Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
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.
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
3.
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
4.
Mol Cell Biochem ; 346(1-2): 57-67, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20872048

ABSTRACT

Heme oxygenase-1 (HO-1) has been well established as a cytoprotective molecule, and has been shown to exert cardioprotective effects in both hypertension and cardiac hypertrophy. However, the precise mechanism of the cardioprotective effect of HO-1 has yet to be fully elucidated. With the natriuretic peptide system (NPS) as a key player in cardiovascular homeostasis and tissue dynamics, we sought to examine the effect of high dietary salt treatment in genetic models of HO-1 expression, and assessed the expression of the NPS in the left ventricle (LV), to determine if the effects of altered HO-1 expression may be due to modified levels of the NPS. Age-matched 12-week old male HO-1 knockout (HO-1(-/-)) and HO-1 cardiomyocyte-specific transgenic overexpressing (HO-1(Tg)) mice were treated with either normal salt (NS; 0.8%) or high salt (HS; 8.0%) chow for 5 weeks. LV mRNA expression was determined using quantitative real-time PCR. ANP peptide level was measured in the LV and plasma using radioimmunoassay, and LV cyclic 3'-5' guanosine monophosphate level was measured using an enzyme immunoassay kit. HO-1(-/-) fed HS diet had significantly higher left ventricle-to-body weight ratio (LV/BW) compared to HO-1(+/+) mice fed NS diet. HO-1(-/-) mice had significantly reduced expression of the NPS compared to controls, and these mice did not exhibit a salt-induced increase in ANP expression. HS treatment had no noticeable effect on LV/BW in HO-1(Tg) mice compared to controls. HO-1(Tg) mice had significantly higher ANP and BNP expression compared to controls. There were no differences in LV cGMP levels among all genotypes and dietary treatments. HO-1 ablation resulted in significantly lower mRNA expression of the NPS, whereas HO-1 overexpression resulted in higher mRNA expression of the NPS. Both were substantiated by peptide levels as measured by RIA. These data indicate that the detrimental effect of reduced HO-1 expression and the cardioprotective effect of increased HO-1 expression may be due, in part, to altered expression of the NPS.


Subject(s)
Atrial Natriuretic Factor/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Natriuretic Peptide, Brain/metabolism , Sodium Chloride, Dietary/administration & dosage , Animals , Atrial Natriuretic Factor/genetics , Base Sequence , DNA Primers , Female , Heme Oxygenase (Decyclizing)/genetics , Male , Mice , Mice, Transgenic , Natriuretic Peptide, Brain/genetics , RNA, Messenger/genetics , Radioimmunoassay
5.
Stem Cells ; 27(4): 971-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19353525

ABSTRACT

Administration of mesenchymal stem cells (MSCs) is an effective therapy to repair cardiac damage after myocardial infarction (MI) in experimental models. However, the mechanisms of action still need to be elucidated. Our group has recently suggested that MSCs mediate their therapeutic effects primarily via paracrine cytoprotective action. Furthermore, we have shown that MSCs overexpressing Akt1 (Akt-MSCs) exert even greater cytoprotection than unmodified MSCs. So far, little has been reported on the metabolic characteristics of infarcted hearts treated with stem cells. Here, we hypothesize that Akt-MSC administration may influence the metabolic processes involved in cardiac adaptation and repair after MI. MI was performed in rats randomized in four groups: sham group and animals treated with control MSCs, Akt-MSCs, or phosphate-buffered saline (PBS). High energy metabolism and basal 2-deoxy-glucose (2-DG) uptake were evaluated on isolated hearts using phosphorus-31 nuclear magnetic resonance spectroscopy at 72 hours and 2 weeks after MI. Treatment with Akt-MSCs spared phosphocreatine stores and significantly limited the increase in 2-DG uptake in the residual intact myocardium compared with the PBS- or the MSC-treated animals. Furthermore, Akt-MSC-treated hearts had normal pH, whereas low pH was measured in the PBS and MSC groups. Correlative analysis indicated that functional recovery after MI was inversely related to the rate of 2-DG uptake. We conclude that administration of MSCs overexpressing Akt at the time of infarction results in preservation of normal metabolism and pH in the surviving myocardium.


Subject(s)
Mesenchymal Stem Cell Transplantation , Myocardial Infarction/metabolism , Myocardial Infarction/therapy , Myocardium/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Bone Marrow Cells/metabolism , Deoxyglucose/metabolism , Female , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Male , Mesenchymal Stem Cells/metabolism , Rats , Rats, Sprague-Dawley , Transduction, Genetic
6.
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
7.
Methods Mol Biol ; 482: 281-94, 2009.
Article in English | MEDLINE | ID: mdl-19089363

ABSTRACT

Mesenchymal stem cells (MSCs) are defined as self-renewing and multipotent cells capable of differentiating into multiple cell types, including osteocytes, chondrocytes, adipocytes, hepatocytes, myocytes, neurons, and cardiomyocytes. MSCs were originally isolated from the bone marrow stroma but they have recently been identified also in other tissues, such as fat, epidermis, and cord blood. Several methods have been used for MSC isolation. The most common method is based on the ability of the MSCs to selectively adhere to plastic surfaces. Phenotypic characterization of MSCs is usually carried out using immunocytochemical detection or fluorescence-activated cell sorting (FACS) analysis of cell surface molecule expression. However, the lack of specific markers renders the characterization of MSCs difficult and sometimes ambiguous. MSCs posses remarkable expansion potential in culture and are highly amenable to genetic modification with various viral vectors rendering them optimal vehicles for cell-based gene therapy. Most importantly, MSC plasticity and the possibility to use them as autologous cells render MSCs suitable for cell therapy and tissue engineering. Furthermore, it is known that MSCs produce and secrete a great variety of cytokines and chemokines that play beneficial paracrine actions when MSCs are used for tissue repair. In this chapter, we describe methods for isolation, ex vivo expansion, phenotypic characterization, and viral infection of MSCs from mouse bone marrow. We also describe a method for preparation of conditioned and concentrated conditioned medium from MSCs. The conditioned medium can be easily tested both in vitro and in vivo when a particular paracrine effect (i.e., cytoprotection) is hypothesized to be an important mechanism of action of the MSCs and/or screened to identify a target paracrine/autocrine mediator.


Subject(s)
Bone Marrow Cells/cytology , Cell Separation/methods , Mesenchymal Stem Cells/cytology , Retroviridae/genetics , Transduction, Genetic/methods , Animals , Cell Proliferation , Cells, Cultured , Culture Media, Conditioned , Immunophenotyping , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence
8.
Am J Pathol ; 172(3): 592-602, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18276782

ABSTRACT

Cysteinyl leukotrienes (CysLTs) have been implicated as inflammatory mediators of cardiovascular disease. Three distinct CysLT receptor subtypes transduce the actions of CysLTs but the role of the endothelial CysLT2 receptor (CysLT2R) in cardiac function is unknown. Here, we investigated the role of CysLT2R in myocardial ischemia-reperfusion (I/R) injury using transgenic (tg) mice overexpressing human CysLT2R in vascular endothelium and nontransgenic (ntg) littermates. Infarction size in tg mice increased 114% compared with ntg mice 48 hours after I/R; this increase was blocked by the CysLT receptor antagonist BAY-u9773. Injection of 125 I-albumin into the systemic circulation revealed significantly enhanced extravasation of the label in tg mice, indicating increased leakage of the coronary endothelium, combined with increased incidence of hemorrhage and cardiomyocyte apoptosis. Expression of proinflammatory genes such as Egr-1, VCAM-1, and ICAM was significantly increased in tg mice relative to ntg controls. Echocardiographic assessment 2 weeks after I/R revealed decreased anterior wall thickness in tg mice. Furthermore, the postreperfusion time constant tau of isovolumic relaxation was significantly increased in tg animals, indicating diastolic dysfunction. These results reveal that endothelium-targeted overexpression of CysLT2R aggravates myocardial I/R injury by increasing endothelial permeability and exacerbating inflammatory gene expression, leading to accelerated left ventricular remodeling, induction of peri-infarct zone cellular apoptosis, and impaired cardiac performance.


Subject(s)
Endothelium, Vascular/metabolism , Membrane Proteins/genetics , Myocardial Reperfusion Injury/genetics , Receptors, Leukotriene/genetics , Animals , Apoptosis/genetics , Cell Membrane Permeability/genetics , Early Growth Response Protein 1/genetics , Intercellular Adhesion Molecule-1/genetics , Leukocyte Common Antigens/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocytes, Cardiac/pathology , Neutrophil Infiltration/genetics , Receptors, Leukotriene/metabolism , Up-Regulation/physiology , Vascular Cell Adhesion Molecule-1/genetics , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/genetics , Ventricular Remodeling/genetics
9.
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
10.
J Mol Cell Cardiol ; 43(5): 580-92, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17920074

ABSTRACT

Heme-oxygenase-1 (HO-1), a stress-inducible protein, is an important cytoprotective agent against ischemia/reperfusion (I/R) injury. However, the role of downstream mediators involved in HO-1-induced cytoprotection is not clear. In the current study we investigated the role of biliverdin reductase, an enzyme involved in the conversion of HO-1-derived biliverdin into bilirubin and the PI3K/Akt pathway in mediating the cytoprotective effects of HO-1 against hypoxia and reoxygenation (H/R) injury in vitro and in vivo. H9c2 cardiomyocytes were transfected with a plasmid expressing HO-1 or LacZ and exposed to 24 h of hypoxia followed by 12 h of reoxygenation. At the end of reoxygenation, reactive oxygen species generation was determined using CM-H(2)DCFDA dye and apoptosis was assessed by TUNEL, caspase activity and Bad phosphorylation. p85 and Akt phosphorylation were determined using cell-based ELISA and phospho-specific antibodies, respectively. HO-1 overexpression increased phosphorylation of the regulatory subunit of the PI3K (p85alpha) and downstream effector Akt in H9c2 cells, leading to decreased ROS and apoptosis. Furthermore, cardiac expression of HO-1 increased basal phosphorylated Akt levels and decreased infarct size in response to LAD ligation and release induced I/R injury. Conversely, PI3K inhibition reversed the effects of HO-1 on Akt phosphorylation, cell death and infarct size. In addition, knockdown of biliverdin reductase (BVR) expression with siRNA attenuated HO-1-induced Akt phosphorylation and increased H/R-induced apoptosis of H9c2 cells. Co-immunoprecipitation revealed protein-protein interaction between BVR and the phosphorylated p85 subunit of the PI3 kinase. Taken together, these results suggest that the enzyme biliverdin reductase plays an important role in mediating cytoprotective effects of HO-1. This effect is mediated, at least in part, via interaction with and activation of the PI3K/Akt pathway.


Subject(s)
Heme Oxygenase-1/metabolism , Hypoxia/prevention & control , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Reperfusion Injury/prevention & control , Animals , Heme Oxygenase-1/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxygen Consumption , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism
11.
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
12.
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
13.
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
14.
FASEB J ; 20(6): 661-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16581974

ABSTRACT

We previously reported that intramyocardial injection of bone marrow-derived mesenchymal stem cells overexpressing Akt (Akt-MSCs) inhibits ventricular remodeling and restores cardiac function measured 2 wk after myocardial infarction. Here, we report that the functional improvement occurs in < 72 h. This early remarkable effect cannot be readily attributed to myocardial regeneration from the donor cells. Thus, we hypothesized that paracrine actions exerted by the cells through the release of soluble factors might be important mechanisms of tissue repair and functional improvement after injection of the Akt-MSCs. Indeed, in the current study we demonstrate that conditioned medium from hypoxic Akt-MSCs markedly inhibits hypoxia-induced apoptosis and triggers vigorous spontaneous contraction of adult rat cardiomyocytes in vitro. When injected into infarcted hearts, the Akt-MSC conditioned medium significantly limits infarct size and improves ventricular function relative to controls. Support to the paracrine hypothesis is provided by data showing that several genes, coding for factors (VEGF, FGF-2, HGF, IGF-I, and TB4) that are potential mediators of the effects exerted by the Akt-MSC conditioned medium, are significantly up-regulated in the Akt-MSCs, particularly in response to hypoxia. Taken together, our data support Akt-MSC-mediated paracrine mechanisms of myocardial protection and functional improvement.


Subject(s)
Cytoprotection/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Paracrine Communication , Proto-Oncogene Proteins c-akt/metabolism , Animals , Female , Fibroblast Growth Factors/metabolism , Gene Expression Regulation , Hepatocyte Growth Factor/metabolism , Insulin-Like Growth Factor I/metabolism , Male , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Proto-Oncogene Proteins c-akt/genetics , Rats , Rats, Sprague-Dawley , Thymosin/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
15.
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
16.
J Am Coll Cardiol ; 47(3): 635-43, 2006 Feb 07.
Article in English | MEDLINE | ID: mdl-16458149

ABSTRACT

OBJECTIVES: We assessed the hypothesis that overexpression of the antioxidant enzyme heme oxygenase (HO)-1 may protect against chronic recurrent ischemia/reperfusion injury. BACKGROUND: Multiple and recurring episodes of myocardial ischemia can result in significant myocardial damage, including myocyte death, fibrosis, and wall thinning, leading to impaired ventricular function and cardiac failure. METHODS: In this study we used a closed-chest rodent model of chronic recurring myocardial ischemia and reperfusion to investigate the efficacy of pre-emptive gene therapy in overexpressing the antioxidant enzyme HO-1, using adeno-associated virus (AAV)-2 as the delivery vector. RESULTS: We show that constitutive overexpression of HO-1 can prevent myocardial wall thinning, inflammation, fibrosis, and deterioration of cardiac function (as measured by echocardiography, histology, and immunohistochemistry) induced by repeated transient myocardial ischemia and reperfusion injury. With HO-1 therapy, there was a significant reduction in apoptosis as determined by levels of markers of survival proteins and terminal deoxynucleotidyltransferase dUTP nick end-labeling staining. This prevention of tissue damage was also associated with reduction in superoxide generation. CONCLUSIONS: Taken together we provide the first evidence of the therapeutic efficacy of pre-emptive AAV-HO-1 delivery for prevention against multiple ischemic injury. This approach protects myocytes by simultaneously activating protective response and inhibiting pathological left ventricular remodeling and, therefore, may be a useful cardio-protective strategy for patients with coronary artery disease at a high risk for recurrent myocardial ischemia.


Subject(s)
Genetic Therapy , Heme Oxygenase-1/genetics , Myocardial Reperfusion Injury/prevention & control , Animals , Apoptosis , Dependovirus/genetics , Genetic Vectors , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/therapeutic use , In Situ Nick-End Labeling , Male , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Rats , Rats, Sprague-Dawley , Recurrence , Superoxides/metabolism
17.
Hypertension ; 46(1): 7-18, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15956118

ABSTRACT

Endothelial dysfunction and cell loss are prominent features in cardiovascular disease. Endothelial progenitor cells (EPCs) originating from the bone marrow play a significant role in neovascularization of ischemic tissues and in re-endothelialization of injured blood vessels. Several studies have shown the therapeutic potential of EPC transplantation in rescue of tissue ischemia and in repair of blood vessels and bioengineering of prosthetic grafts. Recent small-scale trials have provided preliminary evidence of feasibility, safety, and efficacy in patients with myocardial and critical limb ischemia. However, several studies have shown that age and cardiovascular disease risk factors reduce the availability of circulating EPCs (CEPCs) and impair their function to varying degrees. In addition, the relative scarcity of CEPCs limits the ability to expand these cells in sufficient numbers for some therapeutic applications. Priority must be given to the development of strategies to enhance the number and improve the function of CEPCs. Furthermore, alternative sources of EPC such as chord blood need to be explored. Strategies for improvement of cell adhesion, survival, and prevention of cell senescence are also essential to ensure therapeutic viability. Genetic engineering of EPCs may be a useful approach to developing these cells into efficient therapeutic tools. In the clinical arena there is pressing need to standardize the protocols for isolation, culture, and therapeutic application of EPC. Large-scale multi-center randomized trials are required to evaluate the long-term safety and efficacy of EPC therapy. Despite these hurdles, the outlook for EPC-based therapy for cardiovascular disease is promising.


Subject(s)
Cardiovascular Diseases/surgery , Endothelial Cells/transplantation , Stem Cell Transplantation , Aging , Animals , Cell Separation , Genetic Engineering , Humans , Salvage Therapy , Stem Cell Transplantation/adverse effects
18.
Trends Mol Med ; 11(5): 240-50, 2005 May.
Article in English | MEDLINE | ID: mdl-15882612

ABSTRACT

Recent advances in understanding the molecular and cellular basis of cardiovascular diseases, together with the availability of tools for genetic manipulation of the cardiovascular system, offer possibilities for new treatments. Gene therapies have demonstrated potential usefulness for treating complex cardiovascular diseases, such as hypertension, atherosclerosis and myocardial ischemia, in various animal models. Some of these experimental therapies are now undergoing clinical evaluation in patients with cardiovascular disease. However, the successful transition of these therapies into mainstream clinical practice awaits further improvements to vector platforms and delivery tools and the documentation of clinical feasibility, safety and efficacy through multi-center randomized trials.


Subject(s)
Cardiovascular Diseases/therapy , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/therapeutic use , Animals , Cardiovascular Diseases/metabolism , Gene Silencing , Gene Targeting/methods , Gene Transfer Techniques/trends , Genetic Therapy/trends , Humans
20.
Circulation ; 110(14): 2039-46, 2004 Oct 05.
Article in English | MEDLINE | ID: mdl-15451799

ABSTRACT

BACKGROUND: The existence of circulating endothelial progenitor cells (CEPCs) has previously been documented. These cells can be mobilized by cytokines and are recruited to sites of injury, where they may participate in tissue repair. In the present study, we examined the hypothesis that mobilization of CEPCs by exogenous granulocyte-colony stimulating factor (G-CSF) enhances repair of injured arteries by facilitating reendothelialization and inhibiting neointima development. METHODS AND RESULTS: Male rats were injected daily with 50 microg/kg recombinant human G-CSF or 0.9% NaCl SC for 8 days. On the fifth day of treatment, 1 mL of blood was collected for fluorescence-activated cell sorting analysis of mononuclear cells, and the animals underwent balloon angioplasty of the common carotid artery. The animals were killed at 2 or 4 weeks after injury, and the carotid arteries were harvested and processed for immunohistochemistry, scanning electron microscopy (SEM), and morphometric analysis of endothelialization and neointimal formation. G-CSF increased the number of circulating mononuclear cells that express endothelial cell lineage markers several-fold. SEM and immunohistochemical staining with the endothelial marker, platelet and endothelial cell adhesion molecule-1, showed rapid and nearly complete (>90%) reendothelialization of the denuded vessels in the G-CSF-treated animals compared with <20% in the control animals. Reendothelialization was paralleled by a decrease in inflammation in the vessel wall. Neointima thickness was reduced by approximately 60% in the G-CSF-treated animals compared with control animals at 2 and 4 weeks after injury. CONCLUSIONS: We postulate that cytokine-induced mobilization of CEPCs may be a suitable therapeutic strategy for prevention of restenosis after revascularization procedures.


Subject(s)
Angioplasty, Balloon/adverse effects , Carotid Artery Injuries/pathology , Carotid Artery, Common/ultrastructure , Endothelial Cells/physiology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Wound Healing/physiology , Animals , Carotid Artery Injuries/etiology , Cell Differentiation/drug effects , Cell Lineage , E-Selectin/analysis , Endothelial Cells/chemistry , Endothelial Cells/cytology , Hyperplasia , Male , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase Type III , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Splenectomy , Tunica Intima/ultrastructure , Vascular Cell Adhesion Molecule-1/analysis , Vascular Endothelial Growth Factor Receptor-2/analysis , Vasculitis/etiology , Vasculitis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...