Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Am J Physiol Heart Circ Physiol ; 310(2): H290-9, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26566724

ABSTRACT

Marfan syndrome (MFS) is a systemic disorder of connective tissue caused by mutations in fibrillin-1. Cardiac dysfunction in MFS has not been characterized halting the development of therapies of cardiac complication in MFS. We aimed to study the age-dependent cardiac remodeling in the mouse model of MFS FbnC1039G+/- mouse [Marfan heterozygous (HT) mouse] and its association with valvular regurgitation. Marfan HT mice of 2-4 mo demonstrated a mild hypertrophic cardiac remodeling with predominant decline of diastolic function and increased transforming growth factor-ß canonical (p-SMAD2/3) and noncanonical (p-ERK1/2 and p-p38 MAPK) signaling and upregulation of hypertrophic markers natriuretic peptides atrium natriuretic peptide and brain natriuretic peptide. Among older HT mice (6-14 mo), cardiac remodeling was associated with two distinct phenotypes, manifesting either dilated or constricted left ventricular chamber. Dilatation of left ventricular chamber was accompanied by biochemical evidence of greater mechanical stress, including elevated ERK1/2 and p38 MAPK phosphorylation and higher brain natriuretic peptide expression. The aortic valve regurgitation was registered in 20% of the constricted group and 60% of the dilated group, whereas mitral insufficiency was observed in 40% of the constricted group and 100% of the dilated group. Cardiac dysfunction was not associated with the increase of interstitial fibrosis and nonmyocyte proliferation. In the mouse model fibrillin-1, haploinsufficiency results in the early onset of nonfibrotic hypertrophic cardiac remodeling and dysfunction, independently from valvular abnormalities. MFS heart is vulnerable to stress-induced cardiac dilatation in the face of valvular regurgitation, and stress-activated MAPK signals represent a potential target for cardiac management in MFS.


Subject(s)
Marfan Syndrome/pathology , Myocardium/pathology , Animals , Cardiomegaly/diagnostic imaging , Cardiomegaly/pathology , Fibrillin-1 , Fibrillins , Fibrosis/pathology , Hemodynamics , MAP Kinase Signaling System , Male , Marfan Syndrome/diagnostic imaging , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Mitral Valve Insufficiency/etiology , Mitral Valve Insufficiency/pathology , Phenotype , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Ultrasonography , Ventricular Function, Left , p38 Mitogen-Activated Protein Kinases/biosynthesis , p38 Mitogen-Activated Protein Kinases/genetics
2.
Front Cardiovasc Med ; 9: 1096887, 2022.
Article in English | MEDLINE | ID: mdl-36741836

ABSTRACT

Background: Aging is associated with increased levels of reactive oxygen species and inflammation that disrupt proteostasis and mitochondrial function and leads to organism-wide frailty later in life. ARA290 (cibinetide), an 11-aa non-hematopoietic peptide sequence within the cardioprotective domain of erythropoietin, mediates tissue protection by reducing inflammation and fibrosis. Age-associated cardiac inflammation is linked to structural and functional changes in the heart, including mitochondrial dysfunction, impaired proteostasis, hypertrophic cardiac remodeling, and contractile dysfunction. Can ARA290 ameliorate these age-associated cardiac changes and the severity of frailty in advanced age? Methods: We conducted an integrated longitudinal (n = 48) and cross-sectional (n = 144) 15 months randomized controlled trial in which 18-month-old Fischer 344 x Brown Norway rats were randomly assigned to either receive chronic ARA290 treatment or saline. Serial echocardiography, tail blood pressure and body weight were evaluated repeatedly at 4-month intervals. A frailty index was calculated at the final timepoint (33 months of age). Tissues were harvested at 4-month intervals to define inflammatory markers and left ventricular tissue remodeling. Mitochondrial and myocardial cell health was assessed in isolated left ventricular myocytes. Kaplan-Meier survival curves were established. Mixed ANOVA tests and linear mixed regression analysis were employed to determine the effects of age, treatment, and age-treatment interactions. Results: Chronic ARA290 treatment mitigated age-related increases in the cardiac non-myocyte to myocyte ratio, infiltrating leukocytes and monocytes, pro-inflammatory cytokines, total NF-κB, and p-NF-κB. Additionally, ARA290 treatment enhanced cardiomyocyte autophagy flux and reduced cellular accumulation of lipofuscin. The cardiomyocyte mitochondrial permeability transition pore response to oxidant stress was desensitized following chronic ARA290 treatment. Concurrently, ARA290 significantly blunted the age-associated elevation in blood pressure and preserved the LV ejection fraction. Finally, ARA290 preserved body weight and significantly reduced other markers of organism-wide frailty at the end of life. Conclusion: Administration of ARA290 reduces cell and tissue inflammation, mitigates structural and functional changes within the cardiovascular system leading to amelioration of frailty and preserved healthspan.

3.
J Pharmacol Exp Ther ; 337(3): 621-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21363928

ABSTRACT

The vascular form of Ehlers-Danlos syndrome (vEDS), a rare disease with grave complications resulting from rupture of major arteries, is caused by mutations of collagen type III [α1 chain of collagen type III (COL3A1)]. The only, recently proven, preventive strategy consists of the reduction of arterial wall stress by ß-adrenergic blockers. The heterozygous (HT) Col3a1 knockout mouse has reduced expression of collagen III and recapitulates features of a mild presentation of the disease. The objective of this study was to determine whether changing the balance between synthesis and degradation of collagen by chronic treatment with doxycycline, a nonspecific matrix metalloproteinase (MMP) inhibitor, could prevent the development of vascular pathology in HT mice. After 3 months of treatment with doxycycline or placebo, 9-month-old HT or wild-type (WT) mice were subjected to surgical stressing of the aorta. A 3-fold increase in stress-induced aortic lesions found in untreated HT mice 1 week after intervention (cumulative score 4.5 ± 0.87 versus 1.3 ± 0.34 in WT, p < 0.001) was fully prevented in the doxycycline-treated group (1.1 ± 0.56, p < 0.001). Untreated HT mice showed increased MMP-9 activity in the carotid artery and decreased collagen content in the aorta; however, in doxycycline-treated animals there was normalization to the levels observed in WT mice. Doxycycline treatment inhibits the activity of tissue MMP and attenuates the decrease in the collagen content in aortas of mice haploinsufficient for collagen III, as well as prevents the development of stress-induced vessel pathology. The results suggest that doxycycline merits clinical testing as a treatment for vEDS.


Subject(s)
Aortic Diseases/prevention & control , Collagen Type III/metabolism , Doxycycline/pharmacology , Ehlers-Danlos Syndrome/drug therapy , Ehlers-Danlos Syndrome/pathology , Matrix Metalloproteinase Inhibitors , Protease Inhibitors/pharmacology , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Aorta/physiopathology , Aortic Diseases/pathology , Carotid Arteries/drug effects , Carotid Arteries/enzymology , Colon/drug effects , Colon/physiology , Ehlers-Danlos Syndrome/physiopathology , Extracellular Matrix Proteins/metabolism , Female , Matrix Metalloproteinases/blood , Matrix Metalloproteinases/metabolism , Mice , Mice, Knockout , Skin/drug effects , Skin/enzymology
4.
J Pharmacol Exp Ther ; 325(2): 491-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18287209

ABSTRACT

We have reported therapeutic effectiveness of pharmacological stimulation of beta2 adrenoreceptors (ARs) to attenuate the cardiac remodeling and myocardial infarction (MI) expansion in a rat model of dilated cardiomyopathy (DCM) post-MI. Furthermore, the combination of beta2 AR stimulation with beta1 AR blockade exceeded the therapeutic effectiveness of beta1 AR blockade. However, these studies were relatively short (6 weeks). In this study, in the same experimental model, we compared different effects, including survival benefit, of combined therapy with the beta1 AR blocker, metoprolol, plus the beta2 AR agonist, fenoterol (beta1-beta2+), and either therapy alone (beta1- or beta2+) during the 1-year study. Therapy was started 2 weeks after permanent ligation of the left coronary artery. Cardiac remodeling, MI expansion, and left ventricular function were assessed by serial echocardiography and compared with untreated animals (nT). Sixty-seven percent mortality in nT was reduced to 33% in the beta1-beta2+ (p < 0.01). Progressive cardiac remodeling observed in nT and beta1- was significantly attenuated in beta1-beta2+ during the first 6 months of treatment. In beta1-beta2+, MI expansion was completely prevented, and functional decline was significantly attenuated during the entire year. Myocardial apoptosis was significantly reduced in both beta1-beta2+ and beta1-. A reduction of cardiac beta1 AR density and decreases in chronotropic and contractile responses to beta2 AR-specific stimulation in the absence of a reduction of beta2 AR density in nT were precluded in rats receiving combined therapy. The results demonstrate the cardioprotective and survival benefit of long-term combination therapy of beta2 AR agonists and beta1 AR blockers in a model of DCM.


Subject(s)
Adrenergic beta-1 Receptor Antagonists , Adrenergic beta-2 Receptor Agonists , Adrenergic beta-Agonists/therapeutic use , Adrenergic beta-Antagonists/therapeutic use , Cardiomyopathy, Dilated/drug therapy , Cardiotonic Agents/therapeutic use , Fenoterol/therapeutic use , Metoprolol/therapeutic use , Animals , Cardiac Volume/drug effects , Cardiomyopathy, Dilated/physiopathology , Drug Therapy, Combination , Electrocardiography , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Male , Myocardial Infarction/drug therapy , Myocardial Infarction/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Rats , Rats, Wistar , Receptors, Adrenergic, beta-1/physiology , Receptors, Adrenergic, beta-2/physiology , Ventricular Remodeling/drug effects
5.
Circulation ; 110(9): 1083-90, 2004 Aug 31.
Article in English | MEDLINE | ID: mdl-15313944

ABSTRACT

BACKGROUND: Studies in isolated cardiac myocytes have demonstrated that signaling via specific beta1-adrenergic receptor subtypes (beta1ARs) promotes but that signaling via beta2ARs protects from cell death. We hypothesized that prolonged beta(2)AR stimulation or beta1AR blockade would each protect myocytes from death and thereby ameliorate cardiac remodeling in chronic heart failure. METHODS AND RESULTS: A large myocardial infarction (MI) induced in rats by coronary artery ligation resulted in a dilated cardiomyopathy (DCM) characterized by infarct expansion and a progressive increase in left ventricular (LV) end-diastolic volume, accompanied by a reduction in ejection fraction (EF), as assessed by repeated echocardiography. Pressure-volume analysis at 8 weeks after ligation showed that diastolic stiffness (Eed) and arterial elastance (Ea) were increased, end-systolic elastance (Ees) was decreased, and arterioventricular (AV) coupling (Ea/Ees) had deteriorated. Apoptosis was present in both peri-infarct and remote myocardium. Chronic (6-week) administration of the beta2AR agonists fenoterol or zinterol, starting at 2 weeks after MI, reduced the extent of LV dilation, infarct expansion, and EF decline. The beta1AR blocker metoprolol did not affect the former and preserved EF to a lesser extent than did the beta2AR agonists. At 8 weeks after ligation, apoptosis was reduced by all drugs but to a greater extent by beta2AR agonists than by the beta1AR blocker. Both beta2AR agonists and the beta1AR blocker improved AV coupling, the former mainly by reducing Ea and the latter mainly by increasing Ees. Only the beta2AR agonists reduced the Eed and the MI size by reducing infarct expansion. CONCLUSIONS: These results provide proof of concept for the efficacy of chronic beta2AR stimulation in this DCM model.


Subject(s)
Adrenergic beta-1 Receptor Antagonists , Adrenergic beta-2 Receptor Agonists , Adrenergic beta-Agonists/therapeutic use , Adrenergic beta-Antagonists/therapeutic use , Cardiomyopathy, Dilated/drug therapy , Ethanolamines/therapeutic use , Fenoterol/therapeutic use , Metoprolol/therapeutic use , Myocardial Infarction/drug therapy , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Apoptosis/drug effects , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cell Size , Ethanolamines/pharmacology , Fenoterol/pharmacology , Male , Metoprolol/pharmacology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac/pathology , Organ Size , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Ventricular Function, Left/drug effects
6.
Sci Rep ; 1: 70, 2011.
Article in English | MEDLINE | ID: mdl-22355589

ABSTRACT

Sirt1 is an NAD(+)-dependent deacetylase that extends lifespan in lower organisms and improves metabolism and delays the onset of age-related diseases in mammals. Here we show that SRT1720, a synthetic compound that was identified for its ability to activate Sirt1 in vitro, extends both mean and maximum lifespan of adult mice fed a high-fat diet. This lifespan extension is accompanied by health benefits including reduced liver steatosis, increased insulin sensitivity, enhanced locomotor activity and normalization of gene expression profiles and markers of inflammation and apoptosis, all in the absence of any observable toxicity. Using a conditional SIRT1 knockout mouse and specific gene knockdowns we show SRT1720 affects mitochondrial respiration in a Sirt1- and PGC-1α-dependent manner. These findings indicate that SRT1720 has long-term benefits and demonstrate for the first time the feasibility of designing novel molecules that are safe and effective in promoting longevity and preventing multiple age-related diseases in mammals.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/pharmacology , Longevity/drug effects , Obesity/physiopathology , Animals , Apoptosis/drug effects , Body Composition/drug effects , Dietary Fats/administration & dosage , Gene Expression/drug effects , Glucose/metabolism , Homeostasis/drug effects , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Pancreas/drug effects
7.
J Biol Chem ; 282(48): 34984-93, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17925408

ABSTRACT

Pleiotrophin is a development-regulated cytokine and growth factor that can promote angiogenesis, cell proliferation, or differentiation, and it has been reported to have neovasculogenic effects in damaged heart. Developmentally, it is prominently expressed in fetal and neonatal hearts, but it is minimally expressed in normal adult heart. Conversely, we show in a rat model of myocardial infarction and in human dilated cardiomyopathy that pleiotrophin is markedly up-regulated. To elucidate the effects of pleiotrophin on cardiac contractile cells, we employed primary cultures of rat neonatal and adult cardiomyocytes. We show that pleiotrophin is released from cardiomyocytes in vitro in response to hypoxia and that the addition of recombinant pleiotrophin promotes caspase-mediated genomic DNA fragmentation in a dose- and time-dependent manner. Functionally, it potentiates the apoptotic response of neonatal cardiomyocytes to hypoxic stress and to ultraviolet irradiation and of adult cardiomyocytes to hypoxia-reoxygenation. Moreover, UV-induced apoptosis in neonatal cardiomyocytes can be partially inhibited by small interfering RNA-mediated knockdown of endogenous pleiotrophin. Mechanistically, pleiotrophin antagonizes IGF-1 associated Ser-473 phosphorylation of AKT/PKB, and it concomitantly decreases both BAD and GSK3beta phosphorylation. Adenoviral expression of constitutively active AKT and lithium chloride-mediated inhibition of GSK3beta reduce the potentiated programmed cell death elicited by pleiotrophin. These latter data indicate that pleiotrophin potentiates cardiomyocyte cell death, at least partially, through inhibition of AKT signaling. In conclusion, we have uncovered a novel function for pleiotrophin on heart cells following injury. It fosters cardiomyocyte programmed cell death in response to pro-apoptotic stress, which may be critical to myocardial injury repair.


Subject(s)
Carrier Proteins/metabolism , Cytokines/metabolism , Myocytes, Cardiac/metabolism , Neovascularization, Pathologic , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Hypoxia , Male , Myocardium/metabolism , Phosphorylation , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
8.
Cardiovasc Drugs Ther ; 20(4): 245-51, 2006 Aug.
Article in English | MEDLINE | ID: mdl-17019537

ABSTRACT

BACKGROUND: Systemic application of recombinant human erythropoietin (rhEPO) greatly limits cardiac tissue damage and attenuates left ventricular (LV) remodeling after experimentally induced myocardial infarction (MI). However, multiple injections of rhEPO stimulate red blood cell production and elevate the hematocrit (Htc), which might negatively affect the outcome of acute MI. We compared the outcome of experimental MI in rats treated with a single or multiple doses of rhEPO. MATERIALS AND METHODS: Sprague-Dawley male rats were subjected to a permanent ligation of the left descending coronary artery (CL) or sham operation. Immediately after CL animals received either a single i.v. injection of 3,000 IU/kg of rhEPO, or a single injection plus additional injections of the same dose of rhEPO repeated daily for six more days. Echocardiography and blood collection for measurement of Htc were performed prior to, and at 2 and 4 weeks after CL; MI size was measured histologically 4 weeks after CL. RESULTS: A single injection of rhEPO elevated Htc by 11% (p < 0.05) 1 week after CL, but after multiple rhEPO injections Htc increased by 40%. In untreated rats a 140 and 340% expansion in end-diastolic and end-systolic LV volumes, respectively, and 55% decline in ejection fraction (EF) occurred during the 4 week period following CL. A single rhEPO dose attenuated the LV remodeling and EF reduction by 50%. Repeated rhEPO injections did not elicit any additional benefits in respect to LV remodeling. Moreover, at the end of 4 weeks, MI size was significantly reduced (by 40%) by a single injection, while after repeated rhEPO injections the reduction of MI size was not statistically significant. CONCLUSION: The results of this study indicate that multiple dosing of rhEPO after induced myocardial infarction in rats has no added therapeutic benefits over those achieved by a single dose.


Subject(s)
Cardiotonic Agents/therapeutic use , Erythropoietin/therapeutic use , Myocardial Infarction/drug therapy , Animals , Cardiotonic Agents/administration & dosage , Echocardiography , Erythropoietin/administration & dosage , Heart Ventricles/pathology , Hematocrit , Male , Myocardial Infarction/pathology , Rats , Rats, Sprague-Dawley , Recombinant Proteins , Ventricular Remodeling/drug effects
9.
J Pharmacol Exp Ther ; 316(3): 999-1005, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16306273

ABSTRACT

Erythropoietin (EPO), a hematopoietic cytokine, possesses strong antiapoptotic, tissue-protective properties. For clinical applications, it is desirable to separate the hematopoietic and tissue-protective properties. Recently introduced carbamylated erythropoietin (CEPO) does not stimulate the erythropoiesis but retains the antiapoptotic and neuroprotective effects. We tested the ability of CEPO to protect cardiac tissue from toxin-induced and oxidative stress in vitro and ischemic damage in vivo and compared these effects with the effects of EPO. CEPO reduced by 50% the extent of staurosporine-induced apoptosis in isolated rats' cardiomyocytes and increased by 25% the reactive oxygen species threshold for induction of the mitochondrial permeability transition. In an experimental model of myocardial infarction induced by permanent ligation of a coronary artery in rats, similarly to EPO, a single bolus injection of 30 mug/kg b.wt. of CEPO immediately after coronary ligation reduced apoptosis in the myocardial area at risk, examined 24 h later, by 50%. Left ventricular remodeling (ventricular dilation) and functional decline (fall in ejection fraction) assessed by repeated echocardiography were significantly and similarly attenuated in CEPO- and EPO-treated rats. Four weeks after coronary ligation, the myocardial infarction (MI) size in CEPO- and EPO-treated rats was half of that in untreated coronary-ligated animals. Unlike EPO, CEPO had no effect on hematocrit. The antiapoptotic cardioprotective effects of CEPO, shown by its ability to limit both post-MI left ventricular remodeling and the extent of the myocardial scar in the model of permanent coronary artery ligation in rats, demonstrate comparable potency to that of native (nonmodified) EPO.


Subject(s)
Apoptosis/drug effects , Erythropoiesis/drug effects , Erythropoietin/pharmacology , Heart/drug effects , Ventricular Remodeling/drug effects , Animals , Cells, Cultured , Echocardiography , Erythropoietin/analogs & derivatives , Ion Channels/drug effects , Male , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Protective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species
10.
Cardiovasc Drugs Ther ; 19(4): 243-50, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16187008

ABSTRACT

BACKGROUND: Recombinant human erythropoietin (rhEPO) protects tissue from ischemic damage, but translation of this finding into useful guidelines with respect to human trials for myocardial infarction (MI) requires a determination of the minimum effective rhEPO dose and the therapeutic window following MI. METHOD AND RESULTS: Serial echocardiography revealed that during four weeks following MI, induced by a permanent coronary ligation in rats, the LV end-diastolic and end-systolic volumes in untreated rats expanded from 0.35 +/- 0.01 and 0.14 +/- 0.01 ml to 0.84 +/- 0.04 and 0.61 +/- 0.06 ml, respectively, and ejection fraction (EF) reduced by 50%. A single i.v. injection of rhEPO immediately following MI in a dose of 150 IU/kg was as effective as 3,000 IU/kg in causing a 2-fold reduction of the number of apoptotic nuclei in the AAR 24-h later, a 2-fold reduction of the MI size measured 4 weeks later, attenuation of progressive LV dilatation and fall in EF. A 3000 IU/kg dose had similar therapeutic effects when delayed by 4, 8, or 12 h following MI, but was not effective after a 24-h delay. A single dose of 150 IU/kg was effective within 4 h post-MI, but was without effect if administered after an 8-h delay. CONCLUSION: Cell death, final MI size, myocardial remodeling and functional decline are significantly reduced in rats by a single injection of rhEPO in a dose as low as 150 IU/kg if administered during the first 4 h after the ischemic event. Higher doses extend the therapeutic window up to 12 h.


Subject(s)
Cardiotonic Agents/therapeutic use , Erythropoietin/therapeutic use , Myocardial Infarction/drug therapy , Animals , Apoptosis , Cardiotonic Agents/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Echocardiography , Erythropoietin/administration & dosage , Humans , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley , Recombinant Proteins
11.
Am J Pathol ; 167(5): 1429-42, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16251426

ABSTRACT

Increased angiotensin II (Ang II), matrix metalloproteinase type II (MMP2), and sympathetic activity accompany age-associated arterial remodeling. To analyze this relationship, we infused a low subpressor dose of Ang II into young (8 months old) rats. This increased carotid arterial MMP2 transcription, translation, and activation, as well as transforming growth factor-beta1 activity and collagen deposition. A higher Ang II concentration, which increased arterial pressure to that of old (30 months old) untreated rats, produced carotid media thickening and intima infiltration by vascular smooth muscle cells (VSMCs). Ex vivo, Ang II increased MMP2 activity in carotid rings from young rats to that of untreated old rats. Ang II also increased the ability of early passage VSMCs from young rats to invade a synthetic basement membrane, similar to that of untreated VSMCs from old rats. The MMP inhibitor GM6001 and the AT1 receptor antagonist Losartan inhibited these effects. The alpha-adrenoreceptor agonist phenylephrine increased arterial Ang II protein, causing MMP2 activation and intima and media thickening. Exposure of young VSMCs to phenylephrine in vitro increased Ang II protein and MMP2 activity to the levels of old VSMCs; Losartan abolished these effects. Thus, Ang II-induced effects on MMP2, transforming growth factor-beta1, collagen, and VSMCs are central to the arterial remodeling that accompanies advancing age.


Subject(s)
Aging , Angiotensin II/pharmacology , Carotid Arteries/pathology , Matrix Metalloproteinase 2/metabolism , Adrenergic alpha-Agonists/pharmacology , Angiotensin II/administration & dosage , Animals , Antihypertensive Agents/pharmacology , Blood Pressure , Blotting, Western , Carotid Arteries/chemistry , Collagen/analysis , Dipeptides/pharmacology , Immunohistochemistry , Losartan/pharmacology , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/pathology , Organ Culture Techniques , Phenylephrine/pharmacology , Protease Inhibitors/pharmacology , RNA, Messenger/analysis , Rats , Transcription, Genetic , Transforming Growth Factor beta/analysis , Tunica Intima/chemistry , Tunica Intima/pathology , Vasoconstrictor Agents/administration & dosage , Vasoconstrictor Agents/pharmacology
12.
Proc Natl Acad Sci U S A ; 100(20): 11612-7, 2003 Sep 30.
Article in English | MEDLINE | ID: mdl-14500913

ABSTRACT

Erythropoietin (EPO), well known for its role in stimulation of erythropoiesis, has recently been shown to have a dramatic neuroprotective effect in animal models of cerebral ischemia, mechanical trauma of the nervous system, and excitotoxins, mainly by reducing apoptosis. We studied the effect of single systemic administration of recombinant human EPO (rhEPO) on left ventricular (LV) size and function in rats during 8 weeks after the induction of a myocardial infarction (MI) by permanent ligation of the left descending coronary artery. We found that an i.p. injection of 3,000 units/kg of rhEPO immediately after the coronary artery ligation resulted, 24 h later, in a 50% reduction of apoptosis in the myocardial area at risk. Eight weeks after the induction of MI, rats treated with rhEPO had an infarct size 15-25% of the size of that in untreated animals. The reduction in myocardial damage was accompanied by reductions in LV size and functional decline as measured by repeated echocardiography. Thus, a single dose of rhEPO administered around the time of acute, sustained coronary insufficiency merits consideration with respect to its therapeutic potential to limit the extent of resultant MI and contractile dysfunction.


Subject(s)
Coronary Vessels/pathology , Erythropoietin/pharmacology , Heart Ventricles/drug effects , Myocardial Infarction/prevention & control , Animals , Apoptosis , Echocardiography, Doppler , Heart Ventricles/diagnostic imaging , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hematocrit , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL