1
|
Hong MH, Jang YJ, Yoon JJ, Lee HS, Kim HY, Kang DG. Dohongsamul-tang inhibits cardiac remodeling and fibrosis through calcineurin/NFAT and TGF-β/Smad2 signaling in cardiac hypertrophy. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116844. [PMID: 37453625 DOI: 10.1016/j.jep.2023.116844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dohongsammul-tang (DH) is a Korean traditional herbal medicine used to alleviate symptoms caused by extravasated blood. It is known to protect against cardiovascular diseases and promote blood circulation by activating blood circulation to dispel blood stasis. The DH based on the characteristics of its medicinal properties has discovered the potential of alleviating cardiac hypertrophy. Therefore, this study was performed to verify the pharmacological effect of DH on improving cardiovascular disorders and to demonstrate its mutual improvement effect on renal function. Furthermore, aim of this study is founding the new potential beyond the traditional medicinal efficacy of DH, a traditional medicine. AIM OF THE STUDY In cardiovascular disease, cardiac hypertrophy refers to a change in the shape of the heart's structure due to pressure overload. It is known that an increase in myofibrils causes thickening of the heart, resulting in high blood pressure. Therefore, suppressing cardiac hypertrophy may be a major factor in lowering the morbidity, mortality, and heart failure associated with cardiovascular disease. Therefore, the study was performed to investigate whether DH, traditionally used, has effects on improving and alleviating cardiac injury and fibrosis caused by cardiac hypertrophy. MATERIALS AND METHODS Dohongsamul-tang was composed of 6 herbal medicine and each material were boiled with 4 L distilled water for 2 h. The mixture for dohongsamul-tang centrifuged at 3000 rpm for 10 min and concentrated. The concentrated dohongsamul-tang extraction freeze-dried and sotred at 70 °C. The powder of dohongsamul-tang was diluted with distilled water and administered orally. In this study, pressure overload was induced by tying the transverse aortic arch, which is connected to the left ventricle, to the thickness of a 27G needle by performing a surgical operation. The resulting cardiac hypertrophy and heart remodeling was induced and maintained for 8 weeks. RESULTS The study administered propranolol and dohongsamul-tang orally for 10 weeks to investigate their effects on cardiac hypertrophy induced by transverse aortic contraction (TAC) surgery. Results showed that TAC group increased the left ventricle weight and decreased cardiac function, but dohongsamul-tang treatment attenuated these effects. The pressure-volume curve experiment revealed that dohongsamul-tang improved cardiovascular function, which was worsened by TAC group. Dohongsamul-tang treatment also downregulated collagen I and III through the TGF-β/Smad2 signaling pathway and improved hematological biomarkers of cardiac hypertrophy. In addition, dohongsamul-tang treatment improved renal function-related biomarkers, such as blood creatinine, blood urea nitrogen, and neutrophil gelatinase-associated lipocalin, which were increased by TAC-induced cardiac hypertrophy. CONCLUSIONS Taken together, dohongsamul-tang treatment inhibited cardiac remodeling due to pressure overload in the TAC-induced cardiac hypertrophy model, and this effect is thought to be manifested by improving the functional and morphological changes through the calcineurin/NFATc4 and reducing the cardiac fibrosis by suppressing TGF-β/Smad2 signaling pathways.
Collapse
Affiliation(s)
- Mi Hyeon Hong
- Hanbang Cardio-renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea; College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea.
| | - Youn Jae Jang
- Hanbang Cardio-renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea; College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea.
| | - Jung Joo Yoon
- Hanbang Cardio-renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea.
| | - Ho Sub Lee
- Hanbang Cardio-renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea; College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea.
| | - Hye Yoom Kim
- Hanbang Cardio-renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea.
| | - Dae Gill Kang
- Hanbang Cardio-renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea; College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea.
| |
Collapse
|
2
|
Billah M, Naz A, Noor R, Bhindi R, Khachigian LM. Early Growth Response-1: Friend or Foe in the Heart? Heart Lung Circ 2023; 32:e23-e35. [PMID: 37024319 DOI: 10.1016/j.hlc.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 04/07/2023]
Abstract
Cardiovascular disease is a major cause of mortality and morbidity worldwide. Early growth response-1 (Egr-1) plays a critical regulatory role in a range of experimental models of cardiovascular diseases. Egr-1 is an immediate-early gene and is upregulated by various stimuli including shear stress, oxygen deprivation, oxidative stress and nutrient deprivation. However, recent research suggests a new, underexplored cardioprotective side of Egr-1. The main purpose of this review is to explore and summarise the dual nature of Egr-1 in cardiovascular pathobiology.
Collapse
Affiliation(s)
- Muntasir Billah
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, Sydney, NSW, Australia; Sydney Medical School Northern, The University of Sydney, Sydney, NSW, Australia.
| | - Adiba Naz
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Rashed Noor
- School of Environmental and Life Sciences, Independent University Bangladesh, Dhaka, Bangladesh
| | - Ravinay Bhindi
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, Sydney, NSW, Australia; Sydney Medical School Northern, The University of Sydney, Sydney, NSW, Australia
| | - Levon M Khachigian
- Vascular Biology and Translational Research, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
3
|
Altieri DI, Etzion Y, Anderson HD. Cannabinoid receptor agonist attenuates angiotensin II-induced enlargement and mitochondrial dysfunction in rat atrial cardiomyocytes. Front Pharmacol 2023; 14:1142583. [PMID: 37113758 PMCID: PMC10126395 DOI: 10.3389/fphar.2023.1142583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/13/2023] [Indexed: 04/29/2023] Open
Abstract
Pathological remodeling of atrial tissue renders the atria more prone to arrhythmia upon arrival of electrical triggers. Activation of the renin-angiotensin system is an important factor that contributes to atrial remodeling, which may result in atrial hypertrophy and prolongation of P-wave duration. In addition, atrial cardiomyocytes are electrically coupled via gap junctions, and electrical remodeling of connexins may result in dysfunction of coordinated wave propagation within the atria. Currently, there is a lack of effective therapeutic strategies that target atrial remodeling. We previously proposed that cannabinoid receptors (CBR) may have cardioprotective qualities. CB13 is a dual cannabinoid receptor agonist that activates AMPK signaling in ventricular cardiomyocytes. We reported that CB13 attenuates tachypacing-induced shortening of atrial refractoriness and inhibition of AMPK signaling in the rat atria. Here, we evaluated the effects of CB13 on neonatal atrial rat cardiomyocytes (NRAM) stimulated by angiotensin II (AngII) in terms of atrial myocyte enlargement and mitochondrial function. CB13 inhibited AngII-induced enhancement of atrial myocyte surface area in an AMPK-dependent manner. CB13 also inhibited mitochondrial membrane potential deterioration in the same context. However, AngII and CB13 did not affect mitochondrial permeability transition pore opening. We further demonstrate that CB13 increased Cx43 compared to AngII-treated neonatal rat atrial myocytes. Overall, our results support the notion that CBR activation promotes atrial AMPK activation, and prevents myocyte enlargement (an indicator that suggests pathological hypertrophy), mitochondrial depolarization and Cx43 destabilization. Therefore, peripheral CBR activation should be further tested as a novel treatment strategy in the context of atrial remodeling.
Collapse
Affiliation(s)
- Danielle I. Altieri
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine (CCARM), Albrechtsen Research Centre, St Boniface Hospital, Winnipeg, MB, Canada
| | - Yoram Etzion
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hope D. Anderson
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine (CCARM), Albrechtsen Research Centre, St Boniface Hospital, Winnipeg, MB, Canada
- *Correspondence: Hope D. Anderson,
| |
Collapse
|
4
|
Involvement of Histamine 2 Receptor in Alpha 1 Adrenoceptor Mediated Cardiac Hypertrophy and Oxidative Stress in H9c2 Cardio Myoblasts. J Cardiovasc Transl Res 2020; 14:184-194. [PMID: 32385805 DOI: 10.1007/s12265-020-09967-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/06/2020] [Indexed: 01/01/2023]
Abstract
Despite the involvement of ɑ1adrenergic (ɑ1AR) and Histamine 2 receptors (H2R) in cardiac hypertrophy (CH), their relationship is yet to be studied. Our study investigated interrelationship between them using in vitro CH model. H9c2 cardiomyoblasts were exposed to phenylephrine (ɑ1AR agonist-50 μM) in the presence, the absence of famotidine (H2R antagonist-10 μM) and BAY 11-7082 (NF-kB inhibitor-10 μM). The impact of ɑ1AR stimulation on H2R expression and oxidative stress was assessed. Hypertrophic indices were assessed from activities of enzymatic mediators of cardiac hypertrophy, total protein content, BNP levels and cell volume. Additionally, the inverse agonistic property of famotidine and NFkB activity was also studied. ɑ1AR-induced H2R expression, oxidative stress and hypertrophic indices were significantly abolished by famotidine and pharmacological inhibitor of NFkB. Increase in constitutive activity of H2R was noticed correlating with increased receptor population. These results suggest involvement of NFkB-mediated upregulation of H2R in ɑ1AR-mediated CH.
Collapse
|
5
|
Kitsis RN, Leinwand LA. Discordance between gene regulation in vitro and in vivo. Gene Expr 2018; 2:313-8. [PMID: 1472867 PMCID: PMC6057365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- R N Kitsis
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461
| | | |
Collapse
|
6
|
Paudyal A, Dewan S, Ikie C, Whalley BJ, de Tombe PP, Boateng SY. Nuclear accumulation of myocyte muscle LIM protein is regulated by heme oxygenase 1 and correlates with cardiac function in the transition to failure. J Physiol 2016; 594:3287-305. [PMID: 26847743 DOI: 10.1113/jp271809] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/25/2016] [Indexed: 01/19/2023] Open
Abstract
KEY POINTS The present study investigated the mechanism associated with impaired cardiac mechanosensing that leads to heart failure by examining the factors regulating muscle LIM protein subcellular distribution in myocytes. In myocytes, muscle LIM protein subcellular distribution is regulated by cell contractility rather than passive stretch via heme oxygenase-1 and histone deacetylase signalling. The result of the present study provide new insights into mechanotransduction in cardiac myocytes. Myocyte mechanosensitivity, as indicated by the muscle LIM protein ratio, is also correlated with cardiac function in the transition to failure in a guinea-pig model of disease. This shows that the loss mechanosensitivity plays an important role during the transition to failure in the heart. The present study provides the first indication that mechanosensing could be modified pharmacologically during the transition to heart failure. ABSTRACT Impaired mechanosensing leads to heart failure and a decreased ratio of cytoplasmic to nuclear CSRP3/muscle LIM protein (MLP ratio) is associated with a loss of mechanosensitivity. In the present study, we tested whether passive or active stress/strain was important in modulating the MLP ratio and determined whether this correlated with heart function during the transition to failure. We exposed cultured neonatal rat myocytes to a 10% cyclic mechanical stretch at 1 Hz, or electrically paced myocytes at 6.8 V (1 Hz) for 48 h. The MLP ratio decreased by 50% (P < 0.05, n = 4) only in response to electrical pacing, suggesting impaired mechanosensitivity. Inhibition of contractility with 10 μm blebbistatin resulted in an ∼3-fold increase in the MLP ratio (n = 8, P < 0.05), indicating that myocyte contractility regulates nuclear MLP. Inhibition of histone deacetylase (HDAC) signalling with trichostatin A increased nuclear MLP following passive stretch, suggesting that HDACs block MLP nuclear accumulation. Inhibition of heme oxygenase1 (HO-1) activity with protoporphyrin IX zinc(II) blocked MLP nuclear accumulation. To examine how mechanosensitivity changes during the transition to heart failure, we studied a guinea-pig model of angiotensin II infusion (400 ng kg(-1) min(-1) ) over 12 weeks. Using subcellular fractionation, we showed that the MLP ratio increased by 88% (n = 4, P < 0.01) during compensated hypertrophy but decreased significantly during heart failure (P < 0.001, n = 4). The MLP ratio correlated significantly with the E/A ratio (r = 0.71, P < 0.01, n = 12), a clinical measure of diastolic function. These data indicate for the first time that myocyte mechanosensitivity as indicated by the MLP ratio is regulated primarily by myocyte contractility via HO-1 and HDAC signalling.
Collapse
Affiliation(s)
- Anju Paudyal
- Institute of Cardiovascular and Metabolic Research, School of Biological Sciences, Hopkins Building, University of Reading, Whiteknights, Reading, UK
| | - Sukriti Dewan
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Cindy Ikie
- Institute of Cardiovascular and Metabolic Research, School of Biological Sciences, Hopkins Building, University of Reading, Whiteknights, Reading, UK
| | | | - Pieter P de Tombe
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Samuel Y Boateng
- Institute of Cardiovascular and Metabolic Research, School of Biological Sciences, Hopkins Building, University of Reading, Whiteknights, Reading, UK
| |
Collapse
|
7
|
Alpha-1-adrenergic receptors in heart failure: the adaptive arm of the cardiac response to chronic catecholamine stimulation. J Cardiovasc Pharmacol 2014; 63:291-301. [PMID: 24145181 DOI: 10.1097/fjc.0000000000000032] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alpha-1-adrenergic receptors (ARs) are G protein-coupled receptors activated by catecholamines. The alpha-1A and alpha-1B subtypes are expressed in mouse and human myocardium, whereas the alpha-1D protein is found only in coronary arteries. There are far fewer alpha-1-ARs than beta-ARs in the nonfailing heart, but their abundance is maintained or increased in the setting of heart failure, which is characterized by pronounced chronic elevation of catecholamines and beta-AR dysfunction. Decades of evidence from gain and loss-of-function studies in isolated cardiac myocytes and numerous animal models demonstrate important adaptive functions for cardiac alpha-1-ARs to include physiological hypertrophy, positive inotropy, ischemic preconditioning, and protection from cell death. Clinical trial data indicate that blocking alpha-1-ARs is associated with incident heart failure in patients with hypertension. Collectively, these findings suggest that alpha-1-AR activation might mitigate the well-recognized toxic effects of beta-ARs in the hyperadrenergic setting of chronic heart failure. Thus, exogenous cardioselective activation of alpha-1-ARs might represent a novel and viable approach to the treatment of heart failure.
Collapse
|
8
|
Blackmore HL, Niu Y, Fernandez-Twinn DS, Tarry-Adkins JL, Giussani DA, Ozanne SE. Maternal diet-induced obesity programs cardiovascular dysfunction in adult male mouse offspring independent of current body weight. Endocrinology 2014; 155:3970-80. [PMID: 25051449 PMCID: PMC4255219 DOI: 10.1210/en.2014-1383] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 07/12/2014] [Indexed: 02/02/2023]
Abstract
Obese pregnancies are not only associated with adverse consequences for the mother but also the long-term health of her child. Human studies have shown that individuals from obese mothers are at increased risk of premature death from cardiovascular disease (CVD), but are unable to define causality. This study aimed to determine causality using a mouse model of maternal diet-induced obesity. Obesity was induced in female C57BL/6 mice by feeding a diet rich in simple sugars and saturated fat 6 weeks prior to pregnancy and throughout pregnancy and lactation. Control females were fed laboratory chow. Male offspring from both groups were weaned onto chow and studied at 3, 5, 8, and 12 weeks of age for gross cardiac morphometry using stereology, cardiomyocyte cell area by histology, and cardiac fetal gene expression using qRT-PCR. Cardiac function was assessed by isolated Langendorff technology at 12 weeks of age and hearts were analyzed at the protein level for the expression of the β1 adrenergic receptor, muscarinic type-2 acetylcholine receptor, and proteins involved in cardiac contraction. Offspring from obese mothers develop pathologic cardiac hypertrophy associated with re-expression of cardiac fetal genes. By young adulthood these offspring developed severe systolic and diastolic dysfunction and cardiac sympathetic dominance. Importantly, cardiac dysfunction occurred in the absence of any change in corresponding body weight and despite the offspring eating a healthy low-fat diet. These findings provide a causal link to explain human observations relating maternal obesity with premature death from CVD in her offspring.
Collapse
Affiliation(s)
- Heather L Blackmore
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science (H.L.B., D.S.F.-T., J.L.T.-A., S.E.O.), Addenbrookes Hospital, Cambridge CB2 0QQ, United Kingdom; and Department of Physiology, Development, and Neuroscience (Y.N., D.A.G.), University of Cambridge, Cambridge CB2 3EG, United Kingdom
| | | | | | | | | | | |
Collapse
|
9
|
O'Connell TD, Jensen BC, Baker AJ, Simpson PC. Cardiac alpha1-adrenergic receptors: novel aspects of expression, signaling mechanisms, physiologic function, and clinical importance. Pharmacol Rev 2013; 66:308-33. [PMID: 24368739 PMCID: PMC3880467 DOI: 10.1124/pr.112.007203] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Adrenergic receptors (AR) are G-protein-coupled receptors (GPCRs) that have a crucial role in cardiac physiology in health and disease. Alpha1-ARs signal through Gαq, and signaling through Gq, for example, by endothelin and angiotensin receptors, is thought to be detrimental to the heart. In contrast, cardiac alpha1-ARs mediate important protective and adaptive functions in the heart, although alpha1-ARs are only a minor fraction of total cardiac ARs. Cardiac alpha1-ARs activate pleiotropic downstream signaling to prevent pathologic remodeling in heart failure. Mechanisms defined in animal and cell models include activation of adaptive hypertrophy, prevention of cardiac myocyte death, augmentation of contractility, and induction of ischemic preconditioning. Surprisingly, at the molecular level, alpha1-ARs localize to and signal at the nucleus in cardiac myocytes, and, unlike most GPCRs, activate "inside-out" signaling to cause cardioprotection. Contrary to past opinion, human cardiac alpha1-AR expression is similar to that in the mouse, where alpha1-AR effects are seen most convincingly in knockout models. Human clinical studies show that alpha1-blockade worsens heart failure in hypertension and does not improve outcomes in heart failure, implying a cardioprotective role for human alpha1-ARs. In summary, these findings identify novel functional and mechanistic aspects of cardiac alpha1-AR function and suggest that activation of cardiac alpha1-AR might be a viable therapeutic strategy in heart failure.
Collapse
Affiliation(s)
- Timothy D O'Connell
- VA Medical Center (111-C-8), 4150 Clement St., San Francisco, CA 94121. ; or Dr. Timothy D. O'Connell, E-mail:
| | | | | | | |
Collapse
|
10
|
Negative feedback regulation of Homer 1a on norepinephrine-dependent cardiac hypertrophy. Exp Cell Res 2013; 319:1804-1814. [PMID: 23664835 DOI: 10.1016/j.yexcr.2013.04.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 04/22/2013] [Accepted: 04/28/2013] [Indexed: 01/14/2023]
Abstract
Homers are scaffolding proteins that modulate diverse cell functions being able to assemble signalling complexes. In this study, the presence, sub-cellular distribution and function of Homer 1 was investigated. Homer 1a and Homer 1b/c are constitutively expressed in cardiac muscle of both mouse and rat and in HL-1 cells, a cardiac cell line. As judged by confocal immunofluorescence microscopy, Homer 1a displays sarcomeric and peri-nuclear localization. In cardiomyocytes and cultured HL-1 cells, the hypertrophic agonist norepinephrine (NE) induces α1-adrenergic specific Homer 1a over-expression, with a two-to-three-fold increase within 1h, and no up-regulation of Homer 1b/c, as judged by Western blot and qPCR. In HL-1 cells, plasmid-driven over-expression of Homer 1a partially antagonizes activation of ERK phosphorylation and ANF up-regulation, two well-established, early markers of hypertrophy. At the morphometric level, NE-induced increase of cell size is likewise and partially counteracted by exogenous Homer 1a. Under the same experimental conditions, Homer 1b/c does not have any effect on ANF up-regulation nor on cell hypertrophy. Thus, Homer 1a up-regulation is associated to early stages of cardiac hypertrophy and appears to play a negative feedback regulation on molecular transducers of hypertrophy.
Collapse
|
11
|
Ye J, Cardona M, Llovera M, Comella JX, Sanchis D. Translation of Myocyte Enhancer Factor-2 is induced by hypertrophic stimuli in cardiomyocytes through a Calcineurin-dependent pathway. J Mol Cell Cardiol 2012; 53:578-87. [DOI: 10.1016/j.yjmcc.2012.07.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 07/18/2012] [Accepted: 07/19/2012] [Indexed: 10/28/2022]
|
12
|
Sharma S, Liu J, Wei J, Yuan H, Zhang T, Bishopric NH. Repression of miR-142 by p300 and MAPK is required for survival signalling via gp130 during adaptive hypertrophy. EMBO Mol Med 2012; 4:617-32. [PMID: 22367739 PMCID: PMC3407949 DOI: 10.1002/emmm.201200234] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 02/16/2012] [Accepted: 02/20/2012] [Indexed: 11/22/2022] Open
Abstract
An increase in cardiac workload, ultimately resulting in hypertrophy, generates oxidative stress and therefore requires the activation of both survival and growth signal pathways. Here, we wanted to characterize the regulators, targets and mechanistic roles of miR-142, a microRNA (miRNA) negatively regulated during hypertrophy. We show that both miRNA-142-3p and -5p are repressed by serum-derived growth factors in cultured cardiac myocytes, in models of cardiac hypertrophy in vivo and in human cardiomyopathic hearts. Levels of miR-142 are inversely related to levels of acetyltransferase p300 and MAPK activity. When present, miR-142 inhibits both survival and growth pathways by directly targeting nodal regulators p300 and gp130. MiR-142 also potently represses multiple components of the NF-κB pathway, preventing cytokine-mediated NO production and blocks translation of α-actinin. Forced expression of miR-142 during hypertrophic growth induced extensive apoptosis and cardiac dysfunction; conversely, loss of miR-142 fully rescued cardiac function in a murine heart failure model. Downregulation of miR-142 is required to enable cytokine-mediated survival signalling during cardiac growth in response to haemodynamic stress and is a critical element of adaptive hypertrophy.
Collapse
Affiliation(s)
- Salil Sharma
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | | | | | | | | | | |
Collapse
|
13
|
Faul C, Amaral AP, Oskouei B, Hu MC, Sloan A, Isakova T, Gutiérrez OM, Aguillon-Prada R, Lincoln J, Hare JM, Mundel P, Morales A, Scialla J, Fischer M, Soliman EZ, Chen J, Go AS, Rosas SE, Nessel L, Townsend RR, Feldman HI, St John Sutton M, Ojo A, Gadegbeku C, Di Marco GS, Reuter S, Kentrup D, Tiemann K, Brand M, Hill JA, Moe OW, Kuro-O M, Kusek JW, Keane MG, Wolf M. FGF23 induces left ventricular hypertrophy. J Clin Invest 2011; 121:4393-408. [PMID: 21985788 DOI: 10.1172/jci46122] [Citation(s) in RCA: 1518] [Impact Index Per Article: 108.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 08/25/2011] [Indexed: 12/11/2022] Open
Abstract
Chronic kidney disease (CKD) is a public health epidemic that increases risk of death due to cardiovascular disease. Left ventricular hypertrophy (LVH) is an important mechanism of cardiovascular disease in individuals with CKD. Elevated levels of FGF23 have been linked to greater risks of LVH and mortality in patients with CKD, but whether these risks represent causal effects of FGF23 is unknown. Here, we report that elevated FGF23 levels are independently associated with LVH in a large, racially diverse CKD cohort. FGF23 caused pathological hypertrophy of isolated rat cardiomyocytes via FGF receptor-dependent activation of the calcineurin-NFAT signaling pathway, but this effect was independent of klotho, the coreceptor for FGF23 in the kidney and parathyroid glands. Intramyocardial or intravenous injection of FGF23 in wild-type mice resulted in LVH, and klotho-deficient mice demonstrated elevated FGF23 levels and LVH. In an established animal model of CKD, treatment with an FGF-receptor blocker attenuated LVH, although no change in blood pressure was observed. These results unveil a klotho-independent, causal role for FGF23 in the pathogenesis of LVH and suggest that chronically elevated FGF23 levels contribute directly to high rates of LVH and mortality in individuals with CKD.
Collapse
Affiliation(s)
- Christian Faul
- Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Jensen BC, O'Connell TD, Simpson PC. Alpha-1-adrenergic receptors: targets for agonist drugs to treat heart failure. J Mol Cell Cardiol 2011; 51:518-28. [PMID: 21118696 PMCID: PMC3085055 DOI: 10.1016/j.yjmcc.2010.11.014] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Accepted: 11/12/2010] [Indexed: 12/19/2022]
Abstract
Evidence from cell, animal, and human studies demonstrates that α1-adrenergic receptors mediate adaptive and protective effects in the heart. These effects may be particularly important in chronic heart failure, when catecholamine levels are elevated and β-adrenergic receptors are down-regulated and dysfunctional. This review summarizes these data and proposes that selectively activating α1-adrenergic receptors in the heart might represent a novel and effective way to treat heart failure. This article is part of a special issue entitled "Key Signaling Molecules in Hypertrophy and Heart Failure."
Collapse
Affiliation(s)
- Brian C. Jensen
- Cardiology Division, VA Medical Center; Cardiovascular Research Institute; and Department of Medicine, Cardiology Division, University of California, San Francisco, CA, USA
- University of North Carolina, Cardiology Division, 160 Dental Circle, Chapel Hill, NC 27599-7075 USA
| | - Timothy D. O'Connell
- Cardiovascular Health Research Center, Sanford Research/University of South Dakota, 2301 E. 60th Street, Sioux Falls, SD 57104, USA
| | - Paul C. Simpson
- Cardiology Division, VA Medical Center; Cardiovascular Research Institute; and Department of Medicine, Cardiology Division, University of California, San Francisco, CA, USA
| |
Collapse
|
15
|
López JE, Myagmar BE, Swigart PM, Montgomery MD, Haynam S, Bigos M, Rodrigo MC, Simpson PC. β-myosin heavy chain is induced by pressure overload in a minor subpopulation of smaller mouse cardiac myocytes. Circ Res 2011; 109:629-38. [PMID: 21778428 DOI: 10.1161/circresaha.111.243410] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Induction of the fetal hypertrophic marker gene β-myosin heavy chain (β-MyHC) is a signature feature of pressure overload hypertrophy in rodents. β-MyHC is assumed present in all or most enlarged myocytes. OBJECTIVE To quantify the number and size of myocytes expressing endogenous β-MyHC by a flow cytometry approach. METHODS AND RESULTS Myocytes were isolated from the left ventricle of male C57BL/6J mice after transverse aortic constriction (TAC), and the fraction of cells expressing endogenous β-MyHC was quantified by flow cytometry on 10,000 to 20,000 myocytes with use of a validated β-MyHC antibody. Side scatter by flow cytometry in the same cells was validated as an index of myocyte size. β-MyHC-positive myocytes constituted 3 ± 1% of myocytes in control hearts (n=12), increasing to 25 ± 10% at 3 days to 6 weeks after TAC (n=24, P<0.01). β-MyHC-positive myocytes did not enlarge with TAC and were smaller at all times than myocytes without β-MyHC (≈70% as large, P<0.001). β-MyHC-positive myocytes arose by addition of β-MyHC to α-MyHC and had more total MyHC after TAC than did the hypertrophied myocytes that had α-MyHC only. Myocytes positive for β-MyHC were found in discrete regions of the left ventricle in 3 patterns: perivascular, in areas with fibrosis, and in apparently normal myocardium. CONCLUSIONS β-MyHC protein is induced by pressure overload in a minor subpopulation of smaller cardiac myocytes. The hypertrophied myocytes after TAC have α-MyHC only. These data challenge the current paradigm of the fetal hypertrophic gene program and identify a new subpopulation of smaller working ventricular myocytes with more myosin.
Collapse
Affiliation(s)
- Javier E López
- VA Medical Center (111-C-8), 4150 Clement St, San Francisco, CA 94121, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Dorn GW. Adrenergic signaling polymorphisms and their impact on cardiovascular disease. Physiol Rev 2010; 90:1013-62. [PMID: 20664078 DOI: 10.1152/physrev.00001.2010] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
This review examines the impact of recent discoveries defining personal genetics of adrenergic signaling polymorphisms on scientific discovery and medical practice related to cardiovascular diseases. The adrenergic system is the major regulator of minute-by-minute cardiovascular function. Inhibition of adrenergic signaling with pharmacological beta-adrenergic receptor antagonists (beta-blockers) is first-line therapy for heart failure and hypertension. Advances in pharmacology, molecular biology, and genetics of adrenergic signaling pathways have brought us to the point where personal genetic differences in adrenergic signaling factors are being assessed as determinants of risk or progression of cardiovascular disease. For a few polymorphisms, functional data generated in cell-based systems, genetic mouse models, and pharmacological provocation of human subjects are concordant with population studies that suggest altered risk of cardiovascular disease or therapeutic response to beta-blockers. For the majority of adrenergic pathway polymorphisms however, published data conflict, and the clinical relevance of individual genotyping remains uncertain. Here, the current state of laboratory and clinical evidence that adrenergic pathway polymorphisms can affect cardiovascular pathophysiology is comprehensively reviewed and compared, with a goal of placing these data in the broad context of potential clinical applicability.
Collapse
Affiliation(s)
- Gerald W Dorn
- Center for Pharmacogenomics, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| |
Collapse
|
17
|
Matkovich SJ, Zhang Y, Van Booven DJ, Dorn GW. Deep mRNA sequencing for in vivo functional analysis of cardiac transcriptional regulators: application to Galphaq. Circ Res 2010; 106:1459-67. [PMID: 20360248 PMCID: PMC2891025 DOI: 10.1161/circresaha.110.217513] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
RATIONALE Transcriptional profiling can detect subclinical heart disease and provide insight into disease etiology and functional status. Current microarray-based methods are expensive and subject to artifact. OBJECTIVE To develop RNA sequencing methodologies using next generation massively parallel platforms for high throughput comprehensive analysis of individual mouse cardiac transcriptomes. To compare the results of sequencing- and array-based transcriptional profiling in the well-characterized Galphaq transgenic mouse hypertrophy/cardiomyopathy model. METHODS AND RESULTS The techniques for preparation of individually bar-coded mouse heart RNA libraries for Illumina Genome Analyzer II resequencing are described. RNA sequencing showed that 234 high-abundance transcripts (>60 copies/cell) comprised 55% of total cardiac mRNA. Parallel transcriptional profiling of Galphaq transgenic and nontransgenic hearts by Illumina RNA sequencing and Affymetrix Mouse Gene 1.0 ST arrays revealed superior dynamic range for mRNA expression and enhanced specificity for reporting low-abundance transcripts by RNA sequencing. Differential mRNA expression in Galphaq and nontransgenic hearts correlated well between microarrays and RNA sequencing for highly abundant transcripts. RNA sequencing was superior to arrays for accurately quantifying lower-abundance genes, which represented the majority of the regulated genes in the Galphaq transgenic model. CONCLUSIONS RNA sequencing is rapid, accurate, and sensitive for identifying both abundant and rare cardiac transcripts, and has significant advantages in time- and cost-efficiencies over microarray analysis.
Collapse
Affiliation(s)
- Scot J Matkovich
- Department of Medicine, Center for Pharmacogenomics, Washington University School of Medicine, St Louis, Mo 63110, USA
| | | | | | | |
Collapse
|
18
|
Vantler M, Karikkineth BC, Naito H, Tiburcy M, Didié M, Nose M, Rosenkranz S, Zimmermann WH. PDGF-BB protects cardiomyocytes from apoptosis and improves contractile function of engineered heart tissue. J Mol Cell Cardiol 2010; 48:1316-23. [PMID: 20307544 DOI: 10.1016/j.yjmcc.2010.03.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 02/26/2010] [Accepted: 03/15/2010] [Indexed: 12/15/2022]
Abstract
Platelet-derived-growth-factor-BB (PDGF-BB) can protect various cell types from apoptotic cell death, and induce hypertrophic growth and proliferation, but little is known about its direct or indirect effects on cardiomyocytes. Cardiac muscle engineering is compromised by a particularly high rate of cardiomyocyte death. Here we hypothesized that PDGF-BB stimulation can (1) protect cardiomyocytes from apoptosis, (2) enhance myocyte content in and (3) consequently optimize contractile performance of engineered heart tissue (EHT). We investigated the effects of PDGF-receptor activation in neonatal rat heart monolayer- and EHT-cultures by isometric contraction experiments, cytomorphometry, (3)H-thymidine and (3)H-phenylalanine incorporation assays, quantitative PCR (calsequestrin 2, alpha-cardiac and skeletal actin, atrial natriuretic factor, alpha- and beta-myosin heavy chain), immunoblotting (activated caspase 3, Akt-phosphorylation), and ELISA (cell death detection). PDGF-BB did not induce hypertrophy or proliferation in cardiomyocytes, but enhanced contractile performance of EHT. This effect was concentration-dependent (E(max) 10 ng/ml) and maximal only after transient PDGF-BB stimulation (culture days 0-7; total culture duration: 12 days). Improvement of contractile function was associated with higher cardiomyocyte content, as a consequence of PDGF-BB mediated protection from apoptosis (lower caspase-3 activity particularly in cardiomyocytes in PDGF-BB treated vs. untreated EHTs). We confirmed the anti-apoptotic effect of PDGF-BB in monolayer cultures and observed that PI3-kinase inhibition with LY294002 attenuated PDGF-BB-mediated cardiomyocyte protection. We conclude that PDGF-BB does not induce hypertrophy or proliferation, but confers an anti-apoptotic effect on cardiomyocytes. Our findings suggest a further exploitation of PDGF-BB in cardiomyocyte protection in vivo and in vitro.
Collapse
Affiliation(s)
- Marius Vantler
- Klinik III für Innere Medizin, Universität zu Köln, Germany
| | | | | | | | | | | | | | | |
Collapse
|
19
|
SHP2 mediates gp130-dependent cardiomyocyte hypertrophy via negative regulation of skeletal alpha-actin gene. J Mol Cell Cardiol 2010; 49:157-64. [PMID: 20226789 DOI: 10.1016/j.yjmcc.2010.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Revised: 02/27/2010] [Accepted: 03/02/2010] [Indexed: 11/21/2022]
Abstract
Morphological and biochemical phenotypes of cardiomyocyte hypertrophy are determined by neurohumoral factors. Stimulation of G protein-coupled receptor (GPCR) results in uniform cell enlargement in all directions with an increase in skeletal alpha-actin (alpha-SKA) gene expression, while stimulation of gp130 receptor by interleukin-6 (IL-6)-related cytokines induces longitudinal elongation with no increase in alpha-SKA gene expression. Thus, alpha-SKA is a discriminating marker for hypertrophic phenotypes; however, regulatory mechanisms of alpha-SKA gene expression remain unknown. Here, we clarified the role of SH2-containing protein tyrosine phosphatase 2 (SHP2) in alpha-SKA gene expression. In neonatal rat cardiomyocytes, endothelin-1 (ET-1), a GPCR agonist, but not leukemia inhibitory factor (LIF), an IL-6-related cytokine, induced RhoA activation and promotes alpha-SKA gene expression via RhoA. In contrast, LIF, but not ET-1, induced activation of SHP2 in cardiomyocytes, suggesting that SHP2 might negatively regulate alpha-SKA gene expression downstream of gp130. Therefore, we examined the effect of adenovirus-mediated overexpression of wild-type SHP2 (SHP2(WT)), dominant-negative SHP2 (SHP2(C/S)), or beta-galactosidase (beta-gal), on alpha-SKA gene expression. LIF did not upregulate alpha-SKA mRNA in cardiomyocytes overexpressing either beta-gal or SHP2(WT). In cardiomyocytes overexpressing SHP2(C/S), LIF induced upregulation of alpha-SKA mRNA, which was abrogated by concomitant overexpression of either C3-toxin or dominant-negative RhoA. RhoA was activated after LIF stimulation in the cardiomyocytes overexpressing SHP2(C/S), but not in myocytes overexpressing beta-gal. Furthermore, SHP2 mediates LIF-induced longitudinal elongation of cardiomyocytes via ERK5 activation. Collectively, these findings indicate that SHP2 negatively regulates alpha-SKA expression via RhoA inactivation and suggest that SHP2 implicates ERK5 in cardiomyocyte elongation downstream of gp130.
Collapse
|
20
|
Driesen RB, Verheyen FK, Debie W, Blaauw E, Babiker FA, Cornelussen RNM, Ausma J, Lenders MH, Borgers M, Chaponnier C, Ramaekers FCS. Re-expression of alpha skeletal actin as a marker for dedifferentiation in cardiac pathologies. J Cell Mol Med 2009; 13:896-908. [PMID: 19538254 PMCID: PMC3823406 DOI: 10.1111/j.1582-4934.2008.00523.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Differentiation of foetal cardiomyocytes is accompanied by sequential actin isoform expression, i.e. down-regulation of the ‘embryonic’ alpha smooth muscle actin, followed by an up-regulation of alpha skeletal actin (αSKA) and a final predominant expression of alpha cardiac actin (αCA). Our objective was to detect whether re-expression of αSKA occurred during cardiomyocyte dedifferentiation, a phenomenon that has been observed in different pathologies characterized by myocardial dysfunction. Immunohistochemistry of αCA, αSKA and cardiotin was performed on left ventricle biopsies from human patients after coronary bypass surgery. Furthermore, actin isoform expression was investigated in left ventricle samples of rabbit hearts suffering from pressure- and volume-overload and in adult rabbit ventricular cardiomyocytes during dedifferentiation in vitro. Atrial goat samples up to 16 weeks of sustained atrial fibrillation (AF) were studied ultrastructurally and were immunostained for αCA and αSKA. Up-regulation of αSKA was observed in human ventricular cardiomyocytes showing down-regulation of αCA and cardiotin. A patchy re-expression pattern of αSKA was observed in rabbit left ventricular tissue subjected to pressure- and volume-overload. Dedifferentiating cardiomyocytes in vitro revealed a degradation of the contractile apparatus and local re-expression of αSKA. Comparable αSKA staining patterns were found in several areas of atrial goat tissue during 16 weeks of AF together with a progressive glycogen accumulation at the same time intervals. The expression of αSKA in adult dedifferentiating cardiomyocytes, in combination with PAS-positive glycogen and decreased cardiotin expression, offers an additional tool in the evaluation of myocardial dysfunction and indicates major changes in the contractile properties of these cells.
Collapse
Affiliation(s)
- Ronald B Driesen
- Department of Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Park-Holohan SJ, Asselin MC, Turton DR, Williams SL, Hume SP, Camici PG, Rimoldi OE. Quantification of [11C]GB67 binding to cardiac alpha1-adrenoceptors with positron emission tomography: validation in pigs. Eur J Nucl Med Mol Imaging 2008; 35:1624-35. [PMID: 18481065 DOI: 10.1007/s00259-008-0805-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2007] [Accepted: 04/05/2008] [Indexed: 11/27/2022]
Abstract
INTRODUCTION An increase in human cardiac alpha(1)-adrenoceptor (alpha(1)-AR) density is associated with various diseases such as myocardial ischemia, congestive heart failure, hypertrophic cardiomyopathy and hypertension. Positron emission tomography (PET) with an appropriate radioligand offers the possibility of imaging receptor function in the normal and diseased heart. [(11)C]GB67, an analogue of prazosin, has been shown in rats to have potential as a PET ligand with high selectivity to alpha(1)-AR. However, alpha(1)-AR density is up to ten times higher in rat heart compared to that in man. The aim of the present preclinical study was to extend the previous evaluation to a large mammal heart, where the alpha(1)-AR density is comparable to man, and to validate a method for quantification before PET studies in man. METHODS Seven [(11)C]GB67 PET studies, with weight-adjusted target dose of either 5.29 MBq kg(-1) (pilot, test-retest and baseline-predose studies) or 8.22 MBq kg(-1) (baseline-displacement studies), were performed in four anaesthetised pigs (39.5 +/- 3.9 kg). Total myocardial volume of distribution (V (T)) was estimated under different pharmacological conditions using compartmental analysis with a radiolabelled metabolite-corrected arterial plasma input function. A maximum possible blocking dose of 0.12 mumol kg(-1) of unlabeled GB67 was given 20 min before [(11)C]GB67 administration in the predose study and 45 min after administration of [(11)C]GB67 in the displacement study. In addition, [(15)O]CO (3,000 MBq) and [(15)O]H(2)O, with weight adjusted target dose of 10.57 MBq kg(-1), were also administered for estimation of blood volume recovery (RC) of the left ventricular cavity and myocardial perfusion (MBF), respectively. RESULTS [(11)C]GB67 V (T) values (in ml cm(-3)) were estimated to be 24.2 +/- 5.5 (range, 17.3-31.3), 10.1 (predose) and 11.6 (displacement). MBF did not differ within each pig, including between baseline and predose conditions. Predose and displacement studies showed that specific binding of [(11)C]GB67 to myocardial alpha(1)-ARs accounts for approximately 50% of V (T). CONCLUSION The present study offers a methodology for using [(11)C]GB67 as a radioligand to quantify human myocardial alpha(1)-ARs in clinical PET studies.
Collapse
Affiliation(s)
- So-Jin Park-Holohan
- Hammersmith Imanet Ltd., GE HealthCare, Cyclotron Building, Hammersmith Hospital, Du Cane Road, London, UK
| | | | | | | | | | | | | |
Collapse
|
22
|
Wei JQ, Shehadeh L, Mitrani J, Pessanha M, Slepak TI, Webster KA, Bishopric NH. Quantitative control of adaptive cardiac hypertrophy by acetyltransferase p300. Circulation 2008; 118:934-46. [PMID: 18697823 PMCID: PMC2726266 DOI: 10.1161/circulationaha.107.760488] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Acetyltransferase p300 is essential for cardiac development and is thought to be involved in cardiac myocyte growth through MEF2- and GATA4-dependent transcription. However, the importance of p300 in the modulation of cardiac growth in vivo is unknown. METHODS AND RESULTS Pressure overload induced by transverse aortic coarctation, postnatal physiological growth, and human heart failure were associated with large increases in p300. Minimal transgenic overexpression of p300 (1.5- to 3.5-fold) induced striking myocyte and cardiac hypertrophy. Both mortality and cardiac mass were directly related to p300 protein dosage. Heterozygous loss of a single p300 allele reduced pressure overload-induced hypertrophy by approximately 50% and rescued the hypertrophic phenotype of p300 overexpressers. Increased p300 expression had no effect on total histone deacetylase activity but was associated with proportional increases in p300 acetyltransferase activity and acetylation of the p300 substrates histone 3 and GATA-4. Remarkably, a doubling of p300 levels was associated with the de novo acetylation of MEF2. Consistent with this, genes specifically upregulated in p300 transgenic hearts were highly enriched for MEF2 binding sites. CONCLUSIONS Small increments in p300 are necessary and sufficient to drive myocardial hypertrophy, possibly through acetylation of MEF2 and upstream of signals promoting phosphorylation or nuclear export of histone deacetylases. We propose that induction of myocardial p300 content is a primary rate-limiting event in the response to hemodynamic loading in vivo and that p300 availability drives and constrains adaptive myocardial growth. Specific reduction of p300 content or activity may diminish stress-induced hypertrophy and forestall the development of heart failure.
Collapse
Affiliation(s)
- Jian Qin Wei
- University of Miami School of Medicine, Departments of Molecular and Cellular Pharmacology, Medicine and Pediatrics
| | - Lina Shehadeh
- University of Miami School of Medicine, Departments of Molecular and Cellular Pharmacology, Medicine and Pediatrics
| | - James Mitrani
- University of Miami School of Medicine, Departments of Molecular and Cellular Pharmacology, Medicine and Pediatrics
| | - Monica Pessanha
- University of Miami School of Medicine, Departments of Molecular and Cellular Pharmacology, Medicine and Pediatrics
| | - Tatiana I. Slepak
- University of Miami School of Medicine, Departments of Molecular and Cellular Pharmacology, Medicine and Pediatrics
| | - Keith A. Webster
- University of Miami School of Medicine, Departments of Molecular and Cellular Pharmacology, Medicine and Pediatrics
| | - Nanette H. Bishopric
- University of Miami School of Medicine, Departments of Molecular and Cellular Pharmacology, Medicine and Pediatrics
| |
Collapse
|
23
|
Parker TG, Chow KL, Schwartz RJ, Schneider MD. TGF-beta 1 and fibroblast growth factors selectively up-regulate tissue-specific fetal genes in cardiac muscle cells. CIBA FOUNDATION SYMPOSIUM 2007; 157:152-60; discussion 161-4. [PMID: 1712696 DOI: 10.1002/9780470514061.ch10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
TGF-beta 1, like basic and acidic fibroblast growth factor (FGF), inhibits differentiated gene expression in skeletal myoblasts. It potentiates FGF-beta 1 down-regulated expression of the alpha-myosin heavy chain gene and the sarcoplasmic reticulum calcium ATPase gene, yet up-regulated expression of the genes for beta-myosin heavy chain, atrial natriuretic factor, and both skeletal and smooth muscle alpha-actin-four transcripts associated with the embryonic heart. TGF-beta 1 did not affect cardiac alpha-actin gene expression. These responses resemble the generalized 'fetal' phenotype seen during hypertrophy triggered by a haemodynamic load. Chick skeletal and cardiac alpha-actin promoter-driven reported genes were transfected into neonatal rat cardiac myocytes. TGF-beta 1 stimulated skeletal alpha-actin transcription, but not transcription from the cardiac alpha-actin promoter. Basic FGF produced the same results as TGF-beta 1, but acidic FGF suppressed expression of both alpha-actin genes; these results were true for purified and recombinant FGFs. Modulation of alpha-actin transcription by growth factors corresponded accurately to control of the endogenous genes. Three positive cis-acting elements were critical for skeletal alpha-actin transcription in cardiac, as well as skeletal, myocytes, particularly the downstream CCAAT box-associated repeat. Thus, TGF-beta 1 and FGFs selectively induce an ensemble of 'fetal' genes and differentially regulate alpha-actin transcription in cardiac muscle cells.
Collapse
Affiliation(s)
- T G Parker
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030-3498
| | | | | | | |
Collapse
|
24
|
Frazier DP, Wilson A, Dougherty CJ, Li H, Bishopric NH, Webster KA. PKC-α and TAK-1 are intermediates in the activation ofc-Jun NH2-terminal kinase by hypoxia-reoxygenation. Am J Physiol Heart Circ Physiol 2007; 292:H1675-84. [PMID: 17209006 DOI: 10.1152/ajpheart.01132.2006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
c-Jun NH2-terminal kinase (JNK), a member of the MAPK family of protein kinases, is a stress-response kinase that is activated by proinflammatory cytokines and growth factors coupled to membrane receptors or through nonreceptor pathways by stimuli such as heat shock, UV irradiation, protein synthesis inhibitors, and conditions that elevate the levels of reactive oxygen intermediates (ROI). Ischemia followed by reperfusion or hypoxia with reoxygenation represents a condition of high oxidative stress where JNK activation is associated with elevated ROI. We recently demonstrated that the activation of JNK by this condition is initiated by ROI generated by mitochondrial electron transport and involves sequential activation of the proline-rich kinase 2 and the small GTP-binding factors Rac-1 and Cdc42. Here we present evidence that protein kinase C (PKC) and transforming growth factor-β-activated kinase-1 (TAK-1) are also components of this pathway. Inhibition of PKC with the broad-range inhibitor calphostin C, the PKC-α/β-selective inhibitor Go9367, or adenovirus-expressing dominant-negative PKC-α blocked the phosphorylation of proline-rich kinase 2 and JNK. Reoxygenation activated the mitogen-activated protein kinase kinase kinase, TAK-1, and promoted the formation of a complex containing Rac-1, TAK-1, and JNK but not apoptosis-stimulating kinase-1 or p21-activated kinase-1, which was detected within the first 10 min of reoxygenation. These results identify two new components, PKC and TAK-1, that have not been previously described in this signaling pathway.
Collapse
Affiliation(s)
- Donna P Frazier
- Department of Molecular and Cellular Pharmacology, Vascular Biology Institute, University of Miami School of Medicine, Miami, Florida 33136, USA
| | | | | | | | | | | |
Collapse
|
25
|
O’Connell TD, Swigart PM, Rodrigo M, Ishizaka S, Joho S, Turnbull L, Tecott LH, Baker AJ, Foster E, Grossman W, Simpson PC. Alpha1-adrenergic receptors prevent a maladaptive cardiac response to pressure overload. J Clin Invest 2006; 116:1005-15. [PMID: 16585965 PMCID: PMC1421341 DOI: 10.1172/jci22811] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2004] [Accepted: 01/10/2006] [Indexed: 01/06/2023] Open
Abstract
An alpha1-adrenergic receptor (alpha1-AR) antagonist increased heart failure in the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT), but it is unknown whether this adverse result was due to alpha1-AR inhibition or a nonspecific drug effect. We studied cardiac pressure overload in mice with double KO of the 2 main alpha1-AR subtypes in the heart, alpha 1A (Adra1a) and alpha 1B (Adra1b). At 2 weeks after transverse aortic constriction (TAC), KO mouse survival was only 60% of WT, and surviving KO mice had lower ejection fractions and larger end-diastolic volumes than WT mice. Mechanistically, final heart weight and myocyte cross-sectional area were the same after TAC in KO and WT mice. However, KO hearts after TAC had increased interstitial fibrosis, increased apoptosis, and failed induction of the fetal hypertrophic genes. Before TAC, isolated KO myocytes were more susceptible to apoptosis after oxidative and beta-AR stimulation, and beta-ARs were desensitized. Thus, alpha1-AR deletion worsens dilated cardiomyopathy after pressure overload, by multiple mechanisms, indicating that alpha1-signaling is required for cardiac adaptation. These results suggest that the adverse cardiac effects of alpha1-antagonists in clinical trials are due to loss of alpha1-signaling in myocytes, emphasizing concern about clinical use of alpha1-antagonists, and point to a revised perspective on sympathetic activation in heart failure.
Collapse
Affiliation(s)
- Timothy D. O’Connell
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Philip M. Swigart
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - M.C. Rodrigo
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Shinji Ishizaka
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Shuji Joho
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Lynne Turnbull
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Laurence H. Tecott
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Anthony J. Baker
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Elyse Foster
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - William Grossman
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Paul C. Simpson
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| |
Collapse
|
26
|
Gálvez AS, Brunskill EW, Marreez Y, Benner BJ, Regula KM, Kirschenbaum LA, Dorn GW. Distinct pathways regulate proapoptotic Nix and BNip3 in cardiac stress. J Biol Chem 2005; 281:1442-8. [PMID: 16291751 DOI: 10.1074/jbc.m509056200] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Up-regulation of myocardial Nix and BNip3 is associated with apoptosis in cardiac hypertrophy and ischemia, respectively. To identify mechanisms of gene regulation for these critical cardiac apoptosis effectors, the determinants of Nix and BNip3 promoter activation were elucidated by luciferase reporter gene expression in neonatal rat cardiac myocytes. BNip3 transcription was increased by hypoxia but not by phenylephrine (10 microM), angiotensin II (100 nM), or isoproterenol (10 microM). In contrast, Nix transcription was increased by phenylephrine but not by isoproterenol, angiotensin II, or hypoxia. Since phenylephrine stimulates cardiomyocyte hypertrophy via protein kinase C (PKC), the effects of phorbol myristate acetate (PMA, 10 nM for 24 h) and adenoviral PKC expression were assessed. PMA and PKC alpha, but not PKC epsilon or dominant negative PKC alpha, increased Nix transcription. Multiple Nix promoter GC boxes bound transcription factor Sp-1, and basal and PMA- or PKC alpha-stimulated Nix promoter activity was suppressed by mithramycin inhibition of Sp1-DNA interactions. In vivo determinants of Nix expression were evaluated in Nix promoter-luciferase (NixP) transgenic mice that underwent ischemia-reperfusion (1 h/24 h), transverse aortic coarctation (TAC), or cross-breeding with the G(q) overexpression model of hypertrophy. Luciferase activity increased in G alpha(q)-NixP hearts 3.2 +/- 0.4-fold and in TAC hearts 2.8 +/- 0.4-fold but did not increase with infarction-reperfusion. NixP activity was proportional to the extent of TAC hypertrophy and was inhibited by mithramycin. These studies revealed distinct mechanisms of transcriptional regulation for cardiac Nix and BNip3. BNip3 is hypoxia-inducible, whereas Nix expression was induced by G alpha(q)-mediated hypertrophic stimuli. PKC alpha, a G(q) effector, transduced Nix transcriptional induction via Sp1.
Collapse
Affiliation(s)
- Anita S Gálvez
- Department of Internal Medicine, University of Cincinnati, Cincinnati Ohio 45267-0542, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Koshimizu TA, Tanoue A, Hirasawa A, Yamauchi J, Tsujimoto G. Recent advances in alpha1-adrenoceptor pharmacology. Pharmacol Ther 2003; 98:235-44. [PMID: 12725871 DOI: 10.1016/s0163-7258(03)00033-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
alpha(1)-Adrenergic receptors (ARs) mediate some of the main actions of the natural catecholamines, adrenaline and noradrenaline. They participate in many essential physiological processes, such as sympathetic neurotransmission, modulation of hepatic metabolism, control of vascular tone, cardiac contraction, and the regulation of smooth muscle activity in the genitourinary system. Here, we review recent progress on subtype-specific subcellular localization, participation in signaling cascades, and the pivotal function of alpha(1)-ARs, as delineated through studies on genetically engineered animals. Together, these findings will provide new insights into the physiological and pathophysiological roles of the alpha(1)-ARs.
Collapse
Affiliation(s)
- Taka-aki Koshimizu
- Department of Molecular Cell Pharmacology, National Research Institute for Child Health and Development, 3-35-31, Taishi-do, Setagaya-ku, 154, Tokyo, Japan
| | | | | | | | | |
Collapse
|
28
|
Kroumpouzou E, Gomatos IP, Kataki A, Karayannis M, Dangas GD, Toutouzas P. Common pathways for primary hypertrophic and dilated cardiomyopathy. HYBRIDOMA AND HYBRIDOMICS 2003; 22:41-5. [PMID: 12713689 DOI: 10.1089/153685903321538071] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Testing the hypothesis that hypertrophic and dilated cardiomyopathy as well as viral myocarditis share a common mitogenic growth response pathway with mitotically competent cell types are the aims of this study. The expression of the c-fos, H-ras and c-myc genes was immunohistochemically determined in biopsies from 12 patients with dilated cardiomyopathy, 24 patients with hypertrophic cardiomyopathy, and 4 patients with myocarditis. Normal myocardium from 9 subjects was used as the control group. Staining results were correlated with patient's demographic data. C-fos, H-ras and c-myc protein overexpression was seen in 15 patients (62.5%) with primary hypertrophic and 4 patients (33.3%) with dilated cardiomyopathy. The majority of hypertrophic and dilated cardiomyopathy patients expressed at least one of the genes studied compared with the control group (p = 0.006). Primary cardiomyopathy patients also showed a statistically significant difference in the gene co-expression compared with the control group (p = 0.042). C-fos, H-ras, and C-myc protein expression did not differ substantially between patients with hypertrophic and dilated cardiomyopathy. Patients with myocarditis expressed only the C-fos protein (n = 2, 50%). C-fos, h-ras and c-myc genes are overexpressed in patients with cardiac hypertrophy and cardiac dilation. Cardiac myocytes respond to biomechanical stress by initiating several different processes. One of them is oncogene expression. This results in a hypertrophy of the myocytes proportional in length and width (hypertrophic cardiomyopathy or with a relatively greater increase in length than in the width (dilated cardiomyopathy).
Collapse
Affiliation(s)
- E Kroumpouzou
- Department of Cardiology, Hippokration Hospital, Athens Medical School, Greece
| | | | | | | | | | | |
Collapse
|
29
|
Tsoporis JN, Marks A, Zimmer DB, McMahon C, Parker TG. The myocardial protein S100A1 plays a role in the maintenance of normal gene expression in the adult heart. Mol Cell Biochem 2003; 242:27-33. [PMID: 12619862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
S100A1 and S100B are members of a family of 20 kDa Ca2+-binding homodimers that play a role in signal transduction in mammalian cells. S100A1 is the major isoform in normal heart and S100B, normally a brain protein, is induced in hypertrophic myocardium and functions as an intrinsic negative modulator of the hypertrophic response. In order to examine the function of S100A1, we first showed that, in contrast to S100B, S100A1 was downregulated in rat experimental models of myocardial hypertrophy following myocardial infarction or pressure overload. Second, in co-transfection experiments in cultured neonatal rat cardiac myocytes, S100A1 inhibited the alpha1-adrenergic activation of promoters of genes induced during the hypertrophic response including the fetal genes skeletal alpha actin (skACT), and beta-myosin heavy chain (MHC) and S100B, but not the triiodothyronine (T3) activation of the promoter of the alpha-MHC gene, that is normally expressed in adult myocardium. These results suggest that S100A1 is involved in the maintenance of the genetic program that defines normal myocardial function and that its downregulation is permissive for the induction of genes that underlie myocardial hypertrophy.
Collapse
Affiliation(s)
- James N Tsoporis
- The Centre for Cardiovascular Research, Division of Cardiology, Department of Medicine, The Toronto Hospital, University of Toronto, Toronto, ON, Canada
| | | | | | | | | |
Collapse
|
30
|
Yeh T, Wechsler AS, Graham L, Loesser KE, Sica DA, Wolfe L, Jakoi ER. Central sympathetic blockade ameliorates brain death-induced cardiotoxicity and associated changes in myocardial gene expression. J Thorac Cardiovasc Surg 2002; 124:1087-98. [PMID: 12447173 DOI: 10.1067/mtc.2002.124887] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Brain death results in cardiac injury and hemodynamic instability. After brain death, catecholamine levels surge in concert with increased expression of select myocardial gene products. Sympathetic blockade was used to investigate the effects of the adrenergic nervous system on myocardial gene expression in a rabbit model of brain death. METHODS A balloon expansion model of brain death in rabbits (n = 42) was used with and without sympathetic blockade (xylazine, acetylpromazine, and ketamine). Sham-operated and naive rabbits served as control animals. Over 4 hours, mean arterial pressure, heart rate, electrocardiographic results, catecholamine levels, myocardial histology, and messenger RNA levels were assessed. RESULTS Sympathetic blockade decreased basal catecholamine levels and blocked the catecholamine surge accompanying brain death. The typical hemodynamic instability, adverse electrocardiographic changes, and myocellular injury associated with brain death were all significantly decreased. Sympathetic blockade not only suppressed the previously reported increases in myocardial gene expression (cardiac and skeletal alpha-actin, egr-1, and heat shock protein 70) but also suppressed the expression of multiple other genes (alpha and beta myosin heavy chain, calcium ATPase [sarcoplasmic reticulum Ca(2+)-adenosine triphosphatase pump, SERCA-2a], phospholamban [ryanodine receptor], and c-jun). CONCLUSION Central sympathetic blockade minimizes the hemodynamic instability associated with brain death and neutralizes the increased expression of multiple myocardial gene products associated with brain death.
Collapse
Affiliation(s)
- Thomas Yeh
- Jewish Hospital Cardiovascular Research Center at University of Louisville, Department of Surgery, Division of Cardiothoracic Surgery, University of Louisville, Louisville, KY 40202, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Koshimizu TA, Yamauchi J, Hirasawa A, Tanoue A, Tsujimoto G. Recent progress in alpha 1-adrenoceptor pharmacology. Biol Pharm Bull 2002; 25:401-8. [PMID: 11995914 DOI: 10.1248/bpb.25.401] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The adrenoceptors (ARs) play a key role in the modulation of sympathetic nervous system activity and are a site of action for many clinically important therapeutic agents. The alpha1-adrenoceptor subtypes (alpha1A-, alpha1B-, and alpha1D-AR) play a prominent role in regulating vascular tone and hypertrophic growth of smooth muscle and cardiac cells. Their functional characteristics with respect to ligand binding and second messenger utilization have been well described. Here, we review recent progress on subtype-specific subcellular localization, participation in signaling cascades, and the pivotal function of alpha1-ARs, as delineated through studies on genetically engineered animals. Together, these findings will provide new insights into the physiological and pathophysiological roles of the alpha1-ARs.
Collapse
Affiliation(s)
- Taka-aki Koshimizu
- Department of Molecular, Cell Pharmacology, National Children's Medical Research Center, Tokyo, Japan
| | | | | | | | | |
Collapse
|
32
|
|
33
|
Hamano T, Kobayashi K, Sakairi T, Hayashi M, Mutai M. Peroxisome proliferator-activated receptor alpha (PPAR alpha) agonist, WY-14,643, increased transcription of myosin light chain-2 in cardiomyocytes. J Toxicol Sci 2001; 26:275-84. [PMID: 11871124 DOI: 10.2131/jts.26.275] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear hormone receptors that can be activated by xenobiotics and natural fatty acids. To assess the potential physiological activity of PPAR ligands on cardiac muscular cells, the effects of PPAR alpha agonist, WY-14,643, on both rat hearts and a rat cardiomyocyte cell line (H9c2 cells) were investigated. Male F344 rats were fed a diet containing WY-14,643 at a concentration of 100 ppm for 26 weeks. Cardiac muscular hypertrophy was revealed by morphometric analysis in which the diameter of the muscular fibers in WY-14,643-treated rats was larger than those of control rats. Using H9c2 cells in vitro, the protein content per cell was increased in a dose-dependent manner with the treatment of WY-14,643. The transcription of myosin light chain-2 (MLC-2), a parameter of myocardial hypertrophy, was increased in H9c2 cells transfected with the rat MLC-2/luciferase fusion gene by WY-14,643 as well as other peroxisome proliferators, clofibrate and di(2-ethylhexyl) phthalate. In addition, accumulation of myosin light chain protein was confirmed in H9c2 cells treated with WY-14,643 at 10 micrograms/ml for 7 days or more by immunohistochemistry. These results suggest that PPAR alpha ligands have a potential to regulate MLC-2, which is a contractile protein in cardiomyocytes and may play a part of role in the pathogenesis of cardiac hypertrophy.
Collapse
Affiliation(s)
- T Hamano
- Toxicology Laboratory, Mitsubishi Pharma Co., 100-5 Yana, Kisarazu-shi, Chiba 292-0812, Japan
| | | | | | | | | |
Collapse
|
34
|
Clément S, Pellieux C, Chaponnier C, Pedrazzini T, Gabbiani G. Angiotensin II stimulates alpha-skeletal actin expression in cadiomyocytes in vitro and in vivo in the absence of hypertension. Differentiation 2001; 69:66-74. [PMID: 11776396 DOI: 10.1046/j.1432-0436.2001.690107.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Using a specific alpha-skeletal actin antibody, we have previously shown, that during hypertension-associated cardiac hypertrophy in the rat, the expression of alpha-skeletal actin in the myocardium is increased, but maintains focal distribution, compared to normotensive animals. In the present study, we have investigated whether alpha-skeletal actin expression can be induced in the absence of hypertension. For this purpose, we have examined transgenic mice overexpressing angiotensinogen exclusively in the heart. These animals are characterized by high cardiac angiotensin II levels and cardiac hypertrophy accompanied or not by high blood pressure depending on their genetic background, i.e. presence of one or two renin genes. Alpha-skeletal actin levels were highly increased in transgenic compared to wild-type myocardium independently of the number of renin genes, indicating that angiotensin II can stimulate alpha-skeletal actin expression in normotensive animals. Additional in vitro experiments using cultured mouse and rat cardiomyocytes showed that angiotension II not only increases alpha-skeletal actin expression but also induces an increase of its incorporation within II-bands compared to control cardiomyocytes. Angiotensin II increases also the expression of alpha-smooth muscle actin in sarcomeres of cardiomyocytes as well as in fibroblastic cells present within the culture.
Collapse
Affiliation(s)
- S Clément
- University of Geneva-CMU, Department of Pathology, Switzerland
| | | | | | | | | |
Collapse
|
35
|
Abstract
Hypertension and aging adversely affect cardiovascular system and the heart is invariably involved. Manifestations of hypertensive heart disease and of the aging heart appear similar; ventricular hypertrophy, myocardial fibrosis, and impairments in ventricular function and coronary hemodynamics characterize both conditions. However, a great deal of evidence suggests that different underlying pathophysiological mechanisms may be involved. This report discusses most recent clinical and experimental findings and focuses on the alterations in nonmyocytic elements that are a part of heart involvement. Particular attention was given to factors that are responsible for exaggerated myocardial deposition of collagen that, by itself, may be responsible for ventricular dysfunction and impaired coronary hemodynamics in hypertensive and aging hearts. Newly developed therapeutical strategies, based on the most recent experimental and clinical studies, are also discussed.
Collapse
Affiliation(s)
- J Varagic
- Hypertension Research Laboratory, Alton Ochsner Medical Foundation, New Orleans, Louisiana 70121, USA
| | | | | |
Collapse
|
36
|
Luodonpää M, Vuolteenaho O, Eskelinen S, Ruskoaho H. Effects of adrenomedullin on hypertrophic responses induced by angiotensin II, endothelin-1 and phenylephrine. Peptides 2001; 22:1859-66. [PMID: 11754973 DOI: 10.1016/s0196-9781(01)00505-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We examined whether adrenomedullin (AM), a vasoactive peptide with significant expression and binding sites in the heart, modulates the hypertrophic response in cultured neonatal rat ventricular myocytes. Myocyte hypertrophy was induced by treating the cells with angiotensin II (Ang II), endothelin-1 (ET-1) or alpha-adrenergic agonist, L-phenylephrine (PHE). All treatments resulted in a hypertrophic response as reflected by increased protein synthesis and expression of atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP) genes. AM treatment resulted in a complete inhibition of the Ang II-induced increase in ANP and BNP gene expression and secretion. In contrast, no inhibitory effect was seen in either ET-1-induced natriuretic peptide gene expression or PHE-induced ANP and BNP gene expression and secretion. AM had only a modest effect on basal levels of natriuretic peptide secretion and gene expression. When AM mRNA levels in isolated neonatal rat myocytes treated for 48 h with Ang II, ET-1 or PHE were measured, only Ang II induced a consistent increase in AM gene expression. These results indicate that AM is not invariably associated with attenuation of the hypertrophic response but its effect is dependent on the stimulus activating myocyte hypertrophy. AM may form an important autocrine/paracrine growth-inhibitory loop in Ang II-induced myocyte hypertrophy.
Collapse
Affiliation(s)
- M Luodonpää
- Departments of Pharmacology and Toxicology, Biocenter Oulu, 90014 University of Oulu, Oulu, Finland
| | | | | | | |
Collapse
|
37
|
Borgarelli M, Tarducci A, Tidholm A, Häggström J. Canine idiopathic dilated cardiomyopathy. Part II: pathophysiology and therapy. Vet J 2001; 162:182-95. [PMID: 11681869 DOI: 10.1053/tvjl.2001.0616] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Dilated cardiomyopathy (DCM) in dogs is characterized by ventricular and atrial enlargement, and systolic and diastolic dysfunction, with congestive heart failure (CHF) often developing at some stage. With greater understanding of the impact of neuroendocrine stimulation in heart disease, the understanding of the pathophysiology for CHF has changed considerably. It is no longer considered only to be a simple haemodynamic consequence of pump dysfunction, but is now characterized as a complex clinical syndrome with release of many neurohormones, which are believed to have impact on the progression of disease. This change in our understanding of the pathophysiology of CHF has important therapeutic implications. There are strong indications, although not yet proven, that drugs designed to influence the neuroendocrine activity, such as Angiotensin Converting Enzyme (ACE) inhibitors and beta-receptors antagonists, are efficacious as adjunct therapy of heart failure attributable to DCM in dogs. The benefits of drugs designed to influence the myocardial contractile state (positive inotropes) have not been fully evaluated. However, evidence has emerged in recent years indicating that new types of positive inotropes may be beneficial in dogs with DCM. This review focuses on the neuroendocrine aspects of DCM and their possible therapeutic implications and the place for long-term inotropic support in dogs with DCM.
Collapse
Affiliation(s)
- M Borgarelli
- Department of Animal Pathology, Faculty of Veterinary Medicine, University of Torino, Via Leonardo da Vinci 44, 10095 Grugliasco, (To), Italy
| | | | | | | |
Collapse
|
38
|
Miyashita T, Takeishi Y, Takahashi H, Kato S, Kubota I, Tomoike H. Role of calcineurin in insulin-like growth factor-1-induced hypertrophy of cultured adult rat ventricular myocytes. JAPANESE CIRCULATION JOURNAL 2001; 65:815-9. [PMID: 11548882 DOI: 10.1253/jcj.65.815] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The present study examined the role of calcineurin in insulin-like growth factor (IGF)-1-induced hypertrophy in primary cultures of adult rat ventricular myocytes (ARVM), prepared from the ventricles of 14-16-week-old male Sprague-Dawley rats. The effects of several humoral factors, including phenylephrine, angiotensin II, endothelin-1, IGF-1 and interleukin-6, on the morphology of ARVM were studied. Myocyte surface area was significantly increased by IGF-1 (2,268 +/- 571 to 3,018 +/- 836 microm2, p < 0.01), but not by other humoral factors. This hypertrophic effect of IGF-1 was blocked by genistein (tyrosine kinase inhibitor), PD98059 (MEK inhibitor). These findings suggest that IGF-1 produces ARVM hypertrophy by a tyrosine kinase-MEK mediated pathway as has been reported in neonatal cardiomyocytes. IGF-1-mediated ARVM hypertrophy was also attenuated by cyclosporine A (calcineurin inhibitor), and staurosporine and chelerythrine (protein kinase C inhibitors). IGF-1 markedly increased calcineurin activity (8.7 +/- 1.2 to 98.0 +/- 54.3 pmol x h(-1) mg(-1), p < 0.01), and this activation was completely blocked by pre-treatment with cyclosporine A (8.5 +/- 11.4pmol x h(-1) x mg(-1), p < 0.01) and chelerythrine (2.3 +/- 2.7 pmol x h(-1) mg(-1), p < 0.01). It appears that IGF-1 activates calcineurin by a protein kinase C-dependent pathway. Increased mRNA expression of atrial natriuretic factor by IGF-1 was inhibited by cyclosporine A (p < 0.01). The findings indicate that IGF-1 induces ARVM hypertrophy by protein kinase C and calcineurin-related mechanisms. The fact that elevated calcineurin activity and induced atrial natriuretic factor mRNA expression by IGF-1 were blocked by cyclosporine A further supports the hypothesis that calcineurin is critically involved in IGF-1-induced ARVM hypertrophy.
Collapse
Affiliation(s)
- T Miyashita
- The First Department of Internal Medicine, Yamagata University School of Medicine, Japan
| | | | | | | | | | | |
Collapse
|
39
|
Perez-Terzic C, Gacy AM, Bortolon R, Dzeja PP, Puceat M, Jaconi M, Prendergast FG, Terzic A. Directed inhibition of nuclear import in cellular hypertrophy. J Biol Chem 2001; 276:20566-71. [PMID: 11283025 DOI: 10.1074/jbc.m101950200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Each nuclear pore is responsible for both nuclear import and export with a finite capacity for bidirectional transport across the nuclear envelope. It remains poorly understood how the nuclear transport pathway responds to increased demands for nucleocytoplasmic communication. A case in point is cellular hypertrophy in which increased amounts of genetic material need to be transported from the nucleus to the cytosol. Here, we report an adaptive down-regulation of nuclear import supporting such an increased demand for nuclear export. The induction of cardiac cell hypertrophy by phenylephrine or angiotensin II inhibited the nuclear translocation of H1 histones. The removal of hypertrophic stimuli reversed the hypertrophic phenotype and restored nuclear import. Moreover, the inhibition of nuclear export by leptomycin B rescued import. Hypertrophic reprogramming increased the intracellular GTP/GDP ratio and promoted the nuclear redistribution of the GTP-binding transport factor Ran, favoring export over import. Further, in hypertrophy, the reduced creatine kinase and adenylate kinase activities limited energy delivery to the nuclear pore. The reduction of activities was associated with the closure of the cytoplasmic phase of the nuclear pore preventing import at the translocation step. Thus, to overcome the limited capacity for nucleocytoplasmic transport, cells requiring increased nuclear export regulate the nuclear transport pathway by undergoing a metabolic and structural restriction of nuclear import.
Collapse
Affiliation(s)
- C Perez-Terzic
- Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Mayo Foundation, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
O'Connell TD, Rokosh DG, Simpson PC. Cloning and characterization of the mouse alpha1C/A-adrenergic receptor gene and analysis of an alpha1C promoter in cardiac myocytes: role of an MCAT element that binds transcriptional enhancer factor-1 (TEF-1). Mol Pharmacol 2001; 59:1225-34. [PMID: 11306707 DOI: 10.1124/mol.59.5.1225] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
alpha1-Adrenergic receptor (AR) subtypes in the heart are expressed by myocytes but not by fibroblasts, a feature that distinguishes alpha1-ARs from beta-ARs. Here we studied myocyte-specific expression of alpha1-ARs, focusing on the subtype alpha1C (also called alpha1A), a subtype implicated in cardiac hypertrophic signaling in rat models. We first cloned the mouse alpha1C-AR gene, which consisted of two exons with an 18 kb intron, similar to the alpha1B-AR gene. The receptor coding sequence was >90% homologous to that of rat and human. alpha1C-AR transcription in mouse heart was initiated from a single Inr consensus sequence at -588 from the ATG; this and a putative polyadenylation sequence 8.5 kb 3' could account for the predominant 11 kb alpha1C mRNA in mouse heart. A 5'-nontranscribed fragment of 4.4 kb was active as a promoter in cardiac myocytes but not in fibroblasts. Promoter activity in myocytes required a single muscle CAT (MCAT) element, and this MCAT bound in vitro to recombinant and endogenous transcriptional enhancer factor-1. Thus, alpha1C-AR transcription in cardiac myocytes shares MCAT dependence with other cardiac-specific genes, including the alpha- and beta-myosin heavy chains, skeletal alpha-actin, and brain natriuretic peptide. However, the mouse alpha1C gene was not transcribed in the neonatal heart and was not activated by alpha1-AR and other hypertrophic agonists in rat myocytes, and thus differed from other MCAT-dependent genes and the rat alpha1C gene.
Collapse
Affiliation(s)
- T D O'Connell
- Cardiology Division and Research Service, Veterans Affairs Medical Center, San Francisco, California, USA
| | | | | |
Collapse
|
41
|
Sakaguchi G, Young RL, Komeda M, Yamanaka K, Buxton BF, Louis WJ. Left ventricular aneurysm repair in rats: structural, functional, and molecular consequences. J Thorac Cardiovasc Surg 2001; 121:750-61. [PMID: 11279418 DOI: 10.1067/mtc.2001.112462] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVES This study examined the effects of aneurysm repair in a rat model of myocardial infarction on functional indices and on the spatiotemporal distribution of cardiac contractile protein and natriuretic peptide messenger RNA. METHODS In a rat infarct model, expanded left ventricular aneurysms were plicated 4 weeks after infarction. At 30 weeks, transverse heart sections were taken at 4 levels (apex [level 1] through base [level 4]) and assessed by in situ hybridization histochemistry to determine regional messenger RNA levels of pre-pro-atrial natriuretic peptide, cardiac alpha-actin, skeletal alpha-actin, myosin light chain-2v, and beta-myosin heavy chain. RESULTS Rats with plicated left ventricular aneurysms had reduced left ventricular endocardial circumference (19%, P <.005), lower heart weight ratio (31%, P <.05), left ventricular end-diastolic pressures (51%, P <.05), and increased +/-dP/dt (34%-38%, P <.05). Cardiac messenger RNA levels of pre-pro-atrial natriuretic peptide were reduced in the septum (levels 2 and 3), and skeletal alpha-actin levels were reduced in the septum and left ventricular free wall of plicated rats (level 3). beta-Myosin heavy chain levels were markedly reduced in peri-infarct regions of the left ventricular free wall, septum, and right ventricle in plicated rats at level 4, whereas myosin light chain-2v levels were reduced at levels 2 and 4 in the left ventricular free wall and at level 4 in the right ventricle. CONCLUSIONS Plication of left ventricular aneurysm after infarction in the rat significantly reduced cardiac hypertrophy, improved cardiac function, and reduced the upregulation of pre-pro-atrial natriuretic peptide and both fetal and adult contractile protein isoforms associated with cardiac hypertrophy.
Collapse
Affiliation(s)
- G Sakaguchi
- Departments of Clinical Pharmacology and Therapeutics and Cardiac Surgery, Austin and Repatriation Medical Centre, Heidelberg, Victoria 3084, Australia
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
The acute contractile function of the heart is controlled by the effects of released nonepinephrine (NE) on cardiac adrenergic receptors. NE can also act in a more chronic fashion to induce cardiomyocyte growth, characterized by cell enlargement (hypertrophy), increased protein synthesis, alterations in gene expression and addition of sarcomeres. These responses enhance cardiomyocyte contractile function and thus allow the heart to compensate for increased stress. The hypertrophic effects of NE are mediated through Gq-coupled alpha(1)-adrenergic receptors and are mimicked by the actions of other neurohormones (endothelin, prostaglandin F(2alpha) angiotensin II) that also act on Gq-coupled receptors. Activation of phospholipase C by Gq is necessary for these responses, and protein kinase C and MAP kinases have also been implicated. Gq stimulated cardiac hypertrophy is also evident in transgenic mouse models. In contrast, stimulation of G(s)-coupled beta-adrenergic receptors or G(i)-coupled receptors do not directly effect cardiomyocyte hypertrophy. Apoptosis is also induced by G-protein-coupled receptor stimulation in cardiomyocytes. Sustained or excessive activation of either Gq- or Gs-signaling pathways results in apoptotic loss of cardiomyocytes both in vitro and in vivo. Apoptosis is associated with decreased ventricular function in the failing heart. Cardiomyocytes provide an ideal model system for understanding the basis for G-protein mediated hypertrophy and apoptosis, and the mechanisms responsible for the transition from compensatory to deleterious levels of signaling. This information may prove critical for designing interventions that prevent the pathophysiological consequences of heart failure.
Collapse
Affiliation(s)
- J W Adams
- University of California, San Diego, Department of Pharmacology, 9500 Gilman Drive, 0636, La Jolla, CA 92093-0636, USA
| | | |
Collapse
|
43
|
Zhang TT, Takimoto K, Stewart AF, Zhu C, Levitan ES. Independent regulation of cardiac Kv4.3 potassium channel expression by angiotensin II and phenylephrine. Circ Res 2001; 88:476-82. [PMID: 11249870 DOI: 10.1161/01.res.88.5.476] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hypertrophied cardiac myocytes exhibit prolonged action potentials and decreased transient outward potassium current (I(to)). Because Kv4.3 is a major contributor to I:(to), we studied regulation of its expression in neonatal rat cardiac myocytes in response to the known stimulators of cardiac myocyte hypertrophy, angiotensin II (Ang II) and phenylephrine (PE). RNase protection assays and immunoblots revealed that Ang II and PE each downregulate Kv4.3 mRNA and protein. However, although PE induces a faster and more extensive hypertrophic response than Ang II, the PE effect on Kv4.3 mRNA develops slowly and is sustained, whereas Ang II rapidly and transiently decreases Kv4.3 mRNA expression. Turnover measurements revealed that Kv4.3 mRNA is very stable, with a half-life >20 hours. This suggests that Ang II must destabilize the channel mRNA. In contrast, PE does not affect the rate of Kv4.3 mRNA degradation. To test for transcriptional regulation, the 5' flanking region of the rat Kv4.3 gene was cloned, and Kv4.3 promoter-reporter constructs were expressed in cardiac myocytes. Whereas Ang II was found to have no effect on transcription, PE inhibits Kv4.3 promoter activity. Pharmacological experiments also indicate that PE and Ang II act independently to downregulate Kv4.3 gene expression. Thus, regulation of Kv4.3 gene expression is not a simple secondary response to hypertrophy. Rather, Ang II and PE use different mechanisms to decrease Kv4.3 channel expression in neonatal rat cardiac myocytes.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Angiotensin Receptor Antagonists
- Animals
- Animals, Newborn
- Cells, Cultured
- DNA/genetics
- Drug Synergism
- Gene Expression Regulation/drug effects
- Imidazoles/pharmacology
- Luciferases/genetics
- Luciferases/metabolism
- Myocardium/cytology
- Myocardium/metabolism
- Phenylephrine/pharmacology
- Potassium Channels/drug effects
- Potassium Channels/genetics
- Potassium Channels/metabolism
- Potassium Channels, Voltage-Gated
- Promoter Regions, Genetic/genetics
- RNA, Messenger/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1
- Receptor, Angiotensin, Type 2
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Shal Potassium Channels
- Tetrazoles/pharmacology
- Time Factors
- Transcription, Genetic/drug effects
Collapse
Affiliation(s)
- T T Zhang
- Department of Pharmacology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | |
Collapse
|
44
|
O'Donnell JM, Sumbilla CM, Ma H, Farrance IK, Cavagna M, Klein MG, Inesi G. Tight control of exogenous SERCA expression is required to obtain acceleration of calcium transients with minimal cytotoxic effects in cardiac myocytes. Circ Res 2001; 88:415-21. [PMID: 11230109 DOI: 10.1161/01.res.88.4.415] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Collateral effects of exogenous sarcoendoplasmic reticulum Ca(2+) ATPase (SERCA) expression were characterized in neonatal rat and chicken embryo cardiac myocytes, and the conditions required to produce acceleration of Ca(2+) transients with minimal toxicity were established. Cultured myocytes were infected with adenovirus vector carrying the cDNA of wild-type SERCA1, an inactive SERCA1 mutant, or enhanced green fluorescence protein under control of the cytomegalovirus promoter. Controls were exposed to empty virus vector. Each group was tested with and without phenylephrine (PHE) treatment. Under conditions of limited calf-serum exposure, the infected rat myocytes manifested a more rapid increase in size, protein content, and rate of protein synthesis relative to noninfected controls. These changes were not accompanied by reversal to fetal transcriptional pattern (as observed in hypertrophy triggered by PHE) and may be attributable to facilitated exchange with serum factors. SERCA virus titers >5 to 6 plaque-forming units per cell produced overcrowding of ATPase molecules on intracellular membranes, followed by apoptotic death of a significant number of rat but not chicken myocytes. Enhanced green fluorescence protein virus and empty virus also produced cytotoxic effects but at higher titers than SERCA. Expression of exogenous SERCA and enhancement of Ca(2+) transient kinetics could be obtained with minimal cell damage in rat myocytes if the SERCA virus titer were maintained within 1 to 4 plaque-forming units per cell. Expression of endogenous SERCA was unchanged, but expression of exogenous SERCA was higher in myocytes rendered hypertrophic by treatment with PHE than in nontreated controls.
Collapse
Affiliation(s)
- J M O'Donnell
- Department of Biochemistry, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Varagic J, Susic D, Frohlich ED. Low-dose ACE with alpha- or beta-adrenergic receptor inhibitors have beneficial SHR cardiovascular effects. J Cardiovasc Pharmacol Ther 2001; 6:57-63. [PMID: 11452337 DOI: 10.1177/107424840100600107] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND There are no data regarding the prolonged effect of alpha-1 adrenergic receptor antagonists on ventricular collagen content and coronary hemodynamics in spontaneously hypertensive rats (SHR). This study, therefore, was designed to determine the effects of chronic treatment with the alpha-1 adrenergic receptor inhibitor doxazosin on SHR systemic and regional (especially coronary) hemodynamics, cardiovascular mass, and ventricular collagen. The effects of the combination of doxazosin with low-dose angiotensin-converting enzyme inhibitor were studied versus the alpha-1 antagonist alone. These effects were compared with those of a beta-1 adrenergic receptor inhibitor. METHODS AND RESULTS Systemic and regional hemodynamics (radionuclide-labeled microspheres), left and right ventricular weight, hydroxyproline concentration, and aortic weight were measured at age 35 weeks. Doxazosin reduced arterial pressure and total peripheral resistance without changing left ventricular mass and collagen content, whereas monotherapies with the beta-1 antagonist metoprolol or a subdepressor dose of the ACE inhibitor enalapril were effective in reducing left ventricular mass and hydroxyproline without altering pressure. Doxazosin combined with the same low-dose ACE inhibitor reduced left ventricular mass and hydroxyproline without potentiating the hypotensive effect of doxazosin. By contrast, the combination of beta-1 antagonist with the low-dose ACE inhibitor reduced pressure, unlike either agent alone. Aortic weight index was significantly reduced only by doxazosin whether when used alone or with the ACE inhibitor. Low-dose ACE inhibitor with doxazosin or the beta-1 receptor antagonist as well as doxazosin alone decreased renal vascular resistance. CONCLUSION These data show that the low subdepressor dose ACE inhibitor with an alpha- or beta-adrenergic receptor antagonist provides beneficial cardiovascular effects in SHR.
Collapse
Affiliation(s)
- J Varagic
- Hypertension Research Laboratory, Alton Ochsner Medical Foundation, New Orleans, LA 70121, USA
| | | | | |
Collapse
|
46
|
Deng XF, Rokosh DG, Simpson PC. Autonomous and growth factor-induced hypertrophy in cultured neonatal mouse cardiac myocytes. Comparison with rat. Circ Res 2000; 87:781-8. [PMID: 11055982 DOI: 10.1161/01.res.87.9.781] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cultured neonatal rat cardiac myocytes have been used extensively to study cellular and molecular mechanisms of cardiac hypertrophy. However, there are only a few studies in cultured mouse myocytes despite the increasing use of genetically engineered mouse models of cardiac hypertrophy. Therefore, we characterized hypertrophic responses in low-density, serum-free cultures of neonatal mouse cardiac myocytes and compared them with rat myocytes. In mouse myocyte cultures, triiodothyronine (T3), norepinephrine (NE) through a beta-adrenergic receptor, and leukemia inhibitory factor induced hypertrophy by a 20% to 30% increase in [(3)H]phenylalanine-labeled protein content. T3 and NE also increased alpha-myosin heavy chain (MyHC) mRNA and reduced beta-MyHC. In contrast, hypertrophic stimuli in rat myocytes, including alpha(1)-adrenergic agonists, endothelin-1, prostaglandin F(2alpha), interleukin 1beta, and phorbol 12-myristate 13-acetate (PMA), had no effect on mouse myocyte protein content. In further contrast with the rat, none of these agents increased atrial natriuretic factor or beta-MyHC mRNAs. Acute PMA signaling was intact by extracellular signal-regulated kinase (ERK1/2) and immediate-early gene (fos/jun) activation. Remarkably, mouse but not rat myocytes had hypertrophy in the absence of added growth factors, with increases in cell area, protein content, and the mRNAs for atrial natriuretic factor and beta-MyHC. We conclude that mouse myocytes have a unique autonomous hypertrophy. On this background, T3, NE, and leukemia inhibitory factor activate hypertrophy with different mRNA phenotypes, but certain Gq- and protein kinase C-coupled agonists do not.
Collapse
Affiliation(s)
- X F Deng
- VA Medical Center and the Cardiovascular Research Institute and Department of Medicine, University of California, San Francisco, California, USA
| | | | | |
Collapse
|
47
|
Colomer JM, Means AR. Chronic elevation of calmodulin in the ventricles of transgenic mice increases the autonomous activity of calmodulin-dependent protein kinase II, which regulates atrial natriuretic factor gene expression. Mol Endocrinol 2000; 14:1125-36. [PMID: 10935538 DOI: 10.1210/mend.14.8.0496] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Although isoforms of Ca2+/calmodulin-dependent protein kinase II (CaMKII) have been implicated in the regulation of gene expression in cultured cells, this issue has yet to be addressed in vivo. We report that the overexpression of calmodulin in ventricular myocytes of transgenic mice results in an increase in the Ca2+/calmodulin-independent activity of endogenous CaMKII. The calmodulin transgene is regulated by a 500-bp fragment of the atrial natriuretic factor (ANF) gene promoter which, based on cell transfection studies, is itself known to be regulated by CaMKII. The increased autonomous activity of CaMKII maintains the activity of the transgene and establishes a positive feed-forward loop, which also extends the temporal expression of the endogenous ANF promoter in ventricular myocytes. Both the increased activity of CaMKII and transcriptional activation of ANF are highly selective responses to the chronic overexpression of calmodulin. These results indicate that CaMKII can regulate gene expression in vivo and suggest that this enzyme may represent the Ca2+-dependent target responsible for reactivation of the ANF gene during ventricular hypertrophy.
Collapse
Affiliation(s)
- J M Colomer
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
48
|
Oh J, So HS, Park R, Ryu DK, Moon BS, Park OK, Chung YT. The water extract of Jagamchotang protects the ischemia/reperfusion-induced cytotoxicity of rat neonatal myocardial cells via generation of nitric oxide. Immunopharmacol Immunotoxicol 2000; 22:297-315. [PMID: 10952033 DOI: 10.3109/08923970009016422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Jagamchotang has been used for treatment of ischemic myocardial diseases in Chinese traditional medicine. However, little is known about the mechanism by which Jagamchotang rescues myocardial cells from ischemic damages. To elucidate the protective mechanisms, the effects of Jagamchotang on ischemia/reperfusion-induced cytotoxicity and generation of nitric oxide (NO) are investigated in primary neonatal myocardial cells. Ischemia/reperfusion itself induces severe myocardial cell death in vitro. However, treatment of the cells with Jagamchotang significantly reduces both ischemia/reperfusion-induced myocardial cell death and LDH release. In addition, pretreatment of Jagamchotang before reperfusion recovers the lose of beating rates after ischemia/reperfusion. For a while, the water extract of Jagamchotang stimulates myocardial cells in ischemic condition to produce nitric oxide (NO) in a dose dependent manner and it protects the damage of myocardial cells. Furthermore, the protective effects of the water extract of Jagamchotang is mimicked by treatment of sodium nitroprusside, an exogenous NO donor. NG-monomethyi-L-argine (NGMMA), a specific inhibitor of nitric oxide synthase (NOS), significantly blocks the protective effects of Jagamchotang on the cells after ischemia/reperfusion. Taken together, we suggest that the protective effects of Jagamchotang against ischemia/reperfusion-induced myocardial damages may be mediated by NO production during ischemic condition.
Collapse
Affiliation(s)
- J Oh
- Department of Anatomy, Wonkwang University School of Medicine, Iksan, Chonbuk, South Korea
| | | | | | | | | | | | | |
Collapse
|
49
|
Varma DR, Deng XF. Cardiovascular α1-adrenoceptor subtypes: functions and signaling. Can J Physiol Pharmacol 2000. [DOI: 10.1139/y99-142] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
α1-Adrenoceptors (α1AR) are G protein-coupled receptors and include α1A, α1B, and α1D subtypes corresponding to cloned α1a, α1b, and α1d, respectively. α1AR mediate several cardiovascular actions of sympathomimetic amines such as vasoconstriction and cardiac inotropy, hypertrophy, metabolism, and remodeling. α1AR subtypes are products of separate genes and differ in structure, G protein-coupling, tissue distribution, signaling, regulation, and functions. Both α1AAR and α1BAR mediate positive inotropic responses. On the other hand, cardiac hypertrophy is primarily mediated by α1AAR. The only demonstrated major function of α1DAR is vasoconstriction. α1AR are coupled to phospholipase C, phospholipase D, and phospholipase A2; they increase intracellular Ca2+ and myofibrillar sensitivity to Ca2+ and cause translocation of specific phosphokinase C isoforms to the particulate fraction. Cardiac hypertrophic responses to α1AR agonists might involve activation of phosphokinase C and mitogen-activated protein kinase via Gq. α1AR subtypes might interact with each other and with other receptors and signaling mechanisms.Key words: cardiac hypertrophy, inotropic responses, central α1-adrenoreceptors, arrythmias.
Collapse
|
50
|
Hernandez OM, Discher DJ, Bishopric NH, Webster KA. Rapid activation of neutral sphingomyelinase by hypoxia-reoxygenation of cardiac myocytes. Circ Res 2000; 86:198-204. [PMID: 10666416 DOI: 10.1161/01.res.86.2.198] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Elevated levels of oxygen free radicals have been implicated in the pathways of reperfusion injury to myocardial tissue. The targets for free radicals may include specific as well as random intracellular components, and part of the cellular response is the induction of extracellularly activated and stress-activated kinases. The intermediate signals that initiate these stress responses are not known. Here we show that one of the earliest responses of cardiac myocytes to hypoxia and reoxygenation is the activation of neutral sphingomyelinase and accumulation of ceramide. Ceramide increased abruptly after reoxygenation, peaking at 10 minutes with 225+/-40% of the control level. Neutral sphingomyelinase activity was induced with similar kinetics, and both activities remained elevated for several hours. c-Jun N-terminal kinase (JNK) was also activated within the same time frame. Treatment of cardiac myocytes with extracellular ceramides also activated JNK. Pretreating cells with antioxidants quenched sphingomyelinase activation, ceramide accumulation, and JNK activation. Ceramide did not accumulate in reoxygenated nonmuscle fibroblasts, and JNK was not activated by reoxygenation in these cells. The results identify neutral sphingomyelinase activation as one of the earliest responses of cardiac myocytes to the redox stress imposed by hypoxia-reoxygenation. The results are consistent with a pathway of ceramide-mediated activation of JNK.
Collapse
Affiliation(s)
- O M Hernandez
- Department of Molecular and Cellular Pharmacology, University of Miami Medical Center, FL 33136, USA
| | | | | | | |
Collapse
|