Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Mol Cancer Ther ; 21(4): 568-581, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35149549

ABSTRACT

Clinical use of doxorubicin (Dox) is limited by cumulative myelo- and cardiotoxicity. This research focuses on the detailed characterization of PhAc-ALGP-Dox, a targeted tetrapeptide prodrug with a unique dual-step activation mechanism, designed to circumvent Dox-related toxicities and is ready for upcoming clinical investigation. Coupling Dox to a phosphonoacetyl (PhAc)-capped tetrapeptide forms the cell-impermeable, inactive compound, PhAc-ALGP-Dox. After extracellular cleavage by tumor-enriched thimet oligopeptidase-1 (THOP1), a cell-permeable but still biologically inactive dipeptide-conjugate is formed (GP-Dox), which is further processed intracellularly to Dox by fibroblast activation protein-alpha (FAPα) and/or dipeptidyl peptidase-4 (DPP4). In vitro, PhAc-ALGP-Dox is effective in various 2D- and 3D-cancer models, while showing improved safety toward normal epithelium, hematopoietic progenitors, and cardiomyocytes. In vivo, these results translate into a 10-fold higher tolerability and 5-fold greater retention of Dox in the tumor microenvironment compared with the parental drug. PhAc-ALGP-Dox demonstrates 63% to 96% tumor growth inhibition in preclinical models, an 8-fold improvement in efficacy in patient-derived xenograft (PDX) models, and reduced metastatic burden in a murine model of experimental lung metastasis, improving survival by 30%. The current findings highlight the potential clinical benefit of PhAc-ALGP-Dox, a targeted drug-conjugate with broad applicability, favorable tissue biodistribution, significantly improved tolerability, and tumor growth inhibition at primary and metastatic sites in numerous solid tumor models.


Subject(s)
Antineoplastic Agents , Lung Neoplasms , Prodrugs , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Humans , Lung Neoplasms/drug therapy , Mice , Prodrugs/pharmacology , Prodrugs/therapeutic use , Therapeutic Index , Tissue Distribution , Tumor Microenvironment
2.
Circulation ; 119(3): 408-16, 2009 Jan 27.
Article in English | MEDLINE | ID: mdl-19139381

ABSTRACT

BACKGROUND: Ventricular expression of phosphodiesterase-5 (PDE5), an enzyme responsible for cGMP catabolism, is increased in human right ventricular hypertrophy, but its role in left ventricular (LV) failure remains incompletely understood. We therefore measured LV PDE5 expression in patients with advanced systolic heart failure and characterized LV remodeling after myocardial infarction in transgenic mice with cardiomyocyte-specific overexpression of PDE5 (PDE5-TG). METHODS AND RESULTS: Immunoblot and immunohistochemistry techniques revealed that PDE5 expression was greater in explanted LVs from patients with dilated and ischemic cardiomyopathy than in control hearts. To evaluate the impact of increased ventricular PDE5 levels on cardiac function, PDE5-TG mice were generated. Confocal and immunoelectron microscopy revealed increased PDE5 expression in cardiomyocytes, predominantly localized to Z-bands. At baseline, myocardial cGMP levels, cell shortening, and calcium handling in isolated cardiomyocytes and LV hemodynamic measurements were similar in PDE5-TG and wild-type littermates. Ten days after myocardial infarction, LV cGMP levels had increased to a greater extent in wild-type mice than in PDE5-TG mice (P<0.05). Ten weeks after myocardial infarction, LV end-systolic and end-diastolic volumes were larger in PDE5-TG than in wild-type mice (57+/-5 versus 39+/-4 and 65+/-6 versus 48+/-4 muL, respectively; P<0.01 for both). LV systolic dysfunction and diastolic dysfunction were more marked in PDE5-TG than in wild-type mice, associated with enhanced hypertrophy and reduced contractile function in isolated cardiomyocytes from remote myocardium. CONCLUSIONS: Increased PDE5 expression predisposes mice to adverse LV remodeling after myocardial infarction. Increased myocardial PDE5 expression in patients with advanced cardiomyopathy may contribute to the development of heart failure and represents an important therapeutic target.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Gene Expression Regulation, Enzymologic/physiology , Heart Failure/enzymology , Myocardial Infarction/enzymology , Ventricular Remodeling/genetics , Animals , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Cyclic Nucleotide Phosphodiesterases, Type 5/physiology , Heart Failure/physiopathology , Heart Ventricles/enzymology , Heart Ventricles/physiopathology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Infarction/physiopathology , Myocardium/enzymology , Myocardium/pathology
3.
Basic Res Cardiol ; 105(2): 169-79, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20020305

ABSTRACT

Nitric oxide modulates the severity of myocardial ischemia-reperfusion (I/R) injury. We investigated whether cardioselective nitric oxide synthase 3 (NOS3) gene transfer could confer myocardial protection against I/R injury in pigs and examined potential molecular mechanisms. I/R injury was induced by balloon occlusion of the left anterior descending artery for 45 min followed by 4 or 72 h reperfusion. Hemodynamic and pathological changes were measured in pigs in the absence (n = 11) or presence of prior intracoronary retroinfusion of human NOS3 (AdNOS3, 5 x 10(10) PFU, n = 13) or control vector (AdRR5, 5 x 10(10) PFU, n = 11). Retrograde NOS3 gene transfer selectively increased NOS3 expression and NO bioavailability in the area at risk (AAR) without changing endogenous NOS isoform expression. At 4 h R, LV systolic (dP/dt(max)) and diastolic (dP/dt(min)) function was better preserved in AdNOS3- than in AdRR5-injected pigs (2,539 +/- 165 vs. 1,829 +/- 156 mmHg/s, and -2,781 +/- 340 vs. -2,062 +/- 292 mmHg/s, respectively, P < 0.05 for both). Myocardial infarct size (% AAR) was significantly smaller in AdNOS3 than in control and AdRR5 and associated with a significantly greater reduction in cardiac myeloperoxidase activity, a marker of neutrophil infiltration. The latter effects were sustained at 72 h R in a subset of pigs (n = 7). In the AAR, intercellular endothelial adhesion molecule-1 expression and cardiomyocyte apoptosis were significantly lower in AdNOS3. In conclusion, single myocardial NOS3 retroinfusion attenuates I/R injury, and causes a sustained reduction in myocardial infarct size and inflammatory cell infiltration. Gene-based strategies to increase NO bioavailability may have therapeutic potential in myocardial I/R.


Subject(s)
Genetic Therapy , Myocardial Reperfusion Injury/prevention & control , Nitric Oxide Synthase Type III/genetics , Animals , Apoptosis , Endothelial Cells/physiology , Hemodynamics , Leukocytes/physiology , Myocardial Infarction/etiology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Nitric Oxide Synthase Type III/metabolism , Random Allocation , Swine , Transgenes
4.
Stem Cells ; 26(4): 1017-26, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18258720

ABSTRACT

Circulating endothelial progenitor cells (EPCs) contribute to neovascularization of ischemic tissues and repair of injured endothelium. The role of bone marrow-derived progenitor cells in hypoxia-induced pulmonary vascular remodeling and their tissue-engineering potential in pulmonary hypertension (PH) remain largely unknown. We studied endogenous mobilization and homing of EPCs in green fluorescent protein bone marrow chimeric mice exposed to chronic hypoxia, a common hallmark of PH. Despite increased peripheral mobilization, as shown by flow cytometry and EPC culture, bone marrow-derived endothelial cell recruitment in remodeling lung vessels was limited. Moreover, transfer of vascular endothelial growth factor receptor-2+/Sca-1+/CXCR-4+-cultured early-outgrowth EPCs failed to reverse PH, suggesting hypoxia-induced functional impairment of transferred EPCs. Chronic hypoxia decreased migration to stromal cell-derived factor-1alpha, adhesion to fibronectin, incorporation into a vascular network, and nitric oxide production (-41%, -29%, -30%, and -32%, respectively, vs. normoxic EPCs; p < .05 for all). The dysfunctional phenotype of hypoxic EPCs significantly impaired their neovascularization capacity in chronic hind limb ischemia, contrary to normoxic EPCs cultured in identical conditions. Mechanisms contributing to EPC dysfunction include reduced integrin alphav and beta1 expression, decreased mitochondrial membrane potential, and enhanced senescence. Novel insights from chronic hypoxia-induced EPC dysfunction may provide important cues for improved future cell repair strategies.


Subject(s)
Endothelial Cells/physiology , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Stem Cells/pathology , Animals , Cells, Cultured , Chronic Disease , Endothelial Cells/pathology , Endothelial Cells/transplantation , Hindlimb/blood supply , Hindlimb/surgery , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/surgery , Hypoxia/pathology , Hypoxia/surgery , Mice , Mice, Inbred C57BL , Stem Cells/physiology , Time Factors
5.
Circulation ; 116(8): 936-43, 2007 Aug 21.
Article in English | MEDLINE | ID: mdl-17679618

ABSTRACT

BACKGROUND: Nitric oxide (NO) activates soluble guanylate cyclase (sGC), a heterodimer composed of alpha- and beta-subunits, to produce cGMP. NO reduces pulmonary vascular remodeling, but the role of sGC in vascular responses to acute and chronic hypoxia remains incompletely elucidated. We therefore studied pulmonary vascular responses to acute and chronic hypoxia in wild-type (WT) mice and mice with a nonfunctional alpha1-subunit (sGCalpha1-/-). METHODS AND RESULTS: sGCalpha1-/- mice had significantly reduced lung sGC activity and vasodilator-stimulated phosphoprotein phosphorylation. Right ventricular systolic pressure did not differ between genotypes at baseline and increased similarly in WT (22+/-2 to 34+/-2 mm Hg) and sGCalpha1-/- (23+/-2 to 34+/-1 mm Hg) mice in response to acute hypoxia. Inhaled NO (40 ppm) blunted the increase in right ventricular systolic pressure in WT mice (22+/-2 to 24+/-2 mm Hg, P<0.01 versus hypoxia without NO) but not in sGCalpha1-/- mice (22+/-1 to 33+/-1 mm Hg) and was accompanied by a significant rise in lung cGMP content only in WT mice. In contrast, the NO-donor sodium nitroprusside (1.5 mg/kg) decreased systemic blood pressure similarly in awake WT and sGCalpha1-/- mice as measured by telemetry (-37+/-2 versus -42+/-4 mm Hg). After 3 weeks of hypoxia, the increases in right ventricular systolic pressure, right ventricular hypertrophy, and muscularization of intra-acinar pulmonary vessels were 43%, 135%, and 46% greater, respectively, in sGCalpha1-/- than in WT mice (P<0.01). Increased remodeling in sGCalpha1-/- mice was associated with an increased frequency of 5'-bromo-deoxyuridine-positive vessels after 1 and 3 weeks (P<0.01 versus WT). CONCLUSIONS: Deficiency of sGCalpha1 does not alter hypoxic pulmonary vasoconstriction. sGCalpha1 is essential for NO-mediated pulmonary vasodilation and limits chronic hypoxia-induced pulmonary vascular remodeling.


Subject(s)
Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Nitric Oxide/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Vasodilation/physiology , Acute Disease , Animals , Antimetabolites/pharmacokinetics , Blood Pressure/physiology , Bromodeoxyuridine/pharmacokinetics , Chronic Disease , Cyclic GMP/metabolism , Dimerization , Female , Guanylate Cyclase/chemistry , Hypertension, Pulmonary/metabolism , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/metabolism , Male , Mice , Mice, Mutant Strains , Pulmonary Artery/physiology , Pulmonary Circulation/physiology , Receptors, Cytoplasmic and Nuclear/chemistry , Soluble Guanylyl Cyclase , Ventricular Function, Right/physiology
6.
Circ Res ; 94(9): 1256-62, 2004 May 14.
Article in English | MEDLINE | ID: mdl-15044322

ABSTRACT

Nitric oxide (NO) is an important modulator of cardiac performance and left ventricular (LV) remodeling after myocardial infarction (MI). We tested the effect of cardiomyocyte-restricted overexpression of one NO synthase isoform, NOS3, on LV remodeling after MI in mice. LV structure and function before and after permanent LAD coronary artery ligation were compared in transgenic mice with cardiomyocyte-restricted NOS3 overexpression (NOS3-TG) and their wild-type littermates (WT). Before MI, systemic hemodynamic measurements, echocardiographic assessment of LV fractional shortening (FS), heart weight, and myocyte width (as assessed histologically) did not differ in NOS3-TG and WT mice. The inotropic response to graded doses of isoproterenol was significantly reduced in NOS3-TG mice. One week after LAD ligation, the infarcted fraction of the LV did not differ in WT and NOS3-TG mice (34+/-4% versus 36+/-12%, respectively). Four weeks after MI, however, end-systolic LVID was greater, and fractional shortening and maximum and minimum rates of LV pressure development were less in WT than in NOS3-TG mice. LV weight/body weight ratio was greater in WT than in NOS3-TG mice (5.3+/-0.2 versus 4.6+/-0.5 mg/g; P<0.01). Myocyte width in noninfarcted myocardium was greater in WT than in NOS3-TG mice (18.8+/-2.0 versus 16.6+/-1.6 microm; P<0.05), whereas fibrosis in noninfarcted myocardium was similar in both genotypes. Cardiomyocyte-restricted overexpression of NOS3 limits LV dysfunction and remodeling after MI, in part by decreasing myocyte hypertrophy in noninfarcted myocardium.


Subject(s)
Hypertrophy, Left Ventricular/enzymology , Myocardial Infarction/complications , Myocytes, Cardiac/metabolism , Ventricular Remodeling/physiology , Adrenergic beta-Agonists/pharmacology , Animals , Enzyme Induction , Fibrosis , Humans , Hypertrophy , Hypertrophy, Left Ventricular/diagnostic imaging , Hypertrophy, Left Ventricular/etiology , Isoproterenol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Contraction/drug effects , Nitric Oxide Synthase , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Organ Specificity , Promoter Regions, Genetic , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/physiology , Transgenes , Ultrasonography , Ventricular Function, Left/physiology , Ventricular Myosins/genetics
7.
Hum Gene Ther ; 13(18): 2185-95, 2002 Dec 10.
Article in English | MEDLINE | ID: mdl-12542849

ABSTRACT

The utility of adenoviral vectors, currently used in cardiovascular gene transfer protocols, is limited by the brevity of transgene expression and by antiadenoviral immune responses. The effect of preexisting antiadenoviral immunity on intracardiac gene transfer or its modulation by nitric oxide is unknown. Adenoviral vectors, expressing the firefly luciferase gene (AdLuc) or the human nitric oxide synthase 3 (NOS3) gene (AdNOS3), were infused into the great cardiac vein of naive pigs or immunized pigs. Pigs were immunized by intravenous injection of control virus AdRR5 and the resulting neutralizing antibody titers (median, 1:178; p < 0.0001 vs. baseline) were similar to preexisting titers in 54% of randomly selected coronary artery bypass graft patients. In naive animals distribution of transgene expression in the left ventricular free wall was focal. In immunized pigs myocardial luciferase expression 3 days after AdLuc gene transfer was more than 1000-fold lower than in naive pigs, whereas no change in NOS3 transcript levels was detected after AdNOS3 gene transfer. Severe, grade III-IV mononuclear cell infiltration and myocyte apoptosis were observed in four of five AdLuc-infected, immunized animals, compared with low-level inflammation and apoptosis in five of six AdNOS3-infected pigs. Coinfusion of AdLuc and AdNOS3 in immunized pigs resulted in spatially colocalized transgene expression, reduced T cell-mediated inflammation, and myocyte apoptosis and was associated with 200-fold greater median reporter transgene expression levels in the subendocardium (1.0 x 10(3) light units [LU]/mg protein, n = 8, vs. 4.5 x 10(1) LU/mg protein in AdLuc- and AdRR5-coinfected pigs, n = 7, p = 0.02). Preexisting antiadenoviral immunity abrogates myocardial gene expression in pigs and is associated with severe inflammation and myocyte apoptosis. Intracardiac NOS3 gene transfer may reduce these barriers to adenovirus-mediated myocardial gene transfer.


Subject(s)
Dependovirus/immunology , Genetic Vectors/immunology , Myocardium/immunology , Nitric Oxide/metabolism , Animals , Gene Expression Regulation/immunology , Gene Transfer Techniques , Genes, Reporter , Immunity, Innate , Myocarditis/genetics , Myocarditis/immunology , Myocarditis/therapy , Myocardium/metabolism , Swine
8.
PLoS One ; 8(3): e58841, 2013.
Article in English | MEDLINE | ID: mdl-23527037

ABSTRACT

BACKGROUND: The intracellular second messenger cGMP protects the heart under pathological conditions. We examined expression of phosphodiesterase 5 (PDE5), an enzyme that hydrolyzes cGMP, in human and mouse hearts subjected to sustained left ventricular (LV) pressure overload. We also determined the role of cardiac myocyte-specific PDE5 expression in adverse LV remodeling in mice after transverse aortic constriction (TAC). METHODOLOGY/PRINCIPAL FINDINGS: In patients with severe aortic stenosis (AS) undergoing valve replacement, we detected greater myocardial PDE5 expression than in control hearts. We observed robust expression in scattered cardiac myocytes of those AS patients with higher LV filling pressures and BNP serum levels. Following TAC, we detected similar, focal PDE5 expression in cardiac myocytes of C57BL/6NTac mice exhibiting the most pronounced LV remodeling. To examine the effect of cell-specific PDE5 expression, we subjected transgenic mice with cardiac myocyte-specific PDE5 overexpression (PDE5-TG) to TAC. LV hypertrophy and fibrosis were similar as in WT, but PDE5-TG had increased cardiac dimensions, and decreased dP/dtmax and dP/dtmin with prolonged tau (P<0.05 for all). Greater cardiac dysfunction in PDE5-TG was associated with reduced myocardial cGMP and SERCA2 levels, and higher passive force in cardiac myocytes in vitro. CONCLUSIONS/SIGNIFICANCE: Myocardial PDE5 expression is increased in the hearts of humans and mice with chronic pressure overload. Increased cardiac myocyte-specific PDE5 expression is a molecular hallmark in hypertrophic hearts with contractile failure, and represents an important therapeutic target.


Subject(s)
Cardiomegaly/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Myocytes, Cardiac/enzymology , Ventricular Remodeling , Animals , Aortic Valve Stenosis/complications , Calcium/metabolism , Cardiomegaly/etiology , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Extracellular Matrix , Gene Expression , Heart Ventricles/enzymology , Hemodynamics , Humans , Mice , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Time Factors
9.
J Am Coll Cardiol ; 55(20): 2232-43, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20466204

ABSTRACT

OBJECTIVES: We compared biological repair after acute myocardial infarction (AMI) with selected porcine progenitor cell populations. BACKGROUND: Cell types and mechanisms responsible for myocardial repair after AMI remain uncertain. METHODS: In a blinded, randomized study, we infused autologous late-outgrowth endothelial progenitor cells (EPC) (n = 10, 34 +/- 22 x 10(6) CD29-31-positive, capable of tube formation), allogeneic green fluorescent peptide-labeled mesenchymal stem cells (MSC) (n = 11, 10 +/- 2 x 10(6) CD29-44-90-positive, capable of adipogenic and osteogenic differentiation), or vehicle (CON) (n = 12) in the circumflex artery 1 week after AMI. Systolic function (ejection fraction), left ventricular (LV) end-diastolic and end-systolic volumes, and infarct size were assessed with magnetic resonance imaging at 1 week and 7 weeks. Cell engraftment and vascular density were evaluated on postmortem sections. RESULTS: Recovery of LV ejection fraction from 1 to 7 weeks was similar between groups, but LV remodeling markedly differed with a greater increase of LV end-systolic volume in MSC and CON (+11 +/- 12 ml/m(2) and +7 +/- 8 ml/m(2) vs. -3 +/- 11 ml/m(2) in EPC, respectively, p = 0.04), and a similar trend was noted for LV end-diastolic volume (p = 0.09). After EPC, infarct size decreased more in segments with >50% infarct transmurality (p = 0.02 vs. MSC and CON) and was associated with a greater vascular density (p = 0.01). Late outgrowth EPCs secrete higher levels of the pro-angiogenic placental growth factor (733 [277 to 1,214] pg/10(6) vs. 59 [34 to 88] pg/10(6) cells in MSC, p = 0.03) and incorporate in neovessels in vivo. CONCLUSIONS: Infusion of late-outgrowth EPCs after AMI improves myocardial infarction remodeling via enhanced neovascularization but does not mediate cardiomyogenesis. Endothelial progenitor cell transfer might hold promise for heart failure prevention via pro-angiogenic or paracrine matrix-modulating effects.


Subject(s)
Myocardial Infarction/physiopathology , Ventricular Remodeling/physiology , Animals , Cells, Cultured , Immunohistochemistry , Lac Operon/physiology , Magnetic Resonance Imaging, Cine , Matrix Metalloproteinases/blood , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Myocardial Reperfusion , Neovascularization, Physiologic/physiology , Paracrine Communication/physiology , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Transplantation/methods
10.
Open Cardiovasc Med J ; 3: 98-104, 2009 Aug 31.
Article in English | MEDLINE | ID: mdl-19771174

ABSTRACT

OBJECTIVE: Soluble guanylate cyclase (sGC), a heterodimer composed of alpha and beta subunits, synthesizes cGMP in response to nitric oxide (NO). NO modulates vascular tone and structure but the relative contributions of cGMP-dependent versus cGMP-independent mechanisms remain uncertain. We studied the response to vascular injury in male (M) and female (F) mice with targeted deletion of exon 6 of the sGCα1 subunit (sGCα1(-/-)), resulting in a non-functional heterodimer. METHODS: We measured aortic cGMP levels and mRNA transcripts encoding sGC α1, α2, and ß1 subunits in wild type (WT) and sGCa1(-/-) mice. To study the response to vascular injury, BrdU-incorporation and neointima formation (maximum intima to media (I/M) ratio) were determined 5 and 28 days after carotid artery ligation, respectively. RESULTS: Aortic cGMP levels were 4-fold higher in F than in M mice in both genotypes, and, within each gender, 4-fold higher in WT than in sGCa1(-/-). In contrast, sGCα1, sGCα2, and sGCß1 mRNA expression did not differ between groups. ³H-thymidine incorporation in cultured sGCa1(-/-) smooth muscle cells (SMC) was 27%±12% lower than in WT SMC and BrdU-incorporation in carotid arteries 5 days after ligation was significantly less in sGCa1(-/-) M than in WT M. Neointima area and I/M 28 days after ligation were 65% and 62% lower in sGCa1(-/-) M than in WT M mice (p<0,05 for both) but were not different in F mice. CONCLUSION: Functional deletion of sGCa1 resulted in reduced cGMP levels in male sGCa1(-/-) mice and a gender-specific effect on the adaptive response to vascular injury.

11.
J Am Coll Cardiol ; 50(8): 808-17, 2007 Aug 21.
Article in English | MEDLINE | ID: mdl-17707188

ABSTRACT

OBJECTIVES: The purpose of this study was to test if nitric oxide (NO) could improve microvascular perfusion and decrease tissue injury in a porcine model of myocardial ischemia and reperfusion (I/R). BACKGROUND: Inhaled NO is a selective pulmonary vasodilator with biologic effects in remote vascular beds. METHODS: In 37 pigs, the midportion of the left anterior descending coronary artery was occluded for 50 min followed by 4 h of reperfusion. Pigs were treated with a saline infusion (control; n = 14), intravenous nitroglycerin (IV-NTG) at 2 microg/kg/min (n = 11), or inhaled nitric oxide (iNO) at 80 parts per million (n = 12) beginning 10 min before balloon deflation and continuing throughout reperfusion. RESULTS: Total myocardial oxidized NO species in the infarct core was greater in the iNO pigs than in the control or IV-NTG pigs (0.60 +/- 0.05 nmol/mg tissue vs. 0.40 +/- 0.03 nmol/mg tissue and 0.40 +/- 0.02 nmol/mg tissue, respectively; p < 0.01 for both). Infarct size, expressed as percentage of left ventricle area at risk (AAR), was smaller in the iNO pigs than in the control or IV-NTG pigs (31 +/- 6% AAR vs. 58 +/- 7% AAR and 46 +/- 7% AAR, respectively; p < 0.05 for both) and was associated with less creatine phosphokinase-MB release. Inhaled NO improved endocardial and epicardial blood flow in the infarct zone, as measured using colored microspheres (p < 0.001 vs. control and IV-NTG). Moreover, NO inhalation reduced leukocyte infiltration, as reflected by decreased cardiac myeloperoxidase activity (0.8 +/- 0.2 U/mg tissue vs. 2.3 +/- 0.8 U/mg tissue in control and 1.4 +/- 0.4 U/mg tissue in IV-NTG; p < 0.05 for both) and decreased cardiomyocyte apoptosis in the infarct border zone. CONCLUSIONS: Inhalation of NO just before and during coronary reperfusion significantly improves microvascular perfusion, reduces infarct size, and may offer an attractive and novel treatment of myocardial infarction.


Subject(s)
Coronary Circulation/drug effects , Endothelium-Dependent Relaxing Factors/therapeutic use , Myocardial Ischemia/drug therapy , Myocardial Reperfusion Injury/prevention & control , Nitric Oxide/therapeutic use , Administration, Inhalation , Animals , Endothelium-Dependent Relaxing Factors/pharmacology , Female , Male , Microcirculation/drug effects , Myocardial Infarction/pathology , Myocardial Ischemia/complications , Myocardial Ischemia/physiopathology , Myocardium/enzymology , Myocardium/metabolism , Myocardium/pathology , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Oxidation-Reduction , Peroxidase/blood , Peroxidase/metabolism , Swine , Ventricular Function, Left/drug effects
12.
Hypertension ; 47(4): 762-70, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16505204

ABSTRACT

We assessed pulmonary cytochrome P450 (CYP) epoxygenase expression and activity during hypoxia and explored the effects of modulating epoxygenase activity on pulmonary hypertension. The acute hypoxic vasoconstrictor response was studied in Swiss Webster mice, who express CYP2C29 in their lungs. Animals were pretreated with vehicle, the epoxygenase inhibitor (N-methylsulfonyl-6-[2-propargyloxyphenyl] hexanamide) or an inhibitor of the soluble epoxide hydrolase. Whereas the epoxygenase inhibitor attenuated hypoxic pulmonary constriction (by 52%), the soluble epoxide hydrolase inhibitor enhanced the response (by 39%), indicating that CYP epoxygenase-derived epoxyeicosatrienoic acids elicit pulmonary vasoconstriction. Aerosol gene transfer of recombinant adenovirus containing the human CYP2C9 significantly elevated mean pulmonary artery pressure and total pulmonary resistance indices, both of which were sensitive to the inhibitor sulfaphenazole. The prolonged exposure of mice to hypoxia increased CYP2C29 expression, and transcript levels increased 5-fold after exposure to normobaric hypoxia (FIO2 0.07) for 2 hours. This was followed by a 2-fold increase in protein expression and by a significant increase in epoxyeicosatrienoic acid production after 24 hours. Chronic hypoxia (7 days) elicited pulmonary hypertension and pulmonary vascular remodeling, effects that were significantly attenuated in animals continually treated with N-methylsulfonyl-6-[2-propargyloxyphenyl] hexanamide (-46% and -55%, respectively). Our results indicate that endogenously generated epoxygenase products are associated with hypoxic pulmonary hypertension in mice and that selective epoxygenase inhibition significantly reduces acute hypoxic pulmonary vasoconstriction and chronic hypoxia-induced pulmonary vascular remodeling. These observations indicate potential novel targets for the treatment of pulmonary hypertension and highlight a pivotal role for CYP epoxygenases in pulmonary responses to hypoxia.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Hypoxia/physiopathology , Lung/blood supply , Oxygenases/metabolism , Vasoconstriction , Adenoviridae/genetics , Amides/pharmacology , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Blood Vessels/physiopathology , Chronic Disease , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme System/adverse effects , Cytochrome P-450 Enzyme System/genetics , Cytochrome P450 Family 2 , Gene Transfer Techniques , Genetic Vectors , Hemodynamics , Humans , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/physiopathology , Hypoxia/genetics , Lung/enzymology , Mice , Oxygenases/adverse effects , Oxygenases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL