1
|
Monogiou Belik D, Bernasconi R, Xu L, Della Verde G, Lorenz V, Grüterich V, Balzarolo M, Mochizuki M, Pfister O, Kuster GM. The Flt3-inhibitor quizartinib augments apoptosis and promotes maladaptive remodeling after myocardial infarction in mice. Apoptosis 2024; 29:357-371. [PMID: 37945814 PMCID: PMC10873224 DOI: 10.1007/s10495-023-01911-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Tyrosine kinase inhibitors (TKIs) targeting fms-like tyrosine kinase 3 (Flt3) such as quizartinib were specifically designed for acute myeloid leukemia treatment, but also multi-targeting TKIs applied to solid tumor patients inhibit Flt3. Flt3 is expressed in the heart and its activation is cytoprotective in myocardial infarction (MI) in mice. OBJECTIVES We sought to test whether Flt3-targeting TKI treatment aggravates cardiac injury after MI. METHODS AND RESULTS Compared to vehicle, quizartinib (10 mg/kg/day, gavage) did not alter cardiac dimensions or function in healthy mice after four weeks of therapy. Pretreated mice were randomly assigned to MI or sham surgery while receiving quizartinib or vehicle for one more week. Quizartinib did not aggravate the decline in ejection fraction, but significantly enhanced ventricular dilatation one week after infarction. In addition, apoptotic cell death was significantly increased in the myocardium of quizartinib-treated compared to vehicle-treated mice. In vitro, quizartinib dose-dependently decreased cell viability in neonatal rat ventricular myocytes and in H9c2 cells, and increased apoptosis as assessed in the latter. Together with H2O2, quizartinib potentiated the phosphorylation of the pro-apoptotic mitogen activated protein kinase p38 and augmented H2O2-induced cell death and apoptosis beyond additive degree. Pretreatment with a p38 inhibitor abolished apoptosis under quizartinib and H2O2. CONCLUSION Quizartinib potentiates apoptosis and promotes maladaptive remodeling after MI in mice at least in part via a p38-dependent mechanism. These findings are consistent with the multi-hit hypothesis of cardiotoxicity and make cardiac monitoring in patients with ischemic heart disease under Flt3- or multi-targeting TKIs advisable.
Collapse
Affiliation(s)
- Daria Monogiou Belik
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Riccardo Bernasconi
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Lifen Xu
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Giacomo Della Verde
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Vera Lorenz
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Vivienne Grüterich
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Melania Balzarolo
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Michika Mochizuki
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Otmar Pfister
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Basel, Basel, Switzerland
| | - Gabriela M Kuster
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland.
- Department of Cardiology, University Heart Center, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
2
|
Hume RD, Kanagalingam S, Deshmukh T, Chen S, Mithieux SM, Rashid FN, Roohani I, Lu J, Doan T, Graham D, Clayton ZE, Slaughter E, Kizana E, Stempien-Otero AS, Brown P, Thomas L, Weiss AS, Chong JJ. Tropoelastin Improves Post-Infarct Cardiac Function. Circ Res 2023; 132:72-86. [PMID: 36453283 PMCID: PMC9829044 DOI: 10.1161/circresaha.122.321123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND Myocardial infarction (MI) is among the leading causes of death worldwide. Following MI, necrotic cardiomyocytes are replaced by a stiff collagen-rich scar. Compared to collagen, the extracellular matrix protein elastin has high elasticity and may have more favorable properties within the cardiac scar. We sought to improve post-MI healing by introducing tropoelastin, the soluble subunit of elastin, to alter scar mechanics early after MI. METHODS AND RESULTS We developed an ultrasound-guided direct intramyocardial injection method to administer tropoelastin directly into the left ventricular anterior wall of rats subjected to induced MI. Experimental groups included shams and infarcted rats injected with either PBS vehicle control or tropoelastin. Compared to vehicle treated controls, echocardiography assessments showed tropoelastin significantly improved left ventricular ejection fraction (64.7±4.4% versus 46.0±3.1% control) and reduced left ventricular dyssynchrony (11.4±3.5 ms versus 31.1±5.8 ms control) 28 days post-MI. Additionally, tropoelastin reduced post-MI scar size (8.9±1.5% versus 20.9±2.7% control) and increased scar elastin (22±5.8% versus 6.2±1.5% control) as determined by histological assessments. RNA sequencing (RNAseq) analyses of rat infarcts showed that tropoelastin injection increased genes associated with elastic fiber formation 7 days post-MI and reduced genes associated with immune response 11 days post-MI. To show translational relevance, we performed immunohistochemical analyses on human ischemic heart disease cardiac samples and showed an increase in tropoelastin within fibrotic areas. Using RNA-seq we also demonstrated the tropoelastin gene ELN is upregulated in human ischemic heart disease and during human cardiac fibroblast-myofibroblast differentiation. Furthermore, we showed by immunocytochemistry that human cardiac fibroblast synthesize increased elastin in direct response to tropoelastin treatment. CONCLUSIONS We demonstrate for the first time that purified human tropoelastin can significantly repair the infarcted heart in a rodent model of MI and that human cardiac fibroblast synthesize elastin. Since human cardiac fibroblasts are primarily responsible for post-MI scar synthesis, our findings suggest exciting future clinical translation options designed to therapeutically manipulate this synthesis.
Collapse
Affiliation(s)
- Robert D. Hume
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Sydney Medical School, University of Sydney, NSW, Australia (R.D.H., T.D., F.R., Z.E.C., E.K., J.J.H.C.)
| | - Shaan Kanagalingam
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.)
| | - Tejas Deshmukh
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.).,Sydney Medical School, University of Sydney, NSW, Australia (R.D.H., T.D., F.R., Z.E.C., E.K., J.J.H.C.)
| | - Siqi Chen
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.)
| | - Suzanne M. Mithieux
- Charles Perkins Centre, University of Sydney, NSW, Australia (S.M.M., A.S.W.).,School of Life and Environmental Sciences, University of Sydney, NSW, Australia (S.M.M., A.S.W.)
| | - Fairooj N. Rashid
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.)
| | - Iman Roohani
- School of Biomedical Engineering, University of Sydney, NSW, Australia (I.R.).,School of Chemistry, University of New South Wales, Australia (I.R.)
| | - Juntang Lu
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.)
| | - Tram Doan
- Centre for Cancer Research, Westmead Institute for Medical Research, NSW, Australia (T.D.‚ D.G.)
| | - Dinny Graham
- Centre for Cancer Research, Westmead Institute for Medical Research, NSW, Australia (T.D.‚ D.G.).,Westmead Breast Cancer Institute, NSW, Australia (D.G.).,Westmead Clinical School, University of Sydney, NSW, Australia (D.G., L.T.)
| | - Zoe E. Clayton
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Sydney Medical School, University of Sydney, NSW, Australia (R.D.H., T.D., F.R., Z.E.C., E.K., J.J.H.C.)
| | | | - Eddy Kizana
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.).,Sydney Medical School, University of Sydney, NSW, Australia (R.D.H., T.D., F.R., Z.E.C., E.K., J.J.H.C.)
| | - April S. Stempien-Otero
- Department of Medicine, Division of Cardiology, University of Washington School of Medicine, Seattle, WA (A.S.S.-O.)
| | - Paula Brown
- Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.)
| | - Liza Thomas
- Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.).,Westmead Clinical School, University of Sydney, NSW, Australia (D.G., L.T.)
| | | | - James J.H. Chong
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.).,Sydney Medical School, University of Sydney, NSW, Australia (R.D.H., T.D., F.R., Z.E.C., E.K., J.J.H.C.)
| |
Collapse
|
3
|
Zhao H, Zhang Y, Xu X, Sun Q, Yang C, Wang H, Yang J, Yang Y, Yang X, Liu Y, Zhao Y. Sall4 and Myocd Empower Direct Cardiac Reprogramming From Adult Cardiac Fibroblasts After Injury. Front Cell Dev Biol 2021; 9:608367. [PMID: 33718351 PMCID: PMC7953844 DOI: 10.3389/fcell.2021.608367] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 02/08/2021] [Indexed: 01/14/2023] Open
Abstract
Direct conversion of fibroblasts into induced cardiomyocytes (iCMs) holds promising potential to generate functional cardiomyocytes for drug development and clinical applications, especially for direct in situ heart regeneration by delivery of reprogramming genes into adult cardiac fibroblasts in injured hearts. For a decade, many cocktails of transcription factors have been developed to generate iCMs from fibroblasts of different tissues in vitro and some were applied in vivo. Here, we aimed to develop genetic cocktails that induce cardiac reprogramming directly in cultured cardiac fibroblasts isolated from adult mice with myocardial infarction (MICFs), which could be more relevant to heart diseases. We found that the widely used genetic cocktail, Gata4, Mef2c, and Tbx5 (GMT) were inefficient in reprogramming cardiomyocytes from MICFs. In a whole well of a 12-well plate, less than 10 mCherry+ cells (<0.1%) were observed after 2 weeks of GMT infection with Myh6-reporter transgenic MICFs. By screening 22 candidate transcription factors predicted through analyzing the gene regulatory network of cardiac development, we found that five factors, GMTMS (GMT plus Myocd and Sall4), induced more iCMs expressing the cardiac structural proteins cTnT and cTnI at a frequency of about 22.5 ± 2.7% of the transduced MICFs at day 21 post infection. What is more, GMTMS induced abundant beating cardiomyocytes at day 28 post infection. Specifically, Myocd contributed mainly to inducing the expression of cardiac proteins, while Sall4 accounted for the induction of functional properties, such as contractility. RNA-seq analysis of the iCMs at day 28 post infection revealed that they were reprogrammed to adopt a cardiomyocyte-like gene expression profile. Overall, we show here that Sall4 and Myocd play important roles in cardiac reprogramming from MICFs, providing a cocktail of genetic factors that have potential for further applications in in vivo cardiac reprogramming.
Collapse
Affiliation(s)
- Hong Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, The Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China
| | - Yi Zhang
- Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| | - Xiaochan Xu
- The Center for Models of Life, Niels Bohr Institute, Copenhagen, Denmark
| | - Qiushi Sun
- Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| | - Chunyan Yang
- State Key Laboratory of Natural and Biomimetic Drugs, The Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China
| | - Hao Wang
- State Key Laboratory of Natural and Biomimetic Drugs, The Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Junbo Yang
- State Key Laboratory of Natural and Biomimetic Drugs, The Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China
| | - Yang Yang
- State Key Laboratory of Natural and Biomimetic Drugs, The Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China
| | - Xiaochun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, The Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China
| | - Yi Liu
- Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| | - Yang Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, The Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Plastech Pharmaceutical Technology Co., Ltd., Nanjing, China
| |
Collapse
|
4
|
De Villiers C, Riley PR. Mouse models of myocardial infarction: comparing permanent ligation and ischaemia-reperfusion. Dis Model Mech 2020; 13:13/11/dmm046565. [PMID: 33361140 PMCID: PMC7687859 DOI: 10.1242/dmm.046565] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 10/02/2020] [Indexed: 12/13/2022] Open
Abstract
Myocardial infarction (MI) is a disease of major consequence in the modern world, causing permanent, irreversible damage to the heart. Survivors are at risk for developing further cardiovascular pathologies such as heart failure. Further study of MI injury is crucial to improve the understanding and treatment of the post-MI heart. The most commonly used model for MI in vivo is surgical ligation of the left anterior descending coronary artery (LAD). There are two predominant approaches: permanent ligation (PL), where the LAD is permanently occluded with a suture, or ischaemia-reperfusion (IR), where the LAD is temporarily occluded before removing the suture to restore blood flow and tissue reperfusion. PL results in the majority of the area at risk becoming infarcted, leading to significant apoptotic cell death and a large scar. Conversely, IR salvages some of the area at risk; thus, the scar is smaller and includes reperfusion injury, an additional, albeit smaller, second wave of necrotic damage. PL may be a more appropriate model choice for studies of heart tissue injury and wound healing, owing to the larger, more consistent infarcts, while IR enables the study of reperfusion injury. Both are clinically relevant, and the choice of model depends upon the precise pre-clinical research questions to be addressed. Summary: Permanent ligation and ischaemia-reperfusion are common models for studying myocardial infarction. Here, we summarise the differences between them and outline the strengths of each in addressing distinct questions related to the human condition.
Collapse
Affiliation(s)
- Carla De Villiers
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK.,British Heart Foundation Oxbridge Centre of Regenerative Medicine, University of Oxford, Oxford OX1 3PT, UK
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK .,British Heart Foundation Oxbridge Centre of Regenerative Medicine, University of Oxford, Oxford OX1 3PT, UK
| |
Collapse
|
5
|
Dragasevic N, Jakovljevic V, Zivkovic V, Draginic N, Andjic M, Bolevich S, Jovic S. The role of aldosterone inhibitors in cardiac ischemia-reperfusion injury. Can J Physiol Pharmacol 2020; 99:18-29. [PMID: 32799671 DOI: 10.1139/cjpp-2020-0276] [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] [Indexed: 12/15/2022]
Abstract
Myocardial ischaemia-reperfusion (I/R) injury is a well-known term for exacerbation of cellular destruction and dysfunction after the restoration of blood flow to a previously ischaemic heart. A vast number of studies that have demonstrated that the role of mineralocorticoids in cardiovascular diseases is based on the use of pharmacological mineralocorticoid receptor (MR) antagonists. This review paper aimed to summarize current knowledge on the effects of MR antagonists on myocardial I/R injury as well as postinfarction remodeling. Animal models, predominantly the Langendorff technique and left anterior descending coronary artery occlusion, have confirmed the potency of MR antagonists as preconditioning and postconditioning agents in limiting infarct size and postinfarction remodeling. Several preclinical studies in rodents have established and proved possible mechanisms of cardioprotection by MR antagonists, such as reduction of oxidative stress, reduction of inflammation, and apoptosis, therefore limiting the infarct zone. However, the results of some clinical trials are inconsistent, since they reported no benefit of MR antagonists in acute myocardial infarction. Due to this, further studies and the results of ongoing clinical trials regarding MR antagonist administration in patients with acute myocardial infarction are being awaited with great interest.
Collapse
Affiliation(s)
- Nevena Dragasevic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34 000 Kragujevac, Serbia
| | - Vladimir Jakovljevic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34 000 Kragujevac, Serbia.,1st Moscow State Medical University IM Sechenov, Department of Human Pathology, Trubetskaya street 8, 119991 Moscow, Russia
| | - Vladimir Zivkovic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34 000 Kragujevac, Serbia
| | - Nevena Draginic
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovica 69, 34 000 Kragujevac, Serbia
| | - Marijana Andjic
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovica 69, 34 000 Kragujevac, Serbia
| | - Sergey Bolevich
- 1 Moscow State Medical University IM Sechenov, Department of Human Pathology, Trubetskaya street 8, 119991 Moscow, Russia
| | - Slavoljub Jovic
- University of Belgrade, Department of Physiology and Biochemistry, Faculty of Veterinary Medicine, Bul. Oslobodjenja 18, Belgrade, Serbia
| |
Collapse
|
6
|
EphrinA1-Fc Attenuates Ventricular Remodeling and Dysfunction in Chronically Nonreperfused WT but not EphA2-R-M mice. Int J Mol Sci 2020; 21:ijms21165811. [PMID: 32823610 PMCID: PMC7461052 DOI: 10.3390/ijms21165811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/08/2020] [Accepted: 08/11/2020] [Indexed: 12/21/2022] Open
Abstract
Background: EphrinA1-Fc abolishes acute I/R injury and attenuates nonreperfused cardiac injury 4 days after permanent occlusion in mice. The goal of this study was to assess the capacity of a single intramyocardial administration of ephrinA1-Fc at the time of coronary artery ligation, to determine the degree to which early salvage effects translate to reduced adverse remodeling after 4 weeks of nonreperfused myocardial infarction (MI) in wild-type B6 and EphA2-R-M (EphA2 receptor null) mice. Methods: At 4 weeks post-MI, echocardiography, histologic and immunohistochemical analyses of B6 mouse hearts were performed. Primary mouse cardiac fibroblasts (FBs) isolated from B6 mice cultured in the presence of low and high dose ephrinA1-Fc, both with and without pro-fibrotic TGF-β stimulation and Western blots, were probed for relative expression of remodeling proteins MMP-2, MMP-9 and TIMP-1, in addition to DDR2 and (p)SMAD2/3/totalSMAD2/3. Results: EphrinA1-Fc preserved a significant degree of contractile function, decreased adverse left ventricular remodeling, attenuated excessive compensatory hypertrophy, and decreased interstitial fibrosis in wild-type (WT) B6 mouse hearts. In contrast, most of these parameters were poorer in ephrinA1-Fc-treated EphA2-R-M mice. Of note, fibrosis was proportionately decreased, implying that other EphA receptor(s) are more important in regulating the pro-fibrotic response. Primary FBs showed disparate alteration of MMP-2, MMP-9 and TIMP-1, as well as DDR2 and p-SMAD2/3/totalSMAD2/3, which indicates that matrix remodeling and cardiac fibrosis in the injured heart are influenced by ephrinA1-Fc. Conclusion: This study demonstrates the capacity of a single administration of ephrinA1-Fc at the onset of injury to attenuate long-term nonreperfused post-MI ventricular remodeling that results in progressive heart failure, and the important role of EphA2 in mitigating the deleterious effects.
Collapse
|
7
|
Torres MJ, McLaughlin KL, Renegar RH, Valsaraj S, Whitehurst KS, Sharaf OM, Sharma UM, Horton JL, Sarathy B, Parks JC, Brault JJ, Fisher-Wellman KH, Neufer PD, Virag JAI. Intracardiac administration of ephrinA1-Fc preserves mitochondrial bioenergetics during acute ischemia/reperfusion injury. Life Sci 2019; 239:117053. [PMID: 31733316 DOI: 10.1016/j.lfs.2019.117053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/06/2019] [Accepted: 11/08/2019] [Indexed: 12/18/2022]
Abstract
AIMS Intracardiac injection of recombinant EphrinA1-Fc immediately following coronary artery ligation in mice reduces infarct size in both reperfused and non-reperfused myocardium, but the cellular alterations behind this phenomenon remain unknown. MAIN METHODS Herein, 10 wk-old B6129SF2/J male mice were exposed to acute ischemia/reperfusion (30minI/24hrsR) injury immediately followed by intracardiac injection of either EphrinA1-Fc or IgG-Fc. After 24 h of reperfusion, sections of the infarct margin in the left ventricle were imaged via transmission electron microscopy, and mitochondrial function was assessed in both permeabilized fibers and isolated mitochondria, to examine mitochondrial structure, function, and energetics in the early stages of repair. KEY FINDINGS At a structural level, EphrinA1-Fc administration prevented the I/R-induced loss of sarcomere alignment and mitochondrial organization along the Z disks, as well as disorganization of the cristae and loss of inter-mitochondrial junctions. With respect to bioenergetics, loss of respiratory function induced by I/R was prevented by EphrinA1-Fc. Preservation of cardiac bioenergetics was not due to changes in mitochondrial JH2O2 emitting potential, membrane potential, ADP affinity, efficiency of ATP production, or activity of the main dehydrogenase enzymes, suggesting that EphrinA1-Fc indirectly maintains respiratory function via preservation of the mitochondrial network. Moreover, these protective effects were lost in isolated mitochondria, further emphasizing the importance of the intact cardiomyocyte ultrastructure in mitochondrial energetics. SIGNIFICANCE Collectively, these data suggest that intracardiac injection of EphrinA1-Fc protects cardiac function by preserving cardiomyocyte structure and mitochondrial bioenergetics, thus emerging as a potential therapeutic strategy in I/R injury.
Collapse
Affiliation(s)
- Maria J Torres
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, 27834, USA
| | - Kelsey L McLaughlin
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, 27834, USA; Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Randall H Renegar
- Dept of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Smrithi Valsaraj
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - K'Shylah S Whitehurst
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Omar M Sharaf
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Uma M Sharma
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Julie L Horton
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, 27834, USA; Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Brinda Sarathy
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Justin C Parks
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Jeffrey J Brault
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA; Dept of Kinesiology, College of Health and Human Performance, East Carolina University, Greenville, NC, 27834, USA
| | - Kelsey H Fisher-Wellman
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, 27834, USA; Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - P Darrell Neufer
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, 27834, USA; Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Jitka A I Virag
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA.
| |
Collapse
|
8
|
Loss of secretin results in systemic and pulmonary hypertension with cardiopulmonary pathologies in mice. Sci Rep 2019; 9:14211. [PMID: 31578376 PMCID: PMC6775067 DOI: 10.1038/s41598-019-50634-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/23/2019] [Indexed: 12/16/2022] Open
Abstract
More than 1 billion people globally are suffering from hypertension, which is a long-term incurable medical condition that can further lead to dangerous complications and death if left untreated. In earlier studies, the brain-gut peptide secretin (SCT) was found to be able to control blood pressure by its cardiovascular and pulmonary effects. For example, serum SCT in patients with congestive heart failure was one-third of the normal level. These observations strongly suggest that SCT has a causal role in blood pressure control, and in this report, we used constitutive SCT knockout (SCT−/−) mice and control C57BL/6N mice to investigate differences in the morphology, function, underlying mechanisms and response to SCT treatment. We found that SCT−/− mice suffer from systemic and pulmonary hypertension with increased fibrosis in the lungs and heart. Small airway remodelling and pulmonary inflammation were also found in SCT−/− mice. Serum NO and VEGF levels were reduced and plasma aldosterone levels were increased in SCT−/− mice. Elevated cardiac aldosterone and decreased VEGF in the lungs were observed in the SCT−/− mice. More interestingly, SCT replacement in SCT−/− mice could prevent the development of heart and lung pathologies compared to the untreated group. Taken together, we comprehensively demonstrated the critical role of SCT in the cardiovascular and pulmonary systems and provide new insight into the potential role of SCT in the pathological development of cardiopulmonary and cardiovascular diseases.
Collapse
|
9
|
The Effects of Exercise Training Intensity on the Expression of C/EBPβ and CITED4 in Rats with Myocardial Infarction. Asian J Sports Med 2018. [DOI: 10.5812/asjsm.59300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
10
|
Bøtker HE, Hausenloy D, Andreadou I, Antonucci S, Boengler K, Davidson SM, Deshwal S, Devaux Y, Di Lisa F, Di Sante M, Efentakis P, Femminò S, García-Dorado D, Giricz Z, Ibanez B, Iliodromitis E, Kaludercic N, Kleinbongard P, Neuhäuser M, Ovize M, Pagliaro P, Rahbek-Schmidt M, Ruiz-Meana M, Schlüter KD, Schulz R, Skyschally A, Wilder C, Yellon DM, Ferdinandy P, Heusch G. Practical guidelines for rigor and reproducibility in preclinical and clinical studies on cardioprotection. Basic Res Cardiol 2018; 113:39. [PMID: 30120595 PMCID: PMC6105267 DOI: 10.1007/s00395-018-0696-8] [Citation(s) in RCA: 335] [Impact Index Per Article: 47.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/18/2018] [Accepted: 08/03/2018] [Indexed: 02/07/2023]
Affiliation(s)
- Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Palle-Juul Jensens Boulevard 99, 8200, Aarhus N, Denmark.
| | - Derek Hausenloy
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
- The National Institute of Health Research, University College London Hospitals Biomedial Research Centre, Research and Development, London, UK
- National Heart Research Institute Singapore, National Heart Centre, Singapore, Singapore
- Yon Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore, 169857, Singapore
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Salvatore Antonucci
- Department of Biomedical Sciences, CNR Institute of Neuroscience, University of Padova, Via Ugo Bassi 58/B, 35121, Padua, Italy
| | - Kerstin Boengler
- Institute for Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Soni Deshwal
- Department of Biomedical Sciences, CNR Institute of Neuroscience, University of Padova, Via Ugo Bassi 58/B, 35121, Padua, Italy
| | - Yvan Devaux
- Cardiovascular Research Unit, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Fabio Di Lisa
- Department of Biomedical Sciences, CNR Institute of Neuroscience, University of Padova, Via Ugo Bassi 58/B, 35121, Padua, Italy
| | - Moises Di Sante
- Department of Biomedical Sciences, CNR Institute of Neuroscience, University of Padova, Via Ugo Bassi 58/B, 35121, Padua, Italy
| | - Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - David García-Dorado
- Experimental Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), IIS-Fundación Jiménez Díaz, CIBERCV, Madrid, Spain
| | - Efstathios Iliodromitis
- Second Department of Cardiology, Faculty of Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nina Kaludercic
- Department of Biomedical Sciences, CNR Institute of Neuroscience, University of Padova, Via Ugo Bassi 58/B, 35121, Padua, Italy
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Markus Neuhäuser
- Department of Mathematics and Technology, Koblenz University of Applied Science, Remagen, Germany
- Institute for Medical Informatics, Biometry, and Epidemiology, University Hospital Essen, Essen, Germany
| | - Michel Ovize
- Explorations Fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, Lyon, France
- UMR, 1060 (CarMeN), Université Claude Bernard, Lyon1, Villeurbanne, France
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Michael Rahbek-Schmidt
- Department of Cardiology, Aarhus University Hospital, Palle-Juul Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Marisol Ruiz-Meana
- Experimental Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | | | - Rainer Schulz
- Institute for Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Andreas Skyschally
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Catherine Wilder
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany.
| |
Collapse
|
11
|
Fu Y, Jiang W, Zhao Y, Huang Y, Zhang H, Wang H, Pu J. A Simple and Efficient Method for In Vivo Cardiac-specific Gene Manipulation by Intramyocardial Injection in Mice. J Vis Exp 2018. [PMID: 29708533 DOI: 10.3791/57074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Gene manipulation specifically in the heart significantly potentiate the investigation of cardiac disease pathomechanisms and their therapeutic potential. In vivo cardiac-specific gene delivery is commonly achieved by either systemic or local delivery. Systemic injection via tail vein is easy and efficient in manipulating cardiac gene expression by using recombinant adeno-associated virus 9 (AAV9). However, this method requires a relatively high amount of vector for efficient transduction, and may result in nontarget organ gene transduction. Here, we describe a simple, efficient, and time-saving method of intramyocardial injection for in vivo cardiac-specific gene manipulation in mice. Under anesthesia (without ventilation), the pectoral major and minor muscles were bluntly dissected, and the mouse heart was quickly exposed by manual externalization through a small incision at the fourth intercostal space. Subsequently, adenovirus encoding luciferase (Luc) and vitamin D receptor (VDR), or short hairpin RNA (shRNA) targeting VDR, was injected with a Hamilton syringe into the myocardium. Subsequent in vivo imaging demonstrated that luciferase was successfully overexpressed specifically in the heart. Moreover, Western blot analysis confirmed the successful overexpression or silencing of VDR in the mouse heart. Once mastered, this technique can be used for gene manipulation, as well as injection of cells or other materials such as nanogels in the mouse heart.
Collapse
Affiliation(s)
- Yanan Fu
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College; Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University
| | - Wenlong Jiang
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University
| | - Yichao Zhao
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University
| | - Yuli Huang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College
| | - Heng Zhang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College;
| | - Hongju Wang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College
| | - Jun Pu
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College; Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University;
| |
Collapse
|
12
|
Wright LH, Herr DJ, Brown SS, Kasiganesan H, Menick DR. Angiokine Wisp-1 is increased in myocardial infarction and regulates cardiac endothelial signaling. JCI Insight 2018; 3:95824. [PMID: 29467324 DOI: 10.1172/jci.insight.95824] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 01/10/2018] [Indexed: 12/17/2022] Open
Abstract
Myocardial infarctions (MIs) cause the loss of myocytes due to lack of sufficient oxygenation and latent revascularization. Although the administration of histone deacetylase (HDAC) inhibitors reduces the size of infarctions and improves cardiac physiology in small-animal models of MI injury, the cellular targets of the HDACs, which the drugs inhibit, are largely unspecified. Here, we show that WNT-inducible secreted protein-1 (Wisp-1), a matricellular protein that promotes angiogenesis in cancers as well as cell survival in isolated cardiac myocytes and neurons, is a target of HDACs. Further, Wisp-1 transcription is regulated by HDACs and can be modified by the HDAC inhibitor, suberanilohydroxamic acid (SAHA/vorinostat), after MI injury. We observe that, at 7 days after MI, Wisp-1 is elevated 3-fold greater in the border zone of infarction in mice that experience an MI injury and are injected daily with SAHA, relative to MI alone. Additionally, human coronary artery endothelial cells (HCAECs) produce WISP-1 and are responsive to autocrine WISP-1-mediated signaling, which functionally promotes their proangiogenic behavior. Altering endogenous expression of WISP-1 in HCAECs directly impacts their network density in vitro. Therapeutic interventions after a heart attack define the extent of infarct injury, cell survival, and overall prognosis. Our studies shown here identify a potentially novel cardiac angiokine, Wisp-1, that may contribute to beneficial post-MI treatment modalities.
Collapse
Affiliation(s)
| | | | - Symone S Brown
- College of Graduate Studies, Summer Undergraduate Research Program, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | - Donald R Menick
- Division of Cardiology, and.,Ralph H. Johnson VA Medical Center, Charleston, South Carolina, USA
| |
Collapse
|
13
|
Abstract
Ligation of the left anterior descending (LAD) coronary artery in the mouse heart is a widely used model to simulate myocardial infarction and ischemia-reperfusion injury. Here we describe a ligation technique routinely performed in our laboratory to induce myocardial infarction that may be used to study ischemia-reperfusion injury in the myocardium. The methods described enhance location of the LAD coronary artery to allow for accurate ligation, thus increasing reproducibility of infarct size and location.
Collapse
Affiliation(s)
- Zhaobin Xu
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Kevin E McElhanon
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Eric X Beck
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Noah Weisleder
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
14
|
DuSablon A, Parks J, Whitehurst K, Estes H, Chase R, Vlahos E, Sharma U, Wert D, Virag J. EphrinA1-Fc attenuates myocardial ischemia/reperfusion injury in mice. PLoS One 2017; 12:e0189307. [PMID: 29236774 PMCID: PMC5728502 DOI: 10.1371/journal.pone.0189307] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/22/2017] [Indexed: 12/19/2022] Open
Abstract
EphrinA1, a membrane-bound receptor tyrosine kinase ligand expressed in healthy cardiomyocytes, is lost in injured cells following myocardial infarction. Previously, we have reported that a single intramyocardial injection of chimeric ephrinA1-Fc at the time of ischemia reduced injury in the nonreperfused myocardium by 50% at 4 days post-MI by reducing apoptosis and inflammatory cell infiltration. In a clinically relevant model of acute ischemia (30min)/reperfusion (24hr or 4 days) injury, we now demonstrate that ephrinA1-Fc reduces infarct size by 46% and completely preserves cardiac function (ejection fraction, fractional shortening, and chamber dimensions) in the short-term (24hrs post-MI) as well as long-term (4 days). At 24 hours post-MI, diminished serum inflammatory cell chemoattractants in ephrinA1-Fc-treated mice reduces recruitment of neutrophils and leukocytes into the myocardium. Differences in relative expression levels of EphA-Rs are described in the context of their putative role in mediating cardioprotection. Validation by Western blotting of selected targets from mass spectrometry analyses of pooled samples of left ventricular tissue homogenates from mice that underwent 30min ischemia and 24hr of reperfusion (I/R) indicates that ephrinA1-Fc administration alters several regulators of signaling pathways that attenuate apoptosis, promote autophagy, and shift from FA metabolism in favor of increased glycolysis to optimize anaerobic ATP production. Taken together, reduced injury is due a combination of adaptive metabolic reprogramming, improved cell survival, and decreased inflammatory cell recruitment, suggesting that ephrinA1-Fc enhances the capacity of the heart to withstand an ischemic insult.
Collapse
Affiliation(s)
- Augustin DuSablon
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Justin Parks
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - K’Shylah Whitehurst
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Heather Estes
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Robert Chase
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Eleftherios Vlahos
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Uma Sharma
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - David Wert
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Jitka Virag
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
15
|
Hu F, Zhai N, Gao W, Wu P, Luo Y, Pan D, Liu Y, Li D. Outer Balloon Ligation Increases Success Rate of Ischemia-Reperfusion Injury Model in Mice. PLoS One 2016; 11:e0167631. [PMID: 27907155 PMCID: PMC5132321 DOI: 10.1371/journal.pone.0167631] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 11/17/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Coronary artery disease is a growing public health problem and a major cause of morbidity and mortality. Experimental animal models provide valuable tools for studying myocardial ischemia reperfusion (I/R) injury in vivo. OBJECTIVE The purpose of this study was to describe a new method (outer balloon ligation) to induce myocardial I/R injury in mice. METHODS Ninety-nine male C57BL/6J mice were randomly divided into three groups: sham group, classic method group (I/R-C) and the new method group (I/R-N). The surgical procedure and recovery time were recorded. The levels of TNF-α, IL-6, cTnT and LDH were detected by ELISA kits. Hematoxylin-eosin staining was applied to assess neutrophil infiltration. Moreover, surgical survival, myocardial infarction areas, and cardiac function measurements were also recorded. RESULTS The reperfusion operation time in the I/R-N group were markedly less than the I/R-C group (14.73±2.86 vs. 168.60±33.01 sec, p <0.0001). Similarly, the recovery time in I/R-N group was shorter than the I/R-C group (45.39±15.39 vs. 101.70±19.33 min, p <0.0001). The levels of TNF-α and IL-6 in I/R-N group were also markedly lower than in I/R-C group (136.5±22.21 vs. 170.5±24.79 pg/ml, p <0.05 and 100.3±23.74 vs. 144.40±22.24 pg/ml, p <0.001). Compared I/R-N group with I/R-C group, the levels of neutrophil infiltration, cTnT and LDH had no significant differences. Surgical survival rate was 96.7% in the I/R-N group, which was significantly improved compared to the rate of 80% in the I/R-C group. However, there were no significant differences in the areas of myocardial infarction and cardiac function between the two groups. CONCLUSIONS Compared with the classic method, our new method of inducing myocardial I/R injury has higher efficiency and less tissue damage in mice, but achieves the same modeling effects.
Collapse
Affiliation(s)
- Fengwang Hu
- Institute of Cardiovascular Disease, Xuzhou Medical University Xuzhou, Jiangsu, China
| | - Nana Zhai
- Department of Cardiology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wen Gao
- Institute of Cardiovascular Disease, Xuzhou Medical University Xuzhou, Jiangsu, China
| | - Pei Wu
- Institute of Cardiovascular Disease, Xuzhou Medical University Xuzhou, Jiangsu, China
| | - Yuanyuan Luo
- Department of Cardiology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Defeng Pan
- Department of Cardiology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Liu
- Institute of Cardiovascular Disease, Xuzhou Medical University Xuzhou, Jiangsu, China
| | - Dongye Li
- Institute of Cardiovascular Disease, Xuzhou Medical University Xuzhou, Jiangsu, China
| |
Collapse
|
16
|
Akhnokh MK, Yang FH, Samokhvalov V, Jamieson KL, Cho WJ, Wagg C, Takawale A, Wang X, Lopaschuk GD, Hammock BD, Kassiri Z, Seubert JM. Inhibition of Soluble Epoxide Hydrolase Limits Mitochondrial Damage and Preserves Function Following Ischemic Injury. Front Pharmacol 2016; 7:133. [PMID: 27375480 PMCID: PMC4896112 DOI: 10.3389/fphar.2016.00133] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/09/2016] [Indexed: 12/26/2022] Open
Abstract
Aims: Myocardial ischemia can result in marked mitochondrial damage leading to cardiac dysfunction, as such identifying novel mechanisms to limit mitochondrial injury is important. This study investigated the hypothesis that inhibiting soluble epoxide hydrolase (sEH), responsible for converting epoxyeicosatrienoic acids to dihydroxyeicosatrienoic acids protects mitochondrial from injury caused by myocardial infarction. Methods: sEH null and WT littermate mice were subjected to surgical occlusion of the left anterior descending (LAD) artery or sham operation. A parallel group of WT mice received an sEH inhibitor, trans-4-[4-(3-adamantan-1-y1-ureido)-cyclohexyloxy]-benzoic acid (tAUCB; 10 mg/L) or vehicle in the drinking water 4 days prior and 7 days post-MI. Cardiac function was assessed by echocardiography prior- and 7-days post-surgery. Heart tissues were dissected into infarct, peri-, and non-infarct regions to assess ultrastructure by electron microscopy. Complexes I, II, IV, citrate synthase, PI3K activities, and mitochondrial respiration were assessed in non-infarct regions. Isolated working hearts were used to measure the rates of glucose and palmitate oxidation. Results: Echocardiography revealed that tAUCB treatment or sEH deficiency significantly improved systolic and diastolic function post-MI compared to controls. Reduced infarct expansion and less adverse cardiac remodeling were observed in tAUCB-treated and sEH null groups. EM data demonstrated mitochondrial ultrastructure damage occurred in infarct and peri-infarct regions but not in non-infarct regions. Inhibition of sEH resulted in significant improvements in mitochondrial respiration, ATP content, mitochondrial enzymatic activities and restored insulin sensitivity and PI3K activity. Conclusion: Inhibition or genetic deletion of sEH protects against long-term ischemia by preserving cardiac function and maintaining mitochondrial efficiency.
Collapse
Affiliation(s)
- Maria K Akhnokh
- Faculty of Pharmacy and Pharmaceutical Sciences, 2-020M Katz Group Centre for Pharmacy and Health Research, University of Alberta Edmonton, AB, Canada
| | - Feng Hua Yang
- Guangdong Laboratory Animal Monitoring Institute Guangdong, China
| | - Victor Samokhvalov
- Faculty of Pharmacy and Pharmaceutical Sciences, 2-020M Katz Group Centre for Pharmacy and Health Research, University of Alberta Edmonton, AB, Canada
| | - Kristi L Jamieson
- Faculty of Pharmacy and Pharmaceutical Sciences, 2-020M Katz Group Centre for Pharmacy and Health Research, University of Alberta Edmonton, AB, Canada
| | - Woo Jung Cho
- Imaging Core Facility, Faculty of Medicine and Dentistry, University of Alberta Edmonton, AB, Canada
| | - Cory Wagg
- Mazankowski Alberta Heart Institute, University of AlbertaEdmonton, AB, Canada; Department of Pharmacology, Faculty of Medicine and Dentistry, University of AlbertaEdmonton, AB, Canada
| | - Abhijit Takawale
- Mazankowski Alberta Heart Institute, University of AlbertaEdmonton, AB, Canada; Department of Physiology, Faculty of Medicine and Dentistry, University of AlbertaEdmonton, AB, Canada
| | - Xiuhua Wang
- Mazankowski Alberta Heart Institute, University of AlbertaEdmonton, AB, Canada; Department of Physiology, Faculty of Medicine and Dentistry, University of AlbertaEdmonton, AB, Canada
| | - Gary D Lopaschuk
- Mazankowski Alberta Heart Institute, University of AlbertaEdmonton, AB, Canada; Department of Pharmacology, Faculty of Medicine and Dentistry, University of AlbertaEdmonton, AB, Canada; Department of Pediatrics, Faculty of Medicine and Dentistry, University of AlbertaEdmonton, AB, Canada
| | - Bruce D Hammock
- Department of Entomology and Nematology Comprehensive Cancer Center, University of California, Davis Davis, CA, USA
| | - Zamaneh Kassiri
- Mazankowski Alberta Heart Institute, University of AlbertaEdmonton, AB, Canada; Department of Physiology, Faculty of Medicine and Dentistry, University of AlbertaEdmonton, AB, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, 2-020M Katz Group Centre for Pharmacy and Health Research, University of AlbertaEdmonton, AB, Canada; Mazankowski Alberta Heart Institute, University of AlbertaEdmonton, AB, Canada; Department of Pharmacology, Faculty of Medicine and Dentistry, University of AlbertaEdmonton, AB, Canada
| |
Collapse
|
17
|
Shi H, Drummond CA, Fan X, Haller ST, Liu J, Malhotra D, Tian J. Hiding inside? Intracellular expression of non-glycosylated c-kit protein in cardiac progenitor cells. Stem Cell Res 2016; 16:795-806. [PMID: 27161312 DOI: 10.1016/j.scr.2016.04.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/15/2016] [Accepted: 04/22/2016] [Indexed: 12/23/2022] Open
Abstract
Cardiac progenitor cells including c-kit(+) cells and cardiosphere-derived cells (CDCs) play important roles in cardiac repair and regeneration. CDCs were reported to contain only small subpopulations of c-kit(+) cells and recent publications suggested that depletion of the c-kit(+) subpopulation of cells has no effect on regenerative properties of CDCs. However, our current study showed that the vast majority of CDCs from murine heart actually express c-kit, albeit, in an intracellular and non-glycosylated form. Immunostaining and flow cytometry showed that the fluorescent signal indicative of c-kit immunostaining significantly increased when cell membranes were permeabilized. Western blots further demonstrated that glycosylation of c-kit was increased during endothelial differentiation in a time dependent manner. Glycosylation inhibition by 1-deoxymannojirimycin hydrochloride (1-DMM) blocked c-kit glycosylation and reduced expression of endothelial cell markers such as Flk-1 and CD31 during differentiation. Pretreatment of these cells with a c-kit kinase inhibitor (imatinib mesylate) also attenuated Flk-1 and CD31 expression. These results suggest that c-kit glycosylation and its kinase activity are likely needed for these cells to differentiate into an endothelial lineage. In vivo, we found that intracellular c-kit expressing cells are located in the wall of cardiac blood vessels in mice subjected to myocardial infarction. In summary, our work demonstrated for the first time that c-kit is not only expressed in CDCs but may also directly participate in CDC differentiation into an endothelial lineage.
Collapse
Affiliation(s)
- Huilin Shi
- Department of Medicine, Division of Cardiovascular Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Christopher A Drummond
- Department of Medicine, Division of Cardiovascular Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Xiaoming Fan
- Department of Medicine, Division of Cardiovascular Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Steven T Haller
- Department of Medicine, Division of Cardiovascular Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Jiang Liu
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Deepak Malhotra
- Department of Medicine, Division of Cardiovascular Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Jiang Tian
- Department of Medicine, Division of Cardiovascular Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA.
| |
Collapse
|
18
|
de Andrade JNBM, Tang J, Hensley MT, Vandergriff A, Cores J, Henry E, Allen TA, Caranasos TG, Wang Z, Zhang T, Zhang J, Cheng K. Rapid and Efficient Production of Coronary Artery Ligation and Myocardial Infarction in Mice Using Surgical Clips. PLoS One 2015; 10:e0143221. [PMID: 26599500 PMCID: PMC4658059 DOI: 10.1371/journal.pone.0143221] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/02/2015] [Indexed: 11/18/2022] Open
Abstract
Aims The coronary artery ligation model in rodents mimics human myocardial infarction (MI). Normally mechanical ventilation and prolonged anesthesia period are needed. Recently, a method has been developed to create MI by popping-out the heart (without ventilation) followed by immediate suture ligation. Mortality is high due to the time-consuming suture ligation process while the heart is exposed. We sought to improve this method and reduce mortality by rapid coronary ligation using a surgical clip instead of a suture. Methods and Results Mice were randomized into 3 groups: clip MI (CMI), suture MI (SMI), or sham (SHAM). In all groups, heart was manually exposed without intubation through a small incision on the chest wall. Unlike the conventional SMI method, mice in the CMI group received a metal clip on left anterior descending artery (LAD), quickly dispensed by an AutoSuture Surgiclip™. The CMI method took only 1/3 of ligation time of the standard SMI method and improved post-MI survival rate. TTC staining and Masson’s trichrome staining revealed a similar degree of infarct size in the SMI and CMI groups. Echocardiograph confirmed that both SMI and CMI groups had a similar reduction of ejection fraction and fraction shortening over the time. Histological analysis showed that the numbers of CD68+ macrophages and apoptotic cells (TUNEL-positive) are indistinguishable between the two groups. Conclusion This new method, taking only less than 3 minutes to complete, represents an efficient myocardial infarction model in rodents.
Collapse
Affiliation(s)
- James N. B. M. de Andrade
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Junnan Tang
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Michael Taylor Hensley
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Adam Vandergriff
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, United States of America
| | - Jhon Cores
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, United States of America
| | - Eric Henry
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, United States of America
| | - Tyler A. Allen
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Thomas George Caranasos
- Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Zegen Wang
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu, China
| | - Tianxia Zhang
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Jinying Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- * E-mail: (KC); (JZ)
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, United States of America
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu, China
- * E-mail: (KC); (JZ)
| |
Collapse
|
19
|
Masoud WGT, Abo Al-Rob O, Yang Y, Lopaschuk GD, Clanachan AS. Tolerance to ischaemic injury in remodelled mouse hearts: less ischaemic glycogenolysis and preserved metabolic efficiency. Cardiovasc Res 2015; 107:499-508. [PMID: 26150203 DOI: 10.1093/cvr/cvv195] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 06/11/2015] [Indexed: 11/13/2022] Open
Abstract
AIMS Post-infarction remodelled failing hearts have reduced metabolic efficiency. Paradoxically, they have increased tolerance to further ischaemic injury. This study was designed to investigate the metabolic mechanisms that may contribute to this phenomenon and to examine the relationship between ischaemic tolerance and metabolic efficiency during post-ischaemic reperfusion. METHODS AND RESULTS Male C57BL/6 mice were subjected to coronary artery ligation (CAL) or SHAM surgery. After 4 weeks, in vivo mechanical function was assessed by echocardiography, and then isolated working hearts were perfused in this sequence: 45 min aerobic, 15 min global no-flow ischaemia, and 30 min aerobic reperfusion. Left ventricular (LV) function, metabolic rates, and metabolic efficiency were measured. Relative to SHAM, both in vivo and in vitro CAL hearts had depressed cardiac function under aerobic conditions (45 and 36%, respectively), but they had a greater recovery of LV function during post-ischaemic reperfusion (67 vs. 49%, P < 0.05). While metabolic efficiency (LV work per ATP produced) was 50% lower during reperfusion of SHAM hearts, metabolic efficiency in CAL hearts did not decrease. During ischaemia, glycogenolysis was 28% lower in CAL hearts, indicative of lower ischaemic proton production. There were no differences in mitochondrial abundance, calcium handling proteins, or key metabolic enzymes. CONCLUSION Compared with SHAM, remodelled CAL hearts are more tolerant to ischaemic injury and undergo no further deterioration of metabolic efficiency during reperfusion. Less glycogen utilization in CAL hearts during ischaemia may contribute to increased ischaemic tolerance by limiting ischaemic proton production that may improve ion homeostasis during early reperfusion.
Collapse
Affiliation(s)
- Waleed G T Masoud
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, 9-43 Medical Sciences Building, Edmonton, Alberta, Canada T6G 2H7 Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada Cardiovascular Research Centre, Alberta, Canada Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Osama Abo Al-Rob
- Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada Cardiovascular Research Centre, Alberta, Canada Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Yang Yang
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, 9-43 Medical Sciences Building, Edmonton, Alberta, Canada T6G 2H7
| | - Gary D Lopaschuk
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, 9-43 Medical Sciences Building, Edmonton, Alberta, Canada T6G 2H7 Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada Cardiovascular Research Centre, Alberta, Canada Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Alexander S Clanachan
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, 9-43 Medical Sciences Building, Edmonton, Alberta, Canada T6G 2H7 Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada Cardiovascular Research Centre, Alberta, Canada
| |
Collapse
|
20
|
Chang WT, Fisch S, Chen M, Qiu Y, Cheng S, Liao R. Ultrasound based assessment of coronary artery flow and coronary flow reserve using the pressure overload model in mice. J Vis Exp 2015:e52598. [PMID: 25938185 DOI: 10.3791/52598] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Transthoracic Doppler echocardiography (TTDE) is a clinically useful, noninvasive tool for studying coronary artery flow velocity and coronary flow reserve (CFR) in humans. Reduced CFR is accompanied by marked intramyocardial and pericoronary fibrosis and is used as an indication of the severity of dysfunction. This study explores, step-by-step, the real-time changes measured in the coronary flow velocity, CFR and systolic to diastolic peak velocity (S/D) ratio in the setting of an aortic banding model in mice. By using a Doppler transthoracic imaging technique that yields reproducible and reliable data, the method assesses changes in flow in the septal coronary artery (SCA), for a period of over two weeks in mice, that previously either underwent aortic banding or thoracotomy. During imaging, hyperemia in all mice was induced by isoflurane, an anesthetic that increased coronary flow velocity when compared with resting flow. All images were acquired by a single imager. Two ratios, (1) CFR, the ratio between hyperemic and baseline flow velocities, and (2) systolic (S) to diastolic (D) flow were determined, using a proprietary software and by two independent observers. Importantly, the observed changes in coronary flow preceded LV dysfunction as evidenced by normal LV mass and fractional shortening (FS). The method was benchmarked against the current gold standard of coronary assessment, histopathology. The latter technique showed clear pathologic changes in the coronary artery in the form of peri-coronary fibrosis that correlated to the flow changes as assessed by echocardiography. The study underscores the value of using a non-invasive technique to monitor coronary circulation in mouse hearts. The method minimizes redundant use of research animals and demonstrates that advanced ultrasound-based indices, such as CFR and S/D ratios, can serve as viable diagnostic tools in a variety of investigational protocols including drug studies and the study of genetically modified strains.
Collapse
Affiliation(s)
- Wei-Ting Chang
- Cardiac Muscle Research Laboratory, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School; Division of Cardiovascular Medicine, Chi-Mei Medical Center, Tainan
| | - Sudeshna Fisch
- Cardiac Muscle Research Laboratory, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School
| | - Michael Chen
- Cardiac Muscle Research Laboratory, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School
| | - Yiling Qiu
- Cardiac Muscle Research Laboratory, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School
| | - Susan Cheng
- Cardiac Muscle Research Laboratory, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School
| | - Ronglih Liao
- Cardiac Muscle Research Laboratory, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School;
| |
Collapse
|
21
|
Lörchner H, Pöling J, Gajawada P, Hou Y, Polyakova V, Kostin S, Adrian-Segarra JM, Boettger T, Wietelmann A, Warnecke H, Richter M, Kubin T, Braun T. Myocardial healing requires Reg3β-dependent accumulation of macrophages in the ischemic heart. Nat Med 2015; 21:353-62. [PMID: 25751817 DOI: 10.1038/nm.3816] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 02/06/2015] [Indexed: 12/14/2022]
Abstract
Cardiac healing after myocardial ischemia depends on the recruitment and local expansion of myeloid cells, particularly macrophages. Here we identify Reg3β as an essential regulator of macrophage trafficking to the damaged heart. Using mass spectrometry-based secretome analysis, we found that dedifferentiating cardiomyocytes release Reg3β in response to the cytokine OSM, which signals through Jak1 and Stat3. Loss of Reg3β led to a large decrease in the number of macrophages in the ischemic heart, accompanied by increased ventricular dilatation and insufficient removal of neutrophils. This defect in neutrophil removal in turn caused enhanced matrix degradation, delayed collagen deposition and increased susceptibility to cardiac rupture. Our data indicate that OSM, acting through distinct intracellular pathways, regulates both cardiomyocyte dedifferentiation and cardiomyocyte-dependent regulation of macrophage trafficking. Release of OSM from infiltrating neutrophils and macrophages initiates a positive feedback loop in which OSM-induced production of Reg3β in cardiomyocytes attracts additional OSM-secreting macrophages. The activity of the feedback loop controls the degree of macrophage accumulation in the heart, which is instrumental in myocardial healing.
Collapse
Affiliation(s)
- Holger Lörchner
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Jochen Pöling
- 1] Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany. [2] Department of Cardiac Surgery, Schüchtermann-Clinic, Bad Rothenfelde, Germany
| | - Praveen Gajawada
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Yunlong Hou
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Viktoria Polyakova
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Sawa Kostin
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Juan M Adrian-Segarra
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thomas Boettger
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Astrid Wietelmann
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Henning Warnecke
- Department of Cardiac Surgery, Schüchtermann-Clinic, Bad Rothenfelde, Germany
| | - Manfred Richter
- Department of Cardiac Surgery, Kerckhoff-Klinik, Bad Nauheim, Germany
| | - Thomas Kubin
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
22
|
Darbandi Azar A, Tavakoli F, Moladoust H, Zare A, Sadeghpour A. Echocardiographic evaluation of cardiac function in ischemic rats: value of m-mode echocardiography. Res Cardiovasc Med 2014; 3:e22941. [PMID: 25785251 PMCID: PMC4347793 DOI: 10.5812/cardiovascmed.22941] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/08/2014] [Indexed: 11/16/2022] Open
Abstract
Background: Echocardiography is a well-established diagnostic tool for a safe, reproducible and accurate evaluation of cardiac anatomy, hemodynamics and function in clinical practice. Objectives: We sought to demonstrate the efficacy and feasibility of M-mode echocardiography to evaluate cardiac structure and function in normal and MI-induced adult rats. Materials and Methods: All animal procedures were approved by the ethics committee of Tehran University of Medical Sciences and the investigation conformed to the “Guide for the Care and Use of Laboratory Animals” published by the United States National Institutes of Health. Forty-eight male Wistar rats weighing 280-300 grams were obtained from a single breeding colony. The statistical analyses were performed using SPSS 20.0. Results: Echocardiographic measurements were possible in all rats before and after the operation. In our survey, we studied echocardiographic alterations in rats after MI induction. Changes can be seen in all echocardiographic mean values after myocardial infarction (MI), but significant decrease (P < 0.01) of Fractional shortening and Ejection Fraction as well as significant increase (P < 0.05) of end systolic diameter and systolic volume after left anterior descending coronary artery (LAD) ligation can be good signs of MI induction. Conclusions: In light of our results, it can be concluded that we succeeded in establishing a precise echocardiographic method to confidently assess the success of LAD ligation surgery in rats. It is feasible to thoroughly monitor the functional efficiency of regional therapeutic interventions such as intra-myocardial stem cell injection.
Collapse
Affiliation(s)
- Amir Darbandi Azar
- Rajaie Cardiovascular, Medical and Research Centre, Iran University of Medical Sciences, Tehran, IR Iran
| | - Fatemeh Tavakoli
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Hassan Moladoust
- Department of Medical Physics and Biomedical Engineering, Guilan University of Medical Sciences, Rasht, IR Iran
| | - Asghar Zare
- Rajaie Cardiovascular, Medical and Research Centre, Iran University of Medical Sciences, Tehran, IR Iran
| | - Anita Sadeghpour
- Echocardiography Research Center, Department of Cardiovascular Medicine, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, IR Iran
- Corresponding author: Anita Sadeghpour, Echocardiography Research Center, Department of Cardiovascular Medicine, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, IR Iran. Tel: +98-2123922145, E-mail:
| |
Collapse
|
23
|
DuSablon A, Kent S, Coburn A, Virag J. EphA2-receptor deficiency exacerbates myocardial infarction and reduces survival in hyperglycemic mice. Cardiovasc Diabetol 2014; 13:114. [PMID: 25166508 PMCID: PMC4147179 DOI: 10.1186/s12933-014-0114-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 07/12/2014] [Indexed: 02/04/2023] Open
Abstract
Background We have previously shown that EphrinA1/EphA expression profile changes in response to myocardial infarction (MI), exogenous EphrinA1-Fc administration following MI positively influences wound healing, and that deletion of the EphA2 Receptor (EphA2-R) exacerbates injury and remodeling. To determine whether or not ephrinA1-Fc would be of therapeutic value in the hyperglycemic infarcted heart, it is critical to evaluate how ephrinA1/EphA signaling changes in the hyperglycemic myocardium in response to MI. Methods Streptozotocin (STZ)-induced hyperglycemia in wild type (WT) and EphA2-receptor mutant (EphA2-R-M) mice was initiated by an intraperitoneal injection of STZ (150 mg/kg) 10 days before surgery. MI was induced by permanent ligation of the left anterior descending coronary artery and analyses were performed at 4 days post-MI. ANOVAs with Student-Newman Keuls multiple comparison post-hoc analysis illustrated which groups were significantly different, with significance of at least p < 0.05. Results Both WT and EphA2-R-M mice responded adversely to STZ, but only hyperglycemic EphA2-R-M mice had lower ejection fraction (EF) and fractional shortening (FS). At 4 days post-MI, we observed greater post-MI mortality in EphA2-R-M mice compared with WT and this was greater still in the EphA2-R-M hyperglycemic mice. Although infarct size was greater in hyperglycemic WT mice vs normoglycemic mice, there was no difference between hyperglycemic EphA2-R-M mice and normoglycemic EphA2-R-M mice. The hypertrophic response that normally occurs in viable myocardium remote to the infarct was noticeably absent in epicardial cardiomyocytes and cardiac dysfunction worsened in hyperglycemic EphA2-R-M hearts post-MI. The characteristic interstitial fibrotic response in the compensating myocardium remote to the infarct also did not occur in hyperglycemic EphA2-R-M mouse hearts to the same extent as that observed in the hyperglycemic WT mouse hearts. Differences in neutrophil and pan-leukocyte infiltration and serum cytokines implicate EphA2-R in modulation of injury and the differences in ephrinA1 and EphA6-R expression in governing this are discussed. Conclusions We conclude that EphA2-mutant mice are more prone to hyperglycemia-induced increased injury, decreased survival, and worsened LV remodeling due to impaired wound healing.
Collapse
|
24
|
Grieve SM, Mazhar J, Callaghan F, Kok CY, Tandy S, Bhindi R, Figtree GA. Automated quantification of myocardial salvage in a rat model of ischemia-reperfusion injury using 3D high-resolution magnetic resonance imaging (MRI). J Am Heart Assoc 2014; 3:jah3614. [PMID: 25146703 PMCID: PMC4310382 DOI: 10.1161/jaha.114.000956] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background Quantification of myocardial “area at risk” (AAR) and myocardial infarction (MI) zone is critical for assessing novel therapies targeting myocardial ischemia–reperfusion (IR) injury. Current “gold‐standard” methods perfuse the heart with Evan's Blue and stain with triphenyl tetrazolium chloride (TTC), requiring manual slicing and analysis. We aimed to develop and validate a high‐resolution 3‐dimensional (3D) magnetic resonance imaging (MRI) method for quantifying MI and AAR. Methods and Results Forty‐eight hours after IR was induced, rats were anesthetized and gadopentetate dimeglumine was administered intravenously. After 10 minutes, the coronary artery was re‐ligated and a solution containing iron oxide microparticles and Evan's Blue was infused (for comparison). Hearts were harvested and transversally sectioned for TTC staining. Ex vivo MR images of slices were acquired on a 9.4‐T magnet. T2* data allowed visualization of AAR, with microparticle‐associated signal loss in perfused regions. T1 data demonstrated gadolinium retention in infarcted zones. Close correlation (r=0.92 to 0.94; P<0.05) of MRI and Evan's Blue/TTC measures for both AAR and MI was observed when the combined techniques were applied to the same heart slice. However, 3D MRI acquisition and analysis of whole heart reduced intra‐observer variability compared to assessment of isolated slices, and allowed automated segmentation and analysis, thus reducing interobserver variation. Anatomical resolution of 81 μm3 was achieved (versus ≈2 mm with manual slicing). Conclusions This novel, yet simple, MRI technique allows precise assessment of infarct and AAR zones. It removes the need for tissue slicing and provides opportunity for 3D digital analysis at high anatomical resolution in a streamlined manner accessible for all laboratories already performing IR experiments.
Collapse
Affiliation(s)
- Stuart M Grieve
- North Shore Heart Research Group, Kolling Institute, University of Sydney, Australia (S.M.G., J.M., C.Y.K., S.T., R.B., G.A.F.) Sydney Translational Imaging Laboratory, Sydney Medical School, University of Sydney, Australia (S.M.G.) Department of Radiology, Royal Prince Alfred Hospital, Sydney, Australia (S.M.G., F.C.)
| | - Jawad Mazhar
- North Shore Heart Research Group, Kolling Institute, University of Sydney, Australia (S.M.G., J.M., C.Y.K., S.T., R.B., G.A.F.) Department of Cardiology, Royal North Shore Hospital, Sydney, Australia (J.M., R.B., G.A.F.)
| | - Fraser Callaghan
- Department of Radiology, Royal Prince Alfred Hospital, Sydney, Australia (S.M.G., F.C.)
| | - Cindy Y Kok
- North Shore Heart Research Group, Kolling Institute, University of Sydney, Australia (S.M.G., J.M., C.Y.K., S.T., R.B., G.A.F.)
| | - Sarah Tandy
- North Shore Heart Research Group, Kolling Institute, University of Sydney, Australia (S.M.G., J.M., C.Y.K., S.T., R.B., G.A.F.)
| | - Ravinay Bhindi
- North Shore Heart Research Group, Kolling Institute, University of Sydney, Australia (S.M.G., J.M., C.Y.K., S.T., R.B., G.A.F.) Department of Cardiology, Royal North Shore Hospital, Sydney, Australia (J.M., R.B., G.A.F.)
| | - Gemma A Figtree
- North Shore Heart Research Group, Kolling Institute, University of Sydney, Australia (S.M.G., J.M., C.Y.K., S.T., R.B., G.A.F.) Department of Cardiology, Royal North Shore Hospital, Sydney, Australia (J.M., R.B., G.A.F.)
| |
Collapse
|
25
|
Xu Z, Alloush J, Beck E, Weisleder N. A murine model of myocardial ischemia-reperfusion injury through ligation of the left anterior descending artery. J Vis Exp 2014. [PMID: 24747599 DOI: 10.3791/51329] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Acute or chronic myocardial infarction (MI) are cardiovascular events resulting in high morbidity and mortality. Establishing the pathological mechanisms at work during MI and developing effective therapeutic approaches requires methodology to reproducibly simulate the clinical incidence and reflect the pathophysiological changes associated with MI. Here, we describe a surgical method to induce MI in mouse models that can be used for short-term ischemia-reperfusion (I/R) injury as well as permanent ligation. The major advantage of this method is to facilitate location of the left anterior descending artery (LAD) to allow for accurate ligation of this artery to induce ischemia in the left ventricle of the mouse heart. Accurate positioning of the ligature on the LAD increases reproducibility of infarct size and thus produces more reliable results. Greater precision in placement of the ligature will improve the standard surgical approaches to simulate MI in mice, thus reducing the number of experimental animals necessary for statistically relevant studies and improving our understanding of the mechanisms producing cardiac dysfunction following MI. This mouse model of MI is also useful for the preclinical testing of treatments targeting myocardial damage following MI.
Collapse
Affiliation(s)
- Zhaobin Xu
- Davis Heart & Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University
| | - Jenna Alloush
- Davis Heart & Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University
| | - Eric Beck
- Davis Heart & Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University
| | - Noah Weisleder
- Davis Heart & Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University;
| |
Collapse
|
26
|
Virag JAI, Anderson EJ, Kent SD, Blanton HD, Johnson TL, Moukdar F, DeAntonio JH, Thayne K, Ding JM, Lust RM. Cardioprotection via preserved mitochondrial structure and function in the mPer2-mutant mouse myocardium. Am J Physiol Heart Circ Physiol 2013; 305:H477-83. [PMID: 23771689 DOI: 10.1152/ajpheart.00914.2012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
We have previously shown that myocardial infarct size in nonreperfused hearts of mice with a functional deletion of the circadian rhythm gene mPer2 (mPer2-M) was reduced by 43%. We hypothesized that acute ischemia-reperfusion injury (I/R = 30 min I/2 h R) would also be reduced in these mice and that ischemic preconditioning (IPC) (3 × 5 min cycles) before I/R, which enhances protection in wild-type (WT) hearts, would provide further protection in mPer2-M hearts. We observed a 69 and 75% decrease in infarct size in mPer2-M mouse hearts compared with WT following I/R and IPC, respectively. This was coincident with 67% less neutrophil infiltration and 57% less apoptotic cardiomyocytes. IPC in mPer2-M mice before I/R had 48% less neutrophil density and 46% less apoptosis than their WT counterparts. Macrophage density was not different between WT and mPer2-M I/R, but it was 45% higher in mPer2-M IPC mouse hearts compared with WT IPC. There were no baseline differences in cardiac mitochondrial function between WT and mPer2-M mice, but, following I/R, WT exhibited a marked decrease in maximal O₂ consumption supported by complex I-mediated substrates, whereas mPer2-M did not, despite no difference in complex I content. Moreover, cardiac mitochondria from WT mice exhibited a very robust increase in ADP-stimulated O₂ consumption in response to exogenously added cytochrome c, along with a high rate of reactive oxygen species production, none of which was exhibited by cardiac mitochondria from mPer2-M following I/R. Taken together, these findings suggest that mPer2 deletion preserves mitochondrial membrane structure and functional integrity in heart following I/R injury, the consequence of which is preservation of myocardial viability. Understanding the mechanisms connecting cardiac events, mitochondrial function, and mPer2 could lead to preventative and therapeutic strategies for at risk populations.
Collapse
Affiliation(s)
- Jitka A I Virag
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
O'Neal WT, Griffin WF, Dries-Devlin JL, Kent SD, Chen J, Willis MS, Virag JAI. Ephrin-Eph signaling as a potential therapeutic target for the treatment of myocardial infarction. Med Hypotheses 2013; 80:738-44. [PMID: 23562676 DOI: 10.1016/j.mehy.2013.02.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 01/27/2013] [Accepted: 02/28/2013] [Indexed: 01/22/2023]
Abstract
Although numerous strategies have been developed to reduce the initial ischemic insult and cellular injury that occurs during myocardial infarction (MI), few have progressed into the clinical arena. The epidemiologic and economic impact of MI necessitates the development of innovative therapies to rapidly and effectively reduce the initial injury and subsequent cardiac dysfunction. The Eph receptors and their cognate ligands, the ephrins, are the largest family of receptor tyrosine kinases, and their signaling has been shown to play a diverse role in various cellular processes. The recent advances in the study of ephrin-Eph signaling have shown promising progress in many fields of medicine. They have been implicated in the pathophysiology of various cancers and in the regulation of inflammation and apoptosis. Recent studies have shown that manipulation of ephrin-Eph cell signaling can favorably influence cardiomyocyte viability and ultimately preserve cardiac function post-MI. In this article, we explore the hypothesis that manipulation of ephrin-Eph signaling may potentially be a novel therapeutic target in the treatment of MI through alteration of the cellular processes that govern injury and wound healing.
Collapse
Affiliation(s)
- Wesley T O'Neal
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | | | | | | | | | | | | |
Collapse
|