1
|
Bhullar SK, Dhalla NS. Adaptive and maladaptive roles of different angiotensin receptors in the development of cardiac hypertrophy and heart failure. Can J Physiol Pharmacol 2024; 102:86-104. [PMID: 37748204 DOI: 10.1139/cjpp-2023-0226] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Angiotensin II (Ang II) is formed by the action of angiotensin-converting enzyme (ACE) in the renin-angiotensin system. This hormone is known to induce cardiac hypertrophy and heart failure and its actions are mediated by the interaction of both pro- and antihypertrophic Ang II receptors (AT1R and AT2R). Ang II is also metabolized by ACE 2 to Ang-(1-7), which elicits the activation of Mas receptors (MasR) for inducing antihypertrophic actions. Since heart failure under different pathophysiological situations is preceded by adaptive and maladaptive cardiac hypertrophy, we have reviewed the existing literature to gain some information regarding the roles of AT1R, AT2R, and MasR in both acute and chronic conditions of cardiac hypertrophy. It appears that the activation of AT1R may be involved in the development of adaptive and maladaptive cardiac hypertrophy as well as subsequent heart failure because both ACE inhibitors and AT1R antagonists exert beneficial effects. On the other hand, the activation of both AT2R and MasR may prevent the occurrence of maladaptive cardiac hypertrophy and delay the progression of heart failure, and thus therapy with different activators of these antihypertrophic receptors under chronic pathological stages may prove beneficial. Accordingly, it is suggested that a great deal of effort should be made to develop appropriate activators of both AT2R and MasR for the treatment of heart failure subjects.
Collapse
Affiliation(s)
- Sukhwinder K Bhullar
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
2
|
Subbaiah KCV, Wu J, Tang WHW, Yao P. FAM114A1 influences cardiac pathological remodeling by regulating angiotensin II signaling. JCI Insight 2022; 7:152783. [PMID: 35671117 PMCID: PMC9310534 DOI: 10.1172/jci.insight.152783] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 06/01/2022] [Indexed: 11/30/2022] Open
Abstract
Cardiac pathological remodeling, a primary contributor to heart failure (HF) and death, is an important target for HF therapy. However, the signaling pathways that govern cardiac remodeling are not fully elucidated. Here, we found that a functionally unannotated human myocardial infarction–associated (MI-associated) gene, family with sequence similarity 114 member A1 (FAM114A1), is induced in failing human and mouse hearts compared with nonfailing hearts. Homozygous KO of Fam114a1 (Fam114a1–/–) in the mouse genome reduces cardiomyocyte hypertrophy, inflammation, and cardiac fibrosis while restoring cardiac function in angiotensin II–induced (Ang II–induced) and MI-induced HF mouse models. Cardiac fibroblasts (CFs) exhibit the highest FAM114A1 expression among different cardiac cell types. FAM114A1 is a critical autonomous factor for CF proliferation, activation, and migration. Mechanistically, FAM114A1 interacts with angiotensin receptor–associated protein (AGTRAP) and regulates the expression of angiotensin type 1 receptor (AT1R) and downstream Ang II signaling transduction, and it subsequently influences profibrotic response. Our results indicate that FAM114A1 regulates Ang II signaling, thereby activating CFs and other cardiac cells and augmenting pathological cardiac remodeling. These findings provide potentially novel insights into the regulation of cardiac remodeling and identify FAM114A1 as a therapeutic target for the treatment of heart disease.
Collapse
Affiliation(s)
- Kadiam C Venkata Subbaiah
- Department of Medicine, University of Rochester School of Medicine, Rochester, United States of America
| | - Jiangbin Wu
- Department of Medicine, University of Rochester School of Medicine, Rochester, United States of America
| | - Wai Hong Wilson Tang
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, United States of America
| | - Peng Yao
- Department of Medicine, University of Rochester School of Medicine, Rochester, United States of America
| |
Collapse
|
3
|
Pathophysiology of heart failure and an overview of therapies. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00025-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
4
|
Bloise AC, Dos Santos JA, de Brito IV, Bassaneze V, Gomes LF, Alencar AM. Discriminating aspects of global metabolism of neonatal cardiomyocytes from wild type and KO-CSRP3 rats using proton magnetic resonance spectroscopy of culture media samples. In Vitro Cell Dev Biol Anim 2020; 56:604-613. [PMID: 32914385 DOI: 10.1007/s11626-020-00497-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/10/2020] [Indexed: 12/16/2022]
Abstract
Knockout of multifunction gene cysteine- and glycine-rich protein 3 (CSRP3) in cardiomyocytes (CMs) of mice leads to heart dilation, severely affecting its functions. In humans, CSRP3 mutations are associated with hypertrophic (HCM) and dilated cardiomyopathy (DCM). The absence of the CSRP3 expression produces unknown effects on in vitro neonatal CMs' metabolism. The metabolome changes in culture media conditioned by CSRP3 knockout (KO-CSRP3), and wild type (WT) neonatal cardiomyocytes were investigated under untreated or after metabolic challenging conditions produced by isoproterenol (ISO) stimulation, by in vitro high-resolution proton magnetic resonance spectroscopy (1H-MRS)-based metabolomics. Metabolic differences between neonatal KO-CSRP3 and WT rats' CMs were identified. After 72 h of culture, ISO administration was associated with increased CMs' energy requirements and increased levels of threonine, alanine, and 3-hydroxybutyrate in both neonatal KO-CSRP3 and WT CMs conditioned media. When compared with KO-CSRP3, culture media derived from WT cells presented higher lactate concentrations either under basal or ISO-stimulated conditions. The higher activity of ketogenic biochemical pathways met the elevated energy requirements of the contractile cells. Both cells are considered phenotypically indistinguishable in the neonatal period of animal lives, but the observed metabolic stress responses of KO-CSRP3 and WT CMs to ISO were different. KO-CSRP3 CMs produced less lactate than WT CMs in both basal and stimulated conditions. Mainly, ISO-stimulated conditions produced evidence for lactate overload within KO-CSRP3 CMs, while WT CMs succeeded to manage the metabolic stress. Thus, 1H-MRS-based metabolomics was suitable to identify early inefficient energetic metabolism in neonatal KO-CSRP3 CMs. These results may reflect an apparent lower lactate transport and consumption, in association with protein catabolism.
Collapse
Affiliation(s)
- Antonio Carlos Bloise
- Laboratory of Microrheology and Molecular physiology, Instituto de Física, Universidade de São Paulo, São Paulo, 05508-090, Brazil.
| | - Jennifer Adriane Dos Santos
- Laboratory of Microrheology and Molecular physiology, Instituto de Física, Universidade de São Paulo, São Paulo, 05508-090, Brazil.,Laboratory of Genetics and Molecular Cardiology/LIM 13, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, 05403-000, Brazil
| | - Isis Vasconcelos de Brito
- Laboratory of Microrheology and Molecular physiology, Instituto de Física, Universidade de São Paulo, São Paulo, 05508-090, Brazil
| | - Vinicius Bassaneze
- Laboratory of Genetics and Molecular Cardiology/LIM 13, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, 05403-000, Brazil
| | - Ligia Ferreira Gomes
- Laboratory of Microrheology and Molecular physiology, Instituto de Física, Universidade de São Paulo, São Paulo, 05508-090, Brazil.,Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Adriano Mesquita Alencar
- Laboratory of Microrheology and Molecular physiology, Instituto de Física, Universidade de São Paulo, São Paulo, 05508-090, Brazil
| |
Collapse
|
5
|
Chen H, Wu M, Jiang W, Liu X, Zhang J, Yu C. iTRAQ‑based quantitative proteomics analysis of the potential application of secretoneurin gene therapy for cardiac hypertrophy induced by DL‑isoproterenol hydrochloride in mice. Int J Mol Med 2020; 45:793-804. [PMID: 31985029 PMCID: PMC7015125 DOI: 10.3892/ijmm.2020.4472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 12/17/2019] [Indexed: 02/05/2023] Open
Abstract
A previous study by our group demonstrated a protective role of the neuropeptide secretoneurin (SN) in DL‑isoproterenol hydrochloride (ISO)‑induced cardiac hypertrophy in mice. To further characterize the molecular mechanism of SN treatment, an isobaric tags for relative and absolute quantification (iTRAQ)‑based quantitative proteomic analysis was applied to identify putative target proteins and molecular pathways. An SN expression vector was injected into the myocardial tissues of mice, and the animals were then subcutaneously injected with ISO (5 mg/kg/day) for 7 days to induce cardiac hypertrophy. The results of echocardiography and hemodynamic measurements indicated that the function of the heart impaired by ISO treatment was significantly ameliorated via SN gene injection. The investigation of heart proteomics was performed by iTRAQ‑based liquid chromatography‑tandem mass spectrometry analysis. A total of 2,044 quantified proteins and 15 differentially expressed proteins were associated with SN overexpression in mice with cardiac hypertrophy. Functional enrichment analysis demonstrated that these effects were possibly associated with metabolic processes. A protein‑protein interaction network analysis was constructed and the data indicated that apolipoprotein C‑III (Apoc3) was associated with the positive effect of SN on the induction of cardiac hypertrophy in mice. The present study proposed a potential mechanism of SN action on Apoc3 upregulation that may contribute to the amelioration of cardiac hypertrophy. These findings can aid the clinical application of SN in patients with cardiac hypertrophy.
Collapse
Affiliation(s)
| | - Mingjun Wu
- Institute of Life Science, Chongqing Medical University, Chongqing 400016
| | - Wei Jiang
- State Key Laboratory of Biotherapy, Molecular Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiang Liu
- Institute of Life Science, Chongqing Medical University, Chongqing 400016
| | - Jun Zhang
- Institute of Life Science, Chongqing Medical University, Chongqing 400016
| | | |
Collapse
|
6
|
Zhang J, Wei X, Zhang W, Wang F, Li Q. MiR-326 targets MDK to regulate the progression of cardiac hypertrophy through blocking JAK/STAT and MAPK signaling pathways. Eur J Pharmacol 2020; 872:172941. [PMID: 31972179 DOI: 10.1016/j.ejphar.2020.172941] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 12/25/2019] [Accepted: 01/17/2020] [Indexed: 02/07/2023]
Abstract
Cardiac hypertrophy is a heart reaction to the increase of cardiac load, with the characteristics of increased expression of cardiac hypertrophy markers, enhanced protein synthesis, and enlarged cell area. However, molecular mechanisms in cardiac hypertrophy are still poorly substantiated. It has been reported that miRNAs can modulate human diseases, among which miR-326 has been reported as a biological regulator in human cancers, but its role in cardiac hypertrophy is rarely explored. This study focused on the exploration of the potential of miR-326 in cardiac hypertrophy. Our data revealed the downregulation of miR-326 in the TAC-induced hypertrophic mice and the Ang II-induced hypertrophic H9c2 cells. Functionally, miR-326 attenuated the effect of Ang II on cardiac hypertrophy in vitro. In addition, miR-326 negatively regulated JAK/STAT and MAPK signaling pathways. Mechanistically, miR-326 targeted and inhibited MDK to induce JAK/STAT and MAPK pathways. Rescue assays certified that miR-326 attenuated cardiac hypertrophy through targeting MDK and inhibiting JAK/STAT and MAPK signaling pathways. In brief, our study unveiled that miR-326 targets MDK to regulate the progression of cardiac hypertrophy through blocking JAK/STAT and MAPK signaling pathways, indicating that targeting miR-326 as a potential approach for cardiac hypertrophy treatment.
Collapse
Affiliation(s)
- Jintao Zhang
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Heart Center of Henan Provincial People's Hospital, No. 1, Fu Wai Road, Zhengzhou, Henan, 450000, China
| | - Xinhua Wei
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Heart Center of Henan Provincial People's Hospital, No. 1, Fu Wai Road, Zhengzhou, Henan, 450000, China
| | - Weitao Zhang
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Heart Center of Henan Provincial People's Hospital, No. 1, Fu Wai Road, Zhengzhou, Henan, 450000, China
| | - Fengfeng Wang
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Heart Center of Henan Provincial People's Hospital, No. 1, Fu Wai Road, Zhengzhou, Henan, 450000, China
| | - Qun Li
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Heart Center of Henan Provincial People's Hospital, No. 1, Fu Wai Road, Zhengzhou, Henan, 450000, China.
| |
Collapse
|
7
|
Secretoneurin suppresses cardiac hypertrophy through suppression of oxidant stress. Eur J Pharmacol 2018; 822:13-24. [PMID: 29337195 DOI: 10.1016/j.ejphar.2018.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 12/25/2017] [Accepted: 01/10/2018] [Indexed: 02/05/2023]
Abstract
The neuropeptide secretoneurin (SN) plays protective roles in myocardial ischemia. In the present study, the effect of SN in cardiac hypertrophy was investigated. We observed that, in isoproterenol (ISO) treatment induced cardiac or cardiomyocytes hypertrophy, a marked increase in the expression of endogenous SN in mouse plasma, myocardium and primary-cultured cardiomyocytes occurs. In hypertrophic mice, the heart size, heart weight/body weight (HW/BW) ratio, cardiomyocyte size, and atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) expression were significantly higher than those in controls but were effectively suppressed by SN gene therapy. Similarly, the protective effects of SN were also observed in cultured cardiomyocytes following ISO treatment. SN significantly increased the activity of catalase and superoxide dismutase (SOD) in parallel with the decrease in reactive oxygen species levels in cardiomyocytes. We observed that SN evoked the activation of all of the AMPK, P38/MAPK and ERK/MAPK pathways in cardiomyocytes, but pretreatment with only AMPK inhibitor (compound C) and ERK1/2/MAPK inhibitor (PD98059) counteracted the protective effects of SN against cardiomyocyte hypertrophy and the suppressive effects of SN on oxidant stress in cardiomyocytes. These results indicated that endogenous SN is induced in hypertrophic cardiomyocytes, and may play a protective role in the pathogenesis of cardiac hypertrophy. These results suggest that exogenous SN supplementation protects the cardiac hypertrophy induced by ISO treatment through the activation of AMPK and ERK/MAPK pathways, thus upregulating antioxidants and suppressing oxidative stress.
Collapse
|
8
|
Meng Y, Zhang Y, Ma Z, Zhou H, Ni J, Liao H, Tang Q. Genistein attenuates pathological cardiac hypertrophy in vivo and in vitro. Herz 2017; 44:247-256. [DOI: 10.1007/s00059-017-4635-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 09/01/2017] [Accepted: 09/29/2017] [Indexed: 12/31/2022]
|
9
|
Pan L, Sheng M, Huang Z, Zhu Z, Xu C, Teng L, He L, Gu C, Yi C, Li J. Zinc-finger protein 418 overexpression protects against cardiac hypertrophy and fibrosis. PLoS One 2017; 12:e0186635. [PMID: 29065170 PMCID: PMC5655480 DOI: 10.1371/journal.pone.0186635] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 10/04/2017] [Indexed: 11/18/2022] Open
Abstract
Background This study aimed to investigated the effect and mechanism of zinc-finger protein 418 (ZNF418) on cardiac hypertrophy caused by aortic banding (AB), phenylephrine (PE) or angiotensin II (Ang II) in vivo and in vitro. Methods The expression of ZNF418 in hearts of patients with dilated cardiomyopathy (DCM) or hypertrophic cardiomyopathy (HCM) and AB-induced cardiac hypertrophy mice, as well as in Ang II- or PE-induced hypertrophic primary cardiomyocytes was detected by western blotting. Then, the expression of ZNF418 was up-regulated or down-regulated in AB-induced cardiac hypertrophy mice and Ang II -induced hypertrophic primary cardiomyocytes. The hypertrophic responses and fibrosis were evaluated by echocardiography and histological analysis. The mRNA levels of hypertrophy markers and fibrotic markers were detected by RT-qPCR. Furthermore, the phosphorylation and total levels of c-Jun were measured by western blotting. Results ZNF418 was markedly down-regulated in hearts of cardiac hypertrophy and hypertrophic primary cardiomyocytes. Down-regulated ZNF418 exacerbated the myocyte size and fibrosis, moreover increased the mRNA levels of ANP, BNP, β-MHC, MCIP1.4, collagen 1a, collagen III, MMP-2 and fibronection in hearts of AB-treated ZNF418 knockout mice or Ang II-treated cardiomyocytes with AdshZNF418. Conversely, these hypertrophic responses were reduced in the ZNF418 transgenic (TG) mice treated by AB and the AdZNF418-transfected primary cardiomyocytes treated by Ang II. Additionally, the deficiency of ZNF418 enhanced the phosphorylation level of c-jun, and overexpression of ZNF418 suppressed the phosphorylation level of c-jun in vivo and in vitro. Conclusion ZNF418 maybe attenuate hypertrophic responses by inhibiting the activity of c-jun/AP-1.
Collapse
Affiliation(s)
- Liming Pan
- Department of Cardiology, the People’s Hospital of Three Gorges University/the First People’s Hospital of Yichang, Yichang, China
| | - Mengting Sheng
- Department of Intensive Care Unit(ICU), the People’s Hospital of Three Gorges University/the First People’s Hospital of Yichang, Yichang, China
| | - Zirui Huang
- Department of Cardiology, the People’s Hospital of Three Gorges University/the First People’s Hospital of Yichang, Yichang, China
| | - Zhilin Zhu
- Department of Cardiology, the People’s Hospital of Three Gorges University/the First People’s Hospital of Yichang, Yichang, China
| | - Chunli Xu
- Department of Inspection office, the People’s Hospital of Three Gorges University/the First People’s Hospital of Yichang, Yichang, China
| | - Lin Teng
- Department of Cardiology, the First College of Clinical Medical Sciences of Three Gorges University/ Central People's Hospital of Yichang, Yichang, China
| | - Ling He
- Department of Geriatrics, the People’s Hospital of Three Gorges University/the First People’s Hospital of Yichang, Yichang, China
| | - Chen Gu
- Department of B ultrasound room, the People’s Hospital of Three Gorges University/the First People’s Hospital of Yichang, Yichang, China
| | - Cai Yi
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, China
| | - Junming Li
- Department of Cardiology, the People’s Hospital of Three Gorges University/the First People’s Hospital of Yichang, Yichang, China
- * E-mail:
| |
Collapse
|
10
|
Regulation of podoplanin expression by microRNA-29b associates with its antiapoptotic effect in angiotensin II-induced injury of human podocytes. J Hypertens 2016; 34:323-31. [PMID: 26867059 DOI: 10.1097/hjh.0000000000000799] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Angiotensin (Ang)II is involved in induction of proteinuria, renal injury, and apoptosis and thus a major contributor to the development of chronic kidney disease. Podocytes are of major importance for the pathogenesis of several kidney diseases. Decrease of podoplanin (PDPN) in podocytes and podocyte loss has been associated with the development of proteinuria. Little is known about the regulation and biological function of PDPN in podocytes and its role in AngII-mediated kidney damage. Here, we determined the influence of AngII on the expression of PDPN, microRNA (miRNA)-29b and miRNA-497 in human podocytes. Further, we analyzed the impact of small interfering RNA-mediated downregulation of PDPN on AngII-induced apoptosis and viability. Moreover, we characterized the role of miRNA-29b and miRNA-497 in expression regulation of PDPN. METHODS Cell viability and apoptosis were determined by functional assays. Expression analyses were done via Real-Time PCR and western blot analyses. Dual luciferase assay was performed to characterize miRNA-mediated expression control. RESULTS AngII increased the expression of miRNA-29b and reduced PDPN. Small interfering RNA-mediated downregulation of PDPN increased proapoptotic caspase-3 activation and cytochrome C translocation, whereas cell viability and Akt phosphorylation were reduced in AngII-stimulated podocytes. In contrast to miRNA-497, transfection of cells with miRNA-29b mimics significantly decreased PDPN. Cotransfection of cells with miRNA-29b and a dual luciferase reporter vector decreased the luciferase activity compared with controls. CONCLUSION These data demonstrate the posttranscriptional control of PDPN expression by miRNA-29b and support a role of PDPN as an antiapoptotic prosurvival factor in AngII-induced injury of human podocytes.
Collapse
|
11
|
Bytyçi I, Bajraktari G. Left atrial changes in early stages of heart failure with preserved ejection fraction. Echocardiography 2016; 33:1479-1487. [PMID: 27471047 DOI: 10.1111/echo.13306] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND AIM Increased left atrial (LA) mass was introduced as a compensatory mechanism in heart failure (HF) patients. Furthermore, atrial conduction time and LA emptying fraction is are deteriorated in HF with preserved ejection fraction (HFpEF). The aim of this study was to assess the early LA changes in HFpEF patients. METHODS In 79 consecutive patients with HFpEF (age 61±8 years, NYHA class I-III, LV EF ≥45%), a complete 2-dimensional, M-mode, and Doppler echocardiographic study was performed. According to the diastolic dysfunction (DD), patients were divided into three groups: Group I-29 healthy subjects (control group); Group II-HFpEF patients with mild DD; and Group III-HFpEF patients with moderate DD. RESULTS The LV mass was increased (P<.05), septal s', lateral s', septal and lateral MAPSE were decreased (P<.05, for all), E/e' ratio was increased (P<.001), LA mass and minimal volume were increased (P<.001, P<.05), LA emptying fraction was decreased (P<.05), and LA dyssynchrony was deteriorated (P<.05) in patients with mild DD compared to controls. These changes were of the same nature in patients with moderate LV DD. CONCLUSIONS In early stage of DD, in patients with HFpEF, in addition to LV hypertrophy and compromised LV longitudinal systolic function, the LA emptying fraction is reduced, LA mass and LAV min are increased and LA dyssynchrony is significant, despite normal LA dimensions. These findings suggest early LA function deterioration irrespective of normal cavity measurements, hence a need for optimum therapy.
Collapse
Affiliation(s)
- Ibadete Bytyçi
- Clinic of Cardiology, University Clinical Centre of Kosova, Prishtina, Republic of Kosovo.
| | - Gani Bajraktari
- Clinic of Cardiology, University Clinical Centre of Kosova, Prishtina, Republic of Kosovo
- Medical Faculty, University of Prishtina, Prishtina, Republic of Kosovo
| |
Collapse
|
12
|
Berthiaume J, Kirk J, Ranek M, Lyon R, Sheikh F, Jensen B, Hoit B, Butany J, Tolend M, Rao V, Willis M. Pathophysiology of Heart Failure and an Overview of Therapies. Cardiovasc Pathol 2016. [DOI: 10.1016/b978-0-12-420219-1.00008-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
13
|
Xu N, Guan S, Chen Z, Yu Y, Xie J, Pan FY, Zhao NW, Liu L, Yang ZZ, Gao X, Xu B, Li CJ. The alteration of protein prenylation induces cardiomyocyte hypertrophy through Rheb-mTORC1 signalling and leads to chronic heart failure. J Pathol 2015; 235:672-85. [DOI: 10.1002/path.4480] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 10/25/2014] [Accepted: 11/05/2014] [Indexed: 12/16/2022]
Affiliation(s)
- Na Xu
- Ministry of Education Key Laboratory of Model Animals for Disease Study; Model Animal Research Centre and Medical School of Nanjing University, National Resource Centre for Mutant Mice; Nanjing People's Republic of China
| | - Shan Guan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology; College of Life Science, Nanjing Normal University; Nanjing People's Republic of China
| | - Zhong Chen
- Ministry of Education Key Laboratory of Model Animals for Disease Study; Model Animal Research Centre and Medical School of Nanjing University, National Resource Centre for Mutant Mice; Nanjing People's Republic of China
| | - Yang Yu
- State Key Laboratory of Reproductive Biology; Institute of Zoology/Chinese Academy of Sciences; Beijing People's Republic of China
| | - Jun Xie
- Department of Cardiology; Affiliated Drum Tower Hospital of Nanjing University Medical School; Nanjing People's Republic of China
| | - Fei-Yan Pan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology; College of Life Science, Nanjing Normal University; Nanjing People's Republic of China
| | - Ning-Wei Zhao
- Biomedical Research Laboratory; Shimadzu (China) Co. Ltd; Shanghai People's Republic of China
| | - Li Liu
- Department of Geriatrics; First Affiliated Hospital with Nanjing Medical University; Nanjing People's Republic of China
| | - Zhong-Zhou Yang
- Ministry of Education Key Laboratory of Model Animals for Disease Study; Model Animal Research Centre and Medical School of Nanjing University, National Resource Centre for Mutant Mice; Nanjing People's Republic of China
| | - Xiang Gao
- Ministry of Education Key Laboratory of Model Animals for Disease Study; Model Animal Research Centre and Medical School of Nanjing University, National Resource Centre for Mutant Mice; Nanjing People's Republic of China
| | - Biao Xu
- Department of Cardiology; Affiliated Drum Tower Hospital of Nanjing University Medical School; Nanjing People's Republic of China
| | - Chao-Jun Li
- Ministry of Education Key Laboratory of Model Animals for Disease Study; Model Animal Research Centre and Medical School of Nanjing University, National Resource Centre for Mutant Mice; Nanjing People's Republic of China
| |
Collapse
|
14
|
Cisapride protects against cardiac hypertrophy via inhibiting the up-regulation of calcineurin and NFATc-3. Eur J Pharmacol 2014; 735:202-10. [DOI: 10.1016/j.ejphar.2014.04.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 04/06/2014] [Accepted: 04/09/2014] [Indexed: 11/23/2022]
|
15
|
Li C, Li X, Gao X, Zhang R, Zhang Y, Liang H, Xu C, Du W, Zhang Y, Liu X, Ma N, Xu Z, Wang L, Chen X, Lu Y, Ju J, Yang B, Shan H. MicroRNA-328 as a regulator of cardiac hypertrophy. Int J Cardiol 2014; 173:268-76. [DOI: 10.1016/j.ijcard.2014.02.035] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 01/22/2014] [Accepted: 02/22/2014] [Indexed: 01/23/2023]
|
16
|
Saks V, Schlattner U, Tokarska-Schlattner M, Wallimann T, Bagur R, Zorman S, Pelosse M, Santos PD, Boucher F, Kaambre T, Guzun R. Systems Level Regulation of Cardiac Energy Fluxes Via Metabolic Cycles: Role of Creatine, Phosphotransfer Pathways, and AMPK Signaling. SYSTEMS BIOLOGY OF METABOLIC AND SIGNALING NETWORKS 2014. [DOI: 10.1007/978-3-642-38505-6_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
Nardosinone protects H9c2 cardiac cells from angiotensin II-induced hypertrophy. ACTA ACUST UNITED AC 2013; 33:822-826. [DOI: 10.1007/s11596-013-1205-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 09/12/2013] [Indexed: 10/25/2022]
|
18
|
Intermedin suppresses pressure overload cardiac hypertrophy through activation of autophagy. PLoS One 2013; 8:e64757. [PMID: 23737997 PMCID: PMC3667197 DOI: 10.1371/journal.pone.0064757] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Accepted: 04/18/2013] [Indexed: 02/05/2023] Open
Abstract
Left ventricular hypertrophy is a maladaptive response to pressure overload and an important risk factor for heart failure. Intermedin (IMD), a multi-functional peptide, plays important roles in cardiovascular protection. In this study, we revealed an autophagy-dependent mechanism involved in IMD’s protection against cardiac remodeling and cardiomyocyte death in heart hypertrophy. We observed that transverse aortic contraction (TAC) induction, Ang II or ISO exposure induced remarkable increase in the expression of endogenous IMD and its receptor components, CRLR, RAMP1 and RAMP3, in mouse hearts and H9c2 cell cultures, respectively. Furthermore, the heart size, heart weight/body weight ratios, cardiomyocyte size and apoptosis, interstitial collagen, hypertrophic markers including ANP and BNP expression were also significantly increased, which were effectively suppressed by IMD supplementation. In addition, IMD induced capillary angiogenesis and improved functions in hypertrophic hearts. We further observed that IMD induced strong autophagy in hypertrophic hearts and cultured cells, which was paralleling with the decrease in cardiomyocyte size and apoptosis. Furthermore, an autophagy inhibitor, 3-MA, was used to block the IMD-augmented autophagy level, and then the protection of IMD on cardiomyocyte hypertrophy and apoptosis was almost abrogated. We also observed that IMD supplementation stirred intracellular cAMP production, and augmented the ERK1/2 phosphorylation induced by Ang II/ISO exposure in H9c2 cells. In addition, we inhibited PI3K, PKA and MAPK/ERK1/2 signaling pathways by using wortamannin, H89 and PD98059, respectively, in H9c2 cells co-incubating with both IMD and Ang II or ISO, and observed that these inhibitors effectively reduced IMD-augmented autophagy level, but only H89 and PD98059 pre-incubation abrogated the anti-apoptotic action of IMD. These results indicate that the endogenous IMD and its receptor complexes are induced in hypertrophic cardiomyocytes and proposed to play an important role in the pathogenesis of cardiac hypertrophy, and the autophagy stirred by IMD supplementation is involved in its protection against cardiomyocyte hypertrophy and apoptosis through the activation of both cAMP/PKA and MAPK/ERK1/2 pathways.
Collapse
|
19
|
Deng W, Zong J, Bian Z, Zhou H, Yuan Y, Zhang R, Guo H, Zhang Y, Shen D, Li H, Tang Q. Indole-3-carbinol protects against pressure overload induced cardiac remodeling via activating AMPK-α. Mol Nutr Food Res 2013; 57:1680-7. [PMID: 23625645 DOI: 10.1002/mnfr.201300012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Revised: 02/18/2013] [Accepted: 02/19/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Wei Deng
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Jing Zong
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Zhouyan Bian
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Heng Zhou
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Yuan Yuan
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Rui Zhang
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Haipeng Guo
- The Key Laboratory of Cardiovascular Remodeling and Function Research; Chinese Ministry of Education and Chinese Ministry of Health; Qilu Hospital of Shandong University; Jinan P. R. China
| | - Yan Zhang
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Difei Shen
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Hongliang Li
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Qizhu Tang
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| |
Collapse
|
20
|
Zong J, Deng W, Zhou H, Bian ZY, Dai J, Yuan Y, Zhang JY, Zhang R, Zhang Y, Wu QQ, Guo HP, Li HL, Tang QZ. 3,3'-Diindolylmethane protects against cardiac hypertrophy via 5'-adenosine monophosphate-activated protein kinase-α2. PLoS One 2013; 8:e53427. [PMID: 23326427 PMCID: PMC3541184 DOI: 10.1371/journal.pone.0053427] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 11/29/2012] [Indexed: 11/25/2022] Open
Abstract
Purpose 3,3′-Diindolylmethane (DIM) is a natural component of cruciferous plants. It has strong antioxidant and anti-angiogenic effects and promotes the apoptosis of a variety of tumor cells. However, little is known about the critical role of DIM on cardiac hypertrophy. In the present study, we investigated the effects of DIM on cardiac hypertrophy. Methods Multiple molecular techniques such as Western blot analysis, real-time PCR to determine RNA expression levels of hypertrophic, fibrotic and oxidative stress markers, and histological analysis including H&E for histopathology, PSR for collagen deposition, WGA for myocyte cross-sectional area, and immunohistochemical staining for protein expression were used. Results In pre-treatment and reverse experiments, C57/BL6 mouse chow containing 0.05% DIM (dose 100 mg/kg/d DIM) was administered one week prior to surgery or one week after surgery, respectively, and continued for 8 weeks after surgery. In both experiments, DIM reduced to cardiac hypertrophy and fibrosis induced by aortic banding through the activation of 5′-adenosine monophosphate-activated protein kinase-α2 (AMPKα2) and inhibition of mammalian target of the rapamycin (mTOR) signaling pathway. Furthermore, DIM protected against cardiac oxidative stress by regulating expression of estrogen-related receptor-alpha (ERRα) and NRF2 etc. The cardioprotective effects of DIM were ablated in mice lacking functional AMPKα2. Conclusion DIM significantly improves left ventricular function via the activation of AMPKα2 in a murine model of cardiac hypertrophy.
Collapse
Affiliation(s)
- Jing Zong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Heng Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Zhou-yan Bian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Jia Dai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Yuan Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Jie-yu Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Rui Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Yan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Qing-qing Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Hai-peng Guo
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China
| | - Hong-liang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Qi-zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- * E-mail:
| |
Collapse
|
21
|
Rajapurohitam V, Izaddoustdar F, Martinez-Abundis E, Karmazyn M. Leptin-induced Cardiomyocyte Hypertrophy Reveals both Calcium-dependent and Calcium-independent/RhoA-dependent Calcineurin Activation and NFAT Nuclear Translocation. Cell Signal 2012; 24:2283-90. [DOI: 10.1016/j.cellsig.2012.07.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 07/13/2012] [Accepted: 07/24/2012] [Indexed: 12/29/2022]
|
22
|
Ling S, Sun Q, Li Y, Zhang L, Zhang P, Wang X, Tian C, Li Q, Song J, Liu H, Kan G, Cao H, Huang Z, Nie J, Bai Y, Chen S, Li Y, He F, Zhang L, Li Y. CKIP-1 inhibits cardiac hypertrophy by regulating class II histone deacetylase phosphorylation through recruiting PP2A. Circulation 2012; 126:3028-40. [PMID: 23151343 DOI: 10.1161/circulationaha.112.102780] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Sustained cardiac pressure overload-induced hypertrophy and pathological remodeling frequently leads to heart failure. Casein kinase-2 interacting protein-1 (CKIP-1) has been identified to be an important regulator of cell proliferation, differentiation, and apoptosis. However, the physiological role of CKIP-1 in the heart is unknown. METHODS AND RESULTS The results of echocardiography and histology demonstrate that CKIP-1-deficient mice exhibit spontaneous cardiac hypertrophy with aging and hypersensitivity to pressure overload-induced pathological cardiac hypertrophy, as well. Transgenic mice with cardiac-specific overexpression of CKIP-1 showed resistance to cardiac hypertrophy in response to pressure overload. The results of GST pull-down and coimmunoprecipitation assays showed the interaction between CKIP-1 and histone deacetylase 4 (HDAC4), through which they synergistically inhibited transcriptional activity of myocyte-specific enhancer factor 2C. By directly interacting with the catalytic subunit of phosphatase 2A, CKIP-1 overexpression enhanced the binding of catalytic subunit of phosphatase-2A to HDAC4 and promoted HDAC4 dephosphorylation. CONCLUSIONS CKIP-1 was found to be an inhibitor of cardiac hypertrophy by upregulating the dephosphorylation of HDAC4 through the recruitment of protein phosphatase 2A. These results demonstrated a unique function of CKIP-1, by which it suppresses cardiac hypertrophy through its capacity to regulate HDAC4 dephosphorylation and fetal cardiac genes expression.
Collapse
Affiliation(s)
- Shukuan Ling
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Haidian District, Beiqing Rd, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Hinojosa CA, Mgbemena V, Van Roekel S, Austad SN, Miller RA, Bose S, Orihuela CJ. Enteric-delivered rapamycin enhances resistance of aged mice to pneumococcal pneumonia through reduced cellular senescence. Exp Gerontol 2012; 47:958-65. [PMID: 22981852 DOI: 10.1016/j.exger.2012.08.013] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 08/06/2012] [Accepted: 08/30/2012] [Indexed: 11/18/2022]
Abstract
Rapamycin, a potent immunomodulatory drug, has shown promise in the amelioration of numerous age-associated diseases including cancer, Alzheimer's disease and cardiac hypertrophy. Yet the elderly, the population most likely to receive therapeutic rapamycin, are already at increased risk for infectious disease; thus concern exists that rapamycin may exacerbate age-associated immune dysfunctions and worsen infection outcomes. Herein, we examined the impact of enteric delivered rapamycin monotherapy (eRapa) on the susceptibility of aged (22-24month) C57BL/6 mice to Streptococcus pneumoniae, the leading bacterial cause of community-acquired pneumonia. Following challenge with S. pneumoniae, administration of eRapa conferred modest protection against mortality. Reduced mortality was the result of diminished lung damage rather than reduced bacterial burden. eRapa had no effect on basal levels of Interleukin (IL)-1α, IL-6, IL-10, IL-12p70, KC, Interferon-γ, Tumor necrosis factor α and Monocyte chemotactic protein-1 in whole lung homogenates or during pneumococcal pneumonia. Previously we have demonstrated that cellular senescence enhances permissiveness for bacterial pneumonia through increased expression of the bacterial ligands Laminin receptor (LR), Platelet-activating factor receptor (PAFr) and Cytokeratin 10 (K10). These proteins are co-opted by S. pneumoniae and other respiratory tract pathogens for host cell attachment during lung infection. UM-HET3 mice on eRapa had reduced lung cellular senescence as determined by levels of the senescence markers p21 and pRB, but not mH2A.1. Mice on eRapa also had marked reductions in PAFr, LR, and K10. We conclude that eRapa protected aged mice against pneumonia through reduced lung cellular senescence, which in turn, lowered bacterial ligand expression.
Collapse
Affiliation(s)
- Cecilia A Hinojosa
- Department of Microbiology & Immunology, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, United States
| | | | | | | | | | | | | |
Collapse
|
24
|
Lee HJ, Rho J, Gui SR, Kim MK, Lee YK, Lee YS, Kim JE, Cho E, Cho M, Hwang TH. Effect of aldosterone on the amplification of oncolytic vaccinia virus in human cancer lines. THE KOREAN JOURNAL OF HEPATOLOGY 2012; 17:213-9. [PMID: 22102388 PMCID: PMC3304654 DOI: 10.3350/kjhep.2011.17.3.213] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND/AIMS JX-594 is an oncolytic virus derived from the Wyeth vaccinia strain that causes replication-dependent cytolysis and antitumor immunity. Starting with a cross-examination of clinical-trial samples from advanced hepatocellular carcinoma patients having high levels of aldosterone and virus amplification in JX-594 treatment, we investigated the association between virus amplification and aldosterone in human cancer cell lines. METHODS Cell proliferation was determined by a cell-counting-kit-based colorimetric assay, and vaccinia virus quantitation was performed by quantitative polymerase chain reaction (qPCR) and a viral plaque assay. Also, the intracellular pH was measured using a pH-sensitive dye. RESULTS Simultaneous treatment with JX-594 and aldosterone significantly increased viral replication in A2780, PC-3, and HepG2 cell lines, but not in U2OS cell lines. Furthermore, the aldosterone treatment time altered the JX-594 replication according to the cell line. The JX-594 replication peaked after 48 and 24 hours of treatment in PC-3 and HepG2 cells, respectively. qPCR showed that JX-594 entry across the plasma membrane was increased, however, the changes are not significant by the treatment. This was inhibited by treatment with spironolactone (an aldosterone-receptor inhibitor). JX-594 entry was significantly decreased by treatment with EIPA [5-(N-ethyl-N-isopropyl)amiloride; a Na(+)/H(+)-exchange inhibitor], but aldosterone significantly restored JX-594 entry even in the presence of EIPA. Intracellular alkalization was observed after aldosterone treatment but was acidified by EIPA treatment. CONCLUSIONS Aldosterone stimulates JX-594 amplification via increased virus entry by affecting the H(+) gradient.
Collapse
Affiliation(s)
- Hyun Ju Lee
- Department of Pharmacology, Pusan National University School of Korean Medicine, Busan, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
PPARα activation inhibits endothelin-1-induced cardiomyocyte hypertrophy by prevention of NFATc4 binding to GATA-4. Arch Biochem Biophys 2011; 518:71-8. [PMID: 22198280 DOI: 10.1016/j.abb.2011.11.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Revised: 11/13/2011] [Accepted: 11/14/2011] [Indexed: 11/21/2022]
Abstract
Peroxisome proliferator-activated receptor alpha (PPARα) has been implicated in the pathogenesis of cardiac hypertrophy, although its mechanism of action remains largely unknown. To determine the effect of PPARα activation on endothelin-1 (ET-1)-induced cardiomyocyte hypertrophy and explore its molecular mechanisms, we evaluated the interaction of PPARα with nuclear factor of activated T-cells c4 (NFATc4) in nuclei of cardiomyocytes from neonatal rats in primary culture. In ET-1-stimulated cardiomyocytes, data from electrophoretic mobility-shift assays (EMSA) and co-immunoprecipitation (co-IP) revealed that fenofibrate (Fen), a PPARα activator, in a concentration-dependent manner, enhanced the association of NFATc4 with PPARα and decreased its interaction with GATA-4, in promoter complexes involved in activation of the rat brain natriuretic peptide (rBNP) gene. Effects of PPARα overexpression were similar to those of its activation by Fen. PPARα depletion by small interfering RNA abolished inhibitory effects of Fen on NFATc4 binding to GATA-4 and the rBNP DNA. Quantitative RT-PCR and confocal microscopy confirmed inhibitory effects of PPARα activation on elevation of rBNP mRNA levels and ET-1-induced cardiomyocyte hypertrophy. Our results suggest that activated PPARα can compete with GATA-4 binding to NFATc4, thereby decreasing transactivation of NFATc4, and interfering with ET-1 induced cardiomyocyte hypertrophy.
Collapse
|
26
|
New Targets to Treat the Structural Remodeling of the Myocardium. J Am Coll Cardiol 2011; 58:1833-43. [DOI: 10.1016/j.jacc.2011.06.058] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 06/21/2011] [Indexed: 11/20/2022]
|
27
|
Gastrodin protects against cardiac hypertrophy and fibrosis. Mol Cell Biochem 2011; 359:9-16. [DOI: 10.1007/s11010-011-0992-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2011] [Accepted: 07/06/2011] [Indexed: 10/18/2022]
|