Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Front Biosci (Schol Ed) ; 9(1): 46-54, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27814573

ABSTRACT

Hypoxia and inflammatory cytokine activation (H&I) are common processes in many acute and chronic diseases. Thus, a single vector that responds to both hypoxia and inflammatory cytokines, such as TNF-alpha, is useful for assesing the severity of such diseases. Adaptation to hypoxia is regulated primarily by hypoxia inducible transcription factor (HIF alpha) nuclear proteins that engage genes containing a hypoxia response element (HRE). Inflammation activates a multitude of cytokines, including TNF-alpha, that invariably modulate activation of the nuclear factor kappa B (NF-kB) transcription factor. We constructed a vector that encompassed both a hypoxia response element (HRE), and a NF-kappaB responsive element. We show that this vector was functionally responsive to both hypoxia and TNF-alpha, in vitro and in vivo. Thus, this vector might be suitable for the detection and assessment of hypoxia or TNF-alpha.


Subject(s)
Cell Hypoxia/physiology , Genetic Vectors/genetics , NF-kappa B/genetics , Response Elements , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Hypoxia/genetics , Cell Line, Tumor , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , NF-kappa B/metabolism , Promoter Regions, Genetic
2.
New Dir Stud Leadersh ; 2015(146): 49-60, 2015.
Article in English | MEDLINE | ID: mdl-26894903

ABSTRACT

This chapter is about a unique partnership between Ravenscroft, a pre-K-12th grade independent school in Raleigh, North Carolina, and the Center for Creative Leadership. Starting in pre-K, Ravenscroft students embark on the Lead From Here initiative that inspires and empowers them to become citizen leaders.


Subject(s)
Curriculum , Leadership , Moral Development , Program Development , Students , Adolescent , Adult , Child , Humans , Young Adult
3.
Cardiovasc Pathol ; 23(4): 231-7, 2014.
Article in English | MEDLINE | ID: mdl-24813593

ABSTRACT

INTRODUCTION: Heme oxygenase-1 (HO-1) is a cytoprotective enzyme induced by stress. Heart failure is a condition of chronic stress-induced remodeling and is often accompanied by comorbidities such as age and hypertension. HO-1 is known to be protective in the setting of acute myocardial infarction. The role of HO-1 in heart failure is not known, particularly in the setting of pressure overload. METHODS: Mice with alpha-myosin heavy chain restricted expression of HO-1 were aged for 1 year. In addition, mice underwent transverse aortic constriction (TAC) or were infused with isoproterenol (ISO) to induce heart failure. RESULTS: HO-1 transgenic mice developed spontaneous heart failure after 1 year compared to their wild-type littermates and showed accelerated cardiac dysfunction 2 weeks following TAC. Wild-type mice undergoing pressure overload demonstrated extensive interstitial fibrosis that was prevented by HO-1 overexpression, yet HO-1 transgenic mice had reduced capillary density, contractile reserve, and elevated end-diastolic pressure. However, HO-1 transgenic mice had significantly attenuated ISO-induced cardiac dysfunction, interstitial fibrosis, and hypertrophy compared to control. Isolated cardiomyocytes from HO-1 transgenic mice treated with ISO did not show evidence of hypercontracture/necrosis and had reduced NADH oxidase activity. CONCLUSIONS: HO-1 is an effective mechanism for reducing acute myocardial stress such as excess beta-adrenergic activity. However, in our age and pressure overload models, HO-1 showed detrimental rather than therapeutic effects in the development of heart failure.


Subject(s)
Cardiomyopathies/prevention & control , Heart Failure/etiology , Heme Oxygenase-1/metabolism , Aging/pathology , Aging/physiology , Animals , Cardiomyopathies/chemically induced , Cardiomyopathies/pathology , Disease Models, Animal , Heart Failure/pathology , Heart Failure/physiopathology , Heme Oxygenase-1/genetics , Humans , Hypertension/complications , Isoproterenol/toxicity , Male , Mice , Mice, Transgenic , Myocardium/enzymology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Up-Regulation
4.
Cell Transplant ; 21(7): 1443-61, 2012.
Article in English | MEDLINE | ID: mdl-22776314

ABSTRACT

The aim of this study was to evaluate the overexpression of genes central to cell survival and angiogenesis to enhance the function of human late outgrowth endothelial progenitor cells (EPCs) and their utility for infarct recovery. Ischemic myocardial injury creates a hostile microenvironment, which is characterized by hypoxia, oxidative stress, and inflammation. The infarct microenvironment prevents adhesion, survival, and integration of cell transplants that promote neovascularization. EPCs are dysfunctional as a result of risk factors in cardiovascular patients. Protein kinase B (Akt) and heme-oxygenase-1 (HO-1) are intracellular proteins that play an important role in angiogenesis and cell survival. Late outgrowth EPCs transduced ex vivo with Akt and HO-1 demonstrate improved adhesion to extracellular matrix, improved migration toward human cardiomyocytes, and an improved paracrine profile under stress. Enhanced late outgrowth EPCs reduce the tumor necrosis factor-α (TNF-α) burden both in vitro and in vivo, attenuating nuclear factor-κB (NF-κB) activity and promoting cell survival. Akt and HO-1 enhance late outgrowth EPC neovascularization, resulting in improved cardiac performance and reduced negative remodeling after myocardial infarction in nude mice. Alteration of the infarct microenvironment through gene modification of human late outgrowth EPCs enhances the function and integration of transplanted cells for restoration of cardiac function.


Subject(s)
Endothelial Cells/cytology , Heme Oxygenase-1/genetics , Myocardial Infarction/therapy , Proto-Oncogene Proteins c-akt/genetics , Stem Cells/cytology , Animals , Cell Adhesion , Cell Movement , Cells, Cultured , Coronary Vessels/physiology , Genetic Therapy , Heme Oxygenase-1/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/pathology , Myocytes, Cardiac/cytology , Neovascularization, Physiologic , Phagocytosis , Protein Array Analysis , Proteome/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stem Cell Transplantation , Stem Cells/metabolism , Ventricular Remodeling
5.
Exp Biol Med (Maywood) ; 234(5): 582-94, 2009 May.
Article in English | MEDLINE | ID: mdl-19244544

ABSTRACT

AIMS: Reactive oxygen species (ROS) activate multiple signaling pathways involved in cardiac hypertrophy. Since HO-1 exerts potent antioxidant effects, we hypothesized that this enzyme inhibits ROS-induced cardiomyocyte hypertrophy. METHODS: HL-1 cardiomyocytes were transduced with an adenovirus constitutively expressing HO-1 (AdHO-1) to increase basal HO-1 expression and then exposed to 200 microM hydrogen peroxide (H2O2). Hypertrophy was measured using 3H-leucine incorporation, planar morphometry and cell-size by forward-scatter flow-cytometry. The pro-oxidant effect of H2O2 was assessed by redox sensitive fluorophores. Inducing intracellular redox imbalance resulted in cardiomyocyte hypertrophy through transactivation of nuclear factor kappa B (NF-kappaB). RESULTS: Pre-emptive HO-1 overexpression attenuated the redox imbalance and reduced hypertrophic indices. This is the first time that HO-1 has directly been shown to inhibit oxidant-induced cardiomyocyte hypertrophy by a NF-kappaB-dependent mechanism. CONCLUSION: These results demonstrate that HO-1 inhibits pro-oxidant induced cardiomyocyte hypertrophy and suggest that HO-1 may yield therapeutic potential in treatment of.


Subject(s)
Cardiomegaly/enzymology , Heme Oxygenase (Decyclizing)/metabolism , Hydrogen Peroxide/pharmacology , Myocytes, Cardiac/enzymology , Oxidants/pharmacology , Adenoviridae , Animals , Cardiomegaly/genetics , Cardiomegaly/therapy , Cell Line , Heme Oxygenase (Decyclizing)/genetics , NF-kappa B/metabolism , Oxidation-Reduction , Rats , Transduction, Genetic
6.
Am J Physiol Heart Circ Physiol ; 296(2): H263-71, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19060129

ABSTRACT

Numerous cAMP-elevating agents regulate events required for efficient migration of arterial vascular smooth muscle cells (VSMCs). Interestingly, when the impact of cAMP-elevating agents on individual migration-related events is studied, these agents have been shown to have distinct, and sometimes unexpected, effects. For example, although cAMP-elevating agents inhibit overall migration, they promote VSMC adhesion to extracellular matrix proteins and the formation of membrane extensions, which are both events that are essential for and promote migration. Herein, we extend previous observations that identified phosphodiesterase-4D3 (PDE4D3) as an integral component of a PKA/A kinase-anchoring protein (AKAP) complex in cultured/hypertrophied rat cardiac myocytes to the case for nonhypertrophied cardiac myocytes. Moreover, we show that while rat aortic VSMCs also express PDE4D3, this protein is not detected in PKA/AKAP complexes isolated from these cells. In contrast, we show that another PDE4D splice variant expressed in arterial vascular myocytes, namely, PDE4D8, integrates into PKA/AKAP-based signaling complexes in VSMCs. Consistent with the idea that a PDE4D8/PKA/AKAP complex regulates specific VSMC functions, PKA and PDE4D8 were each recruited to leading-edge structures in migrating VSMCs, and inhibition of PDE4D8 recruitment to pseudopodia of migrating cells caused localized changes in actin dynamics. Our data are presented in the context that cardiac myocytes and arterial VSMCs may use distinct PDE4D variants to regulate selected pools of targeted PKA activity and that disruption of this complex may allow selective regulation of cAMP-dependent events between these two cardiovascular cell types.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Cardiac/enzymology , Myocytes, Smooth Muscle/enzymology , Signal Transduction , Actins/metabolism , Animals , Cell Line , Cell Movement , Cells, Cultured , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Humans , Isoenzymes , Male , Muscle, Smooth, Vascular/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Smooth Muscle/drug effects , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Pseudopodia/enzymology , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Transfection
7.
Mol Ther ; 16(3): 571-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18253156

ABSTRACT

With the goal of devising a non-invasive cell therapy for cardiac repair that may be well tolerated by patients with myocardial infarction (MI), this study evaluated the efficacy of intravenous infusion of genetically modified mesenchymal stem cells (MSCs) overexpressing CXC chemokine receptor 4 (CXCR4). CXCR4 is the cognate receptor for stromal-derived factor-1 (SDF-1), a chemokine required for homing of progenitor cells to ischemic tissues. In this study, retrovirally transduced MSCs constitutively expressing CXCR4 (CXCR4-MSCs) were delivered intravenously 24 hours after coronary occlusion/reperfusion in rats. When compared with untransduced MSCs, CXCR4-MSCs homed in toward the infarct region of the myocardium in greater numbers. In the CXCR4-MSC-treated animals, echocardiographic imaging 30 days after MI showed a decrease in anterior wall thinning and good preservation of left ventricular (LV) chamber dimensions, whereas the animals treated with saline or unmodified MSCs showed significant remodeling. Histochemical analysis showed a decrease in collagen I/III ratio in the infarcted wall of CXCR4-MSC-treated animals, thereby suggesting improved chamber compliance. Assessment revealed post-MI recovery of LV function in the CXCR4-MSC-treated animals, whereas LV function remained depressed in the saline and MSC-treated animals. In summary, intravenous delivery of genetically modified MSCs expressing CXCR4 may be a useful, non-invasive, and safe therapeutic strategy for post-infarction myocardial repair.


Subject(s)
Genetic Vectors/genetics , Mesenchymal Stem Cells/cytology , Myocardial Infarction/therapy , Receptors, CXCR4/physiology , Animals , Cell Proliferation , Cell Survival/genetics , Cell Survival/physiology , Echocardiography , Flow Cytometry , Fluorescent Antibody Technique , Genetic Therapy/methods , Immunohistochemistry , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Rats , Rats, Sprague-Dawley , Receptors, CXCR4/genetics
8.
J Pharmacol Exp Ther ; 321(3): 921-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17377061

ABSTRACT

The effects of the phytoestrogens phloretin and phloridzin on Ca(2+) handling, cell shortening, the action potential, and Ca(2+) and K(+) currents in freshly isolated cardiac myocytes from rat ventricle were examined. Phloretin increased the amplitude and area and decreased the rate of decline of electrically evoked Ca(2+) transients in the myocytes. These effects were accompanied by an increase in the Ca(2+) load of the sarcoplasmic reticulum, as determined by the area of caffeine-evoked Ca(2+) transients. An increase in the extent of shortening of the myocytes in response to electrically evoked action potentials was also observed in the presence of phloretin. To further examine possible mechanisms contributing to the observed changes in Ca(2+) handling and contractility, the effects of phloretin on the cardiac action potential and plasma membrane Ca(2+) and K(+) currents were examined. Phloretin markedly increased the action potential duration in the myocytes, and it inhibited the Ca(2+)-independent transient outward K(+) current (I(to)). The inwardly rectifying K(+) current, the sustained outward delayed rectifier K(+) current, and L-type Ca(2+) currents were not significantly different in the presence and absence of phloretin, nor was there any evidence that the Na(+)/Ca(2+) exchanger was affected. The effects of phloretin on Ca(2+) handling in the myocytes are consistent with its effects on I(to). Phloridzin did not significantly alter the amplitude or area of electrically evoked Ca(2+) transients in the myocytes, nor did it have detectable effects on the sarcoplasmic reticulum Ca(2+) load, cell shortening, or the action potential.


Subject(s)
Action Potentials/drug effects , Calcium Signaling/drug effects , Ion Channels/physiology , Myocytes, Cardiac/drug effects , Phloretin/pharmacology , Phlorhizin/pharmacology , Animals , Caffeine/pharmacology , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Calcium Channels, L-Type/physiology , Cell Shape/drug effects , Cytoplasm/metabolism , Electric Stimulation , Electrophysiology , Ion Channels/metabolism , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Potassium Channels/metabolism , Potassium Channels/physiology , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism
9.
Am J Physiol Heart Circ Physiol ; 293(1): H48-59, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17322421

ABSTRACT

We reported previously that predelivery of heme oxygenase-1 (HO-1) gene to the heart by adeno-associated virus-2 (AAV-2) markedly reduces ischemia and reperfusion (I/R)-induced myocardial injury. However, the effect of preemptive HO-1 gene delivery on long-term survival and prevention of postinfarction heart failure has not been determined. We assessed the effect of HO-1 gene delivery on long-term survival, myocardial function, and left ventricular (LV) remodeling 1 yr after myocardial infarction (MI) using echocardiographic imaging, pressure-volume (PV) analysis, and histomorphometric approaches. Two groups of Lewis rats were injected with 2 x 10(11) particles of AAV-LacZ (control) or AAV-human HO-1 (hHO-1) in the anterior-posterior apical region of the LV wall. Six weeks after gene transfer, animals were subjected to 30 min of ischemia by ligation of the left anterior descending artery followed by reperfusion. Echocardiographic measurements and PV analysis of LV function were obtained at 2 wk and 12 mo after I/R. One year after acute MI, mortality was markedly reduced in the HO-1-treated animals compared with the LacZ-treated animals. PV analysis demonstrated significantly enhanced LV developed pressure, elevated maximal dP/dt, and lower end-diastolic volume in the HO-1 animals compared with the LacZ animals. Echocardiography showed a larger apical anterior-to-posterior wall ratio in HO-1 animals compared with LacZ animals. Morphometric analysis revealed extensive myocardial scarring and fibrosis in the infarcted LV area of LacZ animals, which was reduced by 62% in HO-1 animals. These results suggest that preemptive HO-1 gene delivery may be useful as a therapeutic strategy to reduce post-MI LV remodeling and heart failure.


Subject(s)
Disease Models, Animal , Genetic Therapy/methods , Heme Oxygenase (Decyclizing)/therapeutic use , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Ventricular Dysfunction, Left/prevention & control , Ventricular Dysfunction, Left/physiopathology , Animals , Heme Oxygenase (Decyclizing)/genetics , Humans , Male , Myocardial Infarction/complications , Rats , Rats, Inbred Lew , Survival Analysis , Survival Rate , Transfection/methods , Treatment Outcome , Ventricular Dysfunction, Left/etiology
10.
Methods Mol Med ; 139: 197-210, 2007.
Article in English | MEDLINE | ID: mdl-18287673

ABSTRACT

Endothelial progenitor cells (EPCs) and mesenchymal stem cells (MSCs) have emerged as potentially useful substrates for neovascularization and tissue repair and bioengineering. EPCs are a heterogeneous group of endothelial cell precursors originating in the hematopoietic compartment of the bone marrow. MSCs are a rare population of fibroblast-like cells derived from the bone marrow stroma, constituting approximately 0.001-0.01% of the nucleated cells in the marrow. Both cells types have been isolated from the bone marrow. In addition, EPC can be isolated from peripheral blood as well as the spleen, and MSC has also been isolated from peripheral adipose tissue. Several approaches have been used for the isolation of EPC and MSC, including density centrifugation and magnetic bead selection. Phenotypic characterization of both cell types is carried out using immunohistochemical detection and fluorescence-activated cell sorting analysis of cell-surface molecule expression. However, the lack of specific markers for each cell type renders their characterization difficult and ambiguous. In this chapter, we describe the methods that we use routinely for isolation, characterization, and genetic modification of EPC and MSC from human, rabbit, and mouse peripheral blood and bone marrow.


Subject(s)
Endothelial Cells/physiology , Mesenchymal Stem Cells/physiology , Stem Cells/physiology , Transduction, Genetic , Animals , Antigens, Surface/metabolism , Endothelial Cells/cytology , Humans , Immunophenotyping , Mesenchymal Stem Cells/cytology , Mice , Phenotype , Rabbits , Stem Cells/cytology , Viruses/genetics , Viruses/metabolism
11.
J Virol ; 80(20): 10270-3, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17005706

ABSTRACT

Cultures derived from the cerebral cortices and hippocampi of 17-day-old mouse fetuses infected with the CVS strain of rabies virus showed loss of trypan blue exclusion, morphological apoptotic features, and activated caspase 3 expression, indicating apoptosis. The NMDA (N-methyl-D-aspartate acid) antagonists ketamine (125 microM) and MK-801 (60 microM) were found to have no significant neuroprotective effect on CVS-infected neurons, while the caspase inhibitor Ac-Asp-Glu-Val aspartic acid aldehyde (25 microM) exerted a marked neuroprotective effect. Glutamate-stimulated increases in levels of intracellular calcium were reduced in CVS-infected hippocampal neurons. Ketamine (120 mg/kg of body weight/day intraperitoneally) given to CVS-infected adult mice produced no beneficial effects. We have found no supportive evidence that excitotoxicity plays an important role in rabies virus infection.


Subject(s)
Neurons/virology , Rabies virus/growth & development , Rabies , Animals , Apoptosis , Caspase 3 , Caspase Inhibitors , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Disease Models, Animal , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/administration & dosage , Ketamine/pharmacology , Mice , N-Methylaspartate/antagonists & inhibitors , Oligopeptides/pharmacology
12.
Arterioscler Thromb Vasc Biol ; 26(9): 2027-34, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16840713

ABSTRACT

OBJECTIVE: Oxidative stress (OS) induces smooth muscle cell apoptosis in the atherosclerotic plaque, leading to plaque instability and rupture. Heme oxygenase-1 (HO-1) exerts cytoprotective effects in the vessel wall. Recent evidence suggests that PKB/Akt may modulate HO-1 activity. This study examined the role of Akt in mediating the cytoprotective effects of HO-1 in OS-induced apoptosis of human aortic smooth muscle cells (HASMCs). METHODS AND RESULTS: HASMCs were transduced with retroviral vectors expressing HO-1, Akt, or GFP and exposed to H2O2. Cell viability was assessed by MTT assay. OS was determined by CM-H2DCFDA fluorescence, and apoptosis was assessed by terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL), caspase-3 activity, and Bcl-2/Bad levels. Mitochondrial membrane potential (delta psi(m)) was assessed by fluorescence-activated cell sorter (FACS) using JC-1. HO-1 reduced H2O2-induced OS and apoptosis. Akt knockdown removed the protective effect of HO-1 on delta psi(m) during exposure to H2O2. Conversely, HO-1 knockdown removed the protective effect of Akt on delta psi(m). Inhibition of PI3K-Akt reduced induction of HO-1 protein expression by H2O2 and blocked its anti-apoptotic effects. The Akt-mediated upregulation of HO-1 was dependent on activation of HO-1 promoter by Nrf2. CONCLUSIONS: HO-1 and Akt exert codependent cytoprotective effects against OS-induced apoptosis in HASMCs. These findings may have implications for the design of novel therapeutic strategies for plaque stabilization.


Subject(s)
Apoptosis/physiology , Heme Oxygenase-1/physiology , Hydrogen Peroxide/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Oxidants/pharmacology , Proto-Oncogene Proteins c-akt/physiology , Cell Survival/physiology , Cytoprotection/physiology , Heme Oxygenase-1/pharmacology , Humans , Membrane Potentials/physiology , Mitochondria/physiology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Proteins/metabolism
13.
FASEB J ; 20(2): 207-16, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16449792

ABSTRACT

We reported previously that predelivery of the anti-oxidant gene heme oxygenase-1 (HO-1) to the heart by adeno associated virus (AAV) markedly reduces injury after acute myocardial infarction (MI). However, the effect of HO-1 gene delivery on postinfarction recovery has not been investigated. In the current study, we assessed the effect of HO-1 gene delivery on post-MI left ventricle (LV) remodeling and function using echocardiographic imaging and histomorphometric approaches. Two groups of Sprague-Dawley rats were injected with 4 x 10(11) particles of AAV-LacZ (control) or AAV-hHO-1 in the LV wall. Eight wk after gene transfer, the animals were subjected to 30 min of ischemia by ligation of left anterior descending artery (LAD) followed by reperfusion. Echocardiographic measurements were obtained in a blinded fashion prior and at 1.5 and 3 months after I/R. Ejection fraction (EF) was reduced by 13% and 40% in the HO-1 and LacZ groups, respectively at 1.5 months after MI. Three months after MI, EF recovered fully in the HO-1, but only partially in the LacZ-treated animals. Post-MI LV dimensions were markedly increased and the anterior wall was markedly thinned in the LacZ-treated animals compared with the HO-1-treated animals. Significant myocardial scarring and fibrosis were observed in the LacZ-group in association with elevated levels of interstitial collagen I and III and MMP-2 activity. Post-MI myofibroblast accumulation was reduced in the HO-1-treated animals, and retroviral overexpression of HO-1 reduced proliferation of isolated cardiac fibroblasts. Our data indicate that rAAV-HO-1 gene transfer markedly reduces fibrosis and ventricular remodeling and restores LV function and chamber dimensions after myocardial infarction.


Subject(s)
Genetic Therapy , Heme Oxygenase-1/metabolism , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Ventricular Remodeling/physiology , Animals , Cell Proliferation , Cells, Cultured , Collagen/metabolism , Fibroblasts , Fibrosis/genetics , Fibrosis/pathology , Fibrosis/therapy , Gene Expression Regulation, Enzymologic , Heart Ventricles/anatomy & histology , Heme Oxygenase-1/genetics , Humans , Male , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/therapy , Myocardium/cytology , Myocardium/metabolism , Rats , Rats, Sprague-Dawley , Ventricular Function
14.
J Pharmacol Exp Ther ; 316(2): 628-35, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16227472

ABSTRACT

Phytoestrogens are naturally occurring estrogenic compounds found in plants and plant products. These compounds are also known to exert cellular effects independent of their interactions with estrogen receptors. We studied the effects of the phytoestrogens phloretin, phloridzin, genistein, and biochanin A on Ca(2+) uptake into the cardiac muscle sarcoplasmic reticulum (SR). Genistein and biochanin A did not affect SR Ca(2+) uptake. On the other hand, phloretin and phloridzin decreased the maximum velocity of SR Ca(2+) uptake but did not affect the Hill coefficient or the Ca(2+) sensitivity of uptake. Measurements of the ATPase activity of the cardiac SR Ca(2+) pump (SERCA2a) revealed direct inhibitory effects of phloretin and phloridzin on SERCA2a. Neither compound induced a detectable change in the permeability of the SR membrane to Ca(2+). These results indicate that phloretin and phloridzin inhibit cardiac SR Ca(2+) uptake by directly inhibiting SERCA2a.


Subject(s)
Calcium-Transporting ATPases/metabolism , Calcium/metabolism , Heart/drug effects , Myocardium , Phytoestrogens/pharmacology , Sarcoplasmic Reticulum/drug effects , Animals , Dogs , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/metabolism , Heart Ventricles/drug effects , Heart Ventricles/enzymology , Heart Ventricles/metabolism , In Vitro Techniques , Intracellular Membranes/drug effects , Intracellular Membranes/enzymology , Intracellular Membranes/metabolism , Myocardium/enzymology , Myocardium/metabolism , Sarcoplasmic Reticulum/enzymology , Sarcoplasmic Reticulum/metabolism
15.
Am J Physiol Heart Circ Physiol ; 285(2): H661-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12702490

ABSTRACT

Tamoxifen is an estrogen receptor antagonist used in the treatment of breast cancer. However, tamoxifen has been shown to induce QT prolongation of the electrocardiogram, thereby potentially causing life-threatening polymorphic ventricular arrhythmias. The purpose of the present study was to elucidate the electrophysiological mechanism(s) that underlie the arrhythmogenic effects of tamoxifen. We used standard ruptured whole cell and perforated patch-clamping techniques on rat ventricular myocytes to investigate the effects of tamoxifen on cardiac action potential (AP) waveforms and the underlying K+ currents. Tamoxifen (3 micromol/l) markedly prolonged AP duration, decreased maximal rate of depolarization, and decreased resting membrane potential. At this concentration, tamoxifen significantly depressed the Ca2+-independent transient outward K+ current (Ito), sustained outward delayed rectifier K+ current (Isus), inward rectifier K+ current (IK1), and Na+ current (INa) in the myocytes. Lower concentrations of tamoxifen (1 micromol/l) also decreased the resting membrane potential and significantly depressed IK1 to 79 +/- 5% (n = 5; at -120 mV) of pretreatment values. The results of this study indicate that inhibition of Ito, Isus, and IK1 by tamoxifen may underlie AP prolongation in cardiac myocytes and thereby contribute to prolonged QT interval observed in patients.


Subject(s)
Estrogen Antagonists/pharmacology , Myocytes, Cardiac/metabolism , Potassium/metabolism , Sodium/metabolism , Tamoxifen/pharmacology , Action Potentials/drug effects , Animals , Heart Ventricles/cytology , Long QT Syndrome/chemically induced , Long QT Syndrome/physiopathology , Male , Membrane Potentials/drug effects , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...