate-1 and insulin receptor substrate-1-associated phosphoinositide 3-kinase activity in VSMC treated with IGF-1. IGF-1-induced phosphorylation of ERK1/2 also increased significantly in LAR(-/-) VSMC compared with wild-type cells. Furthermore, LAR directly binds to IGF-1R in glutathione S-transferase-LAR pull-down and IGF-1R immunoprecipitation experiments and recombinant LAR dephosphorylates IGF-1R in vitro. Neointima formation in response to arterial injury and IGF-1R phosphorylation in neointima increased significantly in LAR(-/-) mice compared with wild-type mice. A significant decrease in body weight, fasting insulin, and IGF-1 levels were observed in LAR(-/-) mice compared with wild-type mice. Together, these data indicate that LAR regulates IGF-1R signaling in VSMC and dysregulation of this phosphatase may lead to VSMC hyperplasia.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , MAP Kinase Signaling System , Protein Tyrosine Phosphatases/deficiency , Receptor, IGF Type 1/metabolism , Receptors, Cell Surface/deficiency , Tunica Media/metabolism , Animals , Arteries/injuries , Arteries/metabolism , Arteries/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Butadienes/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Humans , Hyperplasia/genetics , Hyperplasia/metabolism , Hyperplasia/pathology , Insulin Resistance/genetics , Insulin-Like Growth Factor I/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Knockout , Nitriles/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Binding/genetics , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/metabolism , Receptor, IGF Type 1/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4 , Receptors, Cell Surface/metabolism , Tunica Media/pathology
8.
Mol Cell Biol ; 27(15): 5352-64, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17526730

ABSTRACT

To examine a role for focal adhesion kinase (FAK) in cardiac morphogenesis, we generated a line of mice with a conditional deletion of FAK in nkx2-5-expressing cells (herein termed FAKnk mice). FAKnk mice died shortly after birth, likely resulting from a profound subaortic ventricular septal defect and associated malalignment of the outflow tract. Additional less penetrant phenotypes included persistent truncus arteriosus and thickened valve leaflets. Thus, conditional inactivation of FAK in nkx2-5-expressing cells leads to the most common congenital heart defect that is also a subset of abnormalities associated with tetralogy of Fallot and the DiGeorge syndrome. No significant differences in proliferation or apoptosis between control and FAKnk hearts were observed. However, decreased myocardialization was observed for the conal ridges of the proximal outflow tract in FAKnk hearts. Interestingly, chemotaxis was significantly attenuated in isolated FAK-null cardiomyocytes in comparison to genetic controls, and these effects were concomitant with reduced tyrosine phosphorylation of Crk-associated substrate (CAS). Thus, it is possible that ventricular septation and appropriate outflow tract alignment is dependent, at least in part, upon FAK-dependent CAS activation and subsequent induction of polarized myocyte movement into the conal ridges. Future studies will be necessary to determine the precise contributions of the additional nkx2-5-derived lineages to the phenotypes observed.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/deficiency , Gene Deletion , Heart Defects, Congenital/enzymology , Heart Ventricles/abnormalities , Heart Ventricles/anatomy & histology , Animals , Cell Movement , Cell Proliferation , Cell Survival , Crk-Associated Substrate Protein/metabolism , Embryo, Mammalian/abnormalities , Embryo, Mammalian/enzymology , Female , Heart Ventricles/embryology , Heart Ventricles/enzymology , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/metabolism , Male , Mice , Morphogenesis , Myocytes, Cardiac/pathology , Myofibrils/pathology , Phenotype , Phosphorylation , Transcription Factors/metabolism
9.
Circ Res ; 98(10): 1254-63, 2006 May 26.
Article in English | MEDLINE | ID: mdl-16601225

ABSTRACT

To characterize novel signaling pathways that underlie NAD(P)H oxidase-mediated signaling in atherosclerosis, we first examined differences in thrombin-induced gene expression between wild-type and p47phox(-/-) (NAD[P]H oxidase-deficient) VSMC. Of the 9000 genes analyzed by cDNA microarray method at the G1/S transition point, 76 genes were similarly and significantly modulated in both the cell types, whereas another 22 genes that encompass various functional groups were regulated in NAD(P)H oxidase-dependent manner. Among these 22 genes, thrombin-induced NAD(P)H oxidase-mediated regulation of Klf15, Igbp1, Ak4, Adamts5, Ech1, Serp1, Sec61a2, Aox1, Aoh1, Fxyd5, Rai14, and Serpinh1 was shown for the first time in VSMC. The role of NAD(P)H oxidase in the regulation of a subset of these genes (CD44, BMP4, Id1, and Id3) was confirmed using modulators of reactive oxygen species (ROS) generation, a ROS scavenger and in gain-of-function experiments. We then characterized regulation of these genes in restenosis and atherosclerosis. In both apoE(-/-) mice and in a mouse vascular injury model, these genes are regulated in NAD(P)H oxidase-dependent manner during vascular lesion formation. Based on these findings, we propose that NAD(P)H oxidase-dependent gene expression in general, and the CD44 and BMP4-Id signaling pathway in particular, is important in restenosis and atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Bone Morphogenetic Proteins/metabolism , Hyaluronan Receptors/metabolism , Inhibitor of Differentiation Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , NADPH Oxidases/physiology , Thrombin/physiology , Animals , Aorta/cytology , Atherosclerosis/etiology , Bone Morphogenetic Protein 4 , Cells, Cultured , Constriction, Pathologic , Femoral Artery/injuries , Gene Expression Regulation , Hyperplasia , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Proteins/genetics , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Tunica Intima/metabolism , Tunica Intima/pathology , Wounds and Injuries/pathology
10.
Arterioscler Thromb Vasc Biol ; 25(5): 950-6, 2005 May.
Article in English | MEDLINE | ID: mdl-15746439

ABSTRACT

OBJECTIVE: Reactive oxygen species (ROS) integrate cellular signaling pathways involved in aortic smooth muscle cell (SMC) proliferation and migration associated with atherosclerosis. However, the effect of subcellular localization of ROS on SMC mitogenic signaling is not yet fully understood. METHODS AND RESULTS: We used superoxide dismutase (SOD)-deficient mouse aortic SMCs to address the role of subcellular ROS localization on SMC phenotype and mitogenic signaling. Compared with wild-type, a 54% decrease in total SOD activity (almost equal to 50% decrease in SOD1 protein levels) and a 42% reduction in SOD2 activity (approximately equal to 50% decrease in SOD2 protein levels) were observed in SOD1+/- and SOD2+/- SMCs, respectively. Consistent with this, basal and thrombin-induced superoxide levels increased in these SMCs. SOD1+/- and SOD2+/- SMCs exhibit increased basal proliferation and enhanced [3H]-thymidine and [3H]-leucine incorporation in basal and thrombin-stimulated conditions. Our results indicate preferential activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and p38 mitogen-activated protein kinases in SOD1+/- and janus kinase/signal transducer and activator of transcriptase (JAK/STAT) pathway in SOD2+/- SMCs. Pharmacological inhibitors of ERK1/2 p38 and JAK2 confirm the SOD genotype-dependent SMC proliferation. CONCLUSIONS: Our results suggest that SOD1 and SOD2 regulate SMC quiescence by suppressing divergent mitogenic signaling pathways, and dysregulation of these enzymes under pathophysiological conditions may lead to SMC hyperplasia and hypertrophy.


Subject(s)
Atherosclerosis/metabolism , MAP Kinase Signaling System/physiology , Muscle, Smooth, Vascular/enzymology , Superoxide Dismutase/metabolism , Animals , Aorta/enzymology , Aorta/pathology , Atherosclerosis/pathology , Cell Compartmentation/physiology , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Enzymologic , Hypertrophy , Janus Kinase 2 , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle, Smooth, Vascular/pathology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL