1
|
Yehia A, Zaki A, Sadaka M, Azeem AMAE. Incremental prognostic value of speckle tracking echocardiography and early follow-up echo assessment in predicting left ventricular recovery after reperfusion for ST-segment elevation myocardial infarction (STEMI). Echocardiography 2024; 41:e15725. [PMID: 38078679 DOI: 10.1111/echo.15725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/02/2023] [Accepted: 11/26/2023] [Indexed: 01/30/2024] Open
Abstract
PURPOSE Up to 50% of patients do not achieve significant left ventricular ejection fraction (LVEF) recovery after primary percutaneous intervention (PPCI) for STEMI. We aimed to identify the echocardiographic predictors for LVEF recovery and assess the value of early follow-up echocardiography (Echo) in risk assessment of post-myocardial infarction (MI) patients. METHODS One hundred one STEMI patients undergoing PPCI were enrolled provided EF below 50%. Baseline echocardiography assessed LVEF, volumes, wall motion score index (WMSI), global longitudinal strain (GLS), global circumferential strain (GCS), and E/e'. Follow-up echocardiography after 6 weeks reassessed left ventricular volumes, LVEF and GLS.GCS was not assessed at follow up. Patients were classified into recovery and non-recovery groups. Predictors of LVEF recovery and major adverse cardiovascular events (MACE) at 6 months were analysed. RESULTS The mean change of EF was 8.04 ± 3.32% in group I versus -.39 ± 5.09 % in group II (p < .001). Recovered patients had better baseline GLS, baseline GCS, E/e', and follow-up GLS. Multivariate regression analysis revealed E/e', GCS, and follow-up GLS after 6 weeks to be strong independent predictors for LVEF recovery. Composite MACE was considerably higher in group II (32.7% vs. 4.1%, p < .001) mainly driven by higher heart failure hospitalisation Multivariate regression analysis revealed baseline GLS, E/e', and ejection fraction (EF) percentage recovery as strong independent predictors for MACE. CONCLUSIONS Multiparametric echocardiographic approach incorporating LVEF, strain parameters, and diastolic function could allow early optimal risk stratification after STEMI treated with PPCI. Follow-up GLS and LVEF percentage change are the strongest predictors for early LV recovery and long term clinical outcome, respectively.
Collapse
Affiliation(s)
- Ahmed Yehia
- Cardiology and Angiology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Amr Zaki
- Cardiology and Angiology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mohamed Sadaka
- Cardiology and Angiology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | | |
Collapse
|
2
|
Gum Arabic protects the rat heart from ischemia/reperfusion injury through anti-inflammatory and antioxidant pathways. Sci Rep 2022; 12:17235. [PMID: 36241904 PMCID: PMC9568585 DOI: 10.1038/s41598-022-22097-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 10/10/2022] [Indexed: 01/06/2023] Open
Abstract
Gum Arabic (GA) is a plant exudate with antioxidant and anti-inflammatory effects. GA has shown promise in protection from and treatment of kidney failure, however, its role in the protection of the heart from ischemia and reperfusion (I/R) has not been investigated. This study investigated the antioxidant and anti-inflammatory effects of Gum Arabic (GA) in the protection of the heart against ischemia/reperfusion (I/R) injury. Langendorff-perfused Wistar rat hearts were divided into seven groups. One group which was subjected to I/R with no other treatment served as the control group. The second group was subjected to buffer perfusion with no ischemia (sham group). The third group was perfused with GA in the absence of ischemia (sham + GA). The rest of the hearts were isolated from rats that had been treated with GA for 4 or 2 weeks in the drinking water, or GA that had been infused intravenously 2 h before sacrifice or added to perfusion buffer at reperfusion. Hemodynamics data were digitally computed; infarct size was measured using 2,3,5-triphenyltetrazolium chloride (TTC) staining and cardiomyocyte injury was assessed by quantifying creatine kinase (CK) and lactate dehydrogenase (LDH) enzymes. The total oxidants (TOS) and antioxidants (TAS), superoxide dismutase (SOD) and pro- and anti-inflammatory cytokines levels were estimated by ELISA. GA treatment for 2 weeks, 4 weeks or 2 hours before sacrifice resulted in a significant (P < 0.05) improvement in cardiac hemodynamics and reduction in infarct size and cardiac enzyme levels compared to respective controls. However, GA administration at the time of reperfusion did not protect the hearts against I/R injury. Furthermore, GA treatment decreased the pro-inflammatory and anti-inflammatory cytokines levels. The levels of TOS in the effluent were significantly decreased (P < 0.05) and SOD levels were significantly (P < 0.05) increased by GA administration. GA protected the heart against I/R injury when administered for 2 or 4 weeks or when infused 2 hours before sacrifice. GA treatment decreased the total oxidants levels, the pro-inflammatory cytokines TNF-α, IL-1β and IL-6 protein levels and increases SOD and anti-inflammatory cytokine IL-10 protein levels.
Collapse
|
3
|
Canty JM. Myocardial Injury, Troponin Release and Cardiomyocyte Death in Brief Ischemia, Failure and Ventricular Remodeling. Am J Physiol Heart Circ Physiol 2022; 323:H1-H15. [PMID: 35559722 DOI: 10.1152/ajpheart.00093.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Troponin released from irreversibly injured myocytes is the gold standard biomarker for the rapid identification of an acute coronary syndrome. In acute myocardial infarction, necrotic cell death is characterized by sarcolemmal disruption in response to a critical level of energy depletion after more than 15-minutes of ischemia. While troponin I and T are highly specific for cardiomyocyte death, high-sensitivity assays have demonstrated that measurable circulating levels of troponin are present in the majority of normal subjects. In addition, transient as well as chronic elevations have been demonstrated in many disease states not clearly associated with myocardial ischemia. The latter observations have given rise to the clinical concept of myocardial injury. This review will summarize evidence supporting the notion that circulating troponin levels parallel the extent of myocyte apoptosis in normal ventricular remodeling and in pathophysiological conditions not associated with infarction or necrosis. It will review the evidence that myocyte apoptosis can be accelerated by both diastolic strain from elevated ventricular preload as well as systolic strain from dyskinesis after brief episodes of ischemia too short to cause a critical level of myocyte energy depletion. We then show how chronic, low rates of myocyte apoptosis from endogenous myocyte turnover, repetitive ischemia or repetitive elevations in LV diastolic pressure can lead to significant myocyte loss in the absence of neurohormonal stimulation. Finally, we posit that the differential response to strain-induced injury in heart failure may determine whether progressive myocyte loss and HFrEF or interstitial fibrosis and HFpEF become the heart failure phenotype.
Collapse
Affiliation(s)
- John M Canty
- VA WNY Health Care System, the Departments of Medicine, Physiology & Biophysics, Biomedical Engineering and The Clinical and Translational Research Center of the University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
4
|
Fan L, Choy JS, Raissi F, Kassab GS, Lee LC. Optimization of cardiac resynchronization therapy based on a cardiac electromechanics-perfusion computational model. Comput Biol Med 2022; 141:105050. [PMID: 34823858 PMCID: PMC8810745 DOI: 10.1016/j.compbiomed.2021.105050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 02/03/2023]
Abstract
Cardiac resynchronization therapy (CRT) is an established treatment for left bundle branch block (LBBB) resulting in mechanical dyssynchrony. Approximately 1/3 of patients with CRT, however, are non-responders. To understand factors affecting CRT response, an electromechanics-perfusion computational model based on animal-specific left ventricular (LV) geometry and coronary vascular networks located in the septum and LV free wall is developed. The model considers contractility-flow and preload-activation time relationships, and is calibrated to simultaneously match the experimental measurements in terms of the LV pressure, volume waveforms and total coronary flow in the left anterior descending and left circumflex territories from 2 swine models under right atrium and right ventricular pacing. The model is then applied to investigate the responses of CRT indexed by peak LV pressure and (dP/dt)max at multiple pacing sites with different degrees of perfusion in the LV free wall. Without the presence of ischemia, the model predicts that basal-lateral endocardial region is the optimal pacing site that can best improve (dP/dt)max by 20%, and is associated with the shortest activation time. In the presence of ischemia, a non-ischemic region becomes the optimal pacing site when coronary flow in the ischemic region fell below 30% of its original value. Pacing at the ischemic region produces little response at that perfusion level. The optimal pacing site is associated with one that optimizes the LV activation time. These findings suggest that CRT response is affected by both pacing site and coronary perfusion, which may have clinical implication in improving CRT responder rates.
Collapse
Affiliation(s)
- Lei Fan
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, USA.
| | - Jenny S Choy
- California Medical Innovations Institute, San Diego, CA, USA
| | - Farshad Raissi
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | | | - Lik Chuan Lee
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
5
|
Ryan M, Morgan H, Chiribiri A, Nagel E, Cleland J, Perera D. Myocardial viability testing: all STICHed up, or about to be REVIVED? Eur Heart J 2022; 43:118-126. [PMID: 34791132 PMCID: PMC8757581 DOI: 10.1093/eurheartj/ehab729] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/14/2021] [Accepted: 09/30/2021] [Indexed: 01/09/2023] Open
Abstract
Patients with ischaemic left ventricular dysfunction frequently undergo myocardial viability testing. The historical model presumes that those who have extensive areas of dysfunctional-yet-viable myocardium derive particular benefit from revascularization, whilst those without extensive viability do not. These suppositions rely on the theory of hibernation and are based on data of low quality: taking a dogmatic approach may therefore lead to patients being refused appropriate, prognostically important treatment. Recent data from a sub-study of the randomized STICH trial challenges these historical concepts, as the volume of viable myocardium failed to predict the effectiveness of coronary artery bypass grafting. Should the Heart Team now abandon viability testing, or are new paradigms needed in the way we interpret viability? This state-of-the-art review critically examines the evidence base for viability testing, focusing in particular on the presumed interactions between viability, functional recovery, revascularization and prognosis which underly the traditional model. We consider whether viability should relate solely to dysfunctional myocardium or be considered more broadly and explore wider uses of viability testingoutside of revascularization decision-making. Finally, we look forward to ongoing and future randomized trials, which will shape evidence-based clinical practice in the future.
Collapse
Affiliation(s)
- Matthew Ryan
- School of Cardiovascular Medicine and Sciences, King’s College London, Westminster Bridge Road, London SE1 7EH, UK
| | - Holly Morgan
- School of Cardiovascular Medicine and Sciences, King’s College London, Westminster Bridge Road, London SE1 7EH, UK
| | - Amedeo Chiribiri
- School of Biomedical Engineering and Imaging Sciences, King’s College London, Westminster Bridge Road, London SE1 7EH, UK
| | - Eike Nagel
- Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - John Cleland
- Robertson Centre for Biostatistics, University of Glasgow, University Avenue, Glasgow G12 8QQ, UK
| | - Divaka Perera
- School of Cardiovascular Medicine and Sciences, King’s College London, Westminster Bridge Road, London SE1 7EH, UK
| |
Collapse
|
6
|
Brandt MM, Cheng C, Merkus D, Duncker DJ, Sorop O. Mechanobiology of Microvascular Function and Structure in Health and Disease: Focus on the Coronary Circulation. Front Physiol 2022; 12:771960. [PMID: 35002759 PMCID: PMC8733629 DOI: 10.3389/fphys.2021.771960] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
The coronary microvasculature plays a key role in regulating the tight coupling between myocardial perfusion and myocardial oxygen demand across a wide range of cardiac activity. Short-term regulation of coronary blood flow in response to metabolic stimuli is achieved via adjustment of vascular diameter in different segments of the microvasculature in conjunction with mechanical forces eliciting myogenic and flow-mediated vasodilation. In contrast, chronic adjustments in flow regulation also involve microvascular structural modifications, termed remodeling. Vascular remodeling encompasses changes in microvascular diameter and/or density being largely modulated by mechanical forces acting on the endothelium and vascular smooth muscle cells. Whereas in recent years, substantial knowledge has been gathered regarding the molecular mechanisms controlling microvascular tone and how these are altered in various diseases, the structural adaptations in response to pathologic situations are less well understood. In this article, we review the factors involved in coronary microvascular functional and structural alterations in obstructive and non-obstructive coronary artery disease and the molecular mechanisms involved therein with a focus on mechanobiology. Cardiovascular risk factors including metabolic dysregulation, hypercholesterolemia, hypertension and aging have been shown to induce microvascular (endothelial) dysfunction and vascular remodeling. Additionally, alterations in biomechanical forces produced by a coronary artery stenosis are associated with microvascular functional and structural alterations. Future studies should be directed at further unraveling the mechanisms underlying the coronary microvascular functional and structural alterations in disease; a deeper understanding of these mechanisms is critical for the identification of potential new targets for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Maarten M Brandt
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Caroline Cheng
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Walter Brendel Center of Experimental Medicine (WBex), LMU Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
7
|
Yang J, Tang L, Zhang F, Yang T, Lu T, Sun K, Sun N, Ren J, Yan M. Sevoflurane preconditioning promotes mesenchymal stem cells to relieve myocardial ischemia/reperfusion injury via TRPC6-induced angiogenesis. Stem Cell Res Ther 2021; 12:584. [PMID: 34809715 PMCID: PMC8607627 DOI: 10.1186/s13287-021-02649-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/06/2021] [Indexed: 01/09/2023] Open
Abstract
Background Ischemic heart diseases is one of the leading causes of death worldwide. Although revascularization timely is an effective therapeutic intervention to salvage the ischemic myocardium, reperfusion itself causes additional myocardial injury called ischemia/reperfusion (I/R) injury. Bone marrow-derived mesenchymal stem cells (MSCs) is one of the promising cells to alleviate ischemic myocardial injury. However, this cell therapy is limited by poor MSCs survival after transplantation. Here, we investigated whether sevoflurane preconditioning could promote MSCs to attenuate myocardial I/R injury via transient receptor potential canonical channel 6 (TRPC6)-induced angiogenesis. Methods The anti-apoptotic effect of sevoflurane preconditioning on MSCs was determined by Annexin V-FITC/propidium iodide staining. TRPC6, hypoxia-inducible factor-1α (HIF-1α), Chemokine receptor 4 (CXCR4) and vascular endothelial growth factor (VEGF) protein expressions and VEGF release from MSCs were determined after hypoxia and reoxygenation (H/R). Small interfering RNA (siRNA) was used to knock down TRPC6 gene expression in MSCs. The angiogenesis of human umbilical vein endothelial cells (HUVECs) co-cultured with MSCs was determined by Matrigel tube formation. Myocardial I/R mouse model was induced by occluding left anterior descending coronary artery for 30 min and then reperfusion. MSCs or sevoflurane preconditioned MSCs were injected around the ligature border zone 5 min before reperfusion. Left ventricle systolic function, infarction size, serum LDH, cTnI and inflammatory cytokines were determined after reperfusion. Results Sevoflurane preconditioning up-regulated TRPC6, HIF-1α, CXCR4 and VEGF expressions in MSCs and VEGF release from MSCs under H/R, which were reversed by knockdown of TRPC6 gene using siRNA in MSCs. Furthermore, sevoflurane preconditioning promoted the angiogenic and anti-inflammatory effect of HUVECs co-cultured with MSCs. Sevoflurane preconditioned MSCs improved left ventricle systolic function and alleviated myocardial infarction and inflammation in mice subjected to I/R insult. Conclusion The current findings reveal that sevoflurane preconditioned MSCs boost angiogenesis in HUVECs subjected to H/R insult and attenuate myocardial I/R injury, which may be mediated by TRPC6 up-regulated HIF-1α, CXCR4 and VEGF. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02649-3.
Collapse
Affiliation(s)
- Jinting Yang
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Lihui Tang
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Fengjiang Zhang
- Clinical Skill Training Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Tingting Yang
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, 710061, China
| | - Ting Lu
- Clinical Research Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Kai Sun
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Na Sun
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Jinxuan Ren
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Min Yan
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
8
|
Zhou H, Ren J, Toan S, Mui D. Role of mitochondrial quality surveillance in myocardial infarction: From bench to bedside. Ageing Res Rev 2021; 66:101250. [PMID: 33388396 DOI: 10.1016/j.arr.2020.101250] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/10/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
Myocardial infarction (MI) is the irreversible death of cardiomyocyte secondary to prolonged lack of oxygen or fresh blood supply. Historically considered as merely cardiomyocyte powerhouse that manufactures ATP and other metabolites, mitochondrion is recently being identified as a signal regulator that is implicated in the crosstalk and signal integration of cardiomyocyte contraction, metabolism, inflammation, and death. Mitochondria quality surveillance is an integrated network system modifying mitochondrial structure and function through the coordination of various processes including mitochondrial fission, fusion, biogenesis, bioenergetics, proteostasis, and degradation via mitophagy. Mitochondrial fission favors the elimination of depolarized mitochondria through mitophagy, whereas mitochondrial fusion preserves the mitochondrial network upon stress through integration of two or more small mitochondria into an interconnected phenotype. Mitochondrial biogenesis represents a regenerative program to replace old and damaged mitochondria with new and healthy ones. Mitochondrial bioenergetics is regulated by a metabolic switch between glucose and fatty acid usage, depending on oxygen availability. To maintain the diversity and function of mitochondrial proteins, a specialized protein quality control machinery regulates protein dynamics and function through the activity of chaperones and proteases, and induction of the mitochondrial unfolded protein response. In this review, we provide an overview of the molecular mechanisms governing mitochondrial quality surveillance and highlight the most recent preclinical and clinical therapeutic approaches to restore mitochondrial fitness during both MI and post-MI heart failure.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Cardiology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China.
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Sam Toan
- Department of Chemical Engineering, University of Minnesota-Duluth, Duluth, MN 55812, USA
| | - David Mui
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
9
|
Abstract
Unlike acute myocardial infarction with reperfusion, in which infarct size is the end point reflecting irreversible injury, myocardial stunning and hibernation result from reversible myocardial ischaemia-reperfusion injury, and contractile dysfunction is the obvious end point. Stunned myocardium is characterized by a disproportionately long-lasting, yet fully reversible, contractile dysfunction that follows brief bouts of myocardial ischaemia. Reperfusion precipitates a burst of reactive oxygen species formation and alterations in excitation-contraction coupling, which interact and cause the contractile dysfunction. Hibernating myocardium is characterized by reduced regional contractile function and blood flow, which both recover after reperfusion or revascularization. Short-term myocardial hibernation is an adaptation of contractile function to the reduced blood flow such that energy and substrate metabolism recover during the ongoing ischaemia. Chronic myocardial hibernation is characterized by severe morphological alterations and altered expression of metabolic and pro-survival proteins. Myocardial stunning is observed clinically and must be recognized but is rarely haemodynamically compromising and does not require treatment. Myocardial hibernation is clinically identified with the use of imaging techniques, and the myocardium recovers after revascularization. Several trials in the past two decades have challenged the superiority of revascularization over medical therapy for symptomatic relief and prognosis in patients with chronic coronary syndromes. A better understanding of the pathophysiology of myocardial stunning and hibernation is important for a more precise indication of revascularization and its consequences. Therefore, this Review summarizes the current knowledge of the pathophysiology of these characteristic reperfusion phenomena and highlights their clinical implications.
Collapse
|
10
|
Pei J, Wang X, Xing Z, Zheng K, Hu X. Short-term and long-term outcomes of revascularization interventions for patients with severely reduced left ventricular ejection fraction: a meta-analysis. ESC Heart Fail 2020; 8:634-643. [PMID: 33274612 PMCID: PMC7835572 DOI: 10.1002/ehf2.13141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/01/2020] [Accepted: 11/11/2020] [Indexed: 01/11/2023] Open
Abstract
AIMS This meta-analysis aimed to determine whether coronary artery bypass grafting (CABG) or percutaneous coronary intervention (PCI) should be preferred in patients with severely reduced left ventricular (LV) ejection fraction. METHODS AND RESULTS We searched the PubMed, EMBASE, and Cochrane Library databases from the conception of the databases till 1 May 2020 for studies on patients with severely reduced LV ejection fraction undergoing CABG and PCI. The primary clinical endpoints were 30 day and long-term mortalities. The secondary endpoints were 30 day and long-term incidences of myocardial infarction (MI) and stroke, long-term cardiovascular mortality, and repeat revascularization. Eighteen studies involving 11 686 patients were analysed. Compared with PCI, CABG had lower long-term mortality [hazard ratio (HR): 0.70, 95% confidence interval (CI): 0.61-0.80, P < 0.01], cardiovascular mortality (HR: 0.60, 95% CI: 0.43-0.85, P < 0.01), MI (HR: 0.51, 95% CI: 0.36-0.72, P < 0.01), and repeat revascularization (HR: 0.32, 95% CI: 0.23-0.47, P < 0.01) risk. Significant differences were not observed for long-term stroke (HR: 1.18, 95% CI: 0.74-1.87, P = 0.49), 30 day mortality (HR: 1.18, 95% CI: 0.89-1.56, P = 0.25), and MI (HR: 0.42, 95% CI: 0.16-1.11, P = 0.08) risk. CABG was associated with a higher risk of stroke within 30 days (HR: 2.88, 95% CI: 1.07-7.77, P = 0.04). In a subgroup analysis of propensity score-matched studies, CABG was associated with a higher long-term risk of stroke (HR: 1.61, 95% CI: 1.20-2.16, P < 0.01). CONCLUSIONS Among patients with severely reduced LV ejection fraction, CABG resulted in a lower mortality rate and an increased risk of stroke.
Collapse
Affiliation(s)
- Junyu Pei
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Xiaopu Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Zhenhua Xing
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China.,Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Keyang Zheng
- Department of Cardiovascular Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xinqun Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| |
Collapse
|
11
|
Gouda E, Babiker F. Micronized flavonoid fraction Daflon 500 protects heart against ischemia–reperfusion injury: an old medicine for a new target. ALL LIFE 2020. [DOI: 10.1080/26895293.2020.1832921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- Eman Gouda
- Department of Physiology, Faculty of Medicine, Health Science Center, Kuwait University, Kuwait, Kuwait
| | - Fawzi Babiker
- Department of Physiology, Faculty of Medicine, Health Science Center, Kuwait University, Kuwait, Kuwait
| |
Collapse
|
12
|
Vancheri F, Longo G, Vancheri S, Henein M. Coronary Microvascular Dysfunction. J Clin Med 2020; 9:E2880. [PMID: 32899944 PMCID: PMC7563453 DOI: 10.3390/jcm9092880] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/02/2020] [Accepted: 09/02/2020] [Indexed: 01/09/2023] Open
Abstract
Many patients with chest pain undergoing coronary angiography do not show significant obstructive coronary lesions. A substantial proportion of these patients have abnormalities in the function and structure of coronary microcirculation due to endothelial and smooth muscle cell dysfunction. The coronary microcirculation has a fundamental role in the regulation of coronary blood flow in response to cardiac oxygen requirements. Impairment of this mechanism, defined as coronary microvascular dysfunction (CMD), carries an increased risk of adverse cardiovascular clinical outcomes. Coronary endothelial dysfunction accounts for approximately two-thirds of clinical conditions presenting with symptoms and signs of myocardial ischemia without obstructive coronary disease, termed "ischemia with non-obstructive coronary artery disease" (INOCA) and for a small proportion of "myocardial infarction with non-obstructive coronary artery disease" (MINOCA). More frequently, the clinical presentation of INOCA is microvascular angina due to CMD, while some patients present vasospastic angina due to epicardial spasm, and mixed epicardial and microvascular forms. CMD may be associated with focal and diffuse epicardial coronary atherosclerosis, which may reinforce each other. Both INOCA and MINOCA are more common in females. Clinical classification of CMD includes the association with conditions in which atherosclerosis has limited relevance, with non-obstructive atherosclerosis, and with obstructive atherosclerosis. Several studies already exist which support the evidence that CMD is part of systemic microvascular disease involving multiple organs, such as brain and kidney. Moreover, CMD is strongly associated with the development of heart failure with preserved ejection fraction (HFpEF), diabetes, hypertensive heart disease, and also chronic inflammatory and autoimmune diseases. Since coronary microcirculation is not visible on invasive angiography or computed tomographic coronary angiography (CTCA), the diagnosis of CMD is usually based on functional assessment of microcirculation, which can be performed by both invasive and non-invasive methods, including the assessment of delayed flow of contrast during angiography, measurement of coronary flow reserve (CFR) and index of microvascular resistance (IMR), evaluation of angina induced by intracoronary acetylcholine infusion, and assessment of myocardial perfusion by positron emission tomography (PET) and magnetic resonance (CMR).
Collapse
Affiliation(s)
- Federico Vancheri
- Department of Internal Medicine, S.Elia Hospital, 93100 Caltanissetta, Italy
| | - Giovanni Longo
- Cardiovascular and Interventional Department, S.Elia Hospital, 93100 Caltanissetta, Italy;
| | - Sergio Vancheri
- Radiology Department, I.R.C.C.S. Policlinico San Matteo, 27100 Pavia, Italy;
| | - Michael Henein
- Institute of Public Health and Clinical Medicine, Umea University, SE-90187 Umea, Sweden;
- Department of Fluid Mechanics, Brunel University, Middlesex, London UB8 3PH, UK
- Molecular and Nuclear Research Institute, St George’s University, London SW17 0RE, UK
| |
Collapse
|
13
|
Fu Y, Li J, Wu S, Wang H. Electroacupuncture pretreatment promotes angiogenesis via hypoxia-inducible factor 1α and vascular endothelial growth factor in a rat model of chronic myocardial ischemia. Acupunct Med 2020; 39:367-375. [PMID: 32811184 DOI: 10.1177/0964528420938378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Electroacupuncture (EA) pretreatment appears useful in the treatment of chronic myocardial ischemia (CMI). The goal of this study was to investigate the effect of EA preconditioning on the regulation of hypoxia-inducible factor (HIF)-1α and vascular endothelial growth factor (VEGF) proteins in a CMI model of vascular regeneration. METHODS A CMI model was established by subcutaneous injection of isoprinosine hydrochloride (ISO) for 14 days in 45 Wistar rats, which had been randomly divided into a model group (n = 15), a CMI group pretreated with sham EA for 21 days (CMI + Sham group, n = 15) and a CMI group pretreated with verum EA for 21 days (CMI + EA, n = 15) prior to modeling. An additional 15 Wistar rats received 0.9% sodium chloride via intraperitoneal injection for 14 consecutive days (control group). Serum levels of VEGF and HIF-1α were measured by ELISA, while protein expression of VEGF and HIF-1α in the area of myocardial infarction was measured by Western blotting. The area of myocardial infarction and fibrosis of the myocardial tissue in the study groups were visualized by hematoxylin-eosin (HE) staining and Masson staining, respectively. RESULTS EA pretreatment improved cardiac function by regulating left ventricular end-diastolic diameter and left ventricular end-systolic diameter, left ventricular ejection fraction and the ST segment voltage of the electrocardiogram. EA pretreatment promoted vascular regeneration by increasing serum levels of VEGF and HIF-1α and by increasing protein expression of HIF-1α and VEGF in the infarcted region of the myocardium, leading to a reduction in the area of myocardial infarction on HE staining and reduction of myocardial fibrosis on Masson staining. CONCLUSION EA pretreatment promotes protein expression of HIF-1α and VEGF in areas of ischemic myocardium, which may represent useful biomarkers for coronary collateral establishment and offer potential targets for therapeutic angiogenesis in patients with CMI.
Collapse
Affiliation(s)
- Yimeng Fu
- Hubei University of Chinese Medicine, Wuhan, China
| | - Jia Li
- Hubei University of Chinese Medicine, Wuhan, China
| | - Song Wu
- Hubei University of Chinese Medicine, Wuhan, China
| | - Hua Wang
- Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
14
|
Zhang Y, Wang VY, Morgan AE, Kim J, Ge L, Guccione JM, Weinsaft JW, Ratcliffe MB. A Novel MRI-Based Finite Element Modeling Method for Calculation of Myocardial Ischemia Effect in Patients With Functional Mitral Regurgitation. Front Physiol 2020; 11:158. [PMID: 32231584 PMCID: PMC7082816 DOI: 10.3389/fphys.2020.00158] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 02/12/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Functional Mitral Regurgitation (FMR) associated with coronary artery disease affects nearly 3 million patients in the United States. Both myocardial infarction (MI) and ischemia contribute to FMR development but uncertainty as to which patients will respond to revascularization (REVASC) of ischemia alone prevents rational decision making about FMR therapy. The aim of this study was to create patient-specific cardiac MRI (CMR) informed finite element (FE) models of the left ventricle (LV), calculate regional LV systolic contractility and then use optimized systolic material properties to simulate the effect of revascularization (virtual REVASC). METHODS We describe a novel FE method able to predict the effect of myocardial ischemia on regional LV function. CMR was obtained in five patients with multi-vessel coronary disease and FMR before and 3 months after percutaneous REVASC and a single healthy volunteer. Patient-specific FE models were created and divided into 17 sectors where the systolic contractility parameter, T m a x of each sector was a function of regional stress perfusion (SP-CMR) and myocardial infarction (LGE-CMR) scores. Sector-specific circumferential and longitudinal end-systolic strain and LV volume from CSPAMM were used in a formal optimization to determine the sector based myocardial contractility, T m a x and ischemia effect, α. Virtual REVASC was simulated by setting α to zero. RESULTS The FE optimization successfully converged with good agreement between calculated and experimental end-systolic strain and LV volumes. Specifically, the optimized T max for the healthy myocardium for five patients and the volunteer was 495.1, 336.8, 173.5, 227.9, 401.4, and 218.9 kPa. The optimized α was found to be 1.0, 0.44, and 0.08 for Patients 1, 2, and 3, and 0 for Patients 4 and 5. The calculated average of radial strain for Patients 1, 2, and 3 at baseline and after virtual REVASC was 0.23 and 0.25, respectively. CONCLUSION We developed a novel computational method able to predict the effect of myocardial ischemia in patients with FMR. This method can be used to predict the effect of ischemia on the regional myocardium and promises to facilitate better understanding of FMR response to REVASC.
Collapse
Affiliation(s)
- Yue Zhang
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Department of Bioengineering, University of California, San Francisco, San Francisco, CA, United States
| | - Vicky Y. Wang
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Department of Bioengineering, University of California, San Francisco, San Francisco, CA, United States
| | - Ashley E. Morgan
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Jiwon Kim
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Liang Ge
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Department of Bioengineering, University of California, San Francisco, San Francisco, CA, United States
| | - Julius M. Guccione
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Department of Bioengineering, University of California, San Francisco, San Francisco, CA, United States
| | | | - Mark B. Ratcliffe
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Department of Bioengineering, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
15
|
Sorop O, van de Wouw J, Merkus D, Duncker DJ. Coronary Microvascular Dysfunction in Cardiovascular Disease: Lessons from Large Animal Models. Microcirculation 2020. [DOI: 10.1007/978-3-030-28199-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
16
|
Tune JD, Goodwill AG, Kiel AM, Baker HE, Bender SB, Merkus D, Duncker DJ. Disentangling the Gordian knot of local metabolic control of coronary blood flow. Am J Physiol Heart Circ Physiol 2019; 318:H11-H24. [PMID: 31702972 DOI: 10.1152/ajpheart.00325.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Recognition that coronary blood flow is tightly coupled with myocardial metabolism has been appreciated for well over half a century. However, exactly how coronary microvascular resistance is tightly coupled with myocardial oxygen consumption (MV̇o2) remains one of the most highly contested mysteries of the coronary circulation to this day. Understanding the mechanisms responsible for local metabolic control of coronary blood flow has been confounded by continued debate regarding both anticipated experimental outcomes and data interpretation. For a number of years, coronary venous Po2 has been generally accepted as a measure of myocardial tissue oxygenation and thus the classically proposed error signal for the generation of vasodilator metabolites in the heart. However, interpretation of changes in coronary venous Po2 relative to MV̇o2 are quite nuanced, inherently circular in nature, and subject to confounding influences that remain largely unaccounted for. The purpose of this review is to highlight difficulties in interpreting the complex interrelationship between key coronary outcome variables and the arguments that emerge from prior studies performed during exercise, hemodilution, hypoxemia, and alterations in perfusion pressure. Furthermore, potential paths forward are proposed to help to facilitate further dialogue and study to ultimately unravel what has become the Gordian knot of the coronary circulation.
Collapse
Affiliation(s)
- Johnathan D Tune
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Adam G Goodwill
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Alexander M Kiel
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| | - Hana E Baker
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Shawn B Bender
- Biomedical Sciences, University of Missouri, Columbia, Missouri.,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Cardiovascular Research School Erasmus University Rotterdam, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Walter-Brendel Center of Experimental Medicine, University Hospital, Ludwig Maximilian University Munich, Munich, Germany.,German Centre for Cardiovascular Research, Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Cardiovascular Research School Erasmus University Rotterdam, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
17
|
Mayr A, Pamminger M, Reindl M, Greulich S, Reinstadler SJ, Tiller C, Holzknecht M, Nalbach T, Plappert D, Kranewitter C, Klug G, Metzler B. Mitral annular plane systolic excursion by cardiac MR is an easy tool for optimized prognosis assessment in ST-elevation myocardial infarction. Eur Radiol 2019; 30:620-629. [PMID: 31392477 PMCID: PMC6890588 DOI: 10.1007/s00330-019-06393-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/16/2019] [Accepted: 07/24/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVES The purpose of this study was to assess the comparative prognostic value of mitral annular plane systolic excursion (MAPSE) versus left ventricular ejection fraction (LVEF), measured by cardiac magnetic resonance (CMR) imaging in patients with ST-elevation myocardial infarction (STEMI) treated with primary percutaneous coronary intervention (pPCI). METHODS CMR was performed in 255 STEMI patients within 2 days (interquartile range (IQR) 2-4 days) after infarction. CMR included MAPSE measurement on CINE 4-chamber view. Patients were followed for major adverse cardiovascular events (MACE)-death, non-fatal myocardial re-infarction, stroke, and new congestive heart failure. RESULTS Patients with MACE (n = 35, 14%, median follow-up 3 years [IQR 1-4 years]) showed significantly lower MAPSE (8 mm [7-8.8] vs. 9.6 mm [8.1-11.5], p < 0.001). The association between decreased MAPSE (< 9 mm, optimal cut-off value by c-statistics) remained significant after adjustment for independent clinical and CMR predictors of MACE. The AUC of MAPSE for the prediction of MACE was 0.74 (CI 95% 0.65-0.82), significantly higher than that of LVEF (0.61 [CI 95% 0.50-0.71]; p < 0.001). CONCLUSIONS Reduced long-axis function assessed with MAPSE measurement using CINE CMR independently predicts long-term prognosis following STEMI. Moreover, MAPSE provided significantly higher prognostic implication in comparison with conventional LVEF measurement. KEY POINTS • MAPSE determined by CMR independently predicts long-term prognosis following STEMI. • MACE-free survival is significantly higher in patients with MAPSE ≥ 9 mm than < 9 mm. • MAPSE provides significantly higher prognostic implication than conventional LVEF.
Collapse
Affiliation(s)
- Agnes Mayr
- University Clinic of Radiology, Medical University Innsbruck, Anichstraße 35, A-6020, Innsbruck, Austria.
| | - Mathias Pamminger
- University Clinic of Radiology, Medical University Innsbruck, Anichstraße 35, A-6020, Innsbruck, Austria
| | - Martin Reindl
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, Anichstraße 35, A-6020, Innsbruck, Austria
| | - Simon Greulich
- Department of Cardiology and Cardiovascular Diseases, University Hospital Tübingen, Otfried Müller-Straße 10, 72076, Tübingen, Germany
| | - Sebastian J Reinstadler
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, Anichstraße 35, A-6020, Innsbruck, Austria
| | - Christina Tiller
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, Anichstraße 35, A-6020, Innsbruck, Austria
| | - Magdalena Holzknecht
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, Anichstraße 35, A-6020, Innsbruck, Austria
| | - Timo Nalbach
- University Clinic of Radiology, Medical University Innsbruck, Anichstraße 35, A-6020, Innsbruck, Austria
| | - David Plappert
- University Clinic of Radiology, Medical University Innsbruck, Anichstraße 35, A-6020, Innsbruck, Austria
| | - Christof Kranewitter
- University Clinic of Radiology, Medical University Innsbruck, Anichstraße 35, A-6020, Innsbruck, Austria
| | - Gert Klug
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, Anichstraße 35, A-6020, Innsbruck, Austria
| | - Bernhard Metzler
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, Anichstraße 35, A-6020, Innsbruck, Austria
| |
Collapse
|
18
|
Zhang L, Jian LL, Li JY, Jin X, Li LZ, Zhang YL, Gong HY, Cui Y. Possible involvement of alpha B-crystallin in the cardioprotective effect of n-butanol extract of Potentilla anserina L. on myocardial ischemia/reperfusion injury in rat. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 55:320-329. [PMID: 30940361 DOI: 10.1016/j.phymed.2018.10.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/15/2018] [Accepted: 10/18/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND It has been reported that n-butanol extract of Potentilla anserina L (NP) had protective effect against acute myocardial ischemia/reperfusion (I/R) injury in mice. Because of limited phytochemical study on NP, its bioactive compounds and underlying protective mechanisms are largely unclear. PURPOSE The purpose of this study was to investigate the major bioactive compounds and possible mechanism for the cardioprotective effect of NP on rat with I/R injury. METHODS We analyzed the phytochemical isolation of NP and identified the structure of compounds, which was elucidated by a combination of spectroscopic analyses. An I/R model was established by I-30 min/R-2 h in Sprage-Dawley rats. The rats were given intragastric administration of NP (49.3, 98.6, and 197.2 mg•kg-1) continuously for 10 days before I/R operation. The morphological changes and apoptosis of cardiomyocytes were observed by H&E staining, Transmission electron microscope and TUNEL staining respectively. The activities or contents of catalase (CAT), superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione (GSH) in plasma were detected. Apoptosis related factors were also measured by RT-PCR and western blot. In order to discover the underlying mechanism of NP on I/R, we performed proteomic analysis using two-dimensional gel electrophoresis (2D-DIGE) and matrix assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF/MS) to describe differential proteins expression. Potential target protein resulted from 2D-DIGE coupled to MALDI-TOF/MS analysis were further confirmed by immunohistochemical staining, RT-PCR, and western blot. RESULTS We isolated and identified 14 compounds, of which 7 compounds belong to triterpenes. Rats pretreated with NP showed a significant increase on the activities of GSH, SOD and CAT, and remarkable decrease on the content of MDA. NP significantly inhibited the apoptosis of cardiomyocyte and decreased the expression of Cyt C and cleaved-caspase-3. Proteomic analysis revealed that alpha B-crystallin (CryAB) might participate in the NP protective effect against I/R. NP enhanced the level of pCryAB Ser59, whereas the expression of CryAB was decreased. CONCLUSION NP was showed to alleviate I/R injury and inhibit myocardial apoptosis, which might be associated with reduction on oxidative stress and apoptosis. CryAB as a possible target involved in the NP protective effect. This study supplied valuable information to develop novel cardioprotective agents from NP extract.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin, China
| | - Le Le Jian
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin, China; Shanxi Provincial Crops Hospital, Chinese People's Armed Police Forces, Xi'an, Shanxi, China
| | - Jian Yu Li
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin, China
| | - Xin Jin
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin, China
| | - Ling Zhi Li
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin, China; Key Laboratory for Prevention and Control of Occupational and Environmental Hazard, Tianjin, China.
| | - Yong Liang Zhang
- Key Laboratory for Prevention and Control of Occupational and Environmental Hazard, Tianjin, China.
| | - Hai Ying Gong
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin, China
| | - Ying Cui
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin, China
| |
Collapse
|
19
|
Zhang L, Zhang Y, Yu X, Xu H, Sui D, Zhao X. Alprostadil attenuates myocardial ischemia/reperfusion injury by promoting antioxidant activity and eNOS activation in rats. Acta Cir Bras 2019; 33:1067-1077. [PMID: 30624512 DOI: 10.1590/s0102-865020180120000004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/17/2018] [Indexed: 11/21/2022] Open
Abstract
PURPOSE To investigate the effect of alprostadil on myocardial ischemia/reperfusion (I/R) in rats. METHODS Rats were subjected to myocardial ischemia for 30 min followed by 24h reperfusion. Alprostadil (4 or 8 μg/kg) was intravenously administered at the time of reperfusion and myocardial infarct size, levels of troponin T, and the activity of creatine kinase-MB (CK-MB) and lactate dehydrogenase (LDH) in the serum were measured. Antioxidative parameters, nitric oxide (NO) content and phosphorylated endothelial nitric oxide synthase 3 (p-eNOS) expression in the left ventricles were also measured. Histopathological examinations of the left ventricles were also performed. RESULTS Alprostadil treatment significantly reduced myocardial infarct size, serum troponin T levels, and CK-MB and LDH activity (P<0.05). Furthermore, treatment with alprostadil significantly decreased malondialdehyde (MDA) content (P<0.05) and markedly reduced myonecrosis, edema and infiltration of inflammatory cells. Superoxide dismutase and catalase activities (P<0.05), NO level (P<0.01) and p-eNOS (P<0.05) were significantly increased in rats treated with alprostadil compared with control rats. CONCLUSION These results indicate that alprostadil protects against myocardial I/R injury and that these protective effects are achieved, at least in part, via the promotion of antioxidant activity and activation of eNOS.
Collapse
Affiliation(s)
- Liping Zhang
- MD, Department of Cardiovascular Medicine, First Hospital, Jilin University, Changchun, Jilin, P.R. China. Acquisition, analysis and interpretation of data; manuscript writing
| | - Ying Zhang
- Master, Pharmacology, College of Pharmacy, Jilin University, Changchun, Jilin, P.R. China. Acquisition of data
| | - Xiaofeng Yu
- MD, Pharmacology, College of Pharmacy, Jilin University, Changchun, Jilin, P.R. China. Acquisition of data, technical procedures, histopathological examinations
| | - Huali Xu
- MD, Pharmacology, College of Pharmacy, Jilin University, Changchun, Jilin, P.R. China. Analysis and interpretation of data
| | - Dayuan Sui
- MD, Pharmacology, College of Pharmacy, Jilin University, Changchun, Jilin, P.R. China. Conception and design of the study, manuscript writing
| | - Xuezhong Zhao
- MD, Department of Cardiovascular Medicine, First Hospital, Jilin University, Changchun, Jilin, P.R. China. Conception and design of the study, critical revision, final approval
| |
Collapse
|
20
|
Cui K, Zhang D, Lyu S, Song X, Yuan F, Xu F, Zhang M. Meta-Analysis Comparing Percutaneous Coronary Revascularization Using Drug-Eluting Stent Versus Coronary Artery Bypass Grafting in Patients With Left Ventricular Systolic Dysfunction. Am J Cardiol 2018; 122:1670-1676. [PMID: 30220418 DOI: 10.1016/j.amjcard.2018.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 08/05/2018] [Accepted: 08/09/2018] [Indexed: 01/11/2023]
Abstract
The relative safety and efficacy of percutaneous coronary intervention (PCI) with drug-eluting stent (DES) and coronary artery bypass grafting (CABG) in patients with left ventricular (LV) systolic dysfunction remains controversial; therefore we conducted this meta-analysis to identify the optimal strategy for such cohorts. A comprehensive search of the electronic databases including PubMed, EMBASE, and Cochrane Library from January 1, 2003 to March 1, 2018 was performed to identify the eligible adjusted observational studies. The primary end point was all-cause death during the longest follow-up, and the generic inverse variance random-effect model was used to estimate the pooled hazard ratios (HRs) with 95% confidence intervals (CIs). Eight adjusted observational studies involving 10,268 patients were included. Compared with CABG, PCI with DES was associated with higher risk of all-cause mortality (HR 1.36, 95% CI 1.16 to 1.60), cardiac mortality (HR 2.20, 95% CI 1.63 to 2.95), myocardial infarction (HR 1.69, 95% CI 1.28 to 2.24), and repeat revascularization (HR 4.95, 95% CI 3.28 to 7.46) in patients with coronary artery disease and LV systolic dysfunction. Besides, separate analysis of patients with LV ejection fraction <35% or left main and/or multivessel disease obtained similar results compared with the overall analysis. However, DES and CABG shared similar rates of stroke (HR 0.92, 95% CI 0.67 to 1.26). In conclusion, CABG appears to be superior to PCI with DES for patients with coronary artery disease and LV systolic dysfunction, particularly in patients with severe LV systolic dysfunction or those with left main and/or multivessel disease.
Collapse
Affiliation(s)
- Kongyong Cui
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Dongfeng Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Shuzheng Lyu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China.
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Fei Yuan
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Feng Xu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Min Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| |
Collapse
|
21
|
Mytsyk M, Isu G, Cerino G, Grapow MTR, Eckstein FS, Marsano A. Paracrine potential of adipose stromal vascular fraction cells to recover hypoxia-induced loss of cardiomyocyte function. Biotechnol Bioeng 2018; 116:132-142. [PMID: 30171703 DOI: 10.1002/bit.26824] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/02/2018] [Accepted: 08/30/2018] [Indexed: 12/21/2022]
Abstract
Cell-based therapies show promising results in cardiac function recovery mostly through paracrine-mediated processes (as angiogenesis) in chronic ischemia. In this study, we aim to develop a 2D (two-dimensional) in vitro cardiac hypoxia model mimicking severe cardiac ischemia to specifically investigate the prosurvival paracrine effects of adipose tissue-derived stromal vascular fraction (SVF) cell secretome released upon three-dimensional (3D) culture. For the 2D-cardiac hypoxia model, neonatal rat cardiomyocytes (CM) were cultured for 5 days at < 1% (approaching anoxia) oxygen (O2 ) tension. Typical cardiac differentiation hallmarks and contractile ability were used to assess both the cardiomyocyte loss of functionality upon anoxia exposure and its possible recovery following the 5-day-treatment with SVF-conditioned media (collected following 6-day-perfusion-based culture on collagen scaffolds in either normoxia or approaching anoxia). The culture at < 1% O 2 for 5 days mimicked the reversible condition of hibernating myocardium with still living and poorly contractile CM (reversible state). Only SVF-medium conditioned in normoxia expressing a high level of the prosurvival hepatocyte-growth factor (HGF) and insulin-like growth factor (IGF) allowed the partial recovery of the functionality of damaged CM. The secretome generated by SVF-engineered tissues showed a high paracrine potential to rescue the nonfunctional CM, therefore resulting in a promising patch-based treatment of specific low-perfused areas after myocardial infarction.
Collapse
Affiliation(s)
- Myroslava Mytsyk
- Department of Surgery, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Giuseppe Isu
- Department of Surgery, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Giulia Cerino
- Department of Surgery, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Martin T R Grapow
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | | | - Anna Marsano
- Department of Surgery, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
22
|
Bonis A, Anderson L, Talhouarne G, Schueller E, Unke J, Krus C, Stokka J, Koepke A, Lehrer B, Schuh A, Andersen JJ, Cooper S. Cardiovascular resistance to thrombosis in 13-lined ground squirrels. J Comp Physiol B 2018; 189:167-177. [PMID: 30317383 DOI: 10.1007/s00360-018-1186-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/08/2018] [Accepted: 10/06/2018] [Indexed: 12/24/2022]
Abstract
13-lined ground squirrels (Ictidomys tridecemlineatus) enter hibernation as a survival strategy during extreme environmental conditions. Typical ground squirrel hibernation is characterized by prolonged periods of torpor with significantly reduced heart rate, blood pressure, and blood flow, interrupted every few weeks by brief interbout arousals (IBA) during which blood flow fluctuates dramatically. These physiological conditions should increase the risk of stasis-induced blood clots and myocardial ischemia. However, ground squirrels have adapted to survive repeated bouts of torpor and IBA without forming lethal blood clots or sustaining lethal ischemic myocardial damage. The purpose of this study was to determine if ground squirrels are resistant to thrombosis and myocardial ischemia during hibernation. Blood markers of coagulation, fibrinolysis, thrombosis, and ischemia, as well as histological markers of myocardial ischemia were measured throughout the annual hibernation cycle. Hibernating ground squirrels were also treated with isoprenaline to induce myocardial ischemia. Thrombin-antithrombin complex levels were significantly reduced (p < 0.05) during hibernation, while D-dimer level remained unchanged throughout the annual cycle, both consistent with an antithrombotic state. During torpor, the ground squirrels were in a hyperfibrinolytic state with an elevated ratio of tissue plasminogen activator complexed with plasminogen activator inhibitor to total plasminogen activator inhibitor (p < 0.05). Histological markers of myocardial ischemia were reversibly elevated during hibernation with no increase in markers of myocardial cell death in the blood. These data suggest that ground squirrels do not form major blood clots during hibernation through suppression of coagulation and a hyperfibrinolytic state. These animals also demonstrate myocardial resistance to ischemia.
Collapse
Affiliation(s)
- Alison Bonis
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Leah Anderson
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Gaëlle Talhouarne
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Emily Schueller
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Jenna Unke
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Catherine Krus
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Jordan Stokka
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Anna Koepke
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Brittany Lehrer
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Anthony Schuh
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | | | - Scott Cooper
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA.
| |
Collapse
|
23
|
Tavares JGP, Errante PR, Govato TCP, Vasques ÊR, Ferraz RRN, Taha MO, Menezes-Rodrigues FS, Caricati-Neto A. Cardioprotective effect of preconditioning is more efficient than postconditioning in rats submitted to cardiac ischemia and reperfusion1. Acta Cir Bras 2018; 33:588-596. [PMID: 30110060 DOI: 10.1590/s0102-865020180070000004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/24/2018] [Indexed: 01/19/2023] Open
Abstract
PURPOSE To investigate the cardioprotective effects of ischemic preconditioning (preIC) and postconditioning (postIC) in animal model of cardiac ischemia/reperfusion. METHODS Adult rats were submitted to protocol of cardiac ischemia/reperfusion (I/R) and randomized into three experimental groups: cardiac I/R (n=33), preCI + cardiac I/R (n=7) and postCI + cardiac I/R (n=8). After this I/R protocol, the incidence of ventricular arrhythmia (VA), atrioventricular block (AVB) and lethality (LET) was evaluated using the electrocardiogram (ECG) analysis. RESULTS After reestablishment of coronary blood flow, we observed variations of the ECG trace with increased incidence of ventricular arrhythmia (VA) (85%), atrioventricular block (AVB) (79%), and increase of lethality (70%) in cardiac I/R group. The comparison between I/R + preIC group with I/R group demonstrated significant reduction in VA incidence to 28%, AVB to 0% and lethality to 14%. The comparison of I/R + postIC group with I/R group was observed significance reduction in AVB incidence to 25% and lethality to 25%. CONCLUSION The preconditioning strategies produce cardioprotection more efficient that postconditioning against myocardial dysfunctions and lethality by cardiac ischemia and reperfusion.
Collapse
Affiliation(s)
- José Gustavo Padrão Tavares
- Fellow PhD degree, Postgraduate Program in Pharmacology, Universidade Federal de São Paulo (UNIFESP), Brazil. Conception and design of the study, analysis and interpretation of data, manuscript writing
| | - Paolo Ruggero Errante
- Fellow PhD degree, Postgraduate Program in Pharmacology, UNIFESP, Sao Paulo-SP, Brazil. Analysis and interpretation of data, manuscript writing
| | - Tânia Carmem Peñaranda Govato
- Assistant Professor, Department of Pharmacology, Faculdade de Medicina do ABC (FMABC), Santo Andre-SP, Brazil. Statistical analysis
| | - Ênio Rodrigues Vasques
- Fellow PhD degree, Department of Gastroenterology, Faculty of Medicine, Universidade de São Paulo (USP), Brazil. Interpretation of electrocardiogram
| | - Renato Ribeiro Nogueira Ferraz
- Full Professor, Program in Management of Health System (PMPA-GSS), Universidade Nove de Julho (UNINOVE), Sao Paulo-SP, Brazil. Critical revision
| | - Murched Omar Taha
- Associate Professor, Department of Surgery, UNIFESP, Sao Paulo-SP, Brazil. Technical procedures
| | | | - Afonso Caricati-Neto
- Associate Professor, Department of Pharmacology, UNIFESP, Sao Paulo-SP, Brazil. Conception and design of the study, critical revision
| |
Collapse
|
24
|
Iwaszczuk P, Kołodziejczyk B, Kruczek T, Drabik L, Płazak W, Komar M, Podolec P, Musiałek P. Ischemic Versus Non-Ischemic (Neurogenic) Myocardial Contractility Impairment in Acute Coronary Syndromes: Prevalence and Impact on Left Ventricular Systolic Function Recovery. Med Sci Monit 2018; 24:3693-3701. [PMID: 29858549 PMCID: PMC6011807 DOI: 10.12659/msm.907466] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 11/13/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Neurogenic mechanism is believed to contribute to left ventricular (LV) systolic dysfunction in acute coronary syndromes (ACS); its extreme form is known as takotsubo cardiomyopathy. However, the magnitude of neurogenic contribution to LV dysfunction in all-comer first-time ACS remains unknown. MATERIAL AND METHODS In 120 consecutive patients with first-time ACS (age 66.3±12.3years, 40 women) coronary angiograms were individually matched to the echocardiographic left ventricular (LV) segments (17-segment model). Baseline contractility impairment was classified as ischemic (I): confined to the stenotic artery(ies) supply area(s), neurogenic (N): in absence of attributable coronary stenosis, or partially ischemic/partially neurogenic (I&N). Echocardiography was repeated at 6 months to determine LV systolic function recovery. RESULTS Neurogenic component (NC) contribution to myocardial contractility impairment was present in 24.2% of ACS patients, with pure N in 6.7% and I&N in 17.5%. Diabetes/pre-diabetes was present in 38.5% vs. 33.5% vs. 0% (I vs. I&N vs. N; p=0.02). Major stressor preceding symptom onset was reported in 3.3% in I, 9.5% in I&N, and 25.0% in N (p=0.03). The number of LV segments with contractility impairment was 2±4 in I, 17±11 in I&N, and 3±16 in N (p<0.05). NC presence was independently associated with better recovery of global LV systolic function (OR 2.99, 95% CI: 1.16-7.76; p=0.024). CONCLUSIONS Novel findings from this study are: (1) NC may contribute to myocardial contractility impairment in 1 in every 4 first-time ACS patients, (2) NC contribution to contractility impairment in ACS is blunted in diabetes or pre-diabetes, and (3) LV systolic function recovery is better in patients with NC.
Collapse
|
25
|
Touyz RM, Herrmann J. Cardiotoxicity with vascular endothelial growth factor inhibitor therapy. NPJ Precis Oncol 2018; 2:13. [PMID: 30202791 PMCID: PMC5988734 DOI: 10.1038/s41698-018-0056-z] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis inhibitors targeting the vascular endothelial growth factor (VEGF) signaling pathway (VSP) have been important additions in the therapy of various cancers, especially renal cell carcinoma and colorectal cancer. Bevazicumab, the first VSP to receive FDA approval in 2004 targeting all circulating isoforms of VEGF-A, has become one of the best-selling drugs of all times. The second wave of tyrosine kinase inhibitors (TKIs), which target the intracellular site of VEGF receptor kinases, began with the approval of sorafenib in 2005 and sunitinib in 2006. Heart failure was subsequently noted, in 2-4% of patients on bevacizumab and in 3-8% of patients on VSP-TKIs. The very fact that the single-targeted monoclonal antibody bevacizumab can induce cardiotoxicity supports a pathomechanistic role for the VSP and the postulate of the "vascular" nature of VSP inhibitor cardiotoxicity. In this review we will outline this scenario in greater detail, reflecting on hypertension and coronary artery disease as risk factors for VSP inhibitor cardiotoxicity, but also similarities with peripartum and diabetic cardiomyopathy. This leads to the concept that any preexisting or coexisting condition that reduces the vascular reserve or utilizes the vascular reserve for compensatory purposes may pose a risk factor for cardiotoxicity with VSP inhibitors. These conditions need to be carefully considered in cancer patients who are to undergo VSP inhibitor therapy. Such vigilance is not to exclude patients from such prognostically extremely important therapy but to understand the continuum and to recognize and react to any cardiotoxicity dynamics early on for superior overall outcomes.
Collapse
Affiliation(s)
- Rhian M. Touyz
- Institute of Cardiovascular & Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
26
|
Wu H, Li L, Niu P, Huang X, Liu J, Zhang F, Shen W, Tan W, Wu Y, Huo Y. The Structure-function remodeling in rabbit hearts of myocardial infarction. Physiol Rep 2018. [PMID: 28637704 PMCID: PMC5492201 DOI: 10.14814/phy2.13311] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Animal models are of importance to investigate basic mechanisms for ischemic heart failure (HF). The objective of the study was to create a rabbit model through multiple coronary artery ligations to investigate the postoperative structure‐function remodeling of the left ventricle (LV) and coronary arterial trees. Here, we hypothesize that the interplay of the degenerated coronary vasculature and increased ventricle wall stress relevant to cardiac fibrosis in vicinity of myocardial infarction (MI) precipitates the incidence and progression of ischemic HF. Echocardiographic measurements showed an approximately monotonic drop of fractional shortening and ejection fraction from 40% and 73% down to 28% and 58% as well as persistent enlargement of LV cavity and slight mitral regurgitation at postoperative 12 weeks. Micro‐CT and histological measurements showed that coronary vascular rarefaction and cardiac fibrosis relevant to inflammation occurred concurrently in vicinity of MI at postoperative 12 weeks albeit there was compensatory vascular growth at postoperative 6 weeks. These findings validate the proposed rabbit model and prove the hypothesis. The post‐MI rabbit model can serve as a reference to test various drugs for treatment of ischemic HF.
Collapse
Affiliation(s)
- Haotian Wu
- School of Basic Medical Sciences, Nanjing University of Traditional Chinese Medicine, Nanjing, China.,Hebei Yiling Pharmaceutical Research Institute, Shijiazhuang, China
| | - Li Li
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China
| | - Pei Niu
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China.,College of Medicine, Hebei University, Baoding, China
| | - Xu Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China
| | - Jinyi Liu
- College of Medicine, Hebei University, Baoding, China
| | | | - Wenzeng Shen
- College of Medicine, Hebei University, Baoding, China
| | - Wenchang Tan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China.,Shenzhen Graduate School, Peking University, Shenzhen, China.,PKU-HKUST Shenzhen-Hongkong Institution, Shenzhen, China
| | - Yiling Wu
- School of Basic Medical Sciences, Nanjing University of Traditional Chinese Medicine, Nanjing, China .,Hebei Yiling Pharmaceutical Research Institute, Shijiazhuang, China.,Key Laboratory, State Administration of Traditional Chinese Medicine (Cardiovascular and cerebrovascular collateral diseases), Shijiazhuang, China.,Hebei Province Key Laboratory of Collateral Diseases, Shijiazhuang, China
| | - Yunlong Huo
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China .,PKU-HKUST Shenzhen-Hongkong Institution, Shenzhen, China
| |
Collapse
|
27
|
Raghavendra U, Fujita H, Gudigar A, Shetty R, Nayak K, Pai U, Samanth J, Acharya U. Automated technique for coronary artery disease characterization and classification using DD-DTDWT in ultrasound images. Biomed Signal Process Control 2018. [DOI: 10.1016/j.bspc.2017.09.030] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
28
|
Lindsey ML, Bolli R, Canty JM, Du XJ, Frangogiannis NG, Frantz S, Gourdie RG, Holmes JW, Jones SP, Kloner RA, Lefer DJ, Liao R, Murphy E, Ping P, Przyklenk K, Recchia FA, Schwartz Longacre L, Ripplinger CM, Van Eyk JE, Heusch G. Guidelines for experimental models of myocardial ischemia and infarction. Am J Physiol Heart Circ Physiol 2018; 314:H812-H838. [PMID: 29351451 PMCID: PMC5966768 DOI: 10.1152/ajpheart.00335.2017] [Citation(s) in RCA: 357] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myocardial infarction is a prevalent major cardiovascular event that arises from myocardial ischemia with or without reperfusion, and basic and translational research is needed to better understand its underlying mechanisms and consequences for cardiac structure and function. Ischemia underlies a broad range of clinical scenarios ranging from angina to hibernation to permanent occlusion, and while reperfusion is mandatory for salvage from ischemic injury, reperfusion also inflicts injury on its own. In this consensus statement, we present recommendations for animal models of myocardial ischemia and infarction. With increasing awareness of the need for rigor and reproducibility in designing and performing scientific research to ensure validation of results, the goal of this review is to provide best practice information regarding myocardial ischemia-reperfusion and infarction models. Listen to this article’s corresponding podcast at ajpheart.podbean.com/e/guidelines-for-experimental-models-of-myocardial-ischemia-and-infarction/.
Collapse
Affiliation(s)
- Merry L Lindsey
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi.,Research Service, G. V. (Sonny) Montgomery Veterans Affairs Medical Center , Jackson, Mississippi
| | - Roberto Bolli
- Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville , Louisville, Kentucky
| | - John M Canty
- Division of Cardiovascular Medicine, Departments of Biomedical Engineering and Physiology and Biophysics, The Veterans Affairs Western New York Health Care System and Clinical and Translational Science Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo , Buffalo, New York
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital , Würzburg , Germany
| | - Robert G Gourdie
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute , Roanoke, Virginia
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia Health System , Charlottesville, Virginia
| | - Steven P Jones
- Department of Medicine, Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
| | - Robert A Kloner
- HMRI Cardiovascular Research Institute, Huntington Medical Research Institutes , Pasadena, California.,Division of Cardiovascular Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - David J Lefer
- Cardiovascular Center of Excellence, Louisiana State University Health Science Center , New Orleans, Louisiana
| | - Ronglih Liao
- Harvard Medical School , Boston, Massachusetts.,Division of Genetics and Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital , Boston, Massachusetts
| | - Elizabeth Murphy
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, Maryland
| | - Peipei Ping
- National Institutes of Health BD2KBig Data to Knowledge (BD2K) Center of Excellence and Department of Physiology, Medicine and Bioinformatics, University of California , Los Angeles, California
| | - Karin Przyklenk
- Cardiovascular Research Institute and Departments of Physiology and Emergency Medicine, Wayne State University School of Medicine , Detroit, Michigan
| | - Fabio A Recchia
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Fondazione G. Monasterio, Pisa , Italy.,Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University , Philadelphia, Pennsylvania
| | - Lisa Schwartz Longacre
- Heart Failure and Arrhythmias Branch, Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, Maryland
| | - Crystal M Ripplinger
- Department of Pharmacology, School of Medicine, University of California , Davis, California
| | - Jennifer E Van Eyk
- The Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center , Los Angeles, California
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School , Essen , Germany
| |
Collapse
|
29
|
Patel H, Mazur W, Williams KA, Kalra DK. Myocardial viability–State of the art: Is it still relevant and how to best assess it with imaging? Trends Cardiovasc Med 2018; 28:24-37. [DOI: 10.1016/j.tcm.2017.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/30/2017] [Accepted: 07/04/2017] [Indexed: 10/19/2022]
|
30
|
Anavekar NS, Chareonthaitawee P, Narula J, Gersh BJ. Revascularization in Patients With Severe Left Ventricular Dysfunction: Is the Assessment of Viability Still Viable? J Am Coll Cardiol 2017; 67:2874-87. [PMID: 27311527 DOI: 10.1016/j.jacc.2016.03.571] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/18/2016] [Accepted: 03/18/2016] [Indexed: 11/17/2022]
Abstract
Myocardial viability assessment is typically reserved for patients with coronary artery disease and significant left ventricular dysfunction. In this setting, there is myocardial adaptation to an altered physiological state that is potentially reversible. Imaging can characterize different parameters of cardiac function; however, despite previously published appraisals of different imaging modalities, there is still uncertainty regarding the role of these tests in clinical practice. The purpose of this review is to reflect on the physiological basis of myocardial viability, discuss the imaging tests available that characterize myocardial viability, and summarize the current published reports on the use of these tests in clinical practice.
Collapse
Affiliation(s)
- Nandan S Anavekar
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | | | - Jagat Narula
- Division of Cardiovascular Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Bernard J Gersh
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
31
|
Abstract
The heart is uniquely responsible for providing its own blood supply through the coronary circulation. Regulation of coronary blood flow is quite complex and, after over 100 years of dedicated research, is understood to be dictated through multiple mechanisms that include extravascular compressive forces (tissue pressure), coronary perfusion pressure, myogenic, local metabolic, endothelial as well as neural and hormonal influences. While each of these determinants can have profound influence over myocardial perfusion, largely through effects on end-effector ion channels, these mechanisms collectively modulate coronary vascular resistance and act to ensure that the myocardial requirements for oxygen and substrates are adequately provided by the coronary circulation. The purpose of this series of Comprehensive Physiology is to highlight current knowledge regarding the physiologic regulation of coronary blood flow, with emphasis on functional anatomy and the interplay between the physical and biological determinants of myocardial oxygen delivery. © 2017 American Physiological Society. Compr Physiol 7:321-382, 2017.
Collapse
Affiliation(s)
- Adam G Goodwill
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
| | - Gregory M Dick
- California Medical Innovations Institute, 872 Towne Center Drive, Pomona, CA
| | - Alexander M Kiel
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
- Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Drive, Lafayette, IN
| | - Johnathan D Tune
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
32
|
Sharma UC, Mosleh W, Chaudhari MR, Katkar R, Weil B, Evelo C, Cimato TR, Pokharel S, Blankesteijn WM, Suzuki G. Myocardial and Serum Galectin-3 Expression Dynamics Marks Post-Myocardial Infarction Cardiac Remodelling. Heart Lung Circ 2016; 26:736-745. [PMID: 28094123 DOI: 10.1016/j.hlc.2016.11.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 10/11/2016] [Accepted: 11/13/2016] [Indexed: 01/22/2023]
Abstract
BACKGROUND Acute myocardial infarction (MI) causes significant changes in cardiac morphology and function. Galectin-3 is a novel and potentially therapeutically important mediator of cardiac remodelling. Myocardial and serum galectin-3 expression dynamics in response to the early cardiovascular outcomes after acute MI are not fully elucidated. METHODS We first performed a comprehensive longitudinal microarray analyses in mice after acute MI. We then measured the serum levels of galectin-3 in a translational porcine model of coronary microembolism-induced post-ischaemic cardiac remodelling. We validated our pre-clinical studies in humans by measuring serum galectin-3 levels of 52 patients with acute ST-elevation MI (STEMI) and 11 healthy controls. We analysed galectin-3 data in relation to the development of major adverse cardiovascular outcomes (MACO). RESULTS Of the 9,753 genes profiled at infarcted and remote myocardium at eight different time points, dynamic myocardial overexpression of galectin-3 mRNA was detected. In a pig model of diffuse myocardial damage and cardiac remodelling, galectin-3 localised to the areas of tissue damage and myocardial fibrosis, with proportionate increase of their serum galectin-3 expression levels. In humans, increased serum galectin-3 level was associated with in-hospital MACO. CONCLUSIONS In this translational study, we demonstrated that galectin-3 is dynamically overexpressed in response to acute MI-induced cardiac remodelling. Elevated galectin-3 levels are associated with the development of in-hospital MACO.
Collapse
Affiliation(s)
- Umesh C Sharma
- Division of Cardiovascular Medicine, University at Buffalo, Buffalo, NY, USA.
| | - Wassim Mosleh
- Division of Cardiovascular Medicine, University at Buffalo, Buffalo, NY, USA
| | - Milind R Chaudhari
- Division of Cardiovascular Medicine, University at Buffalo, Buffalo, NY, USA
| | - Rujuta Katkar
- Division of Cardiovascular Medicine, University at Buffalo, Buffalo, NY, USA
| | - Brian Weil
- Division of Cardiovascular Medicine, University at Buffalo, Buffalo, NY, USA
| | - Chris Evelo
- Department of Bioinformatics-BiGCaT, University of Maastricht, Maastricht, The Netherlands
| | - Thomas R Cimato
- Division of Cardiovascular Medicine, University at Buffalo, Buffalo, NY, USA
| | - Saraswati Pokharel
- Department of Pathology and Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Gen Suzuki
- Division of Cardiovascular Medicine, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
33
|
Cardiac Strain in a Swine Model of Regional Hibernating Myocardium: Effects of CoQ10 on Contractile Reserve Following Bypass Surgery. J Cardiovasc Transl Res 2016; 9:368-73. [PMID: 27184805 DOI: 10.1007/s12265-016-9696-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/03/2016] [Indexed: 11/27/2022]
Abstract
There is conflicting clinical evidence whether administration of coenzyme Q10 (CoQ10) improves function following coronary artery bypass graft surgery (CABG). Using a swine model of hibernating myocardium, we tested whether daily CoQ10 would improve contractile function by MRI at 4-week post-CABG. Twelve pigs underwent a thoracotomy and had a constrictor placed on the left anterior descending (LAD). At 12 weeks, they underwent off-pump bypass and received daily dietary supplements of either CoQ10 (10 mg/kg/day) or placebo. At 4-week post-CABG, circumferential strain measurements in the hibernating LAD region from placebo and CoQ10 groups were not different and increased to a similar extent with dobutamine (-14.7 ± 0.6 versus -14.8 ± 0.1, respectively (NS)). Post-sacrifice, oxidant stress markers were obtained in the mitochondrial isolates and protein carbonyl in the placebo, and CoQ10 groups were 6.14 ± 0.36 and 5.05 ± 0.32 nmol/mg, respectively (NS). In summary, CoQ10 did not improve contractile reserve or reduce oxidant stress at 4-week post-CABG.
Collapse
|
34
|
Yang BR, Cheung KK, Zhou X, Xie RF, Cheng PP, Wu S, Zhou ZY, Tang JY, Hoi PM, Wang YH, Lee SMY. Amelioration of acute myocardial infarction by saponins from flower buds of Panax notoginseng via pro-angiogenesis and anti-apoptosis. JOURNAL OF ETHNOPHARMACOLOGY 2016; 181:50-58. [PMID: 26806572 DOI: 10.1016/j.jep.2016.01.022] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 12/05/2015] [Accepted: 01/18/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The root of Panax notoginseng is traditionally used as an anti-hemorrhagic agent to promote blood circulation without causing "congealed" blood. Furthermore, the flower of P. notoginseng is a popular, traditional medicine taken daily for the preventing of hypertension and for reducing blood cholesterol profiles. Besides, the flower of P. notoginseng contains a higher level of saponins, particularly protopanaxadiol-type ginsenosides, as compared to the root. However, detailed pharmacological studies on this flower have rarely been conducted. MATERIAL AND METHODS In this study, the saponins extracted from the flower of P. notoginseng (FS) were examined on the endothelial cell migration assay, chemically induced vascular insufficiency model in zebrafish larvae and myocardial infraction (MI) model in rats, for determination of their pro-angiogenic and therapeutic effects on MI treatment. RESULTS Our results demonstrate that FS significantly promoted VEGF-induced migration of human umbilical vein endothelial cells (HUVECs) and partially restored defective intersegmental vessels (ISV) in a chemically induced vascular insufficiency model of zebrafish larvae. When compared to MI group, two weeks post-treatment of FS (25-50mg/kg/day) induced approximately 3-fold upregulation of VEGF mRNA expression and a concomitant increase in blood vessel density in the peri-infarct area of the heart. Moreover, TUNEL analysis indicates a reduction in the mean apoptotic nuclei per field in peri-infarct myocardium upon FS treatment. CONCLUSIONS The pro-angiogenic effects of FS demonstrated in in vitro and in vivo experimental models suggest that the purified saponin preparation from flowers of P. notoginseng may potentially provide preventive and therapeutic agent for cardiovascular diseases.
Collapse
Affiliation(s)
- Bin-Rui Yang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Kwok-Kuen Cheung
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Xin Zhou
- Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Rui-Fang Xie
- Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Pei-Pei Cheng
- Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Shen Wu
- Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Zhong-Yan Zhou
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China; Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jing Yi Tang
- Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - You-Hua Wang
- Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| |
Collapse
|
35
|
Immunotolerant Properties of Mesenchymal Stem Cells: Updated Review. Stem Cells Int 2015; 2016:1859567. [PMID: 26839557 PMCID: PMC4709780 DOI: 10.1155/2016/1859567] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 10/03/2015] [Accepted: 10/11/2015] [Indexed: 12/24/2022] Open
Abstract
Stem cell transplantation is a potential therapeutic option to regenerate damaged myocardium and restore function after infarct. Current research is focused on the use of allogeneic mesenchymal stem cells (MSCs) due to their unique immunomodulatory characteristics and ability to be harvested from young and healthy donors. Both animal and human studies support the immunoprivileged state of MSCs and even demonstrate improvements in cardiac function after transplantation. This research continues to be a topic of interest, as advances will ultimately enable the clinical use of these universal cells for therapy after a myocardial infarction. Updated in vitro, in vivo, and clinical trial studies are discussed in detail in the following review.
Collapse
|
36
|
Overexpression of microRNA-133a inhibits ischemia-reperfusion-induced cardiomyocyte apoptosis by targeting DAPK2. J Hum Genet 2015; 60:709-16. [PMID: 26334104 DOI: 10.1038/jhg.2015.96] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 05/25/2015] [Accepted: 06/30/2015] [Indexed: 01/02/2023]
Abstract
To examine microRNA-133a (miR-133a) endogenous expression in cardiomyocytes after ischemia-reperfusion (I/R) injury and study the effects of miR-133a overexpression on I/R injury-induced cardiomyocyte apoptosis. Dual-Luciferase Reporter Assay detected dynamic expression of miR-133a. In an in vitro hypoxia-reoxygenation (HR) injury model and an in vivo rat model of I/R injury, rat cardiomyocytes were transfected with miR-133a mimic to test the effects of miR-133a overexpression on apoptosis. MiR-133a and Death Associated Protein Kinase 2 (DAPK2) mRNA expression was measured using real-time-PCR, and DAPK2 protein expression was detected by western blotting. Annexin V-fluorescein isothiocyanate/propidium iodide (PI) double-staining measured the apoptosis rate in H9C2 cells and transferase dUTP nick end labeling assay quantified the cardiomyocyte apoptosis rate in tissues obtained from in vivo the rat model. DAPK2 is a target of miR-133a. Both in vitro and in vivo results confirmed that after expression of miR-133a mimics, miR-133a levels increased, which was accompanied by decrease in DAPK2 mRNA and protein expression. In H9C2 cells, HR injury caused a sharp decrease in miR-133a expression and a significant upregualtion of DAPK2 mRNA and protein levels. However, exogenous miR-133a expression led to a significant reduction in DAPK2 mRNA and protein levels despite HR injury. Similar results were obtained from in vivo I/R injury model. After HR injury or I/R injury the apoptosis rate of myocardial cells was highly elevated and decreased significantly only after transfection of miR-133a into cardiomyocytes. MiR-133a overexpression may inhibit I/R injury-mediated cardiomyocyte apoptosis by targeting DAPK2, leading to reduced DAPK2 protein, thus miR-133a may potentially have a high therapeutic value in I/R injury.
Collapse
|
37
|
Weil BR, Suzuki G, Leiker MM, Fallavollita JA, Canty JM. Comparative Efficacy of Intracoronary Allogeneic Mesenchymal Stem Cells and Cardiosphere-Derived Cells in Swine with Hibernating Myocardium. Circ Res 2015; 117:634-44. [PMID: 26271689 DOI: 10.1161/circresaha.115.306850] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 08/12/2015] [Indexed: 01/29/2023]
Abstract
RATIONALE Allogeneic bone marrow-derived mesenchymal stem cells (MSCs) and cardiosphere-derived cells (CDCs) have each entered clinical trials, but a direct comparison of these cell types has not been performed in a large animal model of hibernating myocardium. OBJECTIVE Using completely blinded methodology, we compared the efficacy of global intracoronary allogeneic MSCs (icMSCs, ≈35×10(6)) and CDCs (icCDCs, ≈35×10(6)) versus vehicle in cyclosporine-immunosuppressed swine with a chronic left anterior descending coronary artery stenosis (n=26). METHODS AND RESULTS Studies began 3 months after instrumentation when wall thickening was reduced (left anterior descending coronary artery % wall thickening [mean±SD], 38±11% versus 83±26% in remote; P<0.01) and similar among groups. Four weeks after treatment, left anterior descending coronary artery % wall thickening increased similarly after icCDCs and icMSCs, whereas it remained depressed in vehicle-treated controls (icMSCs, 51±13%; icCDCs, 51±17%; vehicle, 34±3%, treatments P<0.05 versus vehicle). There was no change in myocardial perfusion. Both icMSCs and icCDCs increased left anterior descending coronary artery myocyte nuclear density (icMSCs, 1601±279 nuclei/mm(2); icCDCs, 1569±294 nuclei/mm(2); vehicle, 973±181 nuclei/mm(2); treatments P<0.05 versus vehicle) and reduced myocyte diameter (icMSCs, 16.4±1.5 μm; icCDCs, 16.8±1.2 μm; vehicle, 20.2±3.7 μm; treatments P<0.05 versus vehicle) to the same extent. Similar changes in myocyte nuclear density and diameter were observed in the remote region of cell-treated animals. Cell fate analysis using Y-chromosome fluorescent in situ hybridization demonstrated rare cells from sex-mismatched donors. CONCLUSIONS Allogeneic icMSCs and icCDCs exhibit comparable therapeutic efficacy in a large animal model of hibernating myocardium. Both cell types produced equivalent increases in regional function and stimulated myocyte regeneration in ischemic and remote myocardium. The activation of endogenous myocyte proliferation and regression of myocyte cellular hypertrophy support a common mechanism of cardiac repair.
Collapse
Affiliation(s)
- Brian R Weil
- From the Departments of Medicine, Physiology and Biophysics and Biomedical Engineering, and the Clinical and Translational Research Center, University at Buffalo and the VA WNY Health Care System, NY
| | - Gen Suzuki
- From the Departments of Medicine, Physiology and Biophysics and Biomedical Engineering, and the Clinical and Translational Research Center, University at Buffalo and the VA WNY Health Care System, NY
| | - Merced M Leiker
- From the Departments of Medicine, Physiology and Biophysics and Biomedical Engineering, and the Clinical and Translational Research Center, University at Buffalo and the VA WNY Health Care System, NY
| | - James A Fallavollita
- From the Departments of Medicine, Physiology and Biophysics and Biomedical Engineering, and the Clinical and Translational Research Center, University at Buffalo and the VA WNY Health Care System, NY
| | - John M Canty
- From the Departments of Medicine, Physiology and Biophysics and Biomedical Engineering, and the Clinical and Translational Research Center, University at Buffalo and the VA WNY Health Care System, NY.
| |
Collapse
|
38
|
Cardiomyocyte Remodeling in Atrial Fibrillation and Hibernating Myocardium: Shared Pathophysiologic Traits Identify Novel Treatment Strategies? BIOMED RESEARCH INTERNATIONAL 2015. [PMID: 26221599 PMCID: PMC4499626 DOI: 10.1155/2015/587361] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Atrial fibrillation (AF) is the most common arrhythmia and is associated with a high risk of morbidity and mortality. However, there are limited treatment strategies for prevention of disease onset and progression. Development of novel therapies for primary and secondary prevention of AF is critical and requires improved understanding of the cellular and molecular mechanisms underlying the AF disease process. Translational and clinical studies conducted over the past twenty years have revealed that atrial remodeling in AF shares several important pathophysiologic traits with the remodeling processes exhibited by hibernating myocardium that develop in response to chronic ischemia. These shared features, which include an array of structural, metabolic, and electrophysiologic changes, appear to represent a conserved adaptive myocyte response to chronic stress that involves dedifferentiation towards a fetal phenotype to promote survival. In this review, we discuss the pathophysiology of AF, summarize studies supporting a common remodeling program in AF and hibernating myocardium, and propose future therapeutic implications of this emerging paradigm. Ultimately, better understanding of the molecular mechanisms of atrial myocyte remodeling during the onset of AF and the transition from paroxysmal to persistent stages of the disease may facilitate discovery of new therapeutic targets.
Collapse
|
39
|
The Recovery of Hibernating Hearts Lies on a Spectrum: from Bears in Nature to Patients with Coronary Artery Disease. J Cardiovasc Transl Res 2015; 8:244-52. [DOI: 10.1007/s12265-015-9625-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 04/09/2015] [Indexed: 12/13/2022]
|
40
|
Holley CT, Long EK, Butterick TA, Duffy CM, Lindsey ME, Stone LH, McFalls EO, Kelly RF. Mitochondrial fusion proteins in revascularized hibernating hearts. J Surg Res 2015; 195:29-36. [DOI: 10.1016/j.jss.2014.12.052] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 12/29/2014] [Accepted: 12/31/2014] [Indexed: 12/11/2022]
|
41
|
Page BJ, Banas MD, Suzuki G, Weil BR, Young RF, Fallavollita JA, Palka BA, Canty JM. Revascularization of chronic hibernating myocardium stimulates myocyte proliferation and partially reverses chronic adaptations to ischemia. J Am Coll Cardiol 2015; 65:684-97. [PMID: 25677430 DOI: 10.1016/j.jacc.2014.11.040] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 11/05/2014] [Accepted: 11/11/2014] [Indexed: 12/30/2022]
Abstract
BACKGROUND The time course and extent of recovery after revascularization of viable dysfunctional myocardium are variable. Although fibrosis is a major determinant, myocyte structural and molecular remodeling may also play important roles. OBJECTIVES This study sought to determine whether persistent myocyte loss and/or irreversibility of protein changes that develop in hibernating myocardium have an impact on functional recovery in the absence of infarction. METHODS Swine implanted with a chronic left anterior descending artery (LAD) stenosis to produce hibernating myocardium underwent percutaneous revascularization, with serial functional recovery evaluated for 1 month (n = 12). Myocardial tissue was evaluated to assess myocyte size, nuclear density, and proliferation indexes in comparison with those of normal animals and nonrevascularized controls. Proteomic analysis by 2-dimensional differential in-gel electrophoresis was used to determine the reversibility of molecular adaptations of hibernating myocytes. RESULTS At 3 months, physiological features of hibernating myocardium were confirmed, with depressed LAD wall thickening and no significant infarction. Revascularization normalized LAD flow reserve, with no immediate change in LAD wall thickening. Regional LAD wall thickening slowly improved but remained depressed 1 month post-percutaneous coronary intervention. Surprisingly, revascularization was associated with histological evidence of myocytes re-entering the growth phase of the cell cycle and increases in the number of c-Kit(+) cells. Myocyte nuclear density returned to normal, whereas regional myocyte hypertrophy regressed. Proteomic analysis demonstrated heterogeneous effects of revascularization. Up-regulated stress and cytoskeletal proteins normalized, whereas reduced contractile and metabolic proteins persisted. CONCLUSIONS Delayed recovery of hibernating myocardium in the absence of scar may reflect persistent reductions in the amounts of contractile and metabolic proteins. Although revascularization appeared to stimulate myocyte proliferation, the persistence of small immature myocytes may have contributed to delayed functional recovery.
Collapse
Affiliation(s)
- Brian J Page
- UB Clinical and Translational Research Center and Department of Medicine, University at Buffalo, Buffalo, New York
| | - Michael D Banas
- UB Clinical and Translational Research Center and Department of Medicine, University at Buffalo, Buffalo, New York
| | - Gen Suzuki
- UB Clinical and Translational Research Center and Department of Medicine, University at Buffalo, Buffalo, New York
| | - Brian R Weil
- UB Clinical and Translational Research Center and Department of Medicine, University at Buffalo, Buffalo, New York
| | - Rebeccah F Young
- UB Clinical and Translational Research Center and Department of Medicine, University at Buffalo, Buffalo, New York
| | - James A Fallavollita
- UB Clinical and Translational Research Center and Department of Medicine, University at Buffalo, Buffalo, New York; VA Western New York Health Care System, Buffalo, New York
| | - Beth A Palka
- UB Clinical and Translational Research Center and Department of Medicine, University at Buffalo, Buffalo, New York
| | - John M Canty
- UB Clinical and Translational Research Center and Department of Medicine, University at Buffalo, Buffalo, New York; VA Western New York Health Care System, Buffalo, New York; Department of Physiology and Biophysics and Department of Biomedical Engineering, University at Buffalo, Buffalo, New York.
| |
Collapse
|
42
|
Cardiac function in a long-term follow-up study of moderate and severe porcine model of chronic myocardial infarction. BIOMED RESEARCH INTERNATIONAL 2015; 2015:209315. [PMID: 25802838 PMCID: PMC4352740 DOI: 10.1155/2015/209315] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 10/14/2014] [Indexed: 11/17/2022]
Abstract
BACKGROUND Novel therapies need to be evaluated in a relevant large animal model that mimics the clinical course and treatment in a reasonable time frame. To reliably assess therapeutic efficacy, knowledge regarding the translational model and the course of disease is needed. METHODS Landrace pigs were subjected to a transient occlusion of the proximal left circumflex artery (LCx) (n = 6) or mid-left anterior descending artery (LAD) (n = 6) for 150 min. Cardiac function was evaluated before by 2D echocardiography or 3D echocardiography and pressure-volume loop analysis. At 12 weeks of follow-up the heart was excised for histological analysis and infarct size calculations. RESULTS Directly following AMI, LVEF was severely reduced compared to baseline in the LAD group (-17.1 ± 1.6%, P = 0.009) compared to only a moderate reduction in the LCx group (-5.9 ± 1.5%, P = 0.02) and this effect remained unchanged during 12 weeks of follow-up. CONCLUSION Two models of chronic MI, representative for different patient groups, can reproducibly be created through clinically relevant ischemia-reperfusion of the mid-LAD and proximal LCx.
Collapse
|
43
|
Ayaz O, Howlett SE. Testosterone modulates cardiac contraction and calcium homeostasis: cellular and molecular mechanisms. Biol Sex Differ 2015; 6:9. [PMID: 25922656 PMCID: PMC4411792 DOI: 10.1186/s13293-015-0027-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 03/27/2015] [Indexed: 02/06/2023] Open
Abstract
The incidence of cardiovascular disease rises dramatically with age in both men and women. Because a woman's risk of cardiovascular disease rises markedly after the onset of menopause, there has been growing interest in the effect of estrogen on the heart and its role in the pathophysiology of these diseases. Much less attention has been paid to the impact of testosterone on the heart, even though the levels of testosterone also decline with age and low-testosterone levels are linked to the development of cardiovascular diseases. The knowledge that receptors for all major sex steroid hormones, including testosterone, are present on individual cardiomyocytes suggests that these hormones may influence the heart at the cellular level. Indeed, it is well established that there are male-female differences in intracellular Ca(2+) release and contraction in isolated ventricular myocytes. Growing evidence suggests that these differences arise from effects of sex steroid hormones on processes involved in intracellular Ca(2+) homeostasis. This review considers how myocardial contractile function is modified by testosterone, with a focus on the impact of testosterone on processes that regulate Ca(2+) handling at the level of the ventricular myocyte. The idea that testosterone regulates Ca(2+) handling in the heart is important, as Ca(2+) dysregulation plays a key role in the pathogenesis of a variety of different cardiovascular diseases. A better understanding of sex hormone regulation of myocardial Ca(2+) homeostasis may reveal new targets for the treatment of cardiovascular diseases in all older adults.
Collapse
Affiliation(s)
- Omar Ayaz
- Department of Pharmacology, Dalhousie University, 5850 College Street, Sir Charles Tupper Medical Building, PO Box 15000, Halifax, NS B3H 4R2 Canada
| | - Susan Ellen Howlett
- Department of Pharmacology, Dalhousie University, 5850 College Street, Sir Charles Tupper Medical Building, PO Box 15000, Halifax, NS B3H 4R2 Canada
- Medicine (Geriatric Medicine), Dalhousie University, 5850 College Street, PO Box 15000, Halifax, NS B3H 4R2 Canada
| |
Collapse
|
44
|
Holley CT, Long EK, Lindsey ME, McFalls EO, Kelly RF. Recovery of hibernating myocardium: what is the role of surgical revascularization? J Card Surg 2014; 30:224-31. [PMID: 25470424 DOI: 10.1111/jocs.12477] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Myocardial responses to chronic ischemia represent a continuum of adaptations resulting, over time, in a stress-resistant phenotype. One such adaptation, hibernating myocardium (HM), has increased antioxidant capacity that protects against ischemia-induced oxidative stress. Studies have suggested that revascularization alone may not fully restore cardiac function, highlighting the need for targeted therapies to serve as adjuncts to the innate healing process following revascularization. In our review, we discuss current understanding of HM and the recovery process following surgical revascularization, focusing on animal models of HM to understand implications for human patients.
Collapse
|
45
|
Duncker DJ, Koller A, Merkus D, Canty JM. Regulation of coronary blood flow in health and ischemic heart disease. Prog Cardiovasc Dis 2014; 57:409-22. [PMID: 25475073 DOI: 10.1016/j.pcad.2014.12.002] [Citation(s) in RCA: 153] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The major factors determining myocardial perfusion and oxygen delivery have been elucidated over the past several decades, and this knowledge has been incorporated into the management of patients with ischemic heart disease (IHD). The basic understanding of the fluid mechanical behavior of coronary stenoses has also been translated to the cardiac catheterization laboratory where measurements of coronary pressure distal to a stenosis and coronary flow are routinely obtained. However, the role of perturbations in coronary microvascular structure and function, due to myocardial hypertrophy or coronary microvascular dysfunction, in IHD is becoming increasingly recognized. Future studies should therefore be aimed at further improving our understanding of the integrated coronary microvascular mechanisms that control coronary blood flow, and of the underlying causes and mechanisms of coronary microvascular dysfunction. This knowledge will be essential to further improve the treatment of patients with IHD.
Collapse
Affiliation(s)
- Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Cardiovascular Research Institute COEUR, Erasmus MC, University Medical School, Rotterdam, The Netherlands.
| | - Akos Koller
- Department of Pathophysiology and Gerontology, Medical School, University of Pécs, Hungary; Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Cardiovascular Research Institute COEUR, Erasmus MC, University Medical School, Rotterdam, The Netherlands
| | - John M Canty
- Division of Cardiovascular Medicine, University at Buffalo and the Western New York Department of Veterans Affairs Health System, Buffalo, NY, USA
| |
Collapse
|
46
|
Bayeva M, Sawicki KT, Butler J, Gheorghiade M, Ardehali H. Molecular and cellular basis of viable dysfunctional myocardium. Circ Heart Fail 2014; 7:680-91. [PMID: 25028350 DOI: 10.1161/circheartfailure.113.000912] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Marina Bayeva
- From the Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (M.B., K.T.S., M.G., H.A.); and Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (J.B.)
| | - Konrad Teodor Sawicki
- From the Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (M.B., K.T.S., M.G., H.A.); and Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (J.B.)
| | - Javed Butler
- From the Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (M.B., K.T.S., M.G., H.A.); and Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (J.B.)
| | - Mihai Gheorghiade
- From the Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (M.B., K.T.S., M.G., H.A.); and Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (J.B.)
| | - Hossein Ardehali
- From the Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (M.B., K.T.S., M.G., H.A.); and Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (J.B.).
| |
Collapse
|
47
|
Lim SP, Mc Ardle BA, Beanlands RS, Hessian RC. Myocardial Viability: It is Still Alive. Semin Nucl Med 2014; 44:358-74. [DOI: 10.1053/j.semnuclmed.2014.07.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
48
|
Shen X, Young R, Canty JM, Qu J. Quantitative proteomics in cardiovascular research: global and targeted strategies. Proteomics Clin Appl 2014; 8:488-505. [PMID: 24920501 DOI: 10.1002/prca.201400014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/02/2014] [Accepted: 06/06/2014] [Indexed: 11/05/2022]
Abstract
Extensive technical advances in the past decade have substantially expanded quantitative proteomics in cardiovascular research. This has great promise for elucidating the mechanisms of cardiovascular diseases and the discovery of cardiac biomarkers used for diagnosis and treatment evaluation. Global and targeted proteomics are the two major avenues of quantitative proteomics. While global approaches enable unbiased discovery of altered proteins via relative quantification at the proteome level, targeted techniques provide higher sensitivity and accuracy, and are capable of multiplexed absolute quantification in numerous clinical/biological samples. While promising, technical challenges need to be overcome to enable full utilization of these techniques in cardiovascular medicine. Here, we discuss recent advances in quantitative proteomics and summarize applications in cardiovascular research with an emphasis on biomarker discovery and elucidating molecular mechanisms of disease. We propose the integration of global and targeted strategies as a high-throughput pipeline for cardiovascular proteomics. Targeted approaches enable rapid, extensive validation of biomarker candidates discovered by global proteomics. These approaches provide a promising alternative to immunoassays and other low-throughput means currently used for limited validation.
Collapse
Affiliation(s)
- Xiaomeng Shen
- Department of Biochemistry, University at Buffalo, Buffalo, NY, USA; New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY, USA
| | | | | | | |
Collapse
|
49
|
Quindry JC, Hamilton KL. Exercise and cardiac preconditioning against ischemia reperfusion injury. Curr Cardiol Rev 2014; 9:220-9. [PMID: 23909636 PMCID: PMC3780347 DOI: 10.2174/1573403x113099990033] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Accepted: 06/02/2013] [Indexed: 12/30/2022] Open
Abstract
Cardiovascular disease (CVD), including ischemia reperfusion (IR) injury, remains a major cause of morbidity and mortality in industrialized nations. Ongoing research is aimed at uncovering therapeutic interventions against IR injury. Regular exercise participation is recognized as an important lifestyle intervention in the prevention and treatment of CVD and IR injury. More recent understanding reveals that moderate intensity aerobic exercise is also an important experimental model for understanding the cellular mechanisms of cardioprotection against IR injury. An important discovery in this regard was the observation that one-to-several days of exercise will attenuate IR injury. This phenomenon has been observed in young and old hearts of both sexes. Due to the short time course of exercise induced protection, IR injury prevention must be mediated by acute biochemical alterations within the myocardium. Research over the last decade reveals that redundant mechanisms account for exercise induced cardioprotection against IR. While much is now known about exercise preconditioning against IR injury, many questions remain. Perhaps most pressing, is what mechanisms mediate cardioprotection in aged hearts and what sex-dependent differences exist. Given that that exercise preconditioning is a polygenic effect, it is likely that multiple mediators of exercise induced cardioprotection have yet to be uncovered. Also unknown, is whether post translational modifications due to exercise are responsible for IR injury prevention. This review will provide an overview the major mechanisms of IR injury and exercise preconditioning. The discussion highlights many promising avenues for further research and describes how exercise preconditioning may continue to be an important scientific paradigm in the translation of cardioprotection research to the clinic.
Collapse
Affiliation(s)
- John C Quindry
- Cardioprotection Laboratory, Department of Kinesiology, Auburn University, AL 36849, USA
| | | |
Collapse
|
50
|
Qu J, Young R, Page BJ, Shen X, Tata N, Li J, Duan X, Fallavollita JA, Canty JM. Reproducible ion-current-based approach for 24-plex comparison of the tissue proteomes of hibernating versus normal myocardium in swine models. J Proteome Res 2014; 13:2571-84. [PMID: 24697261 PMCID: PMC4015685 DOI: 10.1021/pr5000472] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Hibernating
myocardium is an adaptive response to repetitive myocardial
ischemia that is clinically common, but the mechanism of adaptation
is poorly understood. Here we compared the proteomes of hibernating
versus normal myocardium in a porcine model with 24 biological replicates.
Using the ion-current-based proteomic strategy optimized in this study
to expand upon previous proteomic work, we identified differentially
expressed proteins in new molecular pathways of cardiovascular interest.
The methodological strategy includes efficient extraction with detergent
cocktail; precipitation/digestion procedure with high, quantitative
peptide recovery; reproducible nano-LC/MS analysis on a long, heated
column packed with small particles; and quantification based on ion-current
peak areas. Under the optimized conditions, high efficiency and reproducibility
were achieved for each step, which enabled a reliable comparison of
24 the myocardial samples. To achieve confident discovery of differentially
regulated proteins in hibernating myocardium, we used highly stringent
criteria to define “quantifiable proteins”. These included
the filtering criteria of low peptide FDR and S/N > 10 for peptide
ion currents, and each protein was quantified independently from ≥2
distinct peptides. For a broad methodological validation, the quantitative
results were compared with a parallel, well-validated 2D-DIGE analysis
of the same model. Excellent agreement between the two orthogonal
methods was observed (R = 0.74), and the ion-current-based
method quantified almost one order of magnitude more proteins. In
hibernating myocardium, 225 significantly altered proteins were discovered
with a low false-discovery rate (∼3%). These proteins are involved
in biological processes including metabolism, apoptosis, stress response,
contraction, cytoskeleton, transcription, and translation. This provides
compelling evidence that hibernating myocardium adapts to chronic
ischemia. The major metabolic mechanisms include a down-regulation
of mitochondrial respiration and an increase in glycolysis. Meanwhile,
cardioprotective and cytoskeletal proteins are increased, while cardiomyocyte
contractile proteins are reduced. These intrinsic adaptations to regional
ischemia maintain long-term cardiomyocyte viability at the expense
of contractile function.
Collapse
Affiliation(s)
- Jun Qu
- Department of Pharmaceutical Sciences, ‡Department of Biochemistry, §Department of Medicine, ∥Department of Physiology and Biophysics, ⊥The Center for Research in Cardiovascular Medicine, and #Center for Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo , Buffalo, New York 14214, United States
| | | | | | | | | | | | | | | | | |
Collapse
|