Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
J Orthop Surg Res ; 16(1): 708, 2021 Dec 07.
Article in English | MEDLINE | ID: mdl-34876172

ABSTRACT

BACKGROUND: To compare lay-off times achieved by treating acute muscle injuries in elite football players with a multimodal therapy approach that includes a specific protocol of almost daily radial extracorporeal shock wave therapy (rESWT) with corresponding data reported in the literature. METHODS: We performed a retrospective analysis of treatments and recovery times of muscle injuries suffered by the players of an elite football team competing in the first/second German Bundesliga during one of the previous seasons. RESULTS: A total of 20 acute muscle injuries were diagnosed and treated in the aforementioned season, of which eight (40%) were diagnosed as Type 1a/muscular tightness injuries, five (25%) as Type 2b/muscle strain injuries, four (20%) as Type 3a/partial muscle tear injuries and three (15%) as contusions. All injuries were treated with the previously mentioned multimodal therapy approach. Compared with data reported by Ekstrand et al. (Br J Sports Med 47:769-774, 2013), lay-off times (median/mean) were shortened by 54% and 58%, respectively, in the case of Type 1a injuries, by 50% and 55%, respectively, in the case of Type 2b injuries as well as by 8% and 21%, respectively, in the case of Type 3a injuries. No adverse reactions were observed. CONCLUSIONS: Overall, the multimodal therapy approach investigated in this study is a safe and effective treatment approach for treating Type 1a and 2b acute muscle injuries amongst elite football players and may help to prevent more severe, structural muscle injuries.


Subject(s)
Athletic Injuries , Extracorporeal Shockwave Therapy , Football , Return to Sport , Athletic Injuries/therapy , Humans , Muscles/injuries , Retrospective Studies , Rupture
2.
Front Pharmacol ; 3: 113, 2012.
Article in English | MEDLINE | ID: mdl-22754526

ABSTRACT

The HER2-PI3K pathway is the one of the most mutated pathways in cancer. Several drugs targeting the major kinases of this pathway have been approved by the Food and Drug Administration and many are being tested in clinical trials for the treatment of various cancers. However, the HER2-PI3K pathway is also pivotal for maintaining the physiological function of the heart, especially in the presence of cardiac stress. Clinical studies have shown that in patients treated with doxorubicin concurrently with Trastuzumab, a monoclonal antibody that blocks the HER2 receptor, the New York Heart Association class III/IV heart failure was significantly increased compared to those who were treated with doxorubicin alone (16 vs. 3%). Studies in transgenic mice have also shown that other key kinases of this pathway, such as PI3Kα, PDK1, Akt, and mTOR, are important for protecting the heart from ischemia-reperfusion and aortic stenosis induced cardiac dysfunction. Studies, however, have also shown that inhibition of PI3Kγ improve cardiac function of a failing heart. In addition, results from transgenic mouse models are not always consistent with the outcome of the pharmacological inhibition of this pathway. Here, we will review these findings and discuss how we can address the cardiac side-effects caused by inhibition of this important pathway in both cancer and cardiac biology.

3.
Curr Pharm Des ; 17(18): 1808-17, 2011.
Article in English | MEDLINE | ID: mdl-21631420

ABSTRACT

Neuregulin1 proteins (NRG1s) are epidermal growth factor (EGF) family members which are ligands for the ErbB receptor tyrosine kinases (RTKs). A decade of research has revealed that the NRG1-ErbB signaling is essential for the cardiac development and pivotal for maintaining the physiological function of the adult heart. The first evidence regarding the protective effect of the ErbB2 signaling in the adult heart came from clinical trials in breast cancer patients using Trastuzumab, a monoclonal antibody that blocks the ErbB2 receptor. The incidence of the New York Heart Association (NYHA) class III/IV heart failure increased five-fold in patients treated concurrently with chemotherapy drug doxorubicin and Trastuzumab compared to those treated with doxorubicin alone. Subsequent studies further show that stimulation of the ErbB2 signaling by NRG1s improves cardiomyocyte survival, growth and proliferation, maintains cardiac myofibril structure, counterbalances excessive ß-adrenergic signaling and promotes angiogenesis in the heart. Injections of recombinant NRG1s improve cardiac function in animal models with myocardial infarction, doxorubicin, viral infection or pacing-induced heart failure. Recent clinical trials show that NRG1s are effective for improving the cardiac function in heart failure patients. These results suggest that NRG1s may become a new drug for the treatment of heart failure. NRG1s stimulate RTKs. This is different from Beta-blockers, ACE inhibitors (Angiotensin-Converting Enzyme) and Angiotensin II receptor blockers which inhibit the excessive activation of G-protein coupled receptors (GPCRs). A clear understanding of how NRG1-ErbB signaling regulates cardiac function is essential for successful use of NRG1s for heart failure. Here, we review the current knowledge of the NRG1-ErbB signaling in the heart and discuss the potential use of NRG1s as novel therapy for heart failure.


Subject(s)
Cardiovascular Agents/pharmacology , Heart Failure/drug therapy , Neuregulin-1/pharmacology , Animals , Cardiovascular Agents/therapeutic use , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Neuregulin-1/genetics , Neuregulin-1/metabolism , Neuregulin-1/therapeutic use , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Signal Transduction/drug effects
4.
J Biomed Biotechnol ; 2011: 141039, 2011.
Article in English | MEDLINE | ID: mdl-21541238

ABSTRACT

The actions of Angiotensin II have been implicated in many cardiovascular conditions. It is widely accepted that the cardiovascular effects of Angiotensin II are mediated by different subtypes of receptors: AT(1) and AT(2). These membrane-bound receptors share a part of their nucleic acid but seem to have different distribution and pathophysiological actions. AT(1) mediates most of the Angiotensin II actions since it is ubiquitously expressed in the cardiovascular system of the normal adult. Moreover AT(2) is highly expressed in the developing fetus but its expression in the cardiovascular system is low and declines after birth. However the expression of AT(2) appears to be modulated by pathological states such as hypertension, myocardial infarction or any pathology associated to tissue remodeling or inflammation. The specific role of this receptor is still unclear and different studies involving in vivo and in vitro experiments have shown conflicting data. It is essential to clarify the role of the AT(2) receptor in the different pathological states as it is a potential site for an effective therapeutic regimen that targets the Angiotensin II system. We will review the different genetically modified mouse models used to study the AT(2) receptor and its association with cardiac hypertrophy and heart failure.


Subject(s)
Cardiomegaly/complications , Cardiomegaly/metabolism , Disease Models, Animal , Heart Failure/complications , Heart Failure/metabolism , Receptor, Angiotensin, Type 2/metabolism , Animals , Mice , Mice, Transgenic
5.
Am J Physiol Cell Physiol ; 301(1): C21-30, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21451102

ABSTRACT

Identification of factors that direct embryonic stem (ES) cell (ESC) differentiation into functional cardiomyocytes is essential for successful use of ESC-based therapy for cardiac repair. Neuregulin-1 (NRG1) and microRNA play important roles in the cardiac differentiation of ESCs. Understanding how NRG1 regulates microRNA will provide new mechanistic insights into the role of NRG1 on ESCs. It may also lead to the discovery of novel microRNAs that are important for ESC cardiac differentiation. The objective of this study was to assess the microRNA expression profile during NRG1-induced ESC cardiac differentiation. Murine ESCs were incubated with a recombinant NRG1ß or an inhibitor of ErbB2 or ErbB4 during hanging drop-induced cardiac differentiation. The expression of cardiac-specific markers and microRNAs was analyzed by RT-PCR and microRNA array, respectively. We found that the expression of NRG1 and the ErbB receptors was increased during hanging drop-induced cardiac differentiation of ESCs. NRG1 stimulation during a specific developmental window enhanced, while inhibition of the ErbB2 or ErbB4 receptor inhibited, cardiac differentiation of ESCs. NRG1 increased the expression of mmu-miR-296-3p and mmu-miR-200c*, and decreased mmu-miR-465b-5p. Inhibition of mmu-miR-296-3p or mmu-miR-200c* decreased, while inhibition of mmu-miR-465-5p increased, the differentiation of ESCs into the cardiac lineage. This is the first report demonstrating that microRNAs are differentially regulated by NRG1-ErbB signaling during cardiac differentiation of ESCs. This study has also identified new microRNAs that are important for ESC cardiac differentiation.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , MicroRNAs/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neuregulin-1/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Embryonic Stem Cells/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Developmental , Heart/embryology , Heart/growth & development , Mesoderm/embryology , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Microarray Analysis , Neuregulin-1/genetics , Oncogene Proteins v-erbB/antagonists & inhibitors , Oncogene Proteins v-erbB/metabolism , Polymerase Chain Reaction , Regeneration , Signal Transduction
6.
J Geriatr Cardiol ; 8(3): 195-200, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22783305

ABSTRACT

Current guidelines for implantable cardioverter-defibrillator (ICD) therapy in heart failure patients were established by multiple device trials; however, very few geriatric patients (patients ≥ 65 years old) were included in these studies. This article explores the controversies of ICD implantation in the geriatric population, management of delivered ICD therapy in this age group, and the end of life care in patients with ICD.

7.
Can J Physiol Pharmacol ; 88(5): 568-75, 2010 May.
Article in English | MEDLINE | ID: mdl-20555426

ABSTRACT

Exposure to tobacco smoke is known to have deleterious cardiovascular effects. In this study, we tested whether exposure to tobacco smoke exacerbates the severity of viral myocarditis in mice. Viral myocarditis was generated in 4-week-old male BALB/c mice by injection of Encephalomyocarditis virus (EMCV). Four groups were studied: (1) control (C, no smoke and no virus); (2) smoke only (S, exposure to cigarette smoke for 90 min/day for 15 days); (3) virus only (V); and (4) exposure to smoke for 5 days before plus 10 days following virus injection (S+V). We found that viral inoculation preceded by smoke exposure increased mortality more than twofold compared with virus inoculation alone. In addition, the mRNA level of atrial natriuretic factor was significantly higher in S+V than among any of the other 3 groups. Virus injection significantly decreased cardiac function compared with controls, with further deterioration observed in the S+V group. We also observed a significantly increased rate of apoptosis, with an increased activation of apoptosis-inducing factor in hearts exposed to S+V compared with those exposed to V alone. Our results suggest that preexposure to smoke significantly exacerbates the severity of viral myocarditis, likely through increased viral load and increased cardiomyocyte cell death.


Subject(s)
Cardiovirus Infections/virology , Heart/virology , Myocarditis/virology , Tobacco Smoke Pollution/adverse effects , Viral Load , Animals , Apoptosis , Apoptosis Inducing Factor/biosynthesis , Atrial Natriuretic Factor/biosynthesis , Blotting, Western , Cardiovirus Infections/metabolism , Cardiovirus Infections/pathology , Cardiovirus Infections/physiopathology , Encephalomyocarditis virus , Hemodynamics/physiology , Male , Mice , Mice, Inbred BALB C , Myocarditis/metabolism , Myocarditis/pathology , Myocarditis/physiopathology , Myocardium/metabolism , Myocardium/pathology , Reverse Transcriptase Polymerase Chain Reaction
8.
Am J Physiol Heart Circ Physiol ; 297(6): H1974-83, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19801490

ABSTRACT

Neuregulin-1 (NRG1) is a potential therapeutic agent for the treatment of doxorubicin (Dox)-induced heart failure. NRG1, however, activates the erbB2 receptor, which is frequently overexpressed in breast cancers. It is, therefore, important to understand how NRG1, via erbB2, protects the heart against Dox cardiotoxicity. Here, we studied NRG1-erbB2 signaling in Dox-treated mice hearts and in isolated neonatal rat ventricular myocytes (NRVM). Male C57BL/6 mice were treated with recombinant NRG1 before and daily after a single dose of Dox. Cardiac function was determined by catheterization. Two-week survival was analyzed by the Kaplan-Meier method. Cardiac troponins [cardiac troponin I (cTnI) and cardiac troponin T (cTnT)] and phosphorylated Akt protein levels were determined in mice hearts and in NRVM by Western blot analysis. Activation of caspases and ubiquitinylation of troponins were determined in NRVM by caspase assay and immunoprecipitation. NRG1 significantly improved survival and cardiac function in Dox-treated mice. NRG1 reduced the decrease in cTnI, cTnT, and cardiac troponin C (cTnC) and maintained Akt phosphorylation in Dox-treated mice hearts. NRG1 reduced the decrease in cTnI and cTnT mRNA and proteins in Dox-treated NRVM. Inhibition of erbB2, phosphoinositide 3-kinase (PI3K), Akt, and mTOR blocked the protective effects of NRG1 on cTnI and cTnT in NRVM. NRG1 significantly reduced Dox-induced caspase activation, which degraded troponins, in NRVM. NRG1 reduced Dox-induced proteasome degradation of cTnI. NRG1 attenuates Dox-induced decrease in cardiac troponins by increasing transcription and translation and by inhibiting caspase activation and proteasome degradation of troponin proteins. NRG1 maintains cardiac troponins by the erbB2-PI3K pathway, which may lessen Dox-induced cardiac dysfunction.


Subject(s)
Cardiovascular Agents/administration & dosage , Heart Diseases/prevention & control , Myocardium/metabolism , Neuregulin-1/administration & dosage , Signal Transduction/drug effects , Troponin I/metabolism , Troponin T/metabolism , Animals , Animals, Newborn , Antibiotics, Antineoplastic , Biomarkers/blood , Caspases/metabolism , Cells, Cultured , Creatine Kinase/blood , Disease Models, Animal , Doxorubicin , Glycoproteins/metabolism , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/physiopathology , Humans , Injections, Subcutaneous , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardium/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptor, ErbB-2 , Recombinant Proteins/administration & dosage , TOR Serine-Threonine Kinases , Time Factors , Transcription, Genetic/drug effects , Troponin I/blood , Troponin I/genetics , Troponin T/blood , Troponin T/genetics , Ubiquitination , Up-Regulation , Ventricular Function, Left/drug effects
9.
PLoS Pathog ; 5(5): e1000427, 2009 May.
Article in English | MEDLINE | ID: mdl-19436702

ABSTRACT

Cytomegalovirus (CMV) infection is a common infection in adults (seropositive 60-99% globally), and is associated with cardiovascular diseases, in line with risk factors such as hypertension and atherosclerosis. Several viral infections are linked to hypertension, including human herpes virus 8 (HHV-8) and HIV-1. The mechanisms of how viral infection contributes to hypertension or increased blood pressure are not defined. In this report, the role of CMV infection as a cause of increased blood pressure and in forming aortic atherosclerotic plaques is examined. Using in vivo mouse model and in vitro molecular biology analyses, we find that CMV infection alone caused a significant increase in arterial blood pressure (ABp) (p<0.01 approximately 0.05), measured by microtip catheter technique. This increase in blood pressure by mouse CMV (MCMV) was independent of atherosclerotic plaque formation in the aorta, defined by histological analyses. MCMV DNA was detected in blood vessel samples of viral infected mice but not in the control mice by nested PCR assay. MCMV significantly increased expression of pro-inflammatory cytokines IL-6, TNF-alpha, and MCP-1 in mouse serum by enzyme-linked immunosorbent assay (ELISA). Using quantitative real time reverse transcriptase PCR (Q-RT-PCR) and Western blot, we find that CMV stimulated expression of renin in mouse and human cells in an infectious dose-dependent manner. Co-staining and immunofluorescent microscopy analyses showed that MCMV infection stimulated renin expression at a single cell level. Further examination of angiotensin-II (Ang II) in mouse serum and arterial tissues with ELISA showed an increased expression of Ang II by MCMV infection. Consistent with the findings of the mouse trial, human CMV (HCMV) infection of blood vessel endothelial cells (EC) induced renin expression in a non-lytic infection manner. Viral replication kinetics and plaque formation assay showed that an active, CMV persistent infection in EC and expression of viral genes might underpin the molecular mechanism. These results show that CMV infection is a risk factor for increased arterial blood pressure, and is a co-factor in aortic atherosclerosis. Viral persistent infection of EC may underlie the mechanism. Control of CMV infection can be developed to restrict hypertension and atherosclerosis in the cardiovascular system.


Subject(s)
Blood Pressure , Cytomegalovirus Infections/physiopathology , Herpesviridae Infections/physiopathology , Muromegalovirus/pathogenicity , Angiotensin II/metabolism , Animals , Aorta/pathology , Aorta/virology , Atherosclerosis/virology , Blood Vessels/virology , Cell Line , Chi-Square Distribution , Cytokines/metabolism , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/virology , DNA, Viral/analysis , Diet, Atherogenic , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Humans , Mice , Mice, Inbred C57BL , Models, Biological , Polymerase Chain Reaction , RNA, Viral/analysis , Renin/metabolism , Virus Replication
10.
Stroke ; 40(7): 2560-3, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19443809

ABSTRACT

BACKGROUND AND PURPOSE: Stroke can lead to cerebrogenic cardiac arrhythmias. We sought to investigate the effect of ischemic stroke on cardiac function in a mouse model of permanent middle cerebral artery occlusion (pMCAO). METHODS: Twenty-four hours after the induction of focal ischemia, cardiac function was measured in mice by endovascular catheterization of the heart. Immediately after hemodynamic measurements, mice were euthanized and brains were excised and sectioned to measure infarct volume and the severity of insular cortex injury. Myocardial damage was evaluated by hematoxylin-eosin staining. Serum and heart levels of norepinephrine (NE) were also determined. RESULTS: Cardiac dysfunction occurred in 9 out of 14 mice that underwent left pMCAO. In these 9 mice, the severity of left insular cortex lesion was greater than the mice with normal heart function. The serum and heart levels of NE were significantly higher in left pMCAO mice with heart dysfunction. Liner regression analysis indicates significant inverse correlation between the severity of left insular cortex damage and heart dysfunction. Mice that underwent right pMCAO did not exhibit cardiac dysfunction. CONCLUSIONS: This study shows that left focal cerebral ischemia can produce cardiac dysfunction, which is associated with the extent of left insular cortex damage. Furthermore, mice exhibiting cardiac dysfunction had elevated levels of NE in the serum and heart.


Subject(s)
Brain Ischemia/complications , Brain Ischemia/physiopathology , Brain/physiopathology , Heart/physiopathology , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/physiopathology , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , Norepinephrine/metabolism , Regression Analysis
11.
J Cell Mol Med ; 13(5): 811-25, 2009 May.
Article in English | MEDLINE | ID: mdl-19382894

ABSTRACT

At present the prevalence of heart failure rises along with aging of the population. Current heart failure therapeutic options are directed towards disease prevention via neurohormonal antagonism (beta-blockers, angiotensin converting enzyme inhibitors and/or angiotensin receptor blockers and aldosterone antagonists), symptomatic treatment with diuretics and digitalis and use of biventricular pacing and defibrillators in a special subset of patients. Despite these therapies and device interventions heart failure remains a progressive disease with high mortality and morbidity rates. The number of patients who survive to develop advanced heart failure is increasing. These patients require new therapeutic strategies. In this review two of emerging therapies in the treatment of heart failure are discussed: metabolic modulation and cellular therapy. Metabolic modulation aims to optimize the myocardial energy utilization via shifting the substrate utilization from free fatty acids to glucose. Cellular therapy on the other hand has the goal to achieve true cardiac regeneration. We review the experimental data that support these strategies as well as the available pharmacological agents for metabolic modulation and clinical application of cellular therapy.


Subject(s)
Heart Failure/metabolism , Heart Failure/therapy , Stem Cell Transplantation , Acetyl-CoA C-Acyltransferase/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Adult Stem Cells/transplantation , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Embryonic Stem Cells/transplantation , Heart Failure/drug therapy , Humans , Mesenchymal Stem Cell Transplantation , Muscle, Skeletal/cytology , Myoblasts/transplantation , Oxidative Phosphorylation , PPAR gamma/agonists , Regeneration
12.
Front Biosci (Landmark Ed) ; 14(8): 2845-56, 2009 01 01.
Article in English | MEDLINE | ID: mdl-19273239

ABSTRACT

TNF-alpha released from ischemic heart after acute MI increases the production of other cytokines such as interleukin-1 (IL-1), interleukin-6 (IL-6) and adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1). Activation of nuclear factor kappa B (NF-kappa B) by TNF-alpha , up-regulates the expression of molecules which are involved in inflammation and cell adhesion. For these reasons, we assessed the extent that treatment of MSC with tumor necrosis factor (TNF)-alpha modifies the characteristics of MSC, important to their engraftment in experimental myocardial infarct. Here, we show that pre-treatment of MSC prior to transplantation with tumor necrosis factor (TNF)-alpha increases adhesiveness, and migration of MSC in vitro and leads to increased expression of bone morphogenetic protein (BMP)-2 by MSC. Moreover, this treatment increases the rate of engraftment of MSC and improves recovery of cardiac function after myocardial infarction. These insights might provide better strategies for the treatment of myocardial infarction.


Subject(s)
Mesenchymal Stem Cells/drug effects , Myocardial Infarction/therapy , Tumor Necrosis Factor-alpha/pharmacology , Animals , Base Sequence , Bone Morphogenetic Protein 2/physiology , Cell Transplantation , Collagen/metabolism , DNA Primers , Enzyme-Linked Immunosorbent Assay , Male , Mesenchymal Stem Cells/cytology , Myocardial Infarction/pathology , NF-kappa B/physiology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/physiology
13.
Biochem Biophys Res Commun ; 382(2): 241-6, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19233128

ABSTRACT

The purpose of this study was to study the effect of calcium, cyclic AMP (cAMP) and cyclic GMP (cGMP) on embryonic stem cell (ESC) motility during TNF-alpha-induced chemotaxis. ESCs were monitored using a chemotaxis chamber, with different concentrations of calcium or cAMP or cGMP added to the medium. Changes in intracellular calcium ([Ca(2+)](i)) were measured with the fluorescent dye fura-2/AM. We combined migratory parameters in a mathematical model and described it as "mobility". After adding calcium, a dose-dependant increase in cell speed was found. Cyclic AMP increased mobility as well as the [Ca(2+)](i). In contrast, adding dbcGMP resulted in a significant decrease in the mobility of the ESCs. During migration ESCs showed an increase in [Ca(2+)](i). Furthermore, TNF-alpha dramatically increased the movement as well as the directionality of ESCs. These results demonstrate that ESCs are highly motile and respond to different concentrations of calcium in a dose-related manner.


Subject(s)
Calcium/pharmacology , Cell Movement/drug effects , Cyclic AMP/pharmacology , Cyclic GMP/pharmacology , Embryonic Stem Cells/drug effects , Animals , Cell Line , Embryonic Stem Cells/physiology , Mice , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/physiology
14.
Eur J Pharmacol ; 592(1-3): 123-7, 2008 Sep 11.
Article in English | MEDLINE | ID: mdl-18627770

ABSTRACT

Clozapine, an atypical antipsychotic, is very effective in the treatment of resistant schizophrenia. However, cardiotoxicity of clozapine, particularly in young patients, has raised concerns about its safety. Increased catecholamines have been postulated to trigger an inflammatory response resulting in myocarditis, dilated cardiomyopathy, and death, although this has not yet been thoroughly studied. Here, we used the mouse to study whether clozapine administration could cause adverse myocarditis associated with an increase in catecholamines. Male Balb/C mice, age ~6 weeks, were administered 5, 10 or 25 mg/kg clozapine daily for 7 and 14 days; one group was administered 25 mg/kg clozapine plus 2 mg/kg propranolol for 14 days. Saline-treated mice served as controls. Heart sections were stained with hematoxylin and eosin for histopathological examination. Plasma catecholamines were measured with HPLC. Myocardial TNF-alpha concentrations were determined by ELISA. Histopathology of clozapine-treated mice showed a significant dose-related increase in myocardial inflammation that correlated with plasma catecholamine levels and release of TNF-alpha. Propranolol significantly attenuated these effects. A hypercatecholaminergic state induced by clozapine could explain the occurrence of myocarditis in some patients. Our data suggest that a beta-adrenergic blocking agent may be effective in reducing the incidence and severity of clozapine-induced myocarditis.


Subject(s)
Antipsychotic Agents/toxicity , Catecholamines/physiology , Clozapine/toxicity , Myocarditis/chemically induced , Myocarditis/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Male , Mice , Mice, Inbred BALB C , Myocarditis/pathology , Myocardium/metabolism , Myocardium/pathology , Propranolol/pharmacology , Tumor Necrosis Factor-alpha/metabolism
16.
Am J Physiol Heart Circ Physiol ; 294(3): H1274-81, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18178728

ABSTRACT

The role of the angiotensin II type 2 (AT2) receptor in cardiac hypertrophy remains controversial. We studied the effects of AT2 receptors on chronic pressure overload-induced cardiac hypertrophy in transgenic mice selectively overexpressing AT2 receptors in ventricular myocytes. Left ventricular (LV) hypertrophy was induced by ascending aorta banding (AS). Transgenic mice overexpressing AT2 (AT2TG-AS) and nontransgenic mice (NTG-AS) were studied after 70 days of aortic banding. Nonbanded NTG mice were used as controls. LV function was determined by catheterization via LV puncture and cardiac magnetic resonance imaging. LV myocyte diameter and interstitial collagen were determined by confocal microscopy. Atrial natriuretic polypeptide (ANP) and brain natriuretic peptide (BNP) were analyzed by Northern blot. Sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA)2, inducible nitric oxide synthase (iNOS), endothelial NOS, ERK1/2, p70S6K, Src-homology 2 domain-containing protein tyrosine phosphatase-1, and protein serine/threonine phosphatase 2A were analyzed by Western blot. LV myocyte diameter and collagen were significantly reduced in AT2TG-AS compared with NTG-AS mice. LV anterior and posterior wall thickness were not different between AT2TG-AS and NTG-AS mice. LV systolic and diastolic dimensions were significantly higher in AT2TG-AS than in NTG-AS mice. LV systolic pressure and end-diastolic pressure were lower in AT2TG-AS than in NTG-AS mice. ANP, BNP, and SERCA2 were not different between AT2TG-AS and NTG-AS mice. Phospholamban (PLB) and the PLB-to-SERCA2 ratio were significantly higher in AT2TG-AS than in NTG-AS mice. iNOS was higher in AT2TG-AS than in NTG-AS mice but not significantly different. Our results indicate that AT2 receptor overexpression modified the pathological hypertrophic response to aortic banding in transgenic mice.


Subject(s)
Blood Pressure/physiology , Cardiomegaly/genetics , Cardiomegaly/pathology , Hypertension/pathology , Myocytes, Cardiac/pathology , Receptor, Angiotensin, Type 2/genetics , Animals , Atrial Natriuretic Factor/metabolism , Blotting, Northern , Blotting, Western , Collagen/metabolism , Hypertension/genetics , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Myocardium/metabolism , Natriuretic Peptide, Brain/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Angiotensin, Type 2/physiology , Survival Analysis , Ventricular Function, Left/physiology
17.
Am J Physiol Heart Circ Physiol ; 291(4): H1653-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16699073

ABSTRACT

Embryonic stem cells (ESCs) overexpressing the vascular endothelial growth factor (VEGF) improve cardiac function in mouse models of myocardial ischemia and infarction by mechanisms that are poorly understood. Here we studied the effects of VEGF on cardiomyocyte differentiation of mouse ESCs in vitro. We used flow cytometry to determine the expression of alpha-myosin heavy chain (alpha-MHC), cardiac troponin I (cTn-I), and Nkx2.5 in differentiated ESCs. VEGF (20 ng/ml) significantly enhanced alpha-MHC, cTn-I, and Nkx2.5 expression in differentiated ESCs. Western blot analysis confirmed these findings. We found that VEGF receptor FMS-like tyrosine kinase-1 (Flt-1) and fetal liver kinase-1 (Flk-1) expression increased during ESC differentiation. Antibodies against Flk-1 totally blocked and against Flt-1 partially blocked VEGF-induced NKx2.5-positive-stained cells. The ERK inhibitor PD-098059 abolished VEGF-induced cardiomyocyte differentiation of ESCs. Our results suggest that VEGF promotes cardiomyocyte differentiation predominantly by ERK-mediated Flk-1 activation and, to a lesser extent, by Flt-1 activation. These findings may be of significance for stem cell and growth factor therapies to regenerate failing cardiomyocytes.


Subject(s)
Cell Differentiation/drug effects , Myocytes, Cardiac/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Animals , Cell Line , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Stem Cell Transplantation , Transcription Factors/genetics , Transcription Factors/metabolism , Troponin I/genetics , Troponin I/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Ventricular Myosins/genetics , Ventricular Myosins/metabolism
18.
Pflugers Arch ; 452(3): 268-75, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16715295

ABSTRACT

The present study was designed to determine whether cardiac inflammation is important for the successful homing of stem cells to the heart after intravenous injection in a murine myocarditis model. Male Bagg albino/c mice were infected with encephalomyocarditis virus (EMCV) to produce myocarditis. Subgroups of mice received single injections by tail vein of embryonic stem cells (ESCs) transfected with green fluorescent protein (GFP) as a marker at days 3, 14, or 60 after infection; other subgroups without stem cell injections were killed at each of these time points to assess the degree of inflammation present. The surviving mice were killed at day 90 after virus infection and hemodynamics, gross pathology, histology, and inflammatory cytokine production in the hearts were measured. Our results indicate that myocardial inflammation was most severe and cytokine production highest at day 14 after EMCV inoculation, and in particular, was strongly positive for interleukin 6. Mice receiving intravenous ESC injections on day 14 after EMCV inoculation showed the largest number of GFP-positive cells at the time of death and the greatest functional improvement compared to uninfected controls without inflammation. We conclude that factors released from myocardium during inflammation are important for enhancing the homing, migration, and implantation of systemically infused stem cells.


Subject(s)
Cytokines/metabolism , Embryonic Stem Cells/physiology , Heart/physiology , Myocarditis/physiopathology , Stem Cell Transplantation/methods , Animals , Green Fluorescent Proteins , Injections, Intravenous , Male , Mice , Mice, Inbred BALB C , Myocarditis/pathology , Myocardium/pathology , Necrosis
19.
J Thorac Cardiovasc Surg ; 131(4): 889-97, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16580449

ABSTRACT

OBJECTIVE: The present study was designed to test whether intravenously infused embryonic stem cell-derived cells could translocate to injured myocardium after myocardial infarction and improve cardiac function. METHODS: Cultured embryonic stem cell-derived cells were transfected with green fluorescent protein. Embryonic stem cell-derived cells were administered through the tail vein (approximately 10(7) cells in 1 mL of medium for each rat) every other day for 6 days in 45 rats after myocardial infarction. Six weeks after myocardial infarction and cell infusion, cardiac function, blood flow, and the numeric density of arterioles were measured to test the benefits of cell therapy. An in vitro Transwell assay was performed to evaluate the embryonic stem cell migration. RESULTS: Ventricular function, regional blood flow, and arteriole density were significantly increased in rats receiving intravenously infused embryonic stem cell-derived cells compared with control rats after myocardial infarction. Histologic analysis demonstrated that infused embryonic stem cell-derived cells formed green fluorescent protein-positive grafts in infarcted myocardium. Additionally, positive immunostaining for cardiac troponin I was found in hearts after myocardial infarction receiving embryonic stem cell-derived cell infusion that corresponded to the green fluorescent protein-positive staining. The Transwell migration assay indicated that cultured neonatal rat cardiomyocytes with overexpression of tumor necrosis factor alpha induced greater migration of embryonic stem cells compared with cardiomyocytes without tumor necrosis factor alpha expression. CONCLUSIONS: Our data demonstrate that intravenously infused embryonic stem cell-derived cells homed to the infarcted heart, improved cardiac function, and enhanced regional blood flow at 6 weeks after myocardial infarction. The in vitro migration assay suggested that such a homing mechanism could be associated with locally released cytokines, such as tumor necrosis factor alpha, that are upregulated in the setting of acute myocardial infarction and heart failure.


Subject(s)
Cell Movement , Myocardial Infarction/therapy , Stem Cell Transplantation/methods , Stem Cells/physiology , Animals , Animals, Newborn , Arterioles , Cell Differentiation , Cells, Cultured , Green Fluorescent Proteins , Infusions, Intravenous , Male , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha , Ventricular Function, Left
20.
Am J Physiol Heart Circ Physiol ; 291(1): H52-60, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16399862

ABSTRACT

The p53-upregulated modulator of apoptosis (Puma), a BH3-only member of the Bcl-2 protein family, is required for p53-dependent and -independent forms of apoptosis and has been implicated in the pathomechanism of several diseases, including cancer, acquired immunodeficiency syndrome, and ischemic brain disease. The role of Puma in cardiomyocyte death, however, has not been analyzed. On the basis of the ability of Puma to integrate diverse cell death stimuli, we hypothesized that Puma might be critical for cardiomyocyte death upon ischemia-reperfusion (I/R) of the heart. Here we show that hypoxia-reoxygenation of isolated cardiomyocytes led to an increase in Puma mRNA and protein levels. Moreover, if Puma was delivered by an adenoviral construct, cardiomyocytes died by apoptosis. Under ATP-depleted conditions, however, Puma overexpression primarily induced necrosis, suggesting that Puma is involved in the development of both types of cell death. Consistent with these findings, targeted deletion of Puma in a mouse model attenuated both apoptosis and necrosis. When the Langendorff ex vivo I/R model was used, infarcts were approximately 50% smaller in Puma(-/-) than in wild-type mice. As a result, after I/R, cardiac function was significantly better preserved in Puma(-/-) mice than in their wild-type littermates. Our study thus establishes Puma as an essential mediator of cardiomyocyte death upon I/R injury and offers a novel therapeutic target to limit cell loss in ischemic heart disease.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Gene Targeting/methods , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins/metabolism , Reperfusion Injury/physiopathology , Ventricular Dysfunction, Left/prevention & control , Ventricular Dysfunction, Left/physiopathology , Animals , Animals, Newborn , Apoptosis , Apoptosis Regulatory Proteins/genetics , Cells, Cultured , Drug Delivery Systems/methods , Gene Deletion , Proto-Oncogene Proteins/genetics , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Reperfusion Injury/therapy , Treatment Outcome , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...