1
|
Wu SJ, He RL, Zhao L, Yu XY, Jiang YN, Guan X, Chen QY, Ren FF, Xie ZY, Wu LP, Li L. Cardiac-Specific Overexpression of Caveolin-1 in Rats With Ischemic Cardiomyopathy Improves Arrhythmogenicity and Cardiac Remodelling. Can J Cardiol 2023; 39:73-86. [PMID: 36240973 DOI: 10.1016/j.cjca.2022.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/16/2022] [Accepted: 10/04/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Ischemic cardiomyopathy (ICM) is associated with electrical and structural remodelling, leading to arrhythmias. Caveolin-1 (Cav1) is a membrane protein involved in the pathogenesis of ischemic injury. Cav1 deficiency has been associated with arrhythmogenicity. The current study aimed to determine how Cav1 overexpression inhibits arrhythmias and cardiac remodelling in ICM. METHODS ICM was modelled using left anterior descending (LAD) artery ligation for 4 weeks. Cardiac-specific Cav1 overexpression in ICM on arrhythmias, excitation-contraction coupling, and cardiac remodelling were investigated using the intramyocardial injection of an adeno-associated virus serotype 9 (AAV-9) system, carrying a specific sequence expressing Cav1 (AAVCav1) under the cardiac troponin T (cTnT) promoter. RESULTS Cav1 overexpression decreased susceptibility to arrhythmias by upregulating gap junction connexin 43 (CX43) and reducing spontaneous irregular proarrhythmogenic Ca2+ waves in ventricular cardiomyocytes. It also alleviated ischemic injury-induced contractility weakness by improving Ca2+ cycling through normalizing Ca2+-handling protein levels and improving Ca2+ homeostasis. Masson stain and immunoblotting revealed that the deposition of excessive fibrosis was attenuated by Cav1 overexpression, inhibiting the transforming growth factor-β (TGF-β)/Smad2 signalling pathway. Coimmunoprecipitation assays demonstrated that the interaction between Cav1 and cSrc modulated CX43 expression and Ca2+-handling protein levels. CONCLUSIONS Cardiac-specific overexpression of Cav1 attenuated ventricular arrhythmia, improved Ca2+ cycling, and attenuated cardiac remodelling. These effects were attributed to modulation of CX43, normalized Ca2+-handling protein levels, improved Ca2+ homeostasis, and attenuated cardiac fibrosis.
Collapse
Affiliation(s)
- Shu-Jie Wu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Rui-Lin He
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Lin Zhao
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Xiao-Yu Yu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Yi-Na Jiang
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Xuan Guan
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Qiao-Ying Chen
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Fang-Fang Ren
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Zuo-Yi Xie
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Lian-Pin Wu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Lei Li
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China.
| |
Collapse
|
2
|
Gkrouzoudi A, Tsingotjidou A, Jirkof P. A systematic review on the reporting quality in mouse telemetry implantation surgery using electrocardiogram recording devices. Physiol Behav 2021; 244:113645. [PMID: 34774869 DOI: 10.1016/j.physbeh.2021.113645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/26/2021] [Accepted: 11/08/2021] [Indexed: 01/31/2023]
Abstract
Telemetric monitoring is used in many scientific fields, such as cardiovascular research, neurology, endocrinology, animal welfare research and many more. Nowadays, implanted electrocardiogram (ECG) radiotelemetry units are the gold standard for monitoring ECG traces, heart rate and heart rate variability in freely moving mice. This technology can be a valuable tool when studies utilise it adequately, while also prioritizing animal welfare. Recently, concerns about the reproducibility of research findings have been raised in many scientific fields with insufficient reporting being one of the underlying causes. A systematic review was performed in three literature databases to include all published studies until 31.12.2019 using surgery that involves the placing of ECG recording telemetry devices in adult mice. Data extracted from the publications included selected items recommended by the ARRIVE guidelines and SYRCLE`s tool for assessing risk of bias. We focused on aspects related to quality of reporting, risk of bias reduction measures and ECG measurements characteristics. In general, the quality of reporting was low to moderate in the 234 analyzed publications regarding the animal, husbandry, statistics, and risk of bias related items, but good for more specific telemetry study characteristics. Based on our analyses we assume that there is no or only slight improvement in the reporting quality since 2010, when the ARRIVE guidelines were published.
Collapse
Affiliation(s)
- Alexandra Gkrouzoudi
- Laboratory of Anatomy, Histology and Embryology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Greece
| | - Anastasia Tsingotjidou
- Laboratory of Anatomy, Histology and Embryology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Greece
| | - Paulin Jirkof
- Division for Surgical Research, University Hospital Zurich, University Zurich, Zurich, Switzerland; Office of Animal Welfare and 3Rs, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
3
|
Polylactide Nanocapsules Attenuate Adverse Cardiac Cellular Effects of Lyso-7, a Pan-PPAR Agonist/Anti-Inflammatory New Thiazolidinedione. Pharmaceutics 2021; 13:pharmaceutics13091521. [PMID: 34575597 PMCID: PMC8470374 DOI: 10.3390/pharmaceutics13091521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 11/17/2022] Open
Abstract
Lyso-7 is a novel synthetic thiazolidinedione, which is a receptor (pan) agonist of PPAR α,β/δ,γ with anti-inflammatory activity. We investigated the cardiotoxicity of free Lyso-7 in vitro (4.5–450 nM), and Lyso-7 loaded in polylactic acid nanocapsules (NC) in vivo (Lyso-7-NC, 1.6 mg/kg). In previous work, we characterized Lyso-7-NC. We administered intravenously Lyso-7, Lyso-7-NC, control, and blank-NC once a day for seven days in mice. We assessed cell contraction and intracellular Ca2+ transients on single mice cardiomyocytes enzymatically isolated. Lyso-7 reduced cell contraction and accelerated relaxation while lowering diastolic Ca2+ and reducing Ca2+ transient amplitude. Lyso-7 also promoted abnormal ectopic diastolic Ca2+ events, which isoproterenol dramatically enhanced. Incorporation of Lyso-7 in NC attenuated drug effects on cell contraction and prevented its impact on relaxation, diastolic Ca2+, Ca2+ transient amplitude, Ca2+ transient decay kinetics, and promotion of diastolic Ca2+ events. Acute effects of Lyso-7 on cardiomyocytes in vitro at high concentrations (450 nM) were globally similar to those observed after repeated administration in vivo. In conclusion, we show evidence for off-target effects of Lyso-7, seen during acute exposure of cardiomyocytes to high concentrations and after repeated treatment in mice. Nano-encapsulation of Lyso-7 in polymeric NC attenuated the unwanted effects, particularly ectopic Ca2+ events known to support life-threatening arrhythmias favored by stress or exercise.
Collapse
|
4
|
Chalikias G, Kikas P, Thomaidis A, Rigopoulos P, Pistola A, Lantzouraki A, Zisimopoulos A, Tziakas D. Effect of Sacubitril/Valsartan on circulating catecholamine levels during a 6-month follow-up in heart failure patients. Timeo Danaos et dona ferentes? Acta Cardiol 2021; 76:396-401. [PMID: 32223369 DOI: 10.1080/00015385.2020.1746094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We assessed the effect of Sacubitril/Valsartan on circulating catecholamine levels in patients with HF in an observational cohort study. We included 108 consecutive HF patients attending our HF Outpatients Clinic who were eligible to Sacubitril/Valsartan according to the PARADIGM-HF inclusion and exclusion criteria. We furthermore included 58 stable HF patients under optimal medical therapy as a control group. Norepinephrine and epinephrine were measured with immunoradiometric assays at baseline, at 3- and at 6-month time follow-up. Compared to baseline levels there was no change at three months in epinephrine (p = 0.177) or norepinephrine (p = 0.815) concentrations. At 6 months norepinephrine remained unchanged (p = 0.359). However, at 6 months we observed a significant increase in epinephrine levels compared to baseline [66 pg/mL (37-93) vs 38 pg/mL (18-74), p < 0.001]. In the control group no change was observed in epinephrine levels compared to baseline (p = 0.838). This study is the first to report on the effect of the new drug Sacubitril/Valsartan on circulating catecholamine levels in HF patients. Our data show a significant increase in epinephrine levels during a 6 month follow up in stable HF patients.
Collapse
Affiliation(s)
- George Chalikias
- Cardiology Department, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Petros Kikas
- Cardiology Department, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Adina Thomaidis
- Cardiology Department, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Panagiotis Rigopoulos
- Cardiology Department, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Anastasia Pistola
- Nuclear Medicine Department, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Asimina Lantzouraki
- Cardiology Department, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Athanasios Zisimopoulos
- Nuclear Medicine Department, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Dimitrios Tziakas
- Cardiology Department, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
5
|
Zhang Z, Fang Q, Du T, Chen G, Wang Y, Wang DW. Cardiac-Specific Caveolin-3 Overexpression Prevents Post-Myocardial Infarction Ventricular Arrhythmias by Inhibiting Ryanodine Receptor-2 Hyperphosphorylation. Cardiology 2020; 145:136-147. [PMID: 32007997 DOI: 10.1159/000505316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 12/05/2019] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Ventricular arrhythmia is the most important risk factor for sudden cardiac death (SCD) after acute myocardial infarction (MI) worldwide. However, the molecular mechanisms underlying these arrhythmias are complex and not completely understood. OBJECTIVE Here, we evaluated whether caveolin-3 (Cav3), the structural protein of caveolae, plays an important role in the therapeutic strategy for ventricular arrhythmias. METHODS A model of cardiac-specific overexpression of Cav3 was established to evaluate the incidence of ventricular arrhythmias after MI in mice. Ca2+ imaging was employed to detect the propensity of adult murine cardiomyocytes to generate arrhythmias, and immunoprecipitation and immunofluorescence were used to determine the relationship of proteins. Additionally, qRT-PCR and western blotting were used to detect the mRNA and protein expression. RESULTS We found that cardiac-specific overexpression of Cav3 delivered by a recombinant adeno-associated viral vector reduced the incidence of ventricular arrhythmias and SCD after MI in mice. Ca2+ imaging and western blotting revealed that overexpression of Cav3 reduced diastolic spontaneous Ca2+ waves by inhibiting the hyperphosphorylation of ryanodine receptor-2 (RyR2) at Ser2814, rather than at Ser2808, compared to in rAAV-red fluorescent protein control mice. Furthermore, we demonstrated that Cav3-regulated RYR2 hyperphosphorylation relied on plakophilin-2 in hypoxia-stimulated cultured cardiomyocytes by western blotting, immunoprecipitation, and immunofluorescence in vitro. CONCLUSIONS Our results suggested a novel role for Cav3 in the prevention of ventricular arrhythmias, thereby identifying a new target for preventing SCD after MI.
Collapse
Affiliation(s)
- Zhihao Zhang
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Fang
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingyi Du
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guangzhi Chen
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,
| | - Yan Wang
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Zhao J, Xu T, Zhou Y, Zhou Y, Xia Y, Li D. B-type natriuretic peptide and its role in altering Ca 2+-regulatory proteins in heart failure-mechanistic insights. Heart Fail Rev 2019; 25:861-871. [PMID: 31820203 DOI: 10.1007/s10741-019-09883-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Heart failure (HF) is a worldwide disease with high levels of morbidity and mortality. The pathogenesis of HF is complicated and involves imbalances in hormone and electrolyte. B-type natriuretic peptide (BNP) has served as a biomarker of HF severity, and in recent years, it has been used to treat the disease, thanks to its cardio-protective effects, such as diuresis, natriuresis, and vasodilatation. In stage C/D HF, symptoms are severe despite elevated BNP. Disturbances in Ca2+ homeostasis are often a dominating feature of the disease, causing Ca2+-regulatory protein dysfunction, including reduced expression and activity of sarcoplasmic reticulum Ca2+-ATPase2a (SERCA2a), impaired ryanodine receptors (RYRs) function, intensive Na+-Ca2+ exchanger (NCX), and downregulation of S100A1. The relationship between natriuretic peptides (NPs) and Ca2+-regulatory proteins has been widely studied and represents important mechanisms in the etiology of HF. In this review, we present evidence that BNP may regulate Ca2+-regulatory proteins, in particular, suppressing SERCA2a and S100A1 expression. However, relationships between BNP and other Ca2+-regulatory proteins remain vague.
Collapse
Affiliation(s)
- Jiaqi Zhao
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Tongda Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Yao Zhou
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - You Zhou
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Yong Xia
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China. .,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
7
|
van Duijvenboden K, de Bakker DEM, Man JCK, Janssen R, Günthel M, Hill MC, Hooijkaas IB, van der Made I, van der Kraak PH, Vink A, Creemers EE, Martin JF, Barnett P, Bakkers J, Christoffels VM. Conserved NPPB+ Border Zone Switches From MEF2- to AP-1-Driven Gene Program. Circulation 2019; 140:864-879. [PMID: 31259610 DOI: 10.1161/circulationaha.118.038944] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Surviving cells in the postinfarction border zone are subjected to intense fluctuations of their microenvironment. Recently, border zone cardiomyocytes have been specifically implicated in cardiac regeneration. Here, we defined their unique transcriptional and regulatory properties, and comprehensively validated new molecular markers, including Nppb, encoding B-type natriuretic peptide, after infarction. METHODS Transgenic reporter mice were used to identify the Nppb-positive border zone after myocardial infarction. Transcriptome analysis of remote, border, and infarct zones and of purified cardiomyocyte nuclei was performed using RNA-sequencing. Top candidate genes displaying border zone spatial specificity were histologically validated in ischemic human hearts. Mice in which Nppb was deleted by genome editing were subjected to myocardial infarction. Chromatin accessibility landscapes of border zone and control cardiomyocyte nuclei were assessed by using assay for transposase-accessible chromatin using sequencing. RESULTS We identified the border zone as a spatially confined region transcriptionally distinct from the remote myocardium. The transcriptional response of the border zone was much stronger than that of the remote ventricular wall, involving acute downregulation of mitochondrial oxidative phosphorylation, fatty acid metabolism, calcium handling, and sarcomere function, and the activation of a stress-response program. Analysis of infarcted human hearts revealed that the transcriptionally discrete border zone is conserved in humans, and led to the identification of novel conserved border zone markers including NPPB, ANKRD1, DES, UCHL1, JUN, and FOXP1. Homozygous Nppb mutant mice developed acute and lethal heart failure after myocardial infarction, indicating that B-type natriuretic peptide is required to preserve postinfarct heart function. Assay for transposase-accessible chromatin using sequencing revealed thousands of cardiomyocyte lineage-specific MEF2-occupied regulatory elements that lost accessibility in the border zone. Putative injury-responsive enhancers that gained accessibility were highly associated with AP-1 (activator protein 1) binding sites. Nuclear c-Jun, a component of AP-1, was observed specifically in border zone cardiomyocytes. CONCLUSIONS Cardiomyocytes in a discrete zone bordering the infarct switch from a MEF2-driven homeostatic lineage-specific to an AP-1-driven injury-induced gene expression program. This program is conserved between mouse and human, and includes Nppb expression, which is required to prevent acute heart failure after infarction.
Collapse
Affiliation(s)
- Karel van Duijvenboden
- Departments of Medical Biology, Amsterdam Cardiovascular Sciences (K.v.D., J.C.K.M., R.J., M.G., I.B.H., P.B., V.M.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - Dennis E M de Bakker
- Hubrecht Institute (D.E.M.d.B., J.B.), University Medical Centre Utrecht, The Netherlands
| | - Joyce C K Man
- Departments of Medical Biology, Amsterdam Cardiovascular Sciences (K.v.D., J.C.K.M., R.J., M.G., I.B.H., P.B., V.M.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - Rob Janssen
- Departments of Medical Biology, Amsterdam Cardiovascular Sciences (K.v.D., J.C.K.M., R.J., M.G., I.B.H., P.B., V.M.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - Marie Günthel
- Departments of Medical Biology, Amsterdam Cardiovascular Sciences (K.v.D., J.C.K.M., R.J., M.G., I.B.H., P.B., V.M.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - Matthew C Hill
- Program in Developmental Biology (M.C.H., J.F.M.), Baylor College of Medicine, Houston, TX
| | - Ingeborg B Hooijkaas
- Departments of Medical Biology, Amsterdam Cardiovascular Sciences (K.v.D., J.C.K.M., R.J., M.G., I.B.H., P.B., V.M.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - Ingeborg van der Made
- Experimental Cardiology (I.v.d.M., E.E.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - Petra H van der Kraak
- Department of Pathology (P.H.v.d.K., A.V.), University Medical Centre Utrecht, The Netherlands
| | - Aryan Vink
- Department of Pathology (P.H.v.d.K., A.V.), University Medical Centre Utrecht, The Netherlands
| | - Esther E Creemers
- Experimental Cardiology (I.v.d.M., E.E.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - James F Martin
- Program in Developmental Biology (M.C.H., J.F.M.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology and Biophysics (J.F.M.), Baylor College of Medicine, Houston, TX
| | - Phil Barnett
- Departments of Medical Biology, Amsterdam Cardiovascular Sciences (K.v.D., J.C.K.M., R.J., M.G., I.B.H., P.B., V.M.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute (D.E.M.d.B., J.B.), University Medical Centre Utrecht, The Netherlands
| | - Vincent M Christoffels
- Departments of Medical Biology, Amsterdam Cardiovascular Sciences (K.v.D., J.C.K.M., R.J., M.G., I.B.H., P.B., V.M.C.), Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Natriuretic Peptide Receptor-C Protects Against Angiotensin II-Mediated Sinoatrial Node Disease in Mice. JACC Basic Transl Sci 2018; 3:824-843. [PMID: 30623142 PMCID: PMC6314975 DOI: 10.1016/j.jacbts.2018.08.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/15/2018] [Accepted: 08/16/2018] [Indexed: 12/19/2022]
Abstract
SAN disease is prevalent in hypertension and heart failure and can be induced by chronic Ang II treatment in mice. Ang II caused SAN disease in mice in association with impaired electrical conduction, reduction in the hyperpolarization-activated current (If) in SAN myocytes, and increased SAN fibrosis. Ang II-induced SAN disease was worsened in mice lacking NPR-C in association with enhanced SAN fibrosis. Mice co-treated with Ang II and an NPR-C agonist (cANF) were protected from SAN disease. NPR-C may represent a new target to protect against Ang II-induced SAN disease.
Sinoatrial node (SAN) disease mechanisms are poorly understood, and therapeutic options are limited. Natriuretic peptide(s) (NP) are cardioprotective hormones whose effects can be mediated partly by the NP receptor C (NPR-C). We investigated the role of NPR-C in angiotensin II (Ang II)-mediated SAN disease in mice. Ang II caused SAN disease due to impaired electrical activity in SAN myocytes and increased SAN fibrosis. Strikingly, Ang II treatment in NPR-C−/− mice worsened SAN disease, whereas co-treatment of wild-type mice with Ang II and a selective NPR-C agonist (cANF) prevented SAN dysfunction. NPR-C may represent a new target to protect against the development of Ang II-induced SAN disease.
Collapse
Key Words
- AP, action potential
- Ang II, angiotensin II
- CV, conduction velocity
- DD, diastolic depolarization
- Gmax, maximum conductance
- HR, heart rate
- ICa,L, L-type calcium current
- ICa,T, T-type calcium current
- INCX, sodium–calcium exchanger current
- IV, current voltage relationship
- If, hyperpolarization-activated current
- NP, natriuretic peptide
- NPR, natriuretic peptide receptor
- NPR-C, natriuretic peptide receptor C
- SAN, sinoatrial node
- SBP, systolic blood pressure
- V1/2(act), voltage for 50% channel activation
- cSNRT, corrected sinoatrial node recovery time
- fibrosis
- hypertension
- ion currents
- natriuretic peptide
- sinoatrial node
Collapse
|
9
|
Houde M, Schwertani A, Touil H, Desbiens L, Sarrhini O, Lecomte R, Lepage M, Gagnon H, Takai S, Pejler G, Jacques D, Gobeil F, Day R, D'Orléans-Juste P. Mouse Mast Cell Protease 4 Deletion Protects Heart Function and Survival After Permanent Myocardial Infarction. Front Pharmacol 2018; 9:868. [PMID: 30233357 PMCID: PMC6127244 DOI: 10.3389/fphar.2018.00868] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/17/2018] [Indexed: 01/08/2023] Open
Abstract
Chymase, a mast cell serine protease involved in the generation of multiple cardiovascular factors, such as angiotensin II and endothelin-1 (ET-1), is elevated and participates in tissue degeneration after permanent myocardial infarction (PMI). Anesthetized 4-month old male wild-type (WT) C57BL/6J mice and mouse mast cell protease-4 knockout (mMCP-4 KO) congeners were subjected to ligation of the left anterior descending (LAD) coronary artery. A group of mice was then subjected to Kaplan-Meier 28-day survival analysis. In another group of mice, 18F-fluorodeoxyglucose positron emission tomography (PET) was performed to evaluate heart function and the infarcted zone 3 days post-PMI surgery. Cardiac morphology following PMI was evaluated on formalin-fixed heart slices and glycoproteomic analysis was performed using mass spectrometry. Finally, cardiac and lung tissue content of immunoreactive ET-1 was determined. PMI caused 60% mortality in WT mice, due to left ventricular wall rupture, and 7% in mMCP-4 KO mice. Cardiac PET analysis revealed a significant reduction in left ventricular volume (systolic and diastolic) and preserved the ejection fraction in mMCP-4 KO compared to WT animals. The infarcted area, apoptotic signaling and wall remodeling were significantly decreased in mMCP-4 KO mice compared to their WT congeners, while collagen deposition was increased. Glycoproteomic analysis showed an increase in apolipoprotein A1, an established chymase substrate in mMCP-4 KO mice compared to WT mice post-PMI. ET-1 levels were increased in the lungs of WT, but not mMCP-4 KO mice, 24 h post-PMI. Thus, the genetic deletion of mMCP-4 improved survival and heart function post-PMI.
Collapse
Affiliation(s)
- Martin Houde
- Department of Pharmacology-Physiology, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Universiteit Leiden, Leiden, Netherlands
| | - Adel Schwertani
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Hanène Touil
- Department of Pharmacology-Physiology, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Louisane Desbiens
- Department of Pharmacology-Physiology, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Otman Sarrhini
- Department of Nuclear Medicine and Radiobiology, Sherbrooke Molecular Imaging Center, CRCHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Roger Lecomte
- Department of Nuclear Medicine and Radiobiology, Sherbrooke Molecular Imaging Center, CRCHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Martin Lepage
- Department of Nuclear Medicine and Radiobiology, Sherbrooke Molecular Imaging Center, CRCHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Hugo Gagnon
- PhenoSwitch Bioscience Inc., Sherbrooke, QC, Canada
| | - Shinji Takai
- Department of Innovative Medicine, Osaka Medical College, Osaka, Japan
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.,Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Danielle Jacques
- Department of Anatomy and Cell Biology, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Fernand Gobeil
- Department of Pharmacology-Physiology, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Robert Day
- Department of Surgery, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Pedro D'Orléans-Juste
- Department of Pharmacology-Physiology, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
10
|
Zhai Y, Luo Y, Wu P, Li D. New insights into SERCA2a gene therapy in heart failure: pay attention to the negative effects of B-type natriuretic peptides. J Med Genet 2018; 55:287-296. [PMID: 29478009 DOI: 10.1136/jmedgenet-2017-105120] [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: 10/17/2017] [Revised: 01/30/2018] [Accepted: 02/05/2018] [Indexed: 12/28/2022]
Abstract
Sarcoplasmic/endoplasmic reticulum calcium ATPase 2a (SERCA2a) is a target of interest in gene therapy for heart failure with reduced ejection fraction (HFrEF). However, the results of an important clinical study, the Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease (CUPID) trial, were controversial. Promising results were observed in the CUPID 1 trial, but the results of the CUPID 2 trial were negative. The factors that caused the controversial results remain unclear. Importantly, enrolled patients were required to have a higher plasma level of B-type natriuretic peptide (BNP) in the CUPID 2 trial. Moreover, BNP was shown to inhibit SERCA2a expression. Therefore, it is possible that high BNP levels interact with treatment effects of SERCA2a gene transfer and accordingly lead to negative results of CUPID 2 trial. From this point of view, effects of SERCA2a gene therapy should be explored in heart failure with preserved ejection fraction, which is characterised by lower BNP levels compared with HFrEF. In this review, we summarise the current knowledge of SERCA2a gene therapy for heart failure, analyse potential interaction between BNP levels and therapeutic effects of SERCA2a gene transfer and provide directions for future research to solve the identified problems.
Collapse
Affiliation(s)
- Yuting Zhai
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuanyuan Luo
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Pei Wu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
11
|
Carvalho LSF, Bogniotti LAC, de Almeida OLR, e Silva JCQ, Nadruz W, Coelho OR, Sposito AC. Change of BNP between admission and discharge after ST-elevation myocardial infarction (Killip I) improves risk prediction of heart failure, death, and recurrent myocardial infarction compared to single isolated measurement in addition to the GRACE score. EUROPEAN HEART JOURNAL-ACUTE CARDIOVASCULAR CARE 2018; 8:643-651. [DOI: 10.1177/2048872617753049] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Objective: In ST-elevation myocardial infarction, 7–15% of patients admitted as Killip I will develop symptomatic heart failure or decreased ejection fraction. However, available clinical scores do not predict the risk of severe outcomes well, such as heart failure, recurrent myocardial infarction, and sudden death in these Killip I individuals. Therefore, we evaluated whether one vs two measurements of BNP would improve prediction of adverse outcomes in addition to the GRACE score in ST-elevation myocardial infarction/Killip I individuals. Methods: Consecutive patients with ST-elevation myocardial infarction/Killip I ( n=167) were admitted and followed for 12 months. The GRACE score was calculated and plasma BNP levels were obtained in the first 12 h after symptom onset (D1) and at the fifth day (D5). Results: Fifteen percent of patients admitted as Killip I developed symptomatic heart failure and/or decreased ejection fraction in 12 months. The risk of developing symptomatic heart failure or ejection fraction <40% at 30 days was increased by 8.7-fold (95% confidence interval: 1.10–662, p=0.046) per each 100 pg/dl increase in BNP-change. Both in unadjusted and adjusted Cox-regressions, BNP-change as a continuous variable was associated with incident sudden death/myocardial infarction at 30 days (odds ratio 1.032 per each increase of 10 pg/dl, 95% confidence interval: 1.013–1.052, p<0.001), but BNP-D1 was not. The GRACE score alone showed a moderate C-statistic=0.709 ( p=0.029), but adding BNP-change improved risk discrimination (C-statistic=0.831, p=0.001). Net reclassification confirmed a significant improvement in individual risk prediction by 33.4% (95% confidence interval: 8–61%, p=0.034). However, GRACE +BNP-D1 did not improve risk reclassification at 30 days compared to GRACE ( p=0.8). At 12 months, BNP-change was strongly associated with incident sudden death/myocardial infarction, but not BNP-D1. Conclusions: Only BNP-change following myocardial infarction was associated with poorer short- and long-term outcomes. BNP-change also improves risk reclassification in addition to the GRACE score.
Collapse
Affiliation(s)
- Luiz Sergio F Carvalho
- Cardiology Division, State University of Campinas (Unicamp), Brazil
- Escola Superior de Ciências da Saúde (ESCS), Brazil
| | | | | | - Jose C Quinaglia e Silva
- Escola Superior de Ciências da Saúde (ESCS), Brazil
- University of Brasilia Medical School (UnB), Brazil
| | - Wilson Nadruz
- Cardiology Division, State University of Campinas (Unicamp), Brazil
| | | | - Andrei C Sposito
- Cardiology Division, State University of Campinas (Unicamp), Brazil
| |
Collapse
|
12
|
Role of sympathetic cotransmitter galanin on autonomic balance in heart failure: an active player or a bystander? Anatol J Cardiol 2017; 18:281-288. [PMID: 28811392 PMCID: PMC5731524 DOI: 10.14744/anatoljcardiol.2017.7831] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Objective: Galanin, a cotransmitter similar to neuropeptide Y (NPY), aggravates autonomic imbalance in systolic heart failure (HF) by attenuating vagal tonus after burst sympathetic activity. In animal HF models, galanin antagonists have improved cardiac function. To determine whether galanin is a promising therapeutic target in HF, we studied its concentrations in HF patients and evaluated its correlation with NPY, markers of humoral activity such as pro-BNP and copeptin, and echocardiographic parameters of HF severity. Methods: After recording demographic and echocardiographic characteristics of 87 individuals (57 HF patients and 30 control subjects), fasting serum concentrations of galanin, NPY, copeptin, and pro-BNP were determined. Results: Unlike pro-BNP, copeptin, and NPY, which were significantly elevated in HF patients (p<0.001, p<0.001, and p=0.001, respectively), galanin was similar in HF patients and control subjects (p=0.9). NPY correlated with the echocardiographic parameters of HF severity (r=–0.22, p=0.03 for EF; r=0.3, p=0.005 for Tei index of RV; r=–0.23, p=0.03 for TAPSE; and r=0.24, p=0.024 for E/e¢) and pro-BNP (r=0.22, p=0.046). NPY levels were also associated with beta blocker (BB) use, wherein BB significantly decreased NPY in both HF patients and control subjects. Galanin correlated with humoral biomarkers, pro-BNP and copeptin (r=0.39, p<0.001 and r=0.41, p<0.001, respectively). Although current smoking, BB therapy, pro-BNP, copeptin, and body mass index were associated with galanin in univariate analyses, the multiple linear regression model revealed that pro-BNP was the only significant determinant of galanin levels in HF patients. Conclusion: Our findings confirmed the role of NPY in autonomic balance and suggest that galanin is associated with the proadrenergic state, but its role in HF in humans remains unclear.
Collapse
|
13
|
Grimm G, Resl M, Heinisch BB, Hülsmann M, Luger A, Clodi M, Vila G. B-type natriuretic peptide increases cortisol and catecholamine concentrations in healthy subjects. J Appl Physiol (1985) 2017; 122:1249-1254. [PMID: 28008098 DOI: 10.1152/japplphysiol.00360.2016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 11/23/2016] [Accepted: 12/16/2016] [Indexed: 11/22/2022] Open
Abstract
B-type natriuretic peptide (BNP) is a hormone released by the heart in response to volume load and exerts natriuretic properties. It is clinically used as a diagnostic and prognostic biomarker and investigated as a pharmacological agent in the therapy of heart failure. Here we investigate the changes in pituitary, adrenal, and thyroid hormones in response to BNP administration in a randomized single-blinded crossover study conducted in ten healthy men aged 21-29 yr. Participants received in two study sessions a continuous intravenous infusion during 4 h (once placebo and once 3 pmol·kg-1·min-1 BNP) and remained in supine position throughout the study. Circulating concentrations of pituitary, adrenal, and thyroid hormones, heart rate, and blood pressure were measured at baseline and hourly afterwards. BNP prevented the physiological decrease in cortisol during the late morning hours leading to elevated serum cortisol levels (P = 0.022) and increased circulating epinephrine and norepinephrine concentrations (P = 0.018 and P = 0.036, respectively). These hormone changes were accompanied by an increase in heart rate (P = 0.019) but no differences in blood pressure. Taken together, the impact of BNP on the endocrine system extends beyond the well-known inhibition of the renin-angiotensin-aldosterone system and includes increased adrenergic activity and cortisol concentrations. This neuroendocrine activation might impact the outcome of therapeutical BNP administrations and should be further investigated in conditions associated with increased BNP secretion.NEW & NOTEWORTHY The heart hormone B-type natriuretic peptide (BNP) is increased in patients with heart failure, where it is thought to have beneficial effects by reducing the preload. Here we report that intravenous administration of BNP in men leads to increases in adrenal hormones cortisol, epinephrine, and norepinephrine. Cortisol and catecholamine levels are independent predictors of increased cardiovascular mortality risk; therefore, drugs targeting the BNP system should be evaluated regarding their effects on the neuroendocrine activation accompanying heart failure.
Collapse
Affiliation(s)
- Gabriele Grimm
- Department of Medical and Chemical Laboratory Diagnostics, Medical University of Vienna, Austria
| | - Michael Resl
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Birgit B Heinisch
- Division of Gastroenterology, Department of Internal Medicine III, Medical University of Vienna, Austria; and
| | - Martin Hülsmann
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Anton Luger
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Martin Clodi
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Austria;
| | - Greisa Vila
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Austria
| |
Collapse
|
14
|
Nesiritide Therapy Is Associated With Better Clinical Outcomes Than Dobutamine Therapy in Heart Failure. Am J Ther 2017; 24:e181-e188. [PMID: 26164026 DOI: 10.1097/mjt.0000000000000278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
To evaluate the therapeutic effects of dobutamine and nesiritide in the treatment of heart failure (HF), a meta-analysis of published studies was conducted. Computerized bibliographic databases in Chinese and English languages were carefully searched to identify the relevant literature. A total of 6 cohort studies were enrolled in current meta-analysis for statistical analyses. The effect of dobutamine and nesiritide in patients with HF was estimated by odds ratios (ORs) and 95% confidence interval (CI). Our results revealed a significantly higher survival rate in nesiritide-treated patients, compared with those treated with dobutamine (OR = 1.97; 95% CI, 1.43-2.71; P < 0.001). In addition, a lower readmission rate was also associated with the nesiritide-treated group in comparison with the dobutamine-treated group (OR = 1.96; 95% CI, 1.39-2.78; P < 0.001). A stratified analysis revealed that the subgroup of patients with HF treated with nesiritide showed higher survival outcomes than those patients with HF treated with dobutamine when follow-up period was greater than 6 months (OR = 1.70; 95% CI, 1.21-2.38; P = 0.002) but not under 6 months (P > 0.05). This indicated that nesiritide treatment had longer term benefits as well. Interestingly, based on the reason for readmission, a subgroup analysis of the HF subgroup and the "all-cause" subgroup showed that higher readmission rates were associated with dobutamine treatment in both subgroups (HF: OR = 2.71; 95% CI, = 1.51-4.83; P = 0.001; all-cause: OR = 1.64; 95% CI, 1.06-2.53; P = 0.026; respectively). Our results suggest that nesiritide therapy is associated with a lower in-hospital mortality rates and decreased readmission rates compared with dobutamine treatment in patients with HF.
Collapse
|
15
|
Lin YK, Chen YC, Chen YA, Yeh YH, Chen SA, Chen YJ. B-Type Natriuretic Peptide Modulates Pulmonary Vein Arrhythmogenesis: A Novel Potential Contributor to the Genesis of Atrial Tachyarrhythmia in Heart Failure. J Cardiovasc Electrophysiol 2016; 27:1462-1471. [PMID: 27571932 DOI: 10.1111/jce.13093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 08/11/2016] [Accepted: 08/25/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND Heart failure (HF) plays a critical role in the genesis of atrial fibrillation (AF). A high B-type natriuretic peptide (BNP) level occurs in patients with HF and in patients with AF. However, the role of BNP in the pathophysiology of AF is not clear. The purposes of this study were to evaluate the effects of BNP on pulmonary vein (PV) arrhythmogenesis. METHODS AND RESULTS Whole-cell patch clamp and fluorescence were used to study the action potential, ionic currents, and calcium homeostasis in isolated single rabbit PV cardiomyocytes before and after a BNP infusion, with or without ODQ (10 μM), milrinone (50 μM), or ouabain (1 μM). BNP increased PV spontaneous activity by 28.2 ± 7.5% at 100 nM and by 23.8 ± 9.1% at 300 nM. Similar to those with BNP, milrinone 50 μM increased the PV beating rate from 3.0 ± 0.2 to 3.6 ± 0.3 Hz (P < 0.0005, n = 7). In the presence of ODQ application, BNP didn't change PV spontaneous activity. BNP (100 nM) increased calcium transients (F/F0 from 1.6 ± 0.1 to 1.9 ± 0.2, n = 20, P < 0.05) and increased the pacemaker current (0.4 ± 0.1 to 1.0 ± 0.2 pA/pF, n = 17, P < 0.0005) in PV cardiomyocytes. Moreover, BNP (100 nM) increased the transient inward current, sodium currents, sodium-calcium exchanger currents, and L-type calcium current; but reduced late sodium currents and the Na-K pump in PV cardiomyocytes. CONCLUSION BNP increases PV arrhythmogenesis, which may contribute to the genesis of atrial tachyarrhythmogenesis in HF. Cyclic GMP activation, phosphodiesterase 3 inhibition and Na+ /K+ -ATPase inhibition might participate in the BNP modulation of PV electrophysiology.
Collapse
Affiliation(s)
- Yung-Kuo Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei, Taiwan.,Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Ann Chen
- Division of Nephrology, Sijhih Cathay General Hospital, New Taipei City, Taiwan
| | - Yung-Hsin Yeh
- The First Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University, Taoyuan, Taiwan
| | - Shih-Ann Chen
- School of Medicine, National Yang-Ming University; Division of Cardiology and Cardiovascular Research Center, Veterans General Hospital-Taipei, Taipei, Taiwan
| | - Yi-Jen Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
16
|
Human myoblast transplantation in mice infarcted heart alters the expression profile of cardiac genes associated with left ventricle remodeling. Int J Cardiol 2016; 202:710-21. [DOI: 10.1016/j.ijcard.2015.09.115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 09/24/2015] [Accepted: 09/27/2015] [Indexed: 01/17/2023]
|
17
|
Roy J, Oger C, Thireau J, Roussel J, Mercier-Touzet O, Faure D, Pinot E, Farah C, Taber DF, Cristol JP, Lee JCY, Lacampagne A, Galano JM, Durand T, Le Guennec JY. Nonenzymatic lipid mediators, neuroprostanes, exert the antiarrhythmic properties of docosahexaenoic acid. Free Radic Biol Med 2015; 86:269-78. [PMID: 25911196 DOI: 10.1016/j.freeradbiomed.2015.04.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/18/2015] [Accepted: 04/11/2015] [Indexed: 12/21/2022]
Abstract
Neuroprostanes are lipid mediators produced by nonenzymatic free radical peroxidation of docosahexaenoic acid (DHA). DHA is associated with a lower atherosclerosis risk, suggesting a beneficial role in cardiovascular diseases. The aim of this study was to investigate the influence of DHA peroxidation on its potentially antiarrhythmic properties (AAP) in isolated ventricular cardiomyocytes and in vivo in post-myocardial infarcted mice. Calcium imaging and biochemical experiments indicate that cardiac arrhythmias induced by isoproterenol are associated with Ca(2+) leak from the sarcoplasmic reticulum after oxidation and phosphorylation of the type 2 ryanodine receptor (RyR2) leading to dissociation of the FKBP12.6/RyR2 complex. Both oxidized DHA and 4(RS)-4-F4t-NeuroP prevented cellular arrhythmias and posttranslational modifications of the RyR2 leading to a stabilized FKBP12.6/RyR2 complex. DHA per se did not have AAP. The AAP of 4(RS)-4-F4t-NeuroP was also observed in vivo. In this study, we challenged the paradigm that spontaneously formed oxygenated metabolites of lipids are undesirable as they are unconditionally toxic. This study reveals that the lipid mediator 4(RS)-4-F4t-neuroprostane derived from nonenzymatic peroxidation of docosahexaenoic acid can counteract such deleterious effects through cardiac antiarrhythmic properties. Our findings demonstrate 4(RS)-4-F4t-NeuroP as a mediator of the cardioprotective AAP of DHA. This discovery opens new perspectives for products of nonenzymatic oxidized ω3 polyunsaturated fatty acids as potent mediators in diseases that involve ryanodine complex destabilization such as ischemic events.
Collapse
Affiliation(s)
- Jérôme Roy
- INSERM U1046-UMR 9214, Centre National de la Recherche Scientifique, Physiologie et Médecine Expérimentale du Coeur et des Muscles, Université de Montpellier, 34295 Montpellier Cedex 5, France
| | - Camille Oger
- Institut des Biomolécules Max Mousseron, UMR 5247, Centre National de la Recherche Scientifique, Université de Montpellier, ENSCM, Montpellier, France
| | - Jérôme Thireau
- Institut des Biomolécules Max Mousseron, UMR 5247, Centre National de la Recherche Scientifique, Université de Montpellier, ENSCM, Montpellier, France
| | - Julien Roussel
- INSERM U1046-UMR 9214, Centre National de la Recherche Scientifique, Physiologie et Médecine Expérimentale du Coeur et des Muscles, Université de Montpellier, 34295 Montpellier Cedex 5, France
| | - Olivia Mercier-Touzet
- INSERM U1046-UMR 9214, Centre National de la Recherche Scientifique, Physiologie et Médecine Expérimentale du Coeur et des Muscles, Université de Montpellier, 34295 Montpellier Cedex 5, France
| | - Delinger Faure
- INSERM U1046-UMR 9214, Centre National de la Recherche Scientifique, Physiologie et Médecine Expérimentale du Coeur et des Muscles, Université de Montpellier, 34295 Montpellier Cedex 5, France; Institut des Biomolécules Max Mousseron, UMR 5247, Centre National de la Recherche Scientifique, Université de Montpellier, ENSCM, Montpellier, France
| | - Edith Pinot
- Institut des Biomolécules Max Mousseron, UMR 5247, Centre National de la Recherche Scientifique, Université de Montpellier, ENSCM, Montpellier, France
| | - Charlotte Farah
- INSERM U1046-UMR 9214, Centre National de la Recherche Scientifique, Physiologie et Médecine Expérimentale du Coeur et des Muscles, Université de Montpellier, 34295 Montpellier Cedex 5, France
| | - Douglass F Taber
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA
| | - Jean-Paul Cristol
- INSERM U1046-UMR 9214, Centre National de la Recherche Scientifique, Physiologie et Médecine Expérimentale du Coeur et des Muscles, Université de Montpellier, 34295 Montpellier Cedex 5, France
| | - Jetty C Y Lee
- School of Biological Sciences, The University of Hong Kong, Hong Kong, SAR
| | - Alain Lacampagne
- INSERM U1046-UMR 9214, Centre National de la Recherche Scientifique, Physiologie et Médecine Expérimentale du Coeur et des Muscles, Université de Montpellier, 34295 Montpellier Cedex 5, France
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron, UMR 5247, Centre National de la Recherche Scientifique, Université de Montpellier, ENSCM, Montpellier, France
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron, UMR 5247, Centre National de la Recherche Scientifique, Université de Montpellier, ENSCM, Montpellier, France
| | - Jean-Yves Le Guennec
- INSERM U1046-UMR 9214, Centre National de la Recherche Scientifique, Physiologie et Médecine Expérimentale du Coeur et des Muscles, Université de Montpellier, 34295 Montpellier Cedex 5, France.
| |
Collapse
|
18
|
Roussel J, Thireau J, Brenner C, Saint N, Scheuermann V, Lacampagne A, Le Guennec JY, Fauconnier J. Palmitoyl-carnitine increases RyR2 oxidation and sarcoplasmic reticulum Ca2+ leak in cardiomyocytes: Role of adenine nucleotide translocase. Biochim Biophys Acta Mol Basis Dis 2015; 1852:749-58. [DOI: 10.1016/j.bbadis.2015.01.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 01/16/2015] [Accepted: 01/18/2015] [Indexed: 12/30/2022]
|
19
|
Bellera N, Barba I, Rodriguez-Sinovas A, Ferret E, Asín MA, Gonzalez-Alujas MT, Pérez-Rodon J, Esteves M, Fonseca C, Toran N, Garcia Del Blanco B, Pérez A, Garcia-Dorado D. Single intracoronary injection of encapsulated antagomir-92a promotes angiogenesis and prevents adverse infarct remodeling. J Am Heart Assoc 2014; 3:e000946. [PMID: 25240056 PMCID: PMC4323815 DOI: 10.1161/jaha.114.000946] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background Small and large preclinical animal models have shown that antagomir‐92a‐based therapy reduces early postischemic loss of function, but its effect on postinfarction remodeling is not known. In addition, the reported remote miR‐92a inhibition in noncardiac organs prevents the translation of nonvectorized miR‐targeted therapy to the clinical setting. We investigated whether a single intracoronary administration of antagomir‐92a encapsulated in microspheres could prevent deleterious remodeling of myocardium 1 month after acute myocardial infarction AUTHOR: Should “acute” be added before “myocardial infarction” (since abbreviation is AMI)? Also check at first mention in main text (AMI) without adverse effects. Methods and Results In a percutaneous pig model of reperfused AMI, a single intracoronary administration of antagomir‐92a encapsulated in specific microspheres (9 μm poly‐d,‐lactide‐co‐glycolide [PLGA]) inhibited miR‐92a in a local, selective, and sustained manner (n=3 pigs euthanized 1, 3, and 10 days after treatment; 8×, 2×, and 5×‐fold inhibition at 1, 3, and 10 days). Downregulation of miR‐92a resulted in significant vessel growth (n=27 adult minipigs randomly allocated to blind receive encapsulated antagomir‐92a, encapsulated placebo, or saline [n=8, 9, 9]; P=0.001), reduced regional wall‐motion dysfunction (P=0.03), and prevented adverse remodeling in the infarct area 1 month after injury (P=0.03). Intracoronary injection of microspheres had no significant adverse effect in downstream myocardium in healthy pigs (n=2), and fluorescein isothiocyanate albumin‐PLGA microspheres were not found in myocardium outside the left anterior descending coronary artery territory (n=4) or in other organs (n=2). Conclusions Early single intracoronary administration of encapsulated antagomir‐92a in an adult pig model of reperfused AMI prevents left ventricular remodeling with no local or distant adverse effects, emerging as a promising therapeutic approach to translate to patients who suffer a large AMI.
Collapse
Affiliation(s)
- Neus Bellera
- Laboratory of Experimental and Molecular Cardiocirculatory Pathology, Institut de Recerca, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain (N.B., I.B., A.R.S., D.G.D.) Department of Cardiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain (N.B., T.G.A., J.R., B.G.B., D.G.D.)
| | - Ignasi Barba
- Laboratory of Experimental and Molecular Cardiocirculatory Pathology, Institut de Recerca, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain (N.B., I.B., A.R.S., D.G.D.)
| | - Antonio Rodriguez-Sinovas
- Laboratory of Experimental and Molecular Cardiocirculatory Pathology, Institut de Recerca, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain (N.B., I.B., A.R.S., D.G.D.)
| | - Eulalia Ferret
- I+D Pierre-Fabre Ibérica S.A., Cerdanyola del Vallès, Spain (E.F., M.A.A., A.)
| | - Miguel Angel Asín
- I+D Pierre-Fabre Ibérica S.A., Cerdanyola del Vallès, Spain (E.F., M.A.A., A.)
| | - M Teresa Gonzalez-Alujas
- Department of Cardiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain (N.B., T.G.A., J.R., B.G.B., D.G.D.)
| | - Jordi Pérez-Rodon
- Department of Cardiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain (N.B., T.G.A., J.R., B.G.B., D.G.D.)
| | - Marielle Esteves
- Department of Animal Housing, Institut de Recerca, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain (M.E., C.F.)
| | - Carla Fonseca
- Department of Animal Housing, Institut de Recerca, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain (M.E., C.F.)
| | - Nuria Toran
- Department of Anatomical Pathology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain (N.T.)
| | - Bruno Garcia Del Blanco
- Department of Cardiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain (N.B., T.G.A., J.R., B.G.B., D.G.D.)
| | - Amadeo Pérez
- I+D Pierre-Fabre Ibérica S.A., Cerdanyola del Vallès, Spain (E.F., M.A.A., A.)
| | - David Garcia-Dorado
- Laboratory of Experimental and Molecular Cardiocirculatory Pathology, Institut de Recerca, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain (N.B., I.B., A.R.S., D.G.D.) Department of Cardiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain (N.B., T.G.A., J.R., B.G.B., D.G.D.)
| |
Collapse
|
20
|
Kim JC, Wang J, Son MJ, Cuong NM, Woo SH. Sensitization of cardiac Ca2+ release sites by protein kinase C signaling: evidence from action of murrayafoline A. Pflugers Arch 2014; 467:1607-1621. [DOI: 10.1007/s00424-014-1589-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/06/2014] [Accepted: 07/28/2014] [Indexed: 11/29/2022]
|
21
|
Plante E, Menaouar A, Danalache BA, Broderick TL, Jankowski M, Gutkowska J. Treatment with brain natriuretic peptide prevents the development of cardiac dysfunction in obese diabetic db/db mice. Diabetologia 2014; 57:1257-67. [PMID: 24595856 DOI: 10.1007/s00125-014-3201-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 02/03/2014] [Indexed: 12/21/2022]
Abstract
AIMS/HYPOTHESIS Obesity and diabetes increase the risk of developing cardiovascular diseases and heart failure. These metabolic disorders are generally reflected by natriuretic peptide system deficiency. Since brain natriuretic peptide (BNP) is known to influence metabolism and cardioprotection, we investigated the effect of chronic exogenous BNP treatment on adverse myocardial consequences related to obesity and diabetes. METHODS Ten-week-old C57BL/KsJ-db/db obese diabetic mice (db/db) and their lean control littermates (db/+) were treated with BNP (0.6 μg kg(-1) h(-1)) or saline for 12 weeks (n = 10/group). Serial blood and tomography analysis were performed. Cardiac function was determined by echocardiography, and biochemical and histological heart and fat analyses were also performed. RESULTS BNP treatment resulted in an average increase in plasma BNP levels of 70 pg/ml. An improvement in the metabolic profile of db/db mice was observed, including a reduction in fat content, increased insulin sensitivity, improved glucose tolerance and lower blood glucose, despite increased food intake. db/db mice receiving saline displayed both early systolic and diastolic dysfunction, whereas these functional changes were prevented by BNP treatment. The cardioprotective effects of BNP were attributed to the inhibition of cardiomyocyte apoptosis, myocardial fibrosis, cardiac hypertrophy and the AGE-receptor for AGE (RAGE) system as well as normalisation of cardiac AMP-activated protein kinase and endothelial nitric oxide synthase activities. CONCLUSIONS/INTERPRETATION Our results indicate that chronic BNP treatment at low dose improves the metabolic profile and prevents the development of myocardial dysfunction in db/db mice.
Collapse
Affiliation(s)
- Eric Plante
- Cardiovascular Biochemistry Laboratory, CRCHUM (7-134), Tour Viger, 900 St-Denis St., Montreal, Quebec, H2X 0A9, Canada
| | | | | | | | | | | |
Collapse
|
22
|
Dewenter M, Vettel C, El-Armouche A. The Janus face of BNP therapy in chronic heart failure: beneficial effects unmasked by β blockers? Heart 2014; 100:819-20. [PMID: 24667282 DOI: 10.1136/heartjnl-2014-305503] [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: 11/04/2022] Open
Affiliation(s)
- Matthias Dewenter
- Institute of Pharmacology, University Medical School Göttingen, , Göttingen, Germany
| | | | | |
Collapse
|
23
|
Thireau J, Karam S, Roberge S, Roussel J, Aimond F, Cassan C, Gac A, Babuty D, Le Guennec JY, Lacampagne A, Fauconnier J, Richard S. Β-adrenergic blockade combined with subcutaneous B-type natriuretic peptide: a promising approach to reduce ventricular arrhythmia in heart failure? Heart 2014; 100:833-41. [PMID: 24667281 DOI: 10.1136/heartjnl-2013-305167] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIMS Clinical studies failed to prove convincingly efficiency of intravenous infusion of neseritide during heart failure and evidence suggested a pro-adrenergic action of B-type natriuretic peptide (BNP). However, subcutaneous BNP therapy was recently proposed in heart failure, thus raising new perspectives over what was considered as a promising treatment. We tested the efficiency of a combination of oral β1-adrenergic receptor blocker metoprolol and subcutaneous BNP infusion in decompensated heart failure. METHODS AND RESULTS The effects of metoprolol or/and BNP were studied on cardiac remodelling, excitation-contraction coupling and arrhythmias in an experimental mouse model of ischaemic heart failure following postmyocardial infarction. We determined the cellular and molecular mechanisms involved in anti-remodelling and antiarrhythmic actions. As major findings, the combination was more effective than metoprolol alone in reversing cardiac remodelling and preventing ventricular arrhythmia. The association of the two molecules improved cardiac function, reduced hypertrophy and fibrosis, and corrected the heart rate, sympatho-vagal balance (low frequencies/high frequencies) and ECG parameters (P to R wave interval (PR), QRS duration, QTc intervals). It also improved altered Ca(2+) cycling by normalising Ca(2+)-handling protein levels (S100A1, SERCA2a, RyR2), and prevented pro-arrhythmogenic Ca(2+) waves derived from abnormal Ca(2+) sparks in ventricular cardiomyocytes. Altogether these effects accounted for decreased occurrence of ventricular arrhythmias. CONCLUSIONS Association of subcutaneous BNP and oral metoprolol appeared to be more effective than metoprolol alone. Breaking the deleterious loop linking BNP and sympathetic overdrive in heart failure could unmask the efficiency of BNP against deleterious damages in heart failure and bring a new potential approach against lethal arrhythmia during heart failure.
Collapse
Affiliation(s)
- Jérôme Thireau
- Inserm U1046, Physiologie & Médecine Expérimentale, Cœur et Muscles, Université Montpellier-1 & 2, , Montpellier, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Chan NYK, Robador PA, Levi R. Natriuretic peptide-induced catecholamine release from cardiac sympathetic neurons: inhibition by histamine H3 and H4 receptor activation. J Pharmacol Exp Ther 2012; 343:568-77. [PMID: 22923736 PMCID: PMC3500538 DOI: 10.1124/jpet.112.198747] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 08/23/2012] [Indexed: 12/20/2022] Open
Abstract
We reported previously that natriuretic peptides, including brain natriuretic peptide (BNP), promote norepinephrine release from cardiac sympathetic nerves and dopamine release from differentiated pheochromocytoma PC12 cells. These proexocytotic effects are mediated by an increase in intracellular calcium secondary to cAMP/protein kinase A (PKA) activation caused by a protein kinase G (PKG)-mediated inhibition of phosphodiesterase type 3 (PDE3). The purpose of the present study was to search for novel means to prevent the proadrenergic effects of natriuretic peptides. For this, we focused our attention on neuronal inhibitory Gα(i/o)-coupled histamine H(3) and H(4) receptors. Our findings show that activation of neuronal H(3) and H(4) receptors inhibits the release of catecholamines elicited by BNP in cardiac synaptosomes and differentiated PC12 cells. This effect results from a decrease in intracellular Ca(2+) due to reduced intracellular cAMP/PKA activity, caused by H(3) and H(4) receptor-mediated PKG inhibition and consequent PDE3-induced increase in cAMP metabolism. Indeed, selective H(3) and H(4) receptor agonists each synergized with a PKG inhibitor and a PDE3 activator in attenuating BNP-induced norepinephrine release from cardiac sympathetic nerve endings. This indicates that PKG inhibition and PDE3 stimulation are pivotal for the H(3) and H(4) receptor-mediated attenuation of BNP-induced catecholamine release. Cardiac sympathetic overstimulation is characteristic of advanced heart failure, which was recently found not to be improved by the administration of recombinant BNP (nesiritide), despite the predicated beneficial effects of natriuretic peptides. Because excessive catecholamine release is likely to offset the desirable effects of natriuretic peptides, our findings suggest novel means to alleviate their adverse effects and improve their therapeutic potential.
Collapse
Affiliation(s)
- Noel Yan-Ki Chan
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | |
Collapse
|
25
|
Fan D, Takawale A, Lee J, Kassiri Z. Cardiac fibroblasts, fibrosis and extracellular matrix remodeling in heart disease. FIBROGENESIS & TISSUE REPAIR 2012; 5:15. [PMID: 22943504 PMCID: PMC3464725 DOI: 10.1186/1755-1536-5-15] [Citation(s) in RCA: 599] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 08/16/2012] [Indexed: 12/30/2022]
Abstract
Fibroblasts comprise the largest cell population in the myocardium. In heart disease, the number of fibroblasts is increased either by replication of the resident myocardial fibroblasts, migration and transformation of circulating bone marrow cells, or by transformation of endothelial/epithelial cells into fibroblasts and myofibroblasts. The primary function of fibroblasts is to produce structural proteins that comprise the extracellular matrix (ECM). This can be a constructive process; however, hyperactivity of cardiac fibroblasts can result in excess production and deposition of ECM proteins in the myocardium, known as fibrosis, with adverse effects on cardiac structure and function. In addition to being the primary source of ECM proteins, fibroblasts produce a number of cytokines, peptides, and enzymes among which matrix metalloproteinases (MMPs) and their inhibitors, tissue inhibitor of metalloproteinases (TIMPs), directly impact the ECM turnover and homeostasis. Function of fibroblasts can also in turn be regulated by MMPs and TIMPs. In this review article, we will focus on the function of cardiac fibroblasts in the context of ECM formation, homeostasis and remodeling in the heart. We will discuss the origins and multiple roles of cardiac fibroblasts in myocardial remodeling in different types of heart disease in patients and in animal models. We will further provide an overview of what we have learned from experimental animal models and genetically modified mice with altered expression of ECM regulatory proteins, MMPs and TIMPs.
Collapse
Affiliation(s)
- Dong Fan
- Department of Physiology, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
| | | | | | | |
Collapse
|