1
|
Bode C, Preissl S, Hein L, Lother A. Catecholamine treatment induces reversible heart injury and cardiomyocyte gene expression. Intensive Care Med Exp 2024; 12:48. [PMID: 38733526 PMCID: PMC11088585 DOI: 10.1186/s40635-024-00632-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/07/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Catecholamines are commonly used as therapeutic drugs in intensive care medicine to maintain sufficient organ perfusion during shock. However, excessive or sustained adrenergic activation drives detrimental cardiac remodeling and may lead to heart failure. Whether catecholamine treatment in absence of heart failure causes persistent cardiac injury, is uncertain. In this experimental study, we assessed the course of cardiac remodeling and recovery during and after prolonged catecholamine treatment and investigated the molecular mechanisms involved. RESULTS C57BL/6N wild-type mice were assigned to 14 days catecholamine treatment with isoprenaline and phenylephrine (IsoPE), treatment with IsoPE and subsequent recovery, or healthy control groups. IsoPE improved left ventricular contractility but caused substantial cardiac fibrosis and hypertrophy. However, after discontinuation of catecholamine treatment, these alterations were largely reversible. To uncover the molecular mechanisms involved, we performed RNA sequencing from isolated cardiomyocyte nuclei. IsoPE treatment resulted in a transient upregulation of genes related to extracellular matrix formation and transforming growth factor signaling. While components of adrenergic receptor signaling were downregulated during catecholamine treatment, we observed an upregulation of endothelin-1 and its receptors in cardiomyocytes, indicating crosstalk between both signaling pathways. To follow this finding, we treated mice with endothelin-1. Compared to IsoPE, treatment with endothelin-1 induced minor but longer lasting changes in cardiomyocyte gene expression. DNA methylation-guided analysis of enhancer regions identified immediate early transcription factors such as AP-1 family members Jun and Fos as key drivers of pathological gene expression following catecholamine treatment. CONCLUSIONS The results from this study show that prolonged catecholamine exposure induces adverse cardiac remodeling and gene expression before the onset of left ventricular dysfunction which has implications for clinical practice. The observed changes depend on the type of stimulus and are largely reversible after discontinuation of catecholamine treatment. Crosstalk with endothelin signaling and the downstream transcription factors identified in this study provide new opportunities for more targeted therapeutic approaches that may help to separate desired from undesired effects of catecholamine treatment.
Collapse
Affiliation(s)
- Christine Bode
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sebastian Preissl
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Interdisciplinary Medical Intensive Care, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
2
|
Díaz-Rosas G, Cruz-Hernández M, Ortega-Camarillo C, Pedraza-Galeana A, López-Torres A, Contreras-Ramos A. The sodium borate relieves the hypertrophic damage induced during pregnancy, it improves contractibility, reduces oxidative stress and stimulates cell proliferation. J Trace Elem Med Biol 2023; 80:127269. [PMID: 37506468 DOI: 10.1016/j.jtemb.2023.127269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 07/13/2023] [Accepted: 07/23/2023] [Indexed: 07/30/2023]
Abstract
INTRODUCTION Fetal and postnatal hypertrophy develop in response to such different exposures or illnesses the mother suffers during gestation as anti-infectious and physical agents, obesity, hypertension, diabetes, and even advanced maternal age. This gives rise to high comorbidities in the newborn; therefore, looking for alternatives that contribute to cardiac homeostasis is quite necessary to inhibit the overgrowth of myocytes. Boron-derivative compounds could play a key role in exerting a repairing effect on chronic cardiac damage induced during gestation. METHODOLOGY The cardiotoxic effect of 6.4, 12 and 100 mg/kg of sodium tetraborate administered by oral delivery route to healthy pregnant mice was assessed. After that, the use of the chemical compound was tested in the treatment of pregnant mice previously subjected to isoproterenol (fetal hypertrophy model) on the fifth day post coitus. Prior to the sacrifice of the pups of mice an electrocardiography (ECG) was done. Morphological and histological changes of heart were assessed in newborn pups. As a damage marker, the concentration of p38 nitrogen-activated protein kinases were evaluated by using Western Blot and the levels of malondialdehyde (MDA) as well as glutathione antioxidants (GSH) and glutathione peroxidase (GPx) were tested by spectrometry. Moreover, the mRNA expression for early response genes (c-jun, c-fos y c-myc), late response (GATA-4, Mef2c, NFAT) and heart damage (ANP and BNP) was measured by qPCR real time. RESULTS The supply of 6,4 and 12 mg/kg-sodium tetraborate favored ventricular remodeling with histological alterations. By comparison, 100 mg/kg of sodium tetraborate administered during the fetal stage did not alter neither the cardiac morphology of six-week old pups nor the p38/P-p38MAPK ratio remained the same and no oxidative stress was observed. When pregnant females treated with isoproterenol were treated with 100 mg/kg sodium tetraborate during the fetal stage, an improvement in contractility was detected in the pups with an actual reduction in myocardial fibrosis and oxidative stress, but cardiac mass increased. In addition, the expression levels of c-jun, c-myc, GATA-4, MEF2c and ANP mRNA declined in comparison with CTR. However, the hypertrophic damage mechanism was sustained by c-fos, NFAT and BNP expressions. CONCLUSIONS The set of results achieved suggests that high concentrations of sodium tetraborate have no cardiotoxic effects. Furthermore, sodium tetraborate mitigates hypertrophy induced during pregnancy, thereby improving contractibility, reducing oxidative stress and stimulating cell proliferation. Therefore, sodium tetraborate could be an excellent prophylactic treatment administered by delivery oral route during pregnancy when there is a risk of developing fetal left ventricular hypertrophy (LVH).
Collapse
Affiliation(s)
- Guadalupe Díaz-Rosas
- Laboratory of Molecular Biology in Congenital Malformations Unit. Children's Hospital of Mexico Federico Gomez (HIMFG), Mexico City, Mexico
| | - Mayra Cruz-Hernández
- Laboratory of Molecular Biology in Congenital Malformations Unit. Children's Hospital of Mexico Federico Gomez (HIMFG), Mexico City, Mexico; Medical Research Unit in Biochemistry, Specialties Hospital, National Medical Center SXXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, Del. Cuauhtémoc CP, 06720 CDMX, Mexico
| | - Clara Ortega-Camarillo
- Medical Research Unit in Biochemistry, Specialties Hospital, National Medical Center SXXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, Del. Cuauhtémoc CP, 06720 CDMX, Mexico
| | - Agustín Pedraza-Galeana
- Laboratory of Molecular Biology in Congenital Malformations Unit. Children's Hospital of Mexico Federico Gomez (HIMFG), Mexico City, Mexico
| | - Adolfo López-Torres
- Center for Scientific Research, Institute of Applied Chemistry, University of Papaloapan, Central Circuit No. 200, Col. Parque Industrial, 68301 Tuxtepec, Oaxaca, Mexico
| | - Alejandra Contreras-Ramos
- Laboratory of Molecular Biology in Congenital Malformations Unit. Children's Hospital of Mexico Federico Gomez (HIMFG), Mexico City, Mexico.
| |
Collapse
|
3
|
Fazio A, Evangelisti C, Cappellini A, Mongiorgi S, Koufi FD, Neri I, Marvi MV, Russo M, Ghigo A, Manzoli L, Fiume R, Ratti S. Emerging Roles of Phospholipase C Beta Isozymes as Potential Biomarkers in Cardiac Disorders. Int J Mol Sci 2023; 24:13096. [PMID: 37685903 PMCID: PMC10487445 DOI: 10.3390/ijms241713096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Phospholipase C (PLC) enzymes represent crucial participants in the plasma membrane of mammalian cells, including the cardiac sarcolemmal (SL) membrane of cardiomyocytes. They are responsible for the hydrolysis of phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2) into 1,2-diacylglycerol (DAG) and inositol (1,4,5) trisphosphate (Ins(1,4,5)P3), both essential lipid mediators. These second messengers regulate the intracellular calcium (Ca2+) concentration, which activates signal transduction cascades involved in the regulation of cardiomyocyte activity. Of note, emerging evidence suggests that changes in cardiomyocytes' phospholipid profiles are associated with an increased occurrence of cardiovascular diseases, but the underlying mechanisms are still poorly understood. This review aims to provide a comprehensive overview of the significant impact of PLC on the cardiovascular system, encompassing both physiological and pathological conditions. Specifically, it focuses on the relevance of PLCβ isoforms as potential cardiac biomarkers, due to their implications for pathological disorders, such as cardiac hypertrophy, diabetic cardiomyopathy, and myocardial ischemia/reperfusion injury. Gaining a deeper understanding of the mechanisms underlying PLCβ activation and regulation is crucial for unraveling the complex signaling networks involved in healthy and diseased myocardium. Ultimately, this knowledge holds significant promise for advancing the development of potential therapeutic strategies that can effectively target and address cardiac disorders by focusing on the PLCβ subfamily.
Collapse
Affiliation(s)
- Antonietta Fazio
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Camilla Evangelisti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Alessandra Cappellini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Sara Mongiorgi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Foteini-Dionysia Koufi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Irene Neri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Maria Vittoria Marvi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Michele Russo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Torino, 10126 Torino, Italy; (M.R.); (A.G.)
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Torino, 10126 Torino, Italy; (M.R.); (A.G.)
| | - Lucia Manzoli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Roberta Fiume
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| |
Collapse
|
4
|
Simonides W, Tijsma A, Boelen A, Jongejan R, de Rijke Y, Peeters R, Dentice M, Salvatore D, Muller A. Divergent Thyroid Hormone Levels in Plasma and Left Ventricle of the Heart in Compensated and Decompensated Cardiac Hypertrophy Induced by Chronic Adrenergic Stimulation in Mice. Metabolites 2023; 13:metabo13020308. [PMID: 36837927 PMCID: PMC9960204 DOI: 10.3390/metabo13020308] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/05/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Chronic hemodynamic overload of the heart induces ventricular hypertrophy that may be either compensatory or progress to decompensation and heart failure. The gradual impairment of ventricular function is, at least in part, the result of a reduction of cardiac thyroid-hormone (TH) action. Here, we examined the proposed roles of increased cardiac expression of the TH-inactivating enzyme deiodinase type 3 (D3) and reduced plasma TH levels in diminishing cardiac TH levels. Using minipumps, mice were infused for one and two weeks with isoproterenol (ISO) alone or in combination with phenylephrine (PE). Remodeling of the heart induced by these adrenergic agonists was assessed by echocardiography. Left ventricular (LV) tissue and plasma TH levels (T4 and T3) were determined using liquid chromatography-tandem mass spectrometry. LV D3 activity was determined by conversion of radiolabeled substrate and quantification following HPLC. The results show that ISO induced compensated LV hypertrophy with maintained cardiac output. Plasma levels of T4 and T3 remained normal, but LV hormone levels were reduced by approximately 30% after two weeks, while LV D3 activity was not significantly increased. ISO + PE induced decompensated LV hypertrophy with diminished cardiac output. Plasma levels of T4 and T3 were substantially reduced after one and two weeks, together with a more than 50% reduction of hormone levels in the LV. D3 activity was increased after one week and returned to control levels after two weeks. These data show for the first time that relative to controls, decompensated LV hypertrophy with diminished cardiac output is associated with a greater reduction of cardiac TH levels than compensated hypertrophy with maintained cardiac output. LV D3 activity is unlikely to account for these reductions after two weeks in either condition. Whereas the mechanism of the mild reduction in compensated hypertrophy is unclear, changes in systemic TH homeostasis appear to determine the marked drop in LV TH levels and associated impairment of ventricular function in decompensated hypertrophy.
Collapse
Affiliation(s)
- Warner Simonides
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, 1081 HZ Amsterdam, The Netherlands
- Correspondence: (W.S.); (A.M.)
| | - Alice Tijsma
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, 1081 HZ Amsterdam, The Netherlands
| | - Anita Boelen
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Rutchanna Jongejan
- Department of Clinical Chemistry, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA Rotterdam, The Netherlands
| | - Yolanda de Rijke
- Department of Clinical Chemistry, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA Rotterdam, The Netherlands
| | - Robin Peeters
- Department of Internal Medicine, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA Rotterdam, The Netherlands
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Domenico Salvatore
- Department of Public Health, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Alice Muller
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, 1081 HZ Amsterdam, The Netherlands
- Correspondence: (W.S.); (A.M.)
| |
Collapse
|
5
|
Pustovit KB, Samoilova DV, Abramochkin DV, Filatova TS, Kuzmin VS. α1-adrenergic receptors accompanied by GATA4 expression are related to proarrhythmic conduction and automaticity in rat interatrial septum. J Physiol Biochem 2022; 78:793-805. [PMID: 35802254 DOI: 10.1007/s13105-022-00902-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 05/19/2022] [Indexed: 11/25/2022]
Abstract
The development of interatrial septum (IAS) is a complicated process, which continues during postnatal life. The hypertrophic signals in developing heart are mediated among others by α-adrenergic pathways. These facts suggest the presence of specific electrophysiological features in developing IAS. This study was aimed to investigate the electrical activity in the tissue preparations of IAS from rat heart in normal conditions and under stimulation of adrenoreceptors. Intracellular recording of electrical activity revealed less negative level of resting membrane potential in IAS if compared to myocardium of left atrium. In normal conditions, non-paced IAS preparations were quiescent, but noradrenaline (10-5 M) and phenylephrine (10-5 M) induced spontaneous action potentials, which could be abolished by α1-blocker prazosin (10-5 M), but not β1-blocker atenolol (10-5 M). Optical mapping showed drastic phenylephrine-induced slowing of conduction in adult rat IAS. The α1-dependent ectopic automaticity of IAS myocardium might be explained by immunohistochemical data indicating the presence of transcription factor GATA4 and abundant α1A-adrenoreceptors in myocytes from adult rat IAS. An elevated sensitivity to adrenergic stimulation due to involvement of α1-adrenergic pathways may underlie increased proarrhythmic potential of adult IAS at least in rats.
Collapse
Affiliation(s)
- Ksenia B Pustovit
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia
| | - Daria V Samoilova
- N. N. Blokhin National Medical Research Centre of Oncology, Kashirskoye sh., 24, Moscow, Russia
| | - Denis V Abramochkin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia.
| | - Tatiana S Filatova
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia.,Laboratory of Cardiac Electrophysiology, National Medical Research Center for Cardiology, 3rd Cherepkovskaya, 15a, Moscow, Russia.,Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova str., 1, Moscow, Russia
| | - Vladislav S Kuzmin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia
| |
Collapse
|
6
|
DEC1 represses cardiomyocyte hypertrophy by recruiting PRP19 as an E3 ligase to promote ubiquitination-proteasome-mediated degradation of GATA4. J Mol Cell Cardiol 2022; 169:96-110. [PMID: 35659652 DOI: 10.1016/j.yjmcc.2022.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 04/18/2022] [Accepted: 05/12/2022] [Indexed: 12/14/2022]
Abstract
Although the pro-hypertrophic role of GATA binding protein 4 (GATA4) during cardiac hypertrophy has been well established, the negative regulatory mechanism to counteract its hyperactivation remains elusive. We hypothesized that the hyperactivation of GATA4 could be a result of loss of interaction between GATA4 with specific suppressors. Using high throughput mass spectrometry technology, we carried out a proteomic screen for endogenous suppressor of GATA4, which disassociated with GATA4 during the hypertrophic response in a cultured cardiac myoblast cell line (H9C2 cells). We identified differentiated embryo chondrocyte 1 (DEC1) negatively regulated the function of GATA4 through physical interaction and negatively regulated cardiac hypertrophy both in vivo and in vitro. Particularly, DEC1 promoted the ubiquitination and proteasome-mediated degradation of GATA4, but did not function as an E3 ligase. Again, using mass spectrometry technology, we systematically identified pre-mRNA processing factor 19 (PRP19) as a newfound E3 ligase, which promoted the K6-linked ubiquitination of GATA4 at its lysine 256. Functional experiments performed in cultured neonatal rat ventricular myocytes and H9C2 cells demonstrated that both DEC1 and PRP19 negatively regulated agonist-induced cardiomyocyte hypertrophic responses. Furthermore, rescue experiments performed in these cells revealed that DEC1 and PRP19 suppressed cardiomyocyte hypertrophy by inhibiting the function of GATA4. Our study thus defined the novel DEC1-PRP19-GATA4 axis to be a previously unknown mechanism in regulating cardiomyocyte hypertrophy. Although GATA4 is indispensable for normal cardiac function, harnessing DEC1- or PRP19-mediated negative regulation to counteract the hyperactivation of GATA4 might serve as a novel therapeutic strategy for pathological cardiac hypertrophy.
Collapse
|
7
|
Muehleman DL, Crocini C, Swearingen AR, Ozeroff CD, Leinwand LA. Regression from pathological hypertrophy in mice is sexually dimorphic and stimulus-specific. Am J Physiol Heart Circ Physiol 2022; 322:H785-H797. [PMID: 35302880 PMCID: PMC8993523 DOI: 10.1152/ajpheart.00644.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pathological cardiac hypertrophy is associated with increased morbidity and mortality. Understanding the mechanisms whereby pathological cardiac growth can be reversed could have therapeutic value. Here, we show that pathways leading to regression of pathological cardiac hypertrophy are strongly dependent on the hypertrophic trigger and are significantly modified by sex. Two pathological stimuli causing hypertrophy via distinct pathways were administered to male and female mice: Angiotensin II [Ang II] or Isoproterenol [Iso]. Stimuli were removed after 7 days of treatment, and left ventricles (LV) were studied at 1, 4, and 7 days. Ang II-treated Females did not show regression after stimulus removal. Iso-treated males showed rapid LV hypertrophy regression. Somewhat surprisingly, RNAseq analysis at day 1 after removal of triggers revealed only 45 differentially regulated genes in common among all groups, demonstrating distinct responses. Ingenuity Pathway Analysis predicted strong downregulation of the TGFβ1 pathway in all groups except for Ang II-treated females. Consistently, we found significant downregulation of Smad signaling after stimulus removal including in Ang II-treated females. Additionally, the ERK1/2 pathway was significantly reduced in the groups showing regression. Finally, protein degradation pathways were significantly activated only in Iso-treated males 1 day after stimulus removal. Our data indicate that TGFβ1 downregulation may play a role in the regression of pathological cardiac hypertrophy via downregulation of the ERK1/2 pathway and activation of autophagy and proteasome activity in Iso-treated males. This work highlights that the reversal of pathological hypertrophy does not utilize universal signaling pathways and that sex potently modifies this process.
Collapse
Affiliation(s)
- Deanna L Muehleman
- BioFrontiers Institute University of Colorado Boulder; Department of Molecular and Cellular Development, University of Colorado Boulder, Boulder, Colorado, United States
| | - Claudia Crocini
- BioFrontiers Institute University of Colorado Boulder; Department of Molecular and Cellular Development, University of Colorado Boulder; Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Neuromuscular and Cardiovascular Cell Biology; German Center for Cardiovascular Research (DZHK) Partner Site Berlin, Boulder, Colorado
| | - Alison R Swearingen
- Department of Molecular and Cellular Development, University of Colorado Boulder, Boulder, Colorado, United States
| | - Christopher D Ozeroff
- BioFrontiers Institute University of Colorado Boulder; Department of Molecular and Cellular Development, University of Colorado Boulder, Boulder, Colorado, United States
| | - Leslie A Leinwand
- BioFrontiers Institute University of Colorado Boulder; Department of Molecular and Cellular Development, University of Colorado Boulder, Boulder, Colorado, United States
| |
Collapse
|
8
|
Sigamani V, Rajasingh S, Gurusamy N, Panda A, Rajasingh J. In-Silico and In-Vitro Analysis of Human SOS1 Protein Causing Noonan Syndrome - A Novel Approach to Explore the Molecular Pathways. Curr Genomics 2021; 22:526-540. [PMID: 35386434 PMCID: PMC8905634 DOI: 10.2174/1389202922666211130144221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 11/22/2022] Open
Abstract
Aims Perform in-silico analysis of human SOS1 mutations to elucidate their pathogenic role in Noonan syndrome (NS). Background NS is an autosomal dominant genetic disorder caused by single nucleotide mutation in PTPN11, SOS1, RAF1, and KRAS genes. NS is thought to affect approximately 1 in 1000. NS patients suffer different pathogenic effects depending on the mutations they carry. Analysis of the mutations would be a promising predictor in identifying the pathogenic effect of NS. Methods We performed computational analysis of the SOS1 gene to identify the pathogenic nonsynonymous single nucleotide polymorphisms (nsSNPs) th a t cause NS. SOS1 variants were retrieved from the SNP database (dbSNP) and analyzed by in-silico tools I-Mutant, iPTREESTAB, and MutPred to elucidate their structural and functional characteristics. Results We found that 11 nsSNPs of SOS1 that were linked to NS. 3D modeling of the wild-type and the 11 nsSNPs of SOS1 showed that SOS1 interacts with cardiac proteins GATA4, TNNT2, and ACTN2. We also found that GRB2 and HRAS act as intermediate molecules between SOS1 and cardiac proteins. Our in-silico analysis findings were further validated using induced cardiomyocytes (iCMCs) derived from NS patients carrying SOS1 gene variant c.1654A>G (NSiCMCs) and compared to control human skin fibroblast-derived iCMCs (C-iCMCs). Our in vitro data confirmed that the SOS1, GRB2 and HRAS gene expressions as well as the activated ERK protein, were significantly decreased in NS-iCMCs when compared to C-iCMCs. Conclusion This is the first in-silico and in vitro study demonstrating that 11 nsSNPs of SOS1 play deleterious pathogenic roles in causing NS.
Collapse
Affiliation(s)
- Vinoth Sigamani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Sheeja Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Narasimman Gurusamy
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Arunima Panda
- Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai, India
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Medicine-Cardiology, University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Microbiology, Immunology & Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
9
|
Exploring Functional Differences between the Right and Left Ventricles to Better Understand Right Ventricular Dysfunction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9993060. [PMID: 34497685 PMCID: PMC8421158 DOI: 10.1155/2021/9993060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/04/2021] [Indexed: 12/16/2022]
Abstract
The right and left ventricles have traditionally been studied as individual entities. Furthermore, modifications found in diseased left ventricles are assumed to influence on right ventricle alterations, but the connection is poorly understood. In this review, we describe the differences between ventricles under physiological and pathological conditions. Understanding the mechanisms that differentiate both ventricles would facilitate a more effective use of therapeutics and broaden our knowledge of right ventricle (RV) dysfunction. RV failure is the strongest predictor of mortality in pulmonary arterial hypertension, but at present, there are no definitive therapies directly targeting RV failure. We further explore the current state of drugs and molecules that improve RV failure in experimental therapeutics and clinical trials to treat pulmonary arterial hypertension and provide evidence of their potential benefits in heart failure.
Collapse
|
10
|
Ul Haq A, Carotenuto F, De Matteis F, Prosposito P, Francini R, Teodori L, Pasquo A, Di Nardo P. Intrinsically Conductive Polymers for Striated Cardiac Muscle Repair. Int J Mol Sci 2021; 22:8550. [PMID: 34445255 PMCID: PMC8395236 DOI: 10.3390/ijms22168550] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
One of the most important features of striated cardiac muscle is the excitability that turns on the excitation-contraction coupling cycle, resulting in the heart blood pumping function. The function of the heart pump may be impaired by events such as myocardial infarction, the consequence of coronary artery thrombosis due to blood clots or plaques. This results in the death of billions of cardiomyocytes, the formation of scar tissue, and consequently impaired contractility. A whole heart transplant remains the gold standard so far and the current pharmacological approaches tend to stop further myocardium deterioration, but this is not a long-term solution. Electrically conductive, scaffold-based cardiac tissue engineering provides a promising solution to repair the injured myocardium. The non-conductive component of the scaffold provides a biocompatible microenvironment to the cultured cells while the conductive component improves intercellular coupling as well as electrical signal propagation through the scar tissue when implanted at the infarcted site. The in vivo electrical coupling of the cells leads to a better regeneration of the infarcted myocardium, reducing arrhythmias, QRS/QT intervals, and scar size and promoting cardiac cell maturation. This review presents the emerging applications of intrinsically conductive polymers in cardiac tissue engineering to repair post-ischemic myocardial insult.
Collapse
Affiliation(s)
- Arsalan Ul Haq
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
- CIMER—Centro di Ricerca Interdipartimentale di Medicina Rigenerativa, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (P.P.); (R.F.); (L.T.)
| | - Felicia Carotenuto
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
- CIMER—Centro di Ricerca Interdipartimentale di Medicina Rigenerativa, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (P.P.); (R.F.); (L.T.)
- Department of Fusion and Technologies for Nuclear Safety and Security, Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA, CR Frascati, 00044 Rome, Italy;
| | - Fabio De Matteis
- CIMER—Centro di Ricerca Interdipartimentale di Medicina Rigenerativa, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (P.P.); (R.F.); (L.T.)
- Dipartimento di Ingegneria Industriale, Università degli Studi di Roma “Tor Vergata”, Via del Politecnico, 00133 Roma, Italy
| | - Paolo Prosposito
- CIMER—Centro di Ricerca Interdipartimentale di Medicina Rigenerativa, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (P.P.); (R.F.); (L.T.)
- Dipartimento di Ingegneria Industriale, Università degli Studi di Roma “Tor Vergata”, Via del Politecnico, 00133 Roma, Italy
| | - Roberto Francini
- CIMER—Centro di Ricerca Interdipartimentale di Medicina Rigenerativa, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (P.P.); (R.F.); (L.T.)
- Dipartimento di Ingegneria Industriale, Università degli Studi di Roma “Tor Vergata”, Via del Politecnico, 00133 Roma, Italy
| | - Laura Teodori
- CIMER—Centro di Ricerca Interdipartimentale di Medicina Rigenerativa, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (P.P.); (R.F.); (L.T.)
- Department of Fusion and Technologies for Nuclear Safety and Security, Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA, CR Frascati, 00044 Rome, Italy;
| | - Alessandra Pasquo
- Department of Fusion and Technologies for Nuclear Safety and Security, Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA, CR Frascati, 00044 Rome, Italy;
| | - Paolo Di Nardo
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
- CIMER—Centro di Ricerca Interdipartimentale di Medicina Rigenerativa, Università degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (P.P.); (R.F.); (L.T.)
- L.L. Levshin Institute of Cluster Oncology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| |
Collapse
|
11
|
Ul Haq A, Carotenuto F, Di Nardo P, Francini R, Prosposito P, Pescosolido F, De Matteis F. Extrinsically Conductive Nanomaterials for Cardiac Tissue Engineering Applications. MICROMACHINES 2021; 12:914. [PMID: 34442536 PMCID: PMC8402139 DOI: 10.3390/mi12080914] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 01/09/2023]
Abstract
Myocardial infarction (MI) is the consequence of coronary artery thrombosis resulting in ischemia and necrosis of the myocardium. As a result, billions of contractile cardiomyocytes are lost with poor innate regeneration capability. This degenerated tissue is replaced by collagen-rich fibrotic scar tissue as the usual body response to quickly repair the injury. The non-conductive nature of this tissue results in arrhythmias and asynchronous beating leading to total heart failure in the long run due to ventricular remodelling. Traditional pharmacological and assistive device approaches have failed to meet the utmost need for tissue regeneration to repair MI injuries. Engineered heart tissues (EHTs) seem promising alternatives, but their non-conductive nature could not resolve problems such as arrhythmias and asynchronous beating for long term in-vivo applications. The ability of nanotechnology to mimic the nano-bioarchitecture of the extracellular matrix and the potential of cardiac tissue engineering to engineer heart-like tissues makes it a unique combination to develop conductive constructs. Biomaterials blended with conductive nanomaterials could yield conductive constructs (referred to as extrinsically conductive). These cell-laden conductive constructs can alleviate cardiac functions when implanted in-vivo. A succinct review of the most promising applications of nanomaterials in cardiac tissue engineering to repair MI injuries is presented with a focus on extrinsically conductive nanomaterials.
Collapse
Affiliation(s)
- Arsalan Ul Haq
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (F.C.); (P.D.N.); (F.P.)
- CIMER, Centre for Regenerative Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.F.); (P.P.); (F.D.M.)
| | - Felicia Carotenuto
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (F.C.); (P.D.N.); (F.P.)
- CIMER, Centre for Regenerative Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.F.); (P.P.); (F.D.M.)
| | - Paolo Di Nardo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (F.C.); (P.D.N.); (F.P.)
- CIMER, Centre for Regenerative Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.F.); (P.P.); (F.D.M.)
- L.L. Levshin Institute of Cluster Oncology, I.M. Sechenov First Moscow State Medical University, 119992 Moscow, Russia
| | - Roberto Francini
- CIMER, Centre for Regenerative Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.F.); (P.P.); (F.D.M.)
- Industrial Engineering Department, University of Rome Tor Vergata, Via del Politecnico 1, 00133 Rome, Italy
| | - Paolo Prosposito
- CIMER, Centre for Regenerative Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.F.); (P.P.); (F.D.M.)
- Industrial Engineering Department, University of Rome Tor Vergata, Via del Politecnico 1, 00133 Rome, Italy
| | - Francesca Pescosolido
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (F.C.); (P.D.N.); (F.P.)
- CIMER, Centre for Regenerative Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.F.); (P.P.); (F.D.M.)
| | - Fabio De Matteis
- CIMER, Centre for Regenerative Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.F.); (P.P.); (F.D.M.)
- Industrial Engineering Department, University of Rome Tor Vergata, Via del Politecnico 1, 00133 Rome, Italy
| |
Collapse
|
12
|
Dixit R, Narasimhan C, Balekundri VI, Agrawal D, Kumar A, Mohapatra B. Functional analysis of novel genetic variants of NKX2-5 associated with nonsyndromic congenital heart disease. Am J Med Genet A 2021; 185:3644-3663. [PMID: 34214246 DOI: 10.1002/ajmg.a.62413] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/04/2021] [Accepted: 06/11/2021] [Indexed: 01/26/2023]
Abstract
NKX2-5, a master cardiac regulatory transcription factor was the first known genetic cause of congenital heart diseases (CHDs). To further investigate its role in CHD pathogenesis, we performed mutational screening of 285 CHD probands and 200 healthy controls. Five coding sequence variants were identified in six CHD cases (2.1%), including three in the N-terminal region (p.A61G, p.R95L, and p.E131K) and one each in homeodomain (HD) (p.A148E) and tyrosine-rich domain (p.P247A). Variant-p.A148E showed tertiary structure changes and differential DNA binding affinity of mutant compared to wild type. Two N-terminal variants-p.A61G and p.E131K along with HD variant p.A148E demonstrated significantly reduced transcriptional activity of Nppa and Actc1 promoters in dual luciferase promoter assay supported by their reduced expression in qRT-PCR. Nonetheless, variant p.R95L affected the synergy of NKX2-5 with serum response factor and TBX5 leading to significantly decreased Actc1 promoter activity depicting a distinctive role of this region. The aberrant expression of other target genes-Irx4, Mef2c, Bmp10, Myh6, Myh7, and Myocd is also observed in response to NKX2-5 variants, possibly due to the defective gene regulatory network. Severely impaired downstream promoter activities and abnormal expression of target genes due to N-terminal variants supports the emerging role of this region during cardiac-developmental pathways.
Collapse
Affiliation(s)
- Ritu Dixit
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Chitra Narasimhan
- Department of Pediatric Cardiology, Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bengaluru, Karnataka, India
| | - Vijayalakshmi I Balekundri
- Super Speciality Hospital, Pradhan Mantri Swasthya Suraksha Yojana (PMSSY), Medical College and Research Institute, Bengaluru, Karnataka, India
| | - Damyanti Agrawal
- Department of Cardiothoracic and Vascular Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ashok Kumar
- Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Bhagyalaxmi Mohapatra
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
13
|
Kuzmin VS, Ivanova AD, Potekhina VM, Samoilova DV, Ushenin KS, Shvetsova AA, Petrov AM. The susceptibility of the rat pulmonary and caval vein myocardium to the catecholamine-induced ectopy changes oppositely in postnatal development. J Physiol 2021; 599:2803-2821. [PMID: 33823063 DOI: 10.1113/jp280485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 03/30/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The developmental changes of the caval (SVC) and pulmonary vein (PV) myocardium electrophysiology are traced throughout postnatal ontogenesis. The myocardium in SVC as well as in PV demonstrate age-dependent differences in the ability to maintain resting membrane potential, to manifest automaticity in a form of ectopic action potentials in basal condition and in responses to the adrenergic stimulation. Electrophysiological characteristics of two distinct types of thoracic vein myocardium change in an opposite manner during early postnatal ontogenesis with increased proarrhythmicity of pulmonary and decreased automaticity in caval veins. Predisposition of PV cardiac tissue to proarrhythmycity develops during ontogenesis in time correlation with the establishment of sympathetic innervation of the tissue. The electrophysiological properties of caval vein cardiac tissue shift from a pacemaker-like phenotype to atrial phenotype in accompaniment with sympathetic nerve growth and adrenergic receptor expression changes. ABSTRACT The thoracic vein myocardium is considered as a main source for atrial fibrillation initiation due to its high susceptibility to ectopic activity. The mechanism by which and when pulmonary (PV) and superior vena cava (SVC) became proarrhythmic during postnatal ontogenesis is still unknown. In this study, we traced postnatal changes of electrophysiology in a correlation with the sympathetic innervation and adrenergic receptor distribution to reveal developmental differences in proarrhythmicity occurrence in PV and SVC myocardium. A standard microelectrode technique was used to assess the changes in ability to maintain resting membrane potential (RMP), generate spontaneous action potentials (SAP) and adrenergically induced ectopy in multicellular SVC and PV preparations of rats of different postnatal ages. Immunofluorescence imaging was used to trace postnatal changes in sympathetic innervation, β1- and α1A-adrenergic receptor (AR) distribution. We revealed that the ability to generate SAP and susceptibility to adrenergic stimulation changes during postnatal ontogenesis in an opposite manner in PV and SVC myocardium. While SAP occurrence decreases with age in SVC myocardium, it significantly increases in PV cardiac tissue. PV myocardium starts to demonstrate RMP instability and proarrhythmic activity from the 14th day of postnatal life which correlates with the appearance of the sympathetic innervation of the thoracic veins. In addition, postnatal attenuation of SVC myocardium automaticity occurs concomitantly with sympathetic innervation establishment and increase in β1-ARs, but not α1A-AR levels. Our results support the contention that SVC and PV myocardium electrophysiology change during postnatal development, resulting in higher PV proarrhythmicity in adults.
Collapse
Affiliation(s)
- Vlad S Kuzmin
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskie gory 1, building 12, Moscow, 119991, Russia.,Pirogov Russian National Research Medical University (RNRMU), Ostrovitjanova 1, Moscow, 117997, Russia.,Laboratory of Cardiac Electrophysiology, National Medical Research Cardiological Complex (NMRCC), Institute of Experimental Cardiology, Moscow, Russia
| | - Alexandra D Ivanova
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskie gory 1, building 12, Moscow, 119991, Russia
| | - Viktoria M Potekhina
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskie gory 1, building 12, Moscow, 119991, Russia
| | - Daria V Samoilova
- N. N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | | | - Anastasia A Shvetsova
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskie gory 1, building 12, Moscow, 119991, Russia
| | - Alexey M Petrov
- Institute of Neuroscience, Kazan State Medial University, Butlerova st. 49, Kazan, 420012, Russia.,Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center 'Kazan Scientific Center of RAS', P. O. Box 30, Lobachevsky Str., 2/31, Kazan, 420111, Russia
| |
Collapse
|
14
|
Xie XJ, Li CQ. Chrysophanol Protects Against Acute Heart Failure by Inhibiting JNK1/2 Pathway in Rats. Med Sci Monit 2020; 26:e926392. [PMID: 33044948 PMCID: PMC7566230 DOI: 10.12659/msm.926392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/24/2020] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Acute heart failure (AHF) usually requires urgent therapy. Myocardial damage, oxidative stress, and inflammation are major components in the pathology of AHF. This study was designed to investigate the effects of chrysophanol on AHF. MATERIAL AND METHODS Sprague-Dawley rats were injected with isoprenaline hydrochloride to construct AHF rat models. AHF rats were treated with normal saline (negative control), chrysophanol, the combination of chrysophanol and SP600125, or benazepril (positive control) using sham rats as blank controls. Echocardiography, histological staining, and enzyme activity analysis were performed to assess the heart functions and myocardial damage. Effects on apoptosis, oxidative stress (OS), and inflammation were evaluated by biochemical analysis, TUNEL staining, and ELISA. RESULTS Chrysophanol improved the parameters of cardiac functions and alleviated the myocardial damage accompanied by the reduction of creatine kinase and lactate dehydrogenase activity. Meanwhile, chrysophanol inhibited the myocardial apoptosis along with the upregulation of Bcl-2 and downregulation of Bax and cleaved caspase-3. AHF-induced abnormal changes of OS parameters (MDA, GPx, CAT, SOD) and inflammatory markers (IL-6, IL-1ß, TNF-alpha, IFN-γ) were alleviated by chrysophanol. Benazepril treatment showed similar results with chrysophanol, while the addition of SP600125 enhanced the chrysophanol-mediated protection effects in AHF rats. Western blot analysis demonstrated that chrysophanol inhibited the phosphorylation of JNK1/2 and its upstream/downstream factors. CONCLUSIONS Chrysophanol improved cardiac functions and protected against myocardial damage, apoptosis, OS, and inflammation by inhibiting activation of the JNK1/2 pathway in AHF rat models. These finding indicate that chrysophanol may be a promising approach for treatment of AHF.
Collapse
Affiliation(s)
- Xiao-Jiang Xie
- Department of Cardiology, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, P.R. China
| | - Chang-Qing Li
- Department of Cardiology, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, P.R. China
| |
Collapse
|
15
|
Salamon I, Serio S, Bianco S, Pagiatakis C, Crasto S, Chiariello AM, Conte M, Cattaneo P, Fiorillo L, Felicetta A, di Pasquale E, Kunderfranco P, Nicodemi M, Papait R, Condorelli G. Divergent Transcription of the Nkx2-5 Locus Generates Two Enhancer RNAs with Opposing Functions. iScience 2020; 23:101539. [PMID: 33083767 PMCID: PMC7509214 DOI: 10.1016/j.isci.2020.101539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 07/09/2020] [Accepted: 09/03/2020] [Indexed: 01/12/2023] Open
Abstract
Enhancer RNAs (eRNAs) are a subset of long noncoding RNA generated from genomic enhancers: they are thought to act as potent promoters of the expression of nearby genes through interaction with the transcriptional and epigenomic machineries. In the present work, we describe two eRNAs transcribed from the enhancer of Nkx2-5—a gene specifying a master cardiomyogenic lineage transcription factor (TF)—which we call Intergenic Regulatory Element Nkx2-5 Enhancers (IRENEs). The IRENEs are encoded, respectively, on the same strand (SS) and in the divergent direction (div) respect to the nearby gene. Of note, these two eRNAs have opposing roles in the regulation of Nkx2-5: IRENE-SS acts as a canonical promoter of transcription, whereas IRENE-div represses the activity of the enhancer through recruitment of the histone deacetylase sirtuin 1. Thus, we have identified an autoregulatory loop controlling expression of the master cardiac TF NKX2-5, in which one eRNA represses transcription. Two eRNAs (IRENE-SS, IRENE-div) with opposing functions are found upstream of Nkx2-5 IRENE-SS works as a classical eRNA, acting as a transcriptional activator IRENE-div acts unconventionally, functioning as a transcriptional repressor IRENEs epigenetically control enhancer status and, subsequently, locus architecture
Collapse
Affiliation(s)
- Irene Salamon
- Humanitas Clinical and Research Center-IRCCS, 20189 Rozzano (MI), Italy
| | - Simone Serio
- Humanitas Clinical and Research Center-IRCCS, 20189 Rozzano (MI), Italy.,Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele (MI), Italy
| | - Simona Bianco
- Department of Physics, Federico II University, 80126 Naples, Italy
| | | | - Silvia Crasto
- Humanitas Clinical and Research Center-IRCCS, 20189 Rozzano (MI), Italy.,Institute of Genetics and Biomedical Research (Milan Unit), National Research Council of Italy, 20189 Rozzano (MI), Italy
| | | | - Mattia Conte
- Department of Physics, Federico II University, 80126 Naples, Italy
| | - Paola Cattaneo
- Humanitas Clinical and Research Center-IRCCS, 20189 Rozzano (MI), Italy.,Institute of Genetics and Biomedical Research (Milan Unit), National Research Council of Italy, 20189 Rozzano (MI), Italy
| | - Luca Fiorillo
- Department of Physics, Federico II University, 80126 Naples, Italy
| | - Arianna Felicetta
- Humanitas Clinical and Research Center-IRCCS, 20189 Rozzano (MI), Italy.,Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele (MI), Italy
| | - Elisa di Pasquale
- Humanitas Clinical and Research Center-IRCCS, 20189 Rozzano (MI), Italy.,Institute of Genetics and Biomedical Research (Milan Unit), National Research Council of Italy, 20189 Rozzano (MI), Italy
| | | | - Mario Nicodemi
- Department of Physics, Federico II University, 80126 Naples, Italy.,Berlin Institute of Health, Max Delbrück Center, 13125 Berlin, Germany
| | - Roberto Papait
- Humanitas Clinical and Research Center-IRCCS, 20189 Rozzano (MI), Italy.,Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Gianluigi Condorelli
- Humanitas Clinical and Research Center-IRCCS, 20189 Rozzano (MI), Italy.,Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele (MI), Italy.,Institute of Genetics and Biomedical Research (Milan Unit), National Research Council of Italy, 20189 Rozzano (MI), Italy
| |
Collapse
|
16
|
Herrmann B, Harder L, Oelkrug R, Chen J, Gachkar S, Nock S, Resch J, Korkowski M, Heuer H, Mittag J. Central Hypothyroidism Impairs Heart Rate Stability and Prevents Thyroid Hormone-Induced Cardiac Hypertrophy and Pyrexia. Thyroid 2020; 30:1205-1216. [PMID: 32188347 DOI: 10.1089/thy.2019.0705] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background: Tachycardia, cardiac hypertrophy, and elevated body temperature are major signs of systemic hyperthyroidism, which are considered to reflect the excessive thyroid hormone (TH) action in the respective peripheral tissues. However, recent observations indicate that the central actions of TH also contribute substantially to cardiovascular regulation and thermogenesis. Methods: In this study, we dissect the individual contributions of peripheral TH action versus the central effects in body temperature regulation and cardiovascular functions by taking advantage of mice lacking the TH transporters monocarboxylate transporter 8 (MCT8) and organic anion transporting polypeptide 1C1 (OATP1C1) (M/O double knock-out [dko]), which exhibit elevated serum triiodothyronine (T3) levels while their brain is in a profoundly hypothyroid state. We compared these animals with wild-type (WT) mice that were treated orally with T3 to achieve similarly elevated serum T3 levels, but are centrally hyperthyroid. For the studies, we used radiotelemetry, infrared thermography, gene expression profiling, Western blot analyses, and enzyme linked immunosorbent assays (ELISA) assays. Results: Our analyses revealed mild hyperthermia and cardiac hypertrophy in T3-treated WT mice but not in M/O dko animals, suggesting that central actions of TH are required for these hyperthyroid phenotypes. Although the average heart rate was unaffected in either model, the M/O dko exhibited an altered heart rate frequency distribution with tachycardic bursts in active periods and bradycardic episodes during resting time, demonstrating that the stabilization of heart rate by the autonomic nervous system can be impaired in centrally hypothyroid animals. Conclusions: Our studies unravel distinct phenotypical traits of hyperthyroidism that depend on an intact central nervous system, and provide valuable insight into the cardiovascular pathology of the Allan-Herndon-Dudley syndrome, a condition caused by the lack of MCT8 in humans.
Collapse
Affiliation(s)
- Beate Herrmann
- Department of Molecular Endocrinology, Institute for Endocrinology and Diabetes, Center of Brain Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Lisbeth Harder
- Department of Molecular Endocrinology, Institute for Endocrinology and Diabetes, Center of Brain Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Rebecca Oelkrug
- Department of Molecular Endocrinology, Institute for Endocrinology and Diabetes, Center of Brain Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Jiesi Chen
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - Sogol Gachkar
- Department of Molecular Endocrinology, Institute for Endocrinology and Diabetes, Center of Brain Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Sebastian Nock
- Department of Molecular Endocrinology, Institute for Endocrinology and Diabetes, Center of Brain Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Julia Resch
- Department of Molecular Endocrinology, Institute for Endocrinology and Diabetes, Center of Brain Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Markus Korkowski
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
- Department of Endocrinology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Heike Heuer
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
- Department of Endocrinology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Jens Mittag
- Department of Molecular Endocrinology, Institute for Endocrinology and Diabetes, Center of Brain Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| |
Collapse
|
17
|
Molecular Mechanisms of Adiponectin-Induced Attenuation of Mechanical Stretch-Mediated Vascular Remodeling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6425782. [PMID: 32566092 DOI: 10.1155/2020/6425782] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/12/2020] [Accepted: 04/17/2020] [Indexed: 12/30/2022]
Abstract
Hypertension induces vascular hypertrophy, which changes blood vessels structurally and functionally, leading to reduced tissue perfusion and further hypertension. It is also associated with dysregulated levels of the circulating adipokines leptin and adiponectin (APN). Leptin is an obesity-associated hormone that promotes vascular smooth muscle cell (VSMC) hypertrophy. APN is a cardioprotective hormone that has been shown to attenuate hypertrophic cardiomyopathy. In this study, we investigated the molecular mechanisms of hypertension-induced VSMC remodeling and the involvement of leptin and APN in this process. To mimic hypertension, the rat portal vein (RPV) was mechanically stretched, and the protective effects of APN on mechanical stretch-induced vascular remodeling and the molecular mechanisms involved were examined by using 10 μg/ml APN. Mechanically stretching the RPV significantly decreased APN protein expression after 24 hours and APN mRNA expression in a time-dependent manner in VSMCs. The mRNA expression of the APN receptors AdipoR1, AdipoR2, and T-cadherin significantly increased after 15 hours of stretch. The ratio of APN/leptin expression in VSMCs significantly decreased after 24 hours of mechanical stretch. Stretching the RPV for 3 days increased the weight and [3H]-leucine incorporation significantly, whereas APN significantly reduced hypertrophy in mechanically stretched vessels. Stretching the RPV for 10 minutes significantly decreased phosphorylation of LKB1, AMPK, and eNOS, while APN significantly increased p-LKB1, p-AMPK, and p-eNOS in stretched vessels. Mechanical stretch significantly increased p-ERK1/2 after 10 minutes, whereas APN significantly reduced stretch-induced ERK1/2 phosphorylation. Stretching the RPV also significantly increased ROS generation after 1 hour, whereas APN significantly decreased mechanical stretch-induced ROS production. Exogenous leptin (3.1 nM) markedly increased GATA-4 nuclear translocation in VSMCs, whereas APN significantly attenuated leptin-induced GATA-4 nuclear translocation. Our results decipher molecular mechanisms of APN-induced attenuation of mechanical stretch-mediated vascular hypertrophy, with the promising potential of ultimately translating this protective hormone into the clinic.
Collapse
|
18
|
Abstract
Various strategies have been applied to replace the loss of cardiomyocytes in order to restore reduced cardiac function and prevent the progression of heart disease. Intensive research efforts in the field of cellular reprogramming and cell transplantation may eventually lead to efficient in vivo applications for the treatment of cardiac injuries, representing a novel treatment strategy for regenerative medicine. Modulation of cardiac transcription factor (TF) networks by chemical entities represents another viable option for therapeutic interventions. Comprehensive screening projects have revealed a number of molecular entities acting on molecular pathways highly critical for cellular lineage commitment and differentiation, including compounds targeting Wnt- and transforming growth factor beta (TGFβ)-signaling. Furthermore, previous studies have demonstrated that GATA4 and NKX2-5 are essential TFs in gene regulation of cardiac development and hypertrophy. For example, both of these TFs are required to fully activate mechanical stretch-responsive genes such as atrial natriuretic peptide and brain natriuretic peptide (BNP). We have previously reported that the compound 3i-1000 efficiently inhibited the synergy of the GATA4-NKX2-5 interaction. Cellular effects of 3i-1000 have been further characterized in a number of confirmatory in vitro bioassays, including rat cardiac myocytes and animal models of ischemic injury and angiotensin II-induced pressure overload, suggesting the potential for small molecule-induced cardioprotection.
Collapse
Affiliation(s)
- Mika J. Välimäki
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of PharmacyUniversity of HelsinkiHelsinki, Finland
| | - Heikki J. Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of PharmacyUniversity of HelsinkiHelsinki, Finland
| |
Collapse
|
19
|
Genetic evolution and codon usage analysis of NKX-2.5 gene governing heart development in some mammals. Genomics 2019; 112:1319-1329. [PMID: 31377427 DOI: 10.1016/j.ygeno.2019.07.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 11/21/2022]
Abstract
NKX-2.5 gene is responsible for cardiac development and its targeted disruption apprehends cardiac development at the linear heart tube stage. Bioinformatic analysis was employed to investigate the codon usage pattern and dN/dS of mammalian NKX-2.5 gene. The relative synonymous codon usage analysis revealed variation in codon usage and two synonymous codons namely ATA (Ile) and GTA (Val) were absent in NKX-2.5 gene across selected mammalian species suggesting that these two codons were possibly selected against during evolution. Parity rule 2 analysis of two and four fold amino acids showed CT bias whereas six-fold amino acids revealed GA bias. Neutrality analysis suggests that selection played a prominent role while mutation had a minor role. The dN/dS analysis suggests synonymous substitution played a significant role and it negatively correlated with p-distance of the gene. Purifying natural selection played a dominant role in the genetic evolution of NKX-2.5 gene in mammals.
Collapse
|
20
|
Li MCH, O'Brien TJ, Todaro M, Powell KL. Acquired cardiac channelopathies in epilepsy: Evidence, mechanisms, and clinical significance. Epilepsia 2019; 60:1753-1767. [PMID: 31353444 DOI: 10.1111/epi.16301] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 07/07/2019] [Accepted: 07/07/2019] [Indexed: 12/13/2022]
Abstract
There is growing evidence that cardiac dysfunction in patients with chronic epilepsy could play a pathogenic role in sudden unexpected death in epilepsy (SUDEP). Recent animal studies have revealed that epilepsy secondarily alters the expression of cardiac ion channels alongside abnormal cardiac electrophysiology and remodeling. These molecular findings represent novel evidence for an acquired cardiac channelopathy in epilepsy, distinct from inherited ion channels mutations associated with cardiocerebral phenotypes. Specifically, seizure activity has been shown to alter the messenger RNA (mRNA) and protein expression of voltage-gated sodium channels (Nav 1.1, Nav 1.5), voltage-gated potassium channels (Kv 4.2, Kv 4.3), sodium-calcium exchangers (NCX1), and nonspecific cation-conducting channels (HCN2, HCN4). The pathophysiology may involve autonomic dysfunction and structural cardiac disease, as both are independently associated with epilepsy and ion channel dysregulation. Indeed, in vivo and in vitro studies of cardiac pathology reveal a complex network of signaling pathways and transcription factors regulating ion channel expression in the setting of sympathetic overactivity, cardiac failure, and hypertrophy. Other mechanisms such as circulating inflammatory mediators or exogenous effects of antiepileptic medications lack evidence. Moreover, an acquired cardiac channelopathy may underlie the electrophysiologic cardiac abnormalities seen in chronic epilepsy, potentially contributing to the increased risk of malignant arrhythmias and sudden death. Therefore, further investigation is necessary to establish whether cardiac ion channel dysregulation similarly occurs in patients with epilepsy, and to characterize any pathogenic relationship with SUDEP.
Collapse
Affiliation(s)
- Michael C H Li
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Marian Todaro
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia.,Department of Neurology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Kim L Powell
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Loonat AA, Curtis MK, Richards MA, Nunez-Alonso G, Michl J, Swietach P. A high-throughput ratiometric method for imaging hypertrophic growth in cultured primary cardiac myocytes. J Mol Cell Cardiol 2019; 130:184-196. [PMID: 30986378 PMCID: PMC6520438 DOI: 10.1016/j.yjmcc.2019.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/14/2019] [Accepted: 04/04/2019] [Indexed: 12/21/2022]
Abstract
Maladaptive hypertrophy of cardiac myocytes increases the risk of heart failure. The underlying signaling can be triggered and interrogated in cultured neonatal ventricular myocytes (NRVMs) using sophisticated pharmacological and genetic techniques. However, the methods for quantifying cell growth are, by comparison, inadequate. The lack of quantitative, calibratable and computationally-inexpensive high-throughput technology has limited the scope for using cultured myocytes in large-scale analyses. We present a ratiometric method for quantifying the hypertrophic growth of cultured myocytes, compatible with high-throughput imaging platforms. Protein biomass was assayed from sulforhodamine B (SRB) fluorescence, and image analysis calculated the quotient of signal from extra-nuclear and nuclear regions. The former readout relates to hypertrophic growth, whereas the latter is a reference for correcting protein-independent (e.g. equipment-related) variables. This ratiometric measure, when normalized to the number of cells, provides a robust quantification of cellular hypertrophy. The method was tested by comparing the efficacy of various chemical agonists to evoke hypertrophy, and verified using independent assays (myocyte area, transcripts of markers). The method's high resolving power and wide dynamic range were confirmed by the ability to generate concentration-response curves, track the time-course of hypertrophic responses with fine temporal resolution, describe drug/agonist interactions, and screen for novel anti-hypertrophic agents. The method can be implemented as an end-point in protocols investigating hypertrophy, and is compatible with automated plate-reader platforms for generating high-throughput data, thereby reducing investigator-bias. Finally, the computationally-minimal workflow required for obtaining measurements makes the method simple to implement in most laboratories. Maladaptive hypertrophy of myocytes can lead to heart failure. Common methods for tracking growth in cultured myocytes are inadequate. We design and test a method for tracking myocyte hypertrophy in vitro. The method provides a ratiometric index of growth for high throughput analyses. Using the method, we characterize further details of (anti)hypertrophic responses.
Collapse
Affiliation(s)
- Aminah A Loonat
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom
| | - M Kate Curtis
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Mark A Richards
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Graciela Nunez-Alonso
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Johanna Michl
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Pawel Swietach
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom.
| |
Collapse
|
22
|
Sun S, Li T, Jin L, Piao ZH, Liu B, Ryu Y, Choi SY, Kim GR, Jeong JE, Wi AJ, Lee SJ, Kee HJ, Jeong MH. Dendropanax morbifera Prevents Cardiomyocyte Hypertrophy by Inhibiting the Sp1/GATA4 Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:1021-1044. [PMID: 29986596 DOI: 10.1142/s0192415x18500532] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
An extract of Dendropanax morbifera branch exerts antioxidant, anti-inflammatory, antithrombotic, and anticancer activities. The purpose of this study was to investigate the effect of the extract in isoproterenol-induced cardiac hypertrophy. Phalloidin staining showed that treatment with the extract dramatically prevents isoproterenol-induced H9c2 cell enlargement and the expression of cardiac hypertrophic marker genes, including atrial natriuretic peptide (ANP) and B-type brain natriuretic peptide (BNP). Further, pretreatment with the extract decreased isoproterenol-induced GATA4 and Sp1 expression in H9c2 cells. Overexpression of Sp1 induced the expression of GATA4. The forced expression of Sp1 or its downstream target GATA4, as well as the co-transfection of Sp1 and GATA4 increased the expression of ANP, which was decreased by treatment with the extract. To further elucidate the regulation of the Sp1/GATA4-mediated expression of ANP, knockdown experiments were performed. Transfection with small interfering RNAs (siRNAs) for Sp1 or GATA4 decreased ANP expression. The extract did not further inhibit the expression of ANP reduced by the transfection of GATA4 siRNA. Sp1 knockdown did not affect the expression of ANP that was induced by the overexpression of GATA4; however, GATA4 knockdown abolished the expression of ANP that had been induced by Sp1 overexpression. The extract treatment also attenuated the isoproterenol-induced activation of p38 MAPK, ERK1/2, and JNK1. Hesperidin, catechin, 2,5-dihydroxybenzoic acid, and salicylic acid are the main phenolic compounds present in the extract as observed by high performance liquid chromatography. Hesperidin and 2,5-dihydroxybenzoic acid attenuated isoproterenol-induced cardiac hypertrophy. These findings suggest that the D. morbifera branch extract prevents cardiac hypertrophy by downregulating the activation of Sp1/GATA4 and MAPK signaling pathways.
Collapse
Affiliation(s)
- Simei Sun
- Heart Research Center of Chonnam National University Hospital, Gwangju 61469, Republic of Korea
- Zhengjiang Rongjun Hospital, 352 Zhongshan Road, Jianxing City, Zhejiang Province 314000, P. R. China
- Molecular Medicine, BK21 Plus, Chonnam National University Graduate School, Gwangju 61469, Republic of Korea
| | - Tianyi Li
- The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Li Jin
- Heart Research Center of Chonnam National University Hospital, Gwangju 61469, Republic of Korea
- The Second Affiliated Hospital and Yuying Children’s Hospital, Jilin 132011, P. R. China
| | - Zhe Hao Piao
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Bin Liu
- The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Yuhee Ryu
- Heart Research Center of Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Sin Young Choi
- Heart Research Center of Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Gwi Ran Kim
- Heart Research Center of Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Ji Eun Jeong
- Jeonnam Forest Resources Research Institute, Naju 58213, Republic of Korea
| | - An Jin Wi
- Jeonnam Forest Resources Research Institute, Naju 58213, Republic of Korea
| | - Song Ju Lee
- Department of Food & Nutrition, Gwangju Health University, Gwangju 62287, Republic of Korea
| | - Hae Jin Kee
- Heart Research Center of Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Myung Ho Jeong
- Heart Research Center of Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| |
Collapse
|
23
|
Roles of PDE1 in Pathological Cardiac Remodeling and Dysfunction. J Cardiovasc Dev Dis 2018; 5:jcdd5020022. [PMID: 29690591 PMCID: PMC6023290 DOI: 10.3390/jcdd5020022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/05/2018] [Accepted: 04/20/2018] [Indexed: 12/16/2022] Open
Abstract
Pathological cardiac hypertrophy and dysfunction is a response to various stress stimuli and can result in reduced cardiac output and heart failure. Cyclic nucleotide signaling regulates several cardiac functions including contractility, remodeling, and fibrosis. Cyclic nucleotide phosphodiesterases (PDEs), by catalyzing the hydrolysis of cyclic nucleotides, are critical in the homeostasis of intracellular cyclic nucleotide signaling and hold great therapeutic potential as drug targets. Recent studies have revealed that the inhibition of the PDE family member PDE1 plays a protective role in pathological cardiac remodeling and dysfunction by the modulation of distinct cyclic nucleotide signaling pathways. This review summarizes recent key findings regarding the roles of PDE1 in the cardiac system that can lead to a better understanding of its therapeutic potential.
Collapse
|
24
|
Li C, Huang D, Tang J, Chen M, Lu Q, Li H, Zhang M, Xu B, Mao J. ClC-3 chloride channel is involved in isoprenaline-induced cardiac hypertrophy. Gene 2017; 642:335-342. [PMID: 29158167 DOI: 10.1016/j.gene.2017.11.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/26/2017] [Accepted: 11/15/2017] [Indexed: 01/01/2023]
Abstract
Isoprenaline, an activator of β-adrenergic receptor, has been found to induce cardiac hypertrophy in vivo and in vitro, but the exact mechanism is still unclear. ClC-3 is a member of the chloride channel family and is highly expressed in mammalian myocardium. In the present study, the role of ClC-3 in isopronaline-induced cardiac hypertrophy was investigated. We found that ClC-3 expression was reduced in isoprenaline-induced hypertrophic H9c2 cells, primary rat neonatal cardiomyocytes and myocardium of C57/BL/6 mice, and this reduction was prevented by the pretreatment of propranolol. Adeno-associated virus 9 (AAV9)-mediated ClC-3 expression in myocardium decreased heart mass index, thinned interventricular septum and left ventricular wall and lowered the mRNA expression of natriuretic peptide type A (ANF) and β-myosin heavy chain (β-MHC). Our results showed that ClC-3 played an important role in β-adrenergic cardiac hypertrophy which could be associated with ANF and β-MHC, and all these findings suggested that ClC-3 may be a novel therapeutic target for the prevention or treatment of myocardiac hypertrophy.
Collapse
Affiliation(s)
- Chunmei Li
- Department of Biochemistry and Molecular Biology, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Dan Huang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Hunan Traditional Chinese Medical College, Zhuzhou, 412012, China
| | - Jing Tang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Mengqing Chen
- Department of Biochemistry and Molecular Biology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qun Lu
- Department of Biochemistry and Molecular Biology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - He Li
- Department of Biochemistry and Molecular Biology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | | | - Bin Xu
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates and School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Jianwen Mao
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
25
|
Identification of Novel Non-secosteroidal Vitamin D Receptor Agonists with Potent Cardioprotective Effects and devoid of Hypercalcemia. Sci Rep 2017; 7:8427. [PMID: 28814738 PMCID: PMC5559458 DOI: 10.1038/s41598-017-08670-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 07/12/2017] [Indexed: 12/22/2022] Open
Abstract
Vitamin D regulates many biological processes, but its clinical utility is limited by its hypercalcemic effect. Using a virtual screening platform to search novel chemical probes that activate the vitamin D signaling, we report discovery of novel non-steroidal small-molecule compounds that activate the vitamin D receptor (VDR), but are devoid of hypercalcemia. A lead compound (known as VDR 4-1) demonstrated potent transcriptional activities in a VDR reporter gene assay, and significantly ameliorated cardiac hypertrophy in cell culture studies and in animal models. VDR 4-1 also effectively suppressed secondary hyperparathyroidism in 1α-hydroxylase knockout mice. In contrast to 1α,25-dihydroxyvitamin D3 (1,25-D3 or calcitriol), a naturally occurring VDR agonist, VDR 4-1 therapy even at high doses did not induce hypercalcemia. These findings were accompanied by a lack of upregulation of calcium transport genes in kidney and in the gut providing a mechanism for the lack of hypercalcemia. Furthermore, VDR 4-1 therapy significantly suppressed cardiac hypertrophy and progression to heart failure in both vitamin D deficient and normal mice without inducing significant hypercalcemia. In conclusion, we have identified a unique VDR agonist compound with beneficial effects in mouse models of hyperparathyroidism and heart failure without inducing significant hypercalcemia.
Collapse
|
26
|
Xu JH, Gu JY, Guo YH, Zhang H, Qiu XB, Li RG, Shi HY, Liu H, Yang XX, Xu YJ, Qu XK, Yang YQ. Prevalence and Spectrum of NKX2-5 Mutations Associated With Sporadic Adult-Onset Dilated Cardiomyopathy. Int Heart J 2017; 58:521-529. [PMID: 28690296 DOI: 10.1536/ihj.16-440] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Dilated cardiomyopathy (DCM), the most common form of primary myocardial disease, is a leading cause of congestive heart failure and the most common indication for heart transplantation. Recently, NKX2-5 mutations have been involved in the pathogenesis of familial DCM. However, the prevalence and spectrum of NKX2-5 mutations associated with sporadic DCM remain to be evaluated. In this study, the coding regions and flanking introns of the NKX2-5 gene, which encodes a cardiac transcription factor pivotal for cardiac development and structural remodeling, were sequenced in 210 unrelated patients with sporadic adult-onset DCM. A total of 300 unrelated healthy individuals used as controls were also genotyped for NKX2-5. The functional effect of the mutant NKX2-5 was investigated using a dual-luciferase reporter assay system. As a result, two novel heterozygous NKX2-5 mutations, p.R139W and p.E167X, were identified in 2 unrelated patients with sporadic adult-onset DCM, with a mutational prevalence of approximately 0.95%. The mutations were absent in 600 referential chromosomes and the altered amino acids were completely conserved evolutionarily across species. Functional assays revealed that the NKX2-5 mutants were associated with significantly reduced transcriptional activity. Furthermore, the mutations abrogated the synergistic activation between NKX2-5 and GATA4 as well as TBX20, two other cardiac key transcription factors that have been causally linked to adult-onset DCM. This study is the first to associate NKX2-5 loss-of-function mutations with enhanced susceptibility to sporadic DCM, which provides novel insight into the molecular etiology underpinning DCM, and suggests the potential implications for the genetic counseling and personalized treatment of the DCM patients.
Collapse
Affiliation(s)
- Jia-Hong Xu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine
| | - Jian-Yun Gu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine
| | - Yu-Han Guo
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine
| | - Hong Zhang
- Department of Pharmacy, Tongji Hospital, Tongji University School of Medicine
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Hong-Yu Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Hua Liu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Xiao-Xiao Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Xin-Kai Qu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University.,Department of Cardiovascular Research Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University.,Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University
| |
Collapse
|
27
|
Jackman CP, Carlson AL, Bursac N. Dynamic culture yields engineered myocardium with near-adult functional output. Biomaterials 2016; 111:66-79. [PMID: 27723557 DOI: 10.1016/j.biomaterials.2016.09.024] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 09/20/2016] [Accepted: 09/29/2016] [Indexed: 01/02/2023]
Abstract
Engineered cardiac tissues hold promise for cell therapy and drug development, but exhibit inadequate function and maturity. In this study, we sought to significantly improve the function and maturation of rat and human engineered cardiac tissues. We developed dynamic, free-floating culture conditions for engineering "cardiobundles", 3-dimensional cylindrical tissues made from neonatal rat cardiomyocytes or human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) embedded in fibrin-based hydrogel. Compared to static culture, 2-week dynamic culture of neonatal rat cardiobundles significantly increased expression of sarcomeric proteins, cardiomyocyte size (∼2.1-fold), contractile force (∼3.5-fold), and conduction velocity of action potentials (∼1.4-fold). The average contractile force per cross-sectional area (59.7 mN/mm2) and conduction velocity (52.5 cm/s) matched or approached those of adult rat myocardium, respectively. The inferior function of statically cultured cardiobundles was rescued by transfer to dynamic conditions, which was accompanied by an increase in mTORC1 activity and decline in AMPK phosphorylation and was blocked by rapamycin. Furthermore, dynamic culture effects did not stimulate ERK1/2 pathway and were insensitive to blockers of mechanosensitive channels, suggesting increased nutrient availability rather than mechanical stimulation as the upstream activator of mTORC1. Direct comparison with phenylephrine treatment confirmed that dynamic culture promoted physiological cardiomyocyte growth rather than pathological hypertrophy. Optimized dynamic culture conditions also augmented function of human cardiobundles made reproducibly from cardiomyocytes derived from multiple hPSC lines, resulting in significantly increased contraction force (∼2.5-fold) and conduction velocity (∼1.4-fold). The average specific force of 23.2 mN/mm2 and conduction velocity of 25.8 cm/s approached the functional metrics of adult human myocardium. In conclusion, we have developed a versatile methodology for engineering cardiac tissues with a near-adult functional output without the need for exogenous electrical or mechanical stimulation, and have identified mTOR signaling as an important mechanism for advancing tissue maturation and function in vitro.
Collapse
Affiliation(s)
| | - Aaron L Carlson
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, United States.
| |
Collapse
|
28
|
Soudani N, Ghantous CM, Farhat Z, Shebaby WN, Zibara K, Zeidan A. Calcineurin/NFAT Activation-Dependence of Leptin Synthesis and Vascular Growth in Response to Mechanical Stretch. Front Physiol 2016; 7:433. [PMID: 27746739 PMCID: PMC5040753 DOI: 10.3389/fphys.2016.00433] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 09/13/2016] [Indexed: 12/12/2022] Open
Abstract
Background and Aims: Hypertension and obesity are important risk factors of cardiovascular disease. They are both associated with high leptin levels and have been shown to promote vascular hypertrophy, through the RhoA/ROCK and ERK1/2 phosphorylation. Calcineurin/NFAT activation also induces vascular hypertrophy by upregulating various genes. This study aimed to decipher whether a crosstalk exists between the RhoA/ROCK pathway, Ca2+/calcineurin/NFAT pathway, and ERK1/2 phosphorylation in the process of mechanical stretch-induced vascular smooth muscle cell (VSMC) hypertrophy and leptin synthesis. Methods and Results: Rat portal vein (RPV) organ culture was used to investigate the effect of mechanical stretch and exogenous leptin (3.1 nM) on VSMC hypertrophy and leptin synthesis. Results showed that stretching the RPV significantly upregulated leptin secretion, mRNA, and protein expression, which were inhibited by the calcium channel blocker nifedipine (10 μM), the selective calcineurin inhibitor FK506 (1 nM), and the ERK1/2 inhibitor PD98059 (1 μM). The transcription inhibitor actinomycin D (0.1 μM) and the translation inhibitor cycloheximide (1 mM) significantly decreased stretch-induced leptin protein expression. Mechanical stretch or leptin caused an increase in wet weight changes and protein synthesis, considered as hypertrophic markers, while they were inhibited by FK506 (0.1 nM; 1 nM). In addition, stretch or exogenous leptin significantly increased calcineurin activity and MCIP1 expression whereas leptin induced NFAT nuclear translocation in VSMCs. Moreover, in response to stretch or exogenous leptin, the Rho inhibitor C3 exoenzyme (30 ng/mL), the ROCK inhibitor Y-27632 (10 μM), and the actin depolymerization agents Latrunculin B (50 nM) and cytochalasin D (1 μM) reduced calcineurin activation and NFAT nuclear translocation. ERK1/2 phosphorylation was inhibited by FK506 and C3. Conclusions: Mechanical stretch-induced VSMC hypertrophy and leptin synthesis and secretion are mediated by Ca2+/calcineurin/NFAT activation. RhoA/ROCK and ERK1/2 activation are critical for mechanical stretch-induced calcineurin activation.
Collapse
Affiliation(s)
- Nadia Soudani
- Department of Anatomy, Cell Biology and Physiology, American University of Beirut Beirut, Lebanon
| | - Crystal M Ghantous
- Department of Anatomy, Cell Biology and Physiology, American University of Beirut Beirut, Lebanon
| | - Zein Farhat
- Department of Anatomy, Cell Biology and Physiology, American University of Beirut Beirut, Lebanon
| | - Wassim N Shebaby
- Department of Natural Sciences, Lebanese American University Byblos, Lebanon
| | - Kazem Zibara
- Laboratory of Stem Cells, Department of Biology, Faculty of Sciences, Lebanese University Beirut, Lebanon
| | - Asad Zeidan
- Department of Anatomy, Cell Biology and Physiology, American University of Beirut Beirut, Lebanon
| |
Collapse
|
29
|
Tang X, Gan XT, Rajapurohitam V, Huang CX, Xue J, Lui EMK, Karmazyn M. North American ginseng (Panax quinquefolius) suppresses β-adrenergic-dependent signalling, hypertrophy, and cardiac dysfunction. Can J Physiol Pharmacol 2016; 94:1325-1335. [PMID: 27797280 DOI: 10.1139/cjpp-2016-0337] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
There is increasing evidence for a beneficial effect of ginseng on cardiac pathology. Here, we determined whether North American ginseng can modulate the deleterious effects of the β-adrenoceptor agonist isoproterenol on cardiac hypertrophy and function using in vitro and in vivo approaches. Isoproterenol was administered for 2 weeks at either 25 mg/kg per day or 50 mg/kg per day (ISO25 or ISO50) via a subcutaneously implanted osmotic mini-pump to either control rats or those receiving ginseng (0.9 g/L in the drinking water ad libitum). Isoproterenol produced time- and dose-dependent left ventricular dysfunction, although these effects were attenuated by ginseng. Improved cardiac functions were associated with reduced heart masses, as well as prevention in the upregulation of the hypertrophy-related fetal gene expression. Lung masses were similarly attenuated, suggesting reduced pulmonary congestion. In in vitro studies, ginseng (10 μg/mL) completely suppressed the hypertrophic response to 1 μmol/L isoproterenol in terms of myocyte surface area, as well as reduction in the upregulation of fetal gene expression. These effects were associated with attenuation in both protein kinase A and cAMP response element-binding protein phosphorylation. Ginseng attenuates adverse cardiac adrenergic responses and, therefore, may be an effective therapy to reduce hypertrophy and heart failure associated with excessive catecholamine production.
Collapse
Affiliation(s)
- Xilan Tang
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Xiaohong Tracey Gan
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Venkatesh Rajapurohitam
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Cathy Xiaoling Huang
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Jenny Xue
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Edmund M K Lui
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Morris Karmazyn
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
30
|
Udoko AN, Johnson CA, Dykan A, Rachakonda G, Villalta F, Mandape SN, Lima MF, Pratap S, Nde PN. Early Regulation of Profibrotic Genes in Primary Human Cardiac Myocytes by Trypanosoma cruzi. PLoS Negl Trop Dis 2016; 10:e0003747. [PMID: 26771187 PMCID: PMC4714843 DOI: 10.1371/journal.pntd.0003747] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 10/16/2015] [Indexed: 11/18/2022] Open
Abstract
The molecular mechanisms of Trypanosoma cruzi induced cardiac fibrosis remains to be elucidated. Primary human cardiomyoctes (PHCM) exposed to invasive T. cruzi trypomastigotes were used for transcriptome profiling and downstream bioinformatic analysis to determine fibrotic-associated genes regulated early during infection process (0 to 120 minutes). The identification of early molecular host responses to T. cruzi infection can be exploited to delineate important molecular signatures that can be used for the classification of Chagasic patients at risk of developing heart disease. Our results show distinct gene network architecture with multiple gene networks modulated by the parasite with an incline towards progression to a fibrogenic phenotype. Early during infection, T. cruzi significantly upregulated transcription factors including activator protein 1 (AP1) transcription factor network components (including FOSB, FOS and JUNB), early growth response proteins 1 and 3 (EGR1, EGR3), and cytokines/chemokines (IL5, IL6, IL13, CCL11), which have all been implicated in the onset of fibrosis. The changes in our selected genes of interest did not all start at the same time point. The transcriptome microarray data, validated by quantitative Real-Time PCR, was also confirmed by immunoblotting and customized Enzyme Linked Immunosorbent Assays (ELISA) array showing significant increases in the protein expression levels of fibrogenic EGR1, SNAI1 and IL 6. Furthermore, phosphorylated SMAD2/3 which induces a fibrogenic phenotype is also upregulated accompanied by an increased nuclear translocation of JunB. Pathway analysis of the validated genes and phospho-proteins regulated by the parasite provides the very early fibrotic interactome operating when T. cruzi comes in contact with PHCM. The interactome architecture shows that the parasite induces both TGF-β dependent and independent fibrotic pathways, providing an early molecular foundation for Chagasic cardiomyopathy. Examining the very early molecular events of T. cruzi cellular infection may provide disease biomarkers which will aid clinicians in patient assessment and identification of patient subpopulation at risk of developing Chagasic cardiomyopathy.
Collapse
Affiliation(s)
- Aniekanabassi N. Udoko
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Candice A. Johnson
- Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Andrey Dykan
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Girish Rachakonda
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Fernando Villalta
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Sammed N. Mandape
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Maria F. Lima
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee, United States of America
- School of Graduate Studies and Research, Bioinformatics and Molecular Biology Core, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Siddharth Pratap
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee, United States of America
- School of Graduate Studies and Research, Bioinformatics and Molecular Biology Core, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Pius N. Nde
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
31
|
Mehta G, Kumarasamy S, Wu J, Walsh A, Liu L, Williams K, Joe B, de la Serna IL. MITF interacts with the SWI/SNF subunit, BRG1, to promote GATA4 expression in cardiac hypertrophy. J Mol Cell Cardiol 2015; 88:101-10. [PMID: 26388265 PMCID: PMC4640968 DOI: 10.1016/j.yjmcc.2015.09.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 09/03/2015] [Accepted: 09/17/2015] [Indexed: 11/26/2022]
Abstract
The transcriptional regulation of pathological cardiac hypertrophy involves the interplay of transcription factors and chromatin remodeling enzymes. The Microphthalmia-Associated Transcription Factor (MITF) is highly expressed in cardiomyocytes and is required for cardiac hypertrophy. However, the transcriptional mechanisms by which MITF promotes cardiac hypertrophy have not been elucidated. In this study, we tested the hypothesis that MITF promotes cardiac hypertrophy by activating transcription of pro-hypertrophy genes through interactions with the SWI/SNF chromatin remodeling complex. In an in vivo model of cardiac hypertrophy, expression of MITF and the BRG1 subunit of the SWI/SNF complex increased coordinately in response to pressure overload. Expression of MITF and BRG1 also increased in vitro when cardiomyocytes were stimulated with angiotensin II or a β-adrenergic agonist. Both MITF and BRG1 were required to increase cardiomyocyte size and activate expression of hypertrophy markers in response to β-adrenergic stimulation. We detected physical interactions between MITF and BRG1 in cardiomyocytes and found that they cooperate to regulate expression of a pro-hypertrophic transcription factor, GATA4. Our data show that MITF binds to the E box element in the GATA4 promoter and facilitates recruitment of BRG1. This is associated with enhanced expression of the GATA4 gene as evidenced by increased Histone3 lysine4 tri-methylation (H3K4me3) on the GATA4 promoter. Thus, in hypertrophic cardiomyoctes, MITF is a key transcriptional activator of a pro-hypertrophic gene, GATA4, and this regulation is dependent upon the BRG1 component of the SWI/SNF complex.
Collapse
Affiliation(s)
- Gaurav Mehta
- University of Toledo College of Medicine and Life Sciences, Department of Biochemistry and Cancer Biology, 3035 Arlington Ave, Toledo, OH 43614, USA
| | - Sivarajan Kumarasamy
- University of Toledo College of Medicine and Life Sciences, Program in Physiological Genomics, Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, 3035 Arlington Ave, Toledo, OH 43614, USA
| | - Jian Wu
- University of Toledo College of Medicine and Life Sciences, Department of Biochemistry and Cancer Biology, 3035 Arlington Ave, Toledo, OH 43614, USA
| | - Aaron Walsh
- University of Toledo College of Medicine and Life Sciences, Department of Biochemistry and Cancer Biology, 3035 Arlington Ave, Toledo, OH 43614, USA
| | - Lijun Liu
- University of Toledo College of Medicine and Life Sciences, Department of Biochemistry and Cancer Biology, 3035 Arlington Ave, Toledo, OH 43614, USA
| | - Kandace Williams
- University of Toledo College of Medicine and Life Sciences, Department of Biochemistry and Cancer Biology, 3035 Arlington Ave, Toledo, OH 43614, USA
| | - Bina Joe
- University of Toledo College of Medicine and Life Sciences, Program in Physiological Genomics, Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, 3035 Arlington Ave, Toledo, OH 43614, USA
| | - Ivana L de la Serna
- University of Toledo College of Medicine and Life Sciences, Department of Biochemistry and Cancer Biology, 3035 Arlington Ave, Toledo, OH 43614, USA.
| |
Collapse
|
32
|
Ghantous CM, Kobeissy FH, Soudani N, Rahman FA, Al-Hariri M, Itani HA, Sabra R, Zeidan A. Mechanical stretch-induced vascular hypertrophy occurs through modulation of leptin synthesis-mediated ROS formation and GATA-4 nuclear translocation. Front Pharmacol 2015; 6:240. [PMID: 26557089 PMCID: PMC4615939 DOI: 10.3389/fphar.2015.00240] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/05/2015] [Indexed: 12/11/2022] Open
Abstract
Background: Obesity and hypertension are associated with increased leptin production contributing to cardiovascular remodeling. Mechanisms involving mechanical stretch-induced leptin production and the cross talk between signaling pathways leading to vascular remodeling have not been fully elucidated. Methods and Results: Rat portal vein (RPV) organ culture was used to investigate the effect of mechanical stretch on leptin protein expression in vascular smooth muscle cells (VSMCs). Moreover, the involvement of reactive oxygen species (ROS), the RhoA/ROCK pathway, actin cytoskeleton dynamics and the transcriptional factor GATA-4 activation in mechanical stretch-induced vascular remodeling were investigated. Stretching the RPV for 1 or 24 h significantly increased leptin protein level and ROS formation in VSMCs, which was prevented by 1 h pretreatment with the ROCK inhibitor Y-27632 and the actin cytoskeleton depolymerization agent cytochalasin D. Moreover, Western blotting and immunohistochemistry revealed that mechanical stretch or treatment with 3.1 nmol/L leptin for 24 h significantly increased actin polymerization, as reflected by an increase in the F-actin to G-actin ratio. Increases in blood vessels’ wet weight and [3H]-leucine incorporation following a 24 h treatment with conditioned media from cultured stretched RPVs indicated RPV hypertrophy. This effect was prevented by 1 h pretreatment with anti-leptin antibody, indicating leptin’s crucial role in promoting VSMC hypertrophy. As an index of GATA-4 activation, GATA-4 nuclear translocation was assessed by immunohistochemistry method. Pretreating VSMC with leptin for 1 h significantly activated GATA-4 nuclear translocation, which was potently attenuated by the NADPH oxidase inhibitor apocynin, Y-27632, and cytochalasin D. Conclusion: Our results demonstrate that ROS formation, RhoA/ROCK pathway, and GATA-4 activation play a pivotal role in mechanical stretch-induced leptin synthesis leading to VSMC remodeling.
Collapse
Affiliation(s)
- Crystal M Ghantous
- Cardiovascular Physiology Lab, Department of Anatomy, Cell Biology and Physiology, American University of Beirut , Beirut, Lebanon
| | - Firas H Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut , Beirut, Lebanon
| | - Nadia Soudani
- Cardiovascular Physiology Lab, Department of Anatomy, Cell Biology and Physiology, American University of Beirut , Beirut, Lebanon
| | - Farah A Rahman
- Cardiovascular Physiology Lab, Department of Anatomy, Cell Biology and Physiology, American University of Beirut , Beirut, Lebanon
| | - Mustafa Al-Hariri
- Department of Biochemistry and Molecular Genetics, American University of Beirut , Beirut, Lebanon
| | - Hana A Itani
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine , Nashville, TN, USA
| | - Ramzi Sabra
- Department of Pharmacology and Toxicology, American University of Beirut , Beirut, Lebanon
| | - Asad Zeidan
- Cardiovascular Physiology Lab, Department of Anatomy, Cell Biology and Physiology, American University of Beirut , Beirut, Lebanon
| |
Collapse
|
33
|
Del Olmo-Turrubiarte A, Calzada-Torres A, Díaz-Rosas G, Palma-Lara I, Sánchez-Urbina R, Balderrábano-Saucedo NA, González-Márquez H, Garcia-Alonso P, Contreras-Ramos A. Mouse models for the study of postnatal cardiac hypertrophy. IJC HEART & VASCULATURE 2015; 7:131-140. [PMID: 28785661 PMCID: PMC5497247 DOI: 10.1016/j.ijcha.2015.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 01/19/2015] [Accepted: 02/01/2015] [Indexed: 12/16/2022]
Abstract
The main objective of this study was to create a postnatal model for cardiac hypertrophy (CH), in order to explain the mechanisms that are present in childhood cardiac hypertrophy. Five days after implantation, intraperitoneal (IP) isoproterenol (ISO) was injected for 7 days to pregnant female mice. The fetuses were obtained at 15, 17 and 19 dpc from both groups, also newborns (NB), neonates (7-15 days) and young adults (6 weeks of age). Histopathological exams were done on the hearts. Immunohistochemistry and western blot demonstrated GATA4 and PCNA protein expression, qPCR real time the mRNA of adrenergic receptors (α-AR and β-AR), alpha and beta myosins (α-MHC, β-MHC) and GATA4. After the administration of ISO, there was no change in the number of offsprings. We observed significant structural changes in the size of the offspring hearts. Morphometric analysis revealed an increase in the size of the left ventricular wall and interventricular septum (IVS). Histopathological analysis demonstrated loss of cellular compaction and presence of left ventricular small fibrous foci after birth. Adrenergic receptors might be responsible for changing a physiological into a pathological hypertrophy. However GATA4 seemed to be the determining factor in the pathology. A new animal model was established for the study of pathologic CH in early postnatal stages.
Collapse
Affiliation(s)
- A Del Olmo-Turrubiarte
- Laboratorio de Investigación de Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez (HIMFG), Mexico.,Posgrado en Biología Experimental, Universidad Autónoma Metropolitana, Mexico
| | - A Calzada-Torres
- Laboratorio de Investigación de Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez (HIMFG), Mexico
| | - G Díaz-Rosas
- Laboratorio de Investigación de Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez (HIMFG), Mexico
| | | | - R Sánchez-Urbina
- Laboratorio de Investigación de Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez (HIMFG), Mexico
| | | | - H González-Márquez
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Mexico
| | | | - A Contreras-Ramos
- Laboratorio de Investigación de Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez (HIMFG), Mexico
| |
Collapse
|
34
|
Cardiac transcription factor Nkx2.5 interacts with p53 and modulates its activity. Arch Biochem Biophys 2015; 569:45-53. [DOI: 10.1016/j.abb.2015.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/01/2015] [Indexed: 01/30/2023]
|
35
|
Yuan F, Qiu XB, Li RG, Qu XK, Wang J, Xu YJ, Liu X, Fang WY, Yang YQ, Liao DN. A novel NKX2-5 loss-of-function mutation predisposes to familial dilated cardiomyopathy and arrhythmias. Int J Mol Med 2014; 35:478-86. [PMID: 25503402 DOI: 10.3892/ijmm.2014.2029] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 12/05/2014] [Indexed: 01/08/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is the most prevalent type of primary myocardial disease, which is the third most common cause of heart failure and the most frequent reason for heart transplantation. Aggregating evidence demonstrates that genetic risk factors are involved in the pathogenesis of idiopathic DCM. Nevertheless, DCM is of remarkable genetic heterogeneity and the genetic defects underpinning DCM in an overwhelming majority of patients remain unknown. In the present study, the whole coding exons and splice junction sites of the NKX2-5 gene, which encodes a homeodomain transcription factor crucial for cardiac development and structural remodeling, were sequenced in 130 unrelated patients with idiopathic DCM. The available relatives of the index patient harboring an identified mutation and 200 unrelated ethnically matched healthy individuals used as controls were genotyped for the NKX2-5 gene. The functional effect of the mutant NKX2-5 was characterized in contrast to its wild-type counterpart using a dual-luciferase reporter assay system. As a result, a novel heterozygous NKX2-5 mutation, p.S146W, was identified in a family with DCM inherited as an autosomal dominant trait, which co-segregated with DCM in the family with complete penetrance. Notably, the mutation carriers also had arrhythmias, such as paroxysmal atrial fibrillation and atrioventricular block. The missense mutation was absent in 400 reference chromosomes and the altered amino acid was completely conserved evolutionarily among species. Functional analysis revealed that the NKX2-5 mutant was associated with a significantly reduced transcriptional activity. The findings expand the mutational spectrum of NKX2-5 linked to DCM and provide novel insight into the molecular mechanisms underlying DCM, contributing to the antenatal prophylaxis and allele-specific management of DCM.
Collapse
Affiliation(s)
- Fang Yuan
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Xin-Kai Qu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Juan Wang
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Xu Liu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Wei-Yi Fang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - De-Ning Liao
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
36
|
Koivisto E, Jurado Acosta A, Moilanen AM, Tokola H, Aro J, Pennanen H, Säkkinen H, Kaikkonen L, Ruskoaho H, Rysä J. Characterization of the regulatory mechanisms of activating transcription factor 3 by hypertrophic stimuli in rat cardiomyocytes. PLoS One 2014; 9:e105168. [PMID: 25136830 PMCID: PMC4138181 DOI: 10.1371/journal.pone.0105168] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 07/18/2014] [Indexed: 01/08/2023] Open
Abstract
Aims Activating transcription factor 3 (ATF3) is a stress-activated immediate early gene suggested to have both detrimental and cardioprotective role in the heart. Here we studied the mechanisms of ATF3 activation by hypertrophic stimuli and ATF3 downstream targets in rat cardiomyocytes. Methods and Results When neonatal rat cardiomyocytes were exposed to endothelin-1 (ET-1, 100 nM) and mechanical stretching in vitro, maximal increase in ATF3 expression occurred at 1 hour. Inhibition of extracellular signal-regulated kinase (ERK) by PD98059 decreased ET-1– and stretch–induced increase of ATF3 protein but not ATF3 mRNA levels, whereas protein kinase A (PKA) inhibitor H89 attenuated both ATF3 mRNA transcription and protein expression in response to ET-1 and stretch. To characterize further the regulatory mechanisms upstream of ATF3, p38 mitogen-activated protein kinase (MAPK) signaling was investigated using a gain-of-function approach. Adenoviral overexpression of p38α, but not p38β, increased ATF3 mRNA and protein levels as well as DNA binding activity. To investigate the role of ATF3 in hypertrophic process, we overexpressed ATF3 by adenovirus-mediated gene transfer. In vitro, ATF3 gene delivery attenuated the mRNA transcription of interleukin-6 (IL-6) and plasminogen activator inhibitor-1 (PAI-1), and enhanced nuclear factor-κB (NF-κB) and Nkx-2.5 DNA binding activities. Reduced PAI-1 expression was also detected in vivo in adult rat heart by direct intramyocardial adenovirus-mediated ATF3 gene delivery. Conclusions These data demonstrate that ATF3 activation by ET-1 and mechanical stretch is partly mediated through ERK and cAMP-PKA pathways, whereas p38 MAPK pathway is involved in ATF3 activation exclusively through p38α isoform. ATF3 activation caused induction of modulators of the inflammatory response NF-κB and Nkx-2.5, as well as attenuation of pro-fibrotic and pro-inflammatory proteins IL-6 and PAI-1, suggesting cardioprotective role for ATF3 in the heart.
Collapse
Affiliation(s)
- Elina Koivisto
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Alicia Jurado Acosta
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Anne-Mari Moilanen
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
- Department of Pathology, Institute of Diagnostics, University of Oulu, Oulu, Finland
| | - Heikki Tokola
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
- Department of Pathology, Institute of Diagnostics, University of Oulu, Oulu, Finland
| | - Jani Aro
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Harri Pennanen
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Hanna Säkkinen
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Leena Kaikkonen
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Heikki Ruskoaho
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- * E-mail:
| | - Jaana Rysä
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
37
|
Garg M, Khanna D. Exploration of pharmacological interventions to prevent isoproterenol-induced myocardial infarction in experimental models. Ther Adv Cardiovasc Dis 2014; 8:155-169. [PMID: 24817146 DOI: 10.1177/1753944714531638] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
High incidences of myocardial infarction associated with high morbidity and mortality, are a major concern and economic burden on industrialized nations. Persistent β-adrenergic receptor stimulation with isoproterenol leads to the development of oxidative stress, myocardial inflammation, thrombosis, platelet aggregation and calcium overload, which ultimately cause myocardial infarction. Therapeutic agents that are presently employed for the prevention and management of myocardial infarction are beta-blockers, antithrombotics, thrombolytics, statins, angiotensin converting enzyme inhibitors, angiotensin II type 1 receptor blockers, calcium channel blockers and nitrovasodilators. In spite of effective available interventions, the mortality rate of myocardial infarction is progressively increasing. Thus, there has been a regular need to develop effective therapies for the prevention and management of this insidious disease. In this review, the authors give an overview of the consequences of isoproterenol in the pathogenesis of cardiac disorders and various therapeutic possibilities to prevent these disorders.
Collapse
Affiliation(s)
- Monika Garg
- Cardiovascular Pharmacology Division Department of Pharmacology Rajendra Institute of Technology and Sciences India
| | - Deepa Khanna
- Department of Pharmacology, Cardiovascular Pharmacology Division, Institute of Pharmacy, Rajendra Institute of Technology and Sciences [RITS], Sirsa-125 055, India
| |
Collapse
|
38
|
Matsuoka K, Asano Y, Higo S, Tsukamoto O, Yan Y, Yamazaki S, Matsuzaki T, Kioka H, Kato H, Uno Y, Asakura M, Asanuma H, Minamino T, Aburatani H, Kitakaze M, Komuro I, Takashima S. Noninvasive and quantitative live imaging reveals a potential stress‐responsive enhancer in the failing heart. FASEB J 2014; 28:1870-9. [DOI: 10.1096/fj.13-245522] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ken Matsuoka
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| | - Yoshihiro Asano
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| | - Shuichiro Higo
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| | - Osamu Tsukamoto
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| | - Yi Yan
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| | - Satoru Yamazaki
- Department of Cell BiologyNational Cerebral and Cardiovascular Center Research InstituteSuitaJapan
| | - Takashi Matsuzaki
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Hidetaka Kioka
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| | - Hisakazu Kato
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| | - Yoshihiro Uno
- Laboratory of Reproductive EngineeringInstitute of Experimental Animal Sciences, Osaka University Graduate School of MedicineSuitaJapan
| | - Masanori Asakura
- Department of Clinical Research and DevelopmentNational Cerebral and Cardiovascular Center Research InstituteSuitaJapan
| | - Hiroshi Asanuma
- Department of Cardiovascular Science and TechnologyKyoto Prefectural University School of MedicineKyotoJapan
| | - Tetsuo Minamino
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and TechnologyUniversity of TokyoTokyoJapan
| | - Masafumi Kitakaze
- Department of Clinical Research and DevelopmentNational Cerebral and Cardiovascular Center Research InstituteSuitaJapan
| | - Issei Komuro
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Seiji Takashima
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| |
Collapse
|
39
|
Early growth response 1 is an early signal inducing Cav3.2 T-type calcium channels during cardiac hypertrophy. Cardiovasc Res 2013; 100:222-30. [DOI: 10.1093/cvr/cvt190] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
40
|
Spangenburg EE, Geiger PC, Leinwand LA, Lowe DA. Regulation of physiological and metabolic function of muscle by female sex steroids. Med Sci Sports Exerc 2013; 44:1653-62. [PMID: 22525764 DOI: 10.1249/mss.0b013e31825871fa] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The ability of female sex steroids to regulate tissue function has long been appreciated; however, their role in the regulation of striated muscle function has received considerably less attention. The purpose of this symposium review was to document recent evidence indicating the role female sex steroids have in defining the functional characteristics of striated muscle. The presentations provide substantial evidence indicating that estrogens are critical to the physiological and metabolic regulations of striated muscle; thus, when considering women's health issues, striated muscle must included as an important target tissue along with other classically thought of estrogen-sensitive tissues.
Collapse
Affiliation(s)
- Espen E Spangenburg
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD 21045, USA.
| | | | | | | |
Collapse
|
41
|
SIRT1 mediates the protective function of Nkx2.5 during stress in cardiomyocytes. Basic Res Cardiol 2013; 108:364. [PMID: 23744058 DOI: 10.1007/s00395-013-0364-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 05/02/2013] [Accepted: 05/21/2013] [Indexed: 10/26/2022]
Abstract
Nkx2.5 plays protective roles in cardiac homeostasis and survival in the postnatal hearts. However, the underlying molecular mechanisms that mediate the protective functions of Nkx2.5 remain unknown. Here, we showed that Nkx2.5 was downregulated in response to various stresses and was required for protection against the stress-induced apoptosis of cardiomyocytes. SIRT1, a member of the sirtuin family of proteins, was found to be a direct transcriptional target of Nkx2.5 and was required for the Nkx2.5-mediated protection of cardiomyocytes from doxorubicin (DOX)-induced apoptosis. Moreover, using chromatin immunoprecipitation assays, we found that Nkx2.5 was able to bind to the SIRT1 promoter and that this binding was significantly decreased in DOX-treated mouse hearts. Furthermore, the cardiac-specific overexpression of SIRT1 decreased the DOX-induced apoptosis of cardiomyocytes in SIRT1 transgenic mouse hearts compared with the hearts of their wild-type littermates. These findings demonstrate that SIRT1 acts as a direct transcriptional target of Nkx2.5 that maintains cardiomyocyte homeostasis and survival.
Collapse
|
42
|
Abstract
Stress-induced hypertrophic growth of the heart predisposes the heart to arrhythmia, contractile dysfunction, and clinical heart failure. FHL2 (four-and-a-half LIM domain protein 2) is expressed predominantly in the heart, and inactivation of the gene coding for FHL2 leads to exaggerated responsiveness to adrenergic stress. Activation of calcineurin occurs downstream of β-adrenergic signaling and is required for isoproterenol-induced myocardial hypertrophy. Based on these facts, we hypothesized that FHL2 suppresses stress-induced activation of calcineurin. FHL2 is upregulated in mouse hearts exposed to isoproterenol, a β-adrenergic agonist, and isoproterenol-induced increases in the NFAT target genes RCAN1.4 and BNP were amplified significantly in FHL2 knockout (FHL2(-/-)) mice compared with levels in wild-type (WT) mice. To determine whether the effect of FHL2 on NFAT target gene transcript levels occurred at the level of transcription, HEK 293 cells and neonatal rat ventricular myocytes (NRVMs) were transfected with a luciferase reporter construct harboring the NFAT-dependent promoters of either RCAN1 or interleukin 2 (IL-2). Consistent with the in vivo data, small interfering RNA (siRNA) knockdown of FHL2 led to increased activation of these promoters by constitutively active calcineurin or the calcium ionophore ionomycin. Importantly, activation of the RCAN1 promoter by ionomycin, in control and FHL2 knockdown cells, was abolished by the calcineurin inhibitor cyclosporine, confirming the calcineurin dependence of the response. Overexpression of FHL2 inhibited activation of both NFAT reporter constructs. Furthermore, NRVMs overexpressing FHL2 exhibited reduced hypertrophic growth in response to constitutively active calcineurin, as measured by cell cross-sectional area and fetal gene expression. Finally, immunostaining in isolated adult cardiomyocytes revealed colocalization of FHL2 and calcineurin predominantly at the sarcomere and activation of calcineurin by endothelin-1-facilitated interaction between FHL2 and calcineurin. FHL2 is an endogenous, agonist-dependent suppressor of calcineurin.
Collapse
|
43
|
Hamad EA, Zhu W, Chan TO, Myers V, Gao E, Li X, Zhang J, Song J, Zhang XQ, Cheung JY, Koch W, Feldman AM. Cardioprotection of controlled and cardiac-specific over-expression of A(2A)-adenosine receptor in the pressure overload. PLoS One 2012; 7:e39919. [PMID: 22792196 PMCID: PMC3391213 DOI: 10.1371/journal.pone.0039919] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 05/29/2012] [Indexed: 11/18/2022] Open
Abstract
Adenosine binds to three G protein-coupled receptors (R) located on the cardiomyocyte (A(1)-R, A(2A)-R and A(3)-R) and provides cardiac protection during both ischemic and load-induced stress. While the role of adenosine receptor-subtypes has been well defined in the setting of ischemia-reperfusion, far less is known regarding their roles in protecting the heart during other forms of cardiac stress. Because of its ability to increase cardiac contractility and heart rate, we hypothesized that enhanced signaling through A(2A)-R would protect the heart during the stress of transverse aortic constriction (TAC). Using a cardiac-specific and inducible promoter, we selectively over-expressed A(2A)-R in FVB mice. Echocardiograms were obtained at baseline, 2, 4, 8, 12, 14 weeks and hearts were harvested at 14 weeks, when WT mice developed a significant decrease in cardiac function, an increase in end systolic and diastolic dimensions, a higher heart weight to body weight ratio (HW/BW), and marked fibrosis when compared with sham-operated WT. More importantly, these changes were significantly attenuated by over expression of the A(2A)-R. Furthermore, WT mice also demonstrated marked increases in the hypertrophic genes β-myosin heavy chain (β-MHC), and atrial natriuretic factor (ANF)--changes that are mediated by activation of the transcription factor GATA-4. Levels of the mRNAs encoding β-MHC, ANP, and GATA-4 were significantly lower in myocardium from A(2A)-R TG mice after TAC when compared with WT and sham-operated controls. In addition, three inflammatory factors genes encoding cysteine dioxygenase, complement component 3, and serine peptidase inhibitor, member 3N, were enhanced in WT TAC mice, but their expression was suppressed in A(2A)-R TG mice. A(2A)-R over-expression is protective against pressure-induced heart failure secondary to TAC. These cardioprotective effects are associated with attenuation of GATA-4 expression and inflammatory factors. The A(2A)-R may provide a novel new target for pharmacologic therapy in patients with cardiovascular disease.
Collapse
Affiliation(s)
- Eman A. Hamad
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Medicine, The Center for Translational Medicine, Jefferson Medical College, Philadelphia, Pennsylvania, United States of America
| | - Weizhong Zhu
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Tung O. Chan
- Department of Medicine, The Center for Translational Medicine, Jefferson Medical College, Philadelphia, Pennsylvania, United States of America
| | - Valerie Myers
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Erhe Gao
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Xue Li
- Department of Medicine, The Center for Translational Medicine, Jefferson Medical College, Philadelphia, Pennsylvania, United States of America
| | - Jin Zhang
- Department of Medicine, The Center for Translational Medicine, Jefferson Medical College, Philadelphia, Pennsylvania, United States of America
| | - Jianliang Song
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Xue-Qian Zhang
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Joseph Y. Cheung
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Walter Koch
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Arthur M. Feldman
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
44
|
Coto E, Reguero JR, Palacín M, Gómez J, Alonso B, Iglesias S, Martín M, Tavira B, Díaz-Molina B, Morales C, Morís C, Rodríguez-Lambert JL, Corao AI, Díaz M, Alvarez V. Resequencing the whole MYH7 gene (including the intronic, promoter, and 3' UTR sequences) in hypertrophic cardiomyopathy. J Mol Diagn 2012; 14:518-24. [PMID: 22765922 DOI: 10.1016/j.jmoldx.2012.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 04/03/2012] [Accepted: 04/11/2012] [Indexed: 01/14/2023] Open
Abstract
MYH7 mutations are found in ~20% of hypertrophic cardiomyopathy (HCM) patients. Currently, mutational analysis is based on the sequencing of the coding exons and a few exon-flanking intronic nucleotides, resulting in omission of single-exon deletions and mutations in internal intronic, promoter, and 3' UTR regions. We amplified and sequenced large MYH7 fragments in 60 HCM patients without previously identified sarcomere mutations. Lack of aberrant PCR fragments excluded single-exon deletions in the patients. Instead, we identified several new rare intronic variants. An intron 26 single nucleotide insertion (-5 insC) was predicted to affect pre-mRNA splicing, but allele frequencies did not differ between patients and controls (n = 150). We found several rare promoter variants in the patients compared to controls, some of which were in binding sites for transcription factors and could thus affect gene expression. Only one rare 3' UTR variant (c.*29T>C) found in the patients was absent among the controls. This nucleotide change would not affect the binding of known microRNAs. Therefore, MYH7 mutations outside the coding exon sequences would be rarely found among HCM patients. However, changes in the promoter region could be linked to the risk of developing HCM. Further research to define the functional effect of these variants on gene expression is necessary to confirm the role of the MYH7 promoter in cardiac hypertrophy.
Collapse
Affiliation(s)
- Eliecer Coto
- Molecular Genetics-Laboratory of Medicine-Renal Foundation (IRSIN-FRIAT), University Central Hospital Asturias (HUCA), Oviedo, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Cardiac mechanoenergetics for understanding isoproterenol-induced rat heart failure. ACTA ACUST UNITED AC 2012; 19:163-70. [PMID: 22687629 DOI: 10.1016/j.pathophys.2012.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 12/06/2011] [Accepted: 12/11/2011] [Indexed: 11/23/2022]
Abstract
Considering from clinical implication, it is often complained that short-term experimental diseased heart models do not mimic long-term diseased hearts that one often clinically encountered. The left ventricle (LV) function in rat cardiac hypertrophy models treated with isoproterenol (ISO) up to 16 weeks was followed up with a non-invasive echocardiography. Infusion of either ISO (1.2mgkg(-1)day(-1) for 3 days-16 weeks) or vehicle (saline 24μlday(-1) for 3 days-16 weeks; SA group) was performed by subcutaneously implanting osmotic minipump. LV and right ventricle (RV) weight ratios to body weight (mgg(-1)) in SA, ISO3d, ISO7d and ISO4w were: 1.94±0.10 and 0.54±0.04 (n=7), 2.56±0.10 and 0.66±0.05 (n=7), 2.50±0.25 and 0.64±0.07 (n=10) and 2.40±0.08 and 0.59±0.08 (n=9), respectively. From echocardiography, the LV function of the hypertrophy models at 3 days, 1 and 2 weeks was unchanged but the model at the longer-term than 4 weeks resulted in prolonged systolic failure. These results indicated that only 3-day ISO infusion induced the hypertrophy model similar in shape and function to that induced by 2-week ISO infusion; the 3-day model sufficiently represents the effects of 2-week ISO infusion. In this review, left ventricular (LV) function was compared between rat cardiac hypertrophy models treated with ISO for 3 days (ISO3d) and 7 days (ISO7d) by analyzing LV mechanical work and energetics. The LV mechanical work and energetics was unchanged in SA, ISO3d and ISO7d groups. The LV relaxation rate at 240bpm in ISO3d and ISO7d groups was significantly slower than that in SA group with unchanged contraction rate. The amounts of expression of sarcoplasmic reticulum Ca(2+)-ATPase (SERCA2a), phospholamban (PLB), phosphorylated-Ser(16) PLB (p-PLB), phospholemman (PLM) and Na(+)-K(+)-ATPase (NKA) are significantly decreased in ISO3d and ISO7d groups. Furthermore, the marked collagen production (types I and III) was observed in ISO3d and ISO7d groups. These results suggested the possibility that physiological LV function is compensated, although molecular changes have been generated even in the short-term hypertrophy model. Although a novel histone deacetylase (HDAC) inhibitor, has some beneficial effects on hemodynamics, it has no effects of anti-hypertrophic modalities in ISO3d model. However, a selective sodium proton exchanger-1 (NHE-1) inhibitor normalized ISO-induced down-regulation of SERCA2a without changes in pPLB/PLB expression in the ISO7d model and ameliorates cardiac Ca(2+) handling impairment and prevents the development of cardiac dysfunction. This result indicated that SERCA2a is a key molecule in the ISO7d model. Slow LV relaxation rate in ISO7d model may be due to down-regulation of SERCA2a. In conclusion, lowering the heart rate make it possible to rescue the impairment of LV mechanical work and energetics in the ISO-induced compensatory hypertrophied rat hearts, providing basic evidence for clinical therapy for patients with some types of cardiac failure.
Collapse
|
46
|
Wang Q, Domenighetti AA, Schäfer SC, Weber J, Simon A, Maillard MP, Pedrazzini T, Chen J, Lehr HA, Burnier M. Impact of salt on cardiac differential gene expression and coronary lesion in normotensive mineralocorticoid-treated mice. Am J Physiol Regul Integr Comp Physiol 2012; 302:R1025-33. [DOI: 10.1152/ajpregu.00387.2011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
We previously reported that excess of deoxycorticosterone-acetate (DOCA)/salt-induced cardiac hypertrophy in the absence of hypertension in one-renin gene mice. This model allows us to study molecular mechanisms of high-salt intake in the development of cardiovascular remodeling, independently of blood pressure in a high mineralocorticoid state. In this study, we compared the effect of 5-wk low- and high-salt intake on cardiovascular remodeling and cardiac differential gene expression in mice receiving the same amount of DOCA. Differential gene and protein expression was measured by high-density cDNA microarray assays, real-time PCR and Western blot analysis in DOCA-high salt (HS) vs. DOCA-low salt (LS) mice. DOCA-HS mice developed cardiac hypertrophy, coronary perivascular fibrosis, and left ventricular dysfunction. Differential gene and protein expression demonstrated that high-salt intake upregulated a subset of genes encoding for proteins involved in inflammation and extracellular matrix remodeling (e.g., Col3a1, Col1a2, Hmox1, and Lcn2). A major subset of downregulated genes encoded for transcription factors, including myeloid differentiation primary response (MyD) genes. Our data provide some evidence that vascular remodeling, fibrosis, and inflammation are important consequences of a high-salt intake in DOCA mice. Our study suggests that among the different pathogenic factors of cardiac and vascular remodeling, such as hypertension and mineralocorticoid excess and sodium intake, the latter is critical for the development of the profibrotic and proinflammatory phenotype observed in the heart of normotensive DOCA-treated mice.
Collapse
Affiliation(s)
- Qing Wang
- Service of Nephrology and Hypertension, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Huazhong University of Science and Technology, Wuhan, China
| | | | | | - Johanns Weber
- The Lausanne DNA Array Facility, University of Lausanne, Switzerland
| | - Alexandra Simon
- Service of Nephrology and Hypertension, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Marc P. Maillard
- Service of Nephrology and Hypertension, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | | | - Ju Chen
- Department of Medicine, University of California, San Diego, La Jolla, California
| | | | - Michel Burnier
- Service of Nephrology and Hypertension, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| |
Collapse
|
47
|
Peters BS, Dornaika R, Hosten N, Hadlich S, Mullins JJ, Peters J, Rettig R. Regression of cardiac hypertrophy in cyp1a1ren-2 transgenic rats. J Magn Reson Imaging 2012; 36:373-8. [PMID: 22517449 DOI: 10.1002/jmri.23661] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 03/07/2012] [Indexed: 11/10/2022] Open
Abstract
PURPOSE To evaluate the usefulness of the cyp1a1ren-2 transgenic rat model of inducible hypertension for studies of the development and regression of cardiac hypertrophy. MATERIALS AND METHODS Cyp1a1ren-2 rats received a diet containing 0% or 0.167% indole-3-carbinonl (I3C) for 4 weeks to induce hypertension. Cardiac magnetic resonance imaging (MRI) at 7 T was performed every second week for 10 weeks to measure left ventricular mass and the ejection fraction. Concomitantly, in six cyp1a1ren-2 rats blood pressure was recorded telemetrically. RESULTS Plasma prorenin concentrations rose from 138 ± 38 to 15,490 ± 3990 ng/angiotensin I/mL/h (P < 0.001) in I3C-treated transgenic rats and returned to basal levels after cessation of I3C. Mean blood pressure increased to a plateau of 169 ± 11 mmHg by the second week of induction. After cessation of I3C (day 28), arterial pressure dropped to values slightly below those prior to induction within 4 days (basal: 106 ± 7 mmHg, day 32: 103 ± 21 mmHg; NS). At day 28, left ventricular mass was increased by 39% vs. 4% in controls (P < 0.001) without changes of the ejection fraction. Cardiac hypertrophy was completely reversed at day 70, as evaluated by MRI. CONCLUSION The cyp1a1ren-2 transgenic rat is a useful model to study reversal and healing in the absence of surgical interventions.
Collapse
Affiliation(s)
- Barbara S Peters
- Department of Cardiovascular Medicine, Institute of Physiology, University of Greifswald, Karlsburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
48
|
Schlossarek S, Schuermann F, Geertz B, Mearini G, Eschenhagen T, Carrier L. Adrenergic stress reveals septal hypertrophy and proteasome impairment in heterozygous Mybpc3-targeted knock-in mice. J Muscle Res Cell Motil 2011; 33:5-15. [PMID: 22076249 DOI: 10.1007/s10974-011-9273-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 10/30/2011] [Indexed: 11/27/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is characterized by asymmetric septal hypertrophy and is often caused by mutations in MYBPC3 gene encoding cardiac myosin-binding protein C. In contrast to humans, who are already affected at the heterozygous state, mouse models develop the phenotype mainly at the homozygous state. Evidence from cell culture work suggested that altered proteasome function contributes to the pathogenesis of HCM. Here we tested in two heterozygous Mybpc3-targeted mouse models whether adrenergic stress unmasks a specific cardiac phenotype and proteasome dysfunction. The first model carries a human Mybpc3 mutation (Het-KI), the second is a heterozygous Mybpc3 knock-out (Het-KO). Both models were compared to wild-type (WT) mice. Mice were treated with a combination of isoprenaline and phenylephrine (ISO/PE) or NaCl for 1 week. Whereas ISO/PE induced left ventricular hypertrophy (LVH) with increased posterior wall thickness to a similar extent in all groups, it increased septum thickness only in Het-KI and Het-KO. ISO/PE did not affect the proteasomal chymotrypsin-like activity or β5-subunit protein level in Het-KO or wild-type mice (WT). In contrast, both parameters were markedly lower in Het-KI and negatively correlated with the degree of LVH in Het-KI only. In conclusion, adrenergic stress revealed septal hypertrophy in both heterozygous mouse models of HCM, but proteasome dysfunction only in Het-KI mice, which carry a mutant allele and closely mimic human HCM. This supports the hypothesis that proteasome impairment contributes to the pathophysiology of HCM.
Collapse
Affiliation(s)
- Saskia Schlossarek
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
49
|
Wang N, Guan P, Zhang JP, Li YQ, Chang YZ, Shi ZH, Wang FY, Chu L. Fasudil hydrochloride hydrate, a Rho-kinase inhibitor, suppresses isoproterenol-induced heart failure in rats via JNK and ERK1/2 pathways. J Cell Biochem 2011; 112:1920-9. [PMID: 21433064 DOI: 10.1002/jcb.23112] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The Rho-kinase (ROCK) plays an important role in the pathogenesis of heart injury. Recent cellular and molecular biology studies indicated a pivotal role of the RhoA/ROCK cascade in many aspects of cardiovascular function such as heart failure, cardiac hypertrophy, and ventricular remodeling following myocardial infarction. However, the signal transduction of RhoA/ROCK and its down-stream signaling pathways remains elusive, and the mechanism of ROCK-mediated isoproterenol (ISO)-induced heart failure is still not thoroughly understood. In the present study, we investigated the effect of the ROCK inhibitor, fasudil hydrochloride hydrate, on ISO-induced heart failure and the potential relationship of RhoA/ROCK to the extracellular signal-regulated kinases (ERK) and the c-jun NH 2-terminal kinase (JNK) pathways. Male Sprague-Dawley (SD) rats, maintained on a normal diet, were randomly divided into four groups given control, ISO alone, ISO with low-dose fasudil, or ISO with high-dose fasudil treatments. Fasudil effectively inhibited ISO-induced heart failure, as evaluated by biometric, hemodynamic, and histological examinations. Consistently, ISO-induced ROCK-1 mRNA expression and myosin phosphatase target subunit-1 (MYPT-1) phosphorylation were markedly suppressed by fasudil. In addition, fasudil significantly decreased ISO-induced JNK activation, ERK translocation to the nucleus and subsequent c-fos, c-jun expression and upregulated c-FLIP(L) expression. Taken together, these results indicate that the RhoA/ROCK pathway is essential for ISO induced heart failure, which can be effectively suppressed by fasudil.
Collapse
Affiliation(s)
- Na Wang
- Department of Pharmacology, School of Basic Medicine, Heibei Medical University, Shijiazhuang 050091, Hebei, China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Ca2+-induced PARP-1 activation and ANF expression are coupled events in cardiomyocytes. Biochem J 2011; 438:337-47. [PMID: 21635224 DOI: 10.1042/bj20101484] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The nuclear protein PARP-1 [poly(ADP-ribose) polymerase-1] is activated in cardiomyocytes exposed to hypoxia causing DNA breaks. Unlike this stress-induced PARP-1 activation, our results provide evidence for Ca(2+)-induced PARP-1 activation in contracting newborn cardiomyocytes treated with growth factors and hormones that increased their contraction rate, induced intracellular Ca(2+) mobilization and its rhythmical and transient translocation into the nucleus. Furthermore, activated PARP-1 up-regulated the activity of phosphorylated ERK (extracellular-signal-regulated kinase) in the nucleus, promoting expression of the Elk1 target gene c-fos. Up-regulation of the transcription factor c-Fos/GATA-4 promoted ANF (atrial natriuretic factor) expression. Given that expression of ANF is known to be implicated in morphological changes, growth and development of cardiomyocytes, these results outline a PARP-1-dependent signal transduction mechanism that links contraction rate and Ca(2+) mobilization with the expression of genes underlying morphological changes in cardiomyocytes.
Collapse
|