1
|
Charbonneau H, Savy S, Savy N, Pasquié M, Mayeur N, Angles O, Balech V, Berthelot AL, Croute-Bayle M, Decramer I, Duterque D, Julien V, Mallet L, M'rini M, Quedreux JF, Richard B, Sidobre L, Taillefer L, Thibaud A, Abouliatim I, Berthoumieu P, Garcia O, Soula P, Vahdat O, Breil C, Brunel P, Sciacca G. Comprehensive perioperative blood management in patients undergoing elective bypass cardiac surgery: Benefit effect of health care education and systematic correction of iron deficiency and anemia on red blood cell transfusion. J Clin Anesth 2024; 98:111560. [PMID: 39146724 DOI: 10.1016/j.jclinane.2024.111560] [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: 05/08/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024]
Abstract
STUDY OBJECTIVE The aim of this study was to investigate the efficacy of a two-step patient blood management (PBM) program in red blood cell (RBC) transfusion requirements among patients undergoing elective cardiopulmonary bypass (CPB) surgery. DESIGN Prospective, non-randomized, two-step protocol design. SETTING Cardiac surgery department of Clinique Pasteur, Toulouse, France. PATIENTS 897 patients undergoing for elective CPB surgery. INTERVENTIONS We conducted a two-steps protocol: PBMe and PBMc. PBMe involved a short quality improvement program for health care workers, while PBMc introduced a systematic approach to pre- and postoperative correction of deficiencies, incorporating iron injections, oral vitamins, and erythropoiesis-stimulating agents. MEASUREMENTS The PBM program's effectiveness was evaluated through comparison with a pre-PBM retrospective cohort after propensity score matching. The primary objective was the proportion of patients requiring RBC transfusions during their hospital stay. Secondary objectives were also analyzed. MAIN RESULTS After matching, 343 patients were included in each group. Primary outcomes were observed in 35.7% (pre-PBM), 26.7% (PBMe), and 21.1% (PBMc) of patients, resulting in a significant reduction (40.6%) in the overall RBC transfusion rate. Both the PBMe and PBMc groups exhibited significantly lower risks of RBC transfusion compared to the pre-PBM group, with adjusted odds ratios of 0.59 [95% CI 0.44-0.79] and 0.44 [95% CI 0.32-0.60], respectively. Secondary endpoints included reductions in transfusions exceeding 2 units, total RBC units transfused, administration of allogeneic blood products, and total bleeding volume recorded on Day 1. There were no significant differences noted in mortality rates or the duration of hospital stays. CONCLUSIONS This study suggests that health care education and systematic deficiency correction are associated with reduced RBC transfusion rates in elective CPB surgery. However, further randomized, controlled studies are needed to validate these findings and refine their clinical application.
Collapse
Affiliation(s)
- Hélène Charbonneau
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - Stéphanie Savy
- Clinical Research Committee, Clinique Pasteur, Toulouse, France.
| | - Nicolas Savy
- Toulouse Institute of Mathematics, UMR5219 - University of Toulouse; CNRS - UPS IMT, Toulouse, France.
| | - Marie Pasquié
- Clinical Research Committee, Clinique Pasteur, Toulouse, France.
| | - Nicolas Mayeur
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - Olivier Angles
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - Vincent Balech
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - Anne-Laure Berthelot
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - Madeleine Croute-Bayle
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - Isabelle Decramer
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - David Duterque
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - Valerie Julien
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - Laurent Mallet
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - Mimoun M'rini
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - Jean-François Quedreux
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - Benoit Richard
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - Laurent Sidobre
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - Laurence Taillefer
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - Adrien Thibaud
- Department of Anesthesiology and Intensive Care Unit, Clinique Pasteur, Toulouse, France.
| | - Issam Abouliatim
- Department of Cardiothoracic and Vascular Surgery, Clinique Pasteur, Toulouse, France.
| | - Pierre Berthoumieu
- Department of Cardiothoracic and Vascular Surgery, Clinique Pasteur, Toulouse, France.
| | - Olivier Garcia
- Department of Cardiothoracic and Vascular Surgery, Clinique Pasteur, Toulouse, France.
| | - Philippe Soula
- Department of Cardiothoracic and Vascular Surgery, Clinique Pasteur, Toulouse, France.
| | - Olivier Vahdat
- Department of Cardiothoracic and Vascular Surgery, Clinique Pasteur, Toulouse, France.
| | - Claude Breil
- Department of Cardiothoracic and Vascular Surgery, Clinique Pasteur, Toulouse, France.
| | - Pierre Brunel
- Department of Cardiothoracic and Vascular Surgery, Clinique Pasteur, Toulouse, France.
| | - Giovanni Sciacca
- Department of Cardiothoracic and Vascular Surgery, Clinique Pasteur, Toulouse, France.
| |
Collapse
|
2
|
Zhu Y, Cai G, Lin L, Fu H, Zhang C, Zeng L, Tu C, Yang Z. Age-associated declined function of endothelial progenitor cells and its correlation with plasma IL-18 or IL-23 concentrations in patients with ST-segment elevation myocardial infarction. Front Cardiovasc Med 2024; 11:1351567. [PMID: 38854655 PMCID: PMC11157231 DOI: 10.3389/fcvm.2024.1351567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/03/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND ST-segment elevation myocardial infarction (STEMI) persists to be prevalent in the elderly with a dismal prognosis. The capacity of endothelial progenitor cells (EPCs) is reduced with aging. Nevertheless, the influence of aging on the functionality of EPCs in STEMI is not fully understood. METHOD This study enrolled 20 younger STEMI patients and 21 older STEMI patients. We assessed the Thrombolysis in Myocardial Infarction (TIMI) and Global Registry of Acute Coronary Events Risk (GRACE) scores in two groups. Then, we detected EPC migration, proliferation, adhesion, and plasma interleukin (IL)-18 and IL-23 concentrations in two groups. In addition, we analyzed the interconnection between age, EPC function, plasma IL-18 and IL-23 concentrations, and GRACE or TIMI scores in STEMI patients. RESULT GRACE and TIMI scores in older STEMI patients were higher than in younger STEMI patients, whereas EPC function declined. GRACE and TIMI scores were found to have an inverse relationship with the EPC function. In older STEMI patients, plasma concentrations of IL-18 and IL-23 increased. Plasma IL-18 and IL-23 concentrations were adversely connected to EPC capacity and positively related to GRACE and TIMI scores. Moreover, age was positively correlated with plasma IL-18 or IL-23 concentrations, as well as GRACE or TIMI scores. However, age was adversely correlated with EPC function. CONCLUSION In patients with STEMI, aging results in declined EPC function, which may be associated with inflammatory cytokines. The current investigation may offer new perception about mechanism and therapeutic targets of aging STEMI.
Collapse
Affiliation(s)
- Yuanting Zhu
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Guoyi Cai
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Luyang Lin
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Hongna Fu
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Cong Zhang
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Gastroenterology, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lijin Zeng
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Chang Tu
- Department of Cardiology, SSL Central Hospital of Dongguan City, Dongguan, China
| | - Zhen Yang
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
3
|
Lee J, Rogers HM, Springer DA, Noguchi CT. Neuronal nitric oxide synthase required for erythropoietin modulation of heart function in mice. Front Physiol 2024; 15:1338476. [PMID: 38628440 PMCID: PMC11019009 DOI: 10.3389/fphys.2024.1338476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/04/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction: Erythropoietin (EPO) acts primarily in regulating red blood cell production mediated by high EPO receptor (EPOR) expression in erythroid progenitor cells. EPO activity in non-erythroid tissue is evident in mice with EPOR restricted to erythroid tissues (ΔEPORE) that become obese, glucose-intolerant, and insulin-resistant. In animal models, nitric oxide synthase (NOS) contributes to EPO activities including erythropoiesis, neuroprotection, and cardioprotection against ischemia-reperfusion injury. However, we found that extended EPO treatment to increase hematocrit compromised heart function, while the loss of neuronal NOS (nNOS) was protective against the deleterious activity of EPO to promote heart failure. Methods: Wild-type (WT) mice, ΔEPORE mice, and nNOS-knockout mice (nNOS-/-) were placed on a high-fat diet to match the ΔEPORE obese phenotype and were treated with EPO for 3 weeks. Hematocrit and metabolic response to EPO treatment were monitored. Cardiac function was assessed by echocardiography and ultrasonography. Results: ΔEPORE mice showed a decrease in the left ventricular outflow tract (LVOT) peak velocity, ejection fraction, and fractional shortening, showing that endogenous non-erythroid EPO response is protective for heart function. EPO treatment increased hematocrit in all mice and decreased fat mass in male WT, demonstrating that EPO regulation of fat mass requires non-erythroid EPOR. EPO treatment also compromised heart function in WT mice, and decreased the pulmonary artery peak velocity (PA peak velocity), LVOT peak velocity, ejection fraction, and fractional shortening, but it had minimal effect in further reducing the heart function in ΔEPORE mice, indicating that the adverse effect of EPO on heart function is not related to EPO-stimulated erythropoiesis. ΔEPORE mice had increased expression of heart failure-associated genes, hypertrophic cardiomyopathy-related genes, and sarcomeric genes that were also elevated with EPO treatment in WT mice. Male and female nNOS-/- mice were protected against diet-induced obesity. EPO treatment in nNOS-/- mice increased the hematocrit that tended to be lower than WT mice and decreased the PA peak velocity but did not affect the LVOT peak velocity, ejection fraction, and fractional shortening, suggesting that nNOS is required for the adverse effect of EPO treatment on heart function in WT mice. EPO treatment did not change expression of heart failure-associated gene expression in nNOS-/- mice. Discussion: Endogenous EPO has a protective effect on heart function. With EPO administration, in contrast to the protective effect to the cardiac injury of acute EPO treatment, extended EPO treatment to increase hematocrit in WT mice adversely affected the heart function with a corresponding increase in expression of heart failure-associated genes. This EPO activity was independent of EPO-stimulated erythropoiesis and required EPOR in non-erythroid tissue and nNOS activity, while nNOS-/- mice were protected from the EPO-associated adverse effect on heart function. These data provide evidence that nNOS contributes to the negative impact on the heart function of high-dose EPO treatment for anemia.
Collapse
Affiliation(s)
- Jeeyoung Lee
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Heather M. Rogers
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Danielle A. Springer
- Murine Phenotyping Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Constance T. Noguchi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
4
|
Miura T, Sato T, Yano T, Takaguri A, Miki T, Tohse N, Nishizawa K. Role of Erythropoiesis-Stimulating Agents in Cardiovascular Protection in CKD Patients: Reappraisal of Their Impact and Mechanisms. Cardiovasc Drugs Ther 2023; 37:1175-1192. [PMID: 35150385 DOI: 10.1007/s10557-022-07321-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/28/2022] [Indexed: 11/28/2022]
Abstract
Erythropoiesis-stimulating agents (ESAs) have markedly reduced the need for blood transfusion for renal anemia and are included in standard therapies for patients with chronic kidney disease (CKD). Various protective effects of ESAs on the cardiovascular system have been discovered through basic research, and the effects have received much attention because the rates of cardiovascular events and mortality are high in CKD patients. However, randomized clinical trials did not provide strong evidence that ESAs exert cardioprotection in humans, including CKD patients. It is difficult to assess the cardioprotective effects of ESAs in CKD patients through the clinical data that has been reported to date because the relationship between hemoglobin level rather than ESA dose and cardiovascular event rates was examined in most studies. Interestingly, recent studies using a rat model of CKD showed that the infarct size-limiting effect of an ESA was lost when its dose was increased to a level that normalized blood hemoglobin levels, suggesting that the optimal dose of an ESA for myocardial protection is less than the dose required to normalize hemoglobin levels. Furthermore, animal models of traditional coronary risk factors or comorbidities were resistant to the cardioprotective effects of ESAs because of interruptions in signal-mediated mechanisms downstream of erythropoietin receptors. In this review, we briefly discuss basic and clinical data on the impact of anemia on coronary and systemic circulation, the effects of CKD on the cardiovascular system, and the multiple pharmacological actions of ESAs to examine whether the ESAs that are prescribed for renal anemia exert any cardioprotection in patients with CKD.
Collapse
Affiliation(s)
- Tetsuji Miura
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 15-4-1, Maeda-7, Teine-ku, Sapporo, Japan.
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshiyuki Yano
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Akira Takaguri
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Japan
| | - Takayuki Miki
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Cardiology and Diabetes, Oji General Hospital, Tomakomai, Japan
| | - Noritsugu Tohse
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Keitaro Nishizawa
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Nephrology, Asahikawa Red Cross, Hospital, Asahikawa, Japan
| |
Collapse
|
5
|
Kittur FS, Hung CY, Li PA, Sane DC, Xie J. Asialo-rhuEPO as a Potential Neuroprotectant for Ischemic Stroke Treatment. Pharmaceuticals (Basel) 2023; 16:610. [PMID: 37111367 PMCID: PMC10143832 DOI: 10.3390/ph16040610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Neuroprotective drugs to protect the brain against cerebral ischemia and reperfusion (I/R) injury are urgently needed. Mammalian cell-produced recombinant human erythropoietin (rhuEPOM) has been demonstrated to have excellent neuroprotective functions in preclinical studies, but its neuroprotective properties could not be consistently translated in clinical trials. The clinical failure of rhuEPOM was thought to be mainly due to its erythropoietic activity-associated side effects. To exploit its tissue-protective property, various EPO derivatives with tissue-protective function only have been developed. Among them, asialo-rhuEPO, lacking terminal sialic acid residues, was shown to be neuroprotective but non-erythropoietic. Asialo-rhuEPO can be prepared by enzymatic removal of sialic acid residues from rhuEPOM (asialo-rhuEPOE) or by expressing human EPO gene in glycoengineered transgenic plants (asialo-rhuEPOP). Both types of asialo-rhuEPO, like rhuEPOM, displayed excellent neuroprotective effects by regulating multiple cellular pathways in cerebral I/R animal models. In this review, we describe the structure and properties of EPO and asialo-rhuEPO, summarize the progress on neuroprotective studies of asialo-rhuEPO and rhuEPOM, discuss potential reasons for the clinical failure of rhuEPOM with acute ischemic stroke patients, and advocate future studies needed to develop asialo-rhuEPO as a multimodal neuroprotectant for ischemic stroke treatment.
Collapse
Affiliation(s)
- Farooqahmed S. Kittur
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| | - Chiu-Yueh Hung
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| | - P. Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| | - David C. Sane
- Carilion Clinic and Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA;
| | - Jiahua Xie
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| |
Collapse
|
6
|
Soldozy S, Dalzell C, Skaff A, Ali Y, Norat P, Yagmurlu K, Park MS, Kalani MYS. Reperfusion injury in acute ischemic stroke: Tackling the irony of revascularization. Clin Neurol Neurosurg 2023; 225:107574. [PMID: 36696846 DOI: 10.1016/j.clineuro.2022.107574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 12/12/2022] [Accepted: 12/23/2022] [Indexed: 01/06/2023]
Abstract
Reperfusion injury is an unfortunate consequence of restoring blood flow to tissue after a period of ischemia. This phenomenon can occur in any organ, although it has been best studied in cardiac cells. Based on cardiovascular studies, neuroprotective strategies have been developed. The molecular biology of reperfusion injury remains to be fully elucidated involving several mechanisms, however these mechanisms all converge on a similar final common pathway: blood brain barrier disruption. This results in an inflammatory cascade that ultimately leads to a loss of cerebral autoregulation and clinical worsening. In this article, the authors present an overview of these mechanisms and the current strategies being employed to minimize injury after restoration of blood flow to compromised cerebral territories.
Collapse
Affiliation(s)
- Sauson Soldozy
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, USA; Department of Neurosurgery, Westchester Medical Center, Valhalla, NY, USA
| | - Christina Dalzell
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, USA
| | - Anthony Skaff
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, USA
| | - Yusuf Ali
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, USA
| | - Pedro Norat
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, USA
| | - Kaan Yagmurlu
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, USA
| | - Min S Park
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, USA
| | - M Yashar S Kalani
- Department of Surgery, University of Oklahoma, and St. John's Neuroscience Institute, Tulsa, OK, USA.
| |
Collapse
|
7
|
Abdu FA, Galip J, Qi P, Zhang W, Mohammed AQ, Liu L, Yin G, Mohammed AA, Mareai RM, Jiang R, Xu Y, Che W. Association of stress hyperglycemia ratio and poor long-term prognosis in patients with myocardial infarction with non-obstructive coronary arteries. Cardiovasc Diabetol 2023; 22:11. [PMID: 36647062 PMCID: PMC9843969 DOI: 10.1186/s12933-023-01742-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Stress hyperglycemia ratio (SHR) is a novel biomarker of true acute hyperglycemia condition and is associated with a worse prognosis in patients with myocardial infarction (MI). However, the effects of SHR in the setting of MI with non-obstructive coronary arteries (MINOCA) have not been investigated. This study aimed to explore the association between SHR and long-term clinical outcomes among MINOCA patients. METHODS A total of 410 MINOCA patients were included in the final analysis of this study. The patients were divided into three groups based on the SHR tertiles: [SHR1 group (SHR ≤ 0.73), (n = 143); SHR2 group (SHR 0.73-0.84), n = 131; and SHR3 group (SHR ≥ 0.84), n = 136]. Follow-up for major adverse cardiovascular events (MACE) was conducted on all patients. Cox regression and Kaplan-Meier curve analysis were used to evaluate the relationship between SHR and MACE. The receiver operating curve (ROC) analysis was applied to obtain the optimal cut-off value of SHR for predicting clinical MACE. RESULTS A total of 92 patients developed MACE during the mean 34 months of follow-up. A significant increase in MACE was observed in the SHR3 group compared to the SHR1 and SHR2 groups (35.3% vs. 15.4% and 16.8%, respectively; P < 0.001). The Kaplan-Meier curves demonstrate that SHR3 patients had the highest MACE risk compared to SHR1 and SHR2 patients (log-rank P < 0.001). In addition, when both SHR tertiles and diabetes status were considered, those with SHR3 and diabetes had the highest hazard of MACE (log-rank P < 0.001). Multivariate Cox regression analysis showed that the SHR3 is associated with a 2.465-fold increase in the risk of MACE (adjusted HR, 2.465; 95% CI 1.461-4.159, P = 0.001). The ROC curve analysis showed that the optimal SHR cut-off value for predicting clinical MACE among MINOCA was 0.86. CONCLUSION Our data indicates, for the first time, that SHR is independently associated with poor long-term prognosis in patients suffering from MINOCA. The optimal SHR cut-off value for predicting clinical MACE among MINOCA patients was 0.86. These findings suggest that SHR may play a potential role in the cardiovascular risk stratification of the MINOCA population.
Collapse
Affiliation(s)
- Fuad A. Abdu
- grid.24516.340000000123704535Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072 China
| | - Jassur Galip
- grid.24516.340000000123704535Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072 China
| | - Penglong Qi
- grid.24516.340000000123704535Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072 China
| | - Wen Zhang
- grid.24516.340000000123704535Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072 China
| | - Abdul-Quddus Mohammed
- grid.24516.340000000123704535Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072 China
| | - Lu Liu
- grid.24516.340000000123704535Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072 China
| | - Guoqing Yin
- grid.24516.340000000123704535Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072 China
| | - Ayman A. Mohammed
- grid.24516.340000000123704535Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072 China
| | - Redhwan M. Mareai
- grid.24516.340000000123704535Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072 China
| | - Rong Jiang
- grid.24516.340000000123704535Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072 China
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China.
| | - Wenliang Che
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China. .,Department of Cardiology, Shanghai Tenth People's Hospital Chongming branch, Shanghai, China.
| |
Collapse
|
8
|
Tan L, Long LZ, Li HZ, Yang WW, Peng YX, Lu JM, Liao FF, Ma XC, Qu H, Fu CG, Zhang SS. Growth factor for therapeutic angiogenesis in ischemic heart disease: A meta-analysis of randomized controlled trials. Front Cell Dev Biol 2022; 10:1095623. [PMID: 36568984 PMCID: PMC9780500 DOI: 10.3389/fcell.2022.1095623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
Aim: This study was designed to systematically evaluate the effects of growth factor (GF) for therapeutic angiogenesis on ischemic heart disease (IHD) by pooling the results of randomized controlled trials (RCTs). Methods and Results: PubMed, EMBASE, and CENTRAL databases were searched from inception to October 2022. RCTs, investigating the effects of GF therapy on IHD, were included. The risk bias of included study was assessed according to Cochrane tool. Weighted mean difference (WMD), calculated with fixed effect model or random effect model, was used to evaluate the effects of GF therapy on left ventricular ejection fraction (LVEF) and Canadian Cardiovascular Society (CCS) angina class. Relative risk (RR) was used to evaluate the effects of GF therapy on all-cause mortality, major adverse cardiovascular events (MACE) and revascularization. Meta-analysis, meta-regression analysis and publication bias analysis were performed by RevMan 5.3 or Stata 15.1 software. Twenty-nine studies involving 2899 IHD patients (1,577 patients in GF group and 1,322 patients in control group) were included. Compared with the control group, GF therapy did not reduce all-cause mortality (RR: 0.82; 95% CI: 0.54-1.24; p = 0.341), MACE [(RR: 0.83; 95% CI: 0.61-1.12; p = 0.227), revascularization (RR: 1.27, 95% CI: 0.82-1.96, p = 0.290) and CCS angina class (WMD: -0.08, 95% CI: -0.36 to 0.20, p = 0.560). However, GF therapy could increase LVEF during short-term follow-up (<1 year). Conclusion: GF for therapeutic angiogenesis was beneficial for increasing LVEF during short-term follow-up (<1 year), however, the therapy was not efficacious in decreasing all-cause mortality, MACE and revascularization.
Collapse
Affiliation(s)
- Ling Tan
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lin-Zi Long
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hong-Zheng Li
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Wen-Wen Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu-Xuan Peng
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Jie-Ming Lu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Fei-Fei Liao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-Chang Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,National Cardiovascular Clinical Medical Research Center of TCM, Beijing, China
| | - Hua Qu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,National Cardiovascular Clinical Medical Research Center of TCM, Beijing, China,*Correspondence: Hua Qu, ; Chang-Geng Fu, ; Shan-Shan Zhang,
| | - Chang-Geng Fu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,National Cardiovascular Clinical Medical Research Center of TCM, Beijing, China,*Correspondence: Hua Qu, ; Chang-Geng Fu, ; Shan-Shan Zhang,
| | - Shan-Shan Zhang
- Beijing Xibeiwang Town Community Health Service Center, Beijing, China,*Correspondence: Hua Qu, ; Chang-Geng Fu, ; Shan-Shan Zhang,
| |
Collapse
|
9
|
Jung C, Rezar R, Wischmann P, Masyuk M, Datz C, Bruno RR, Kelm M, Wernly B. The role of anemia on admission in acute coronary syndrome - An umbrella review of systematic reviews and meta-analyses. Int J Cardiol 2022; 367:1-10. [PMID: 36055474 DOI: 10.1016/j.ijcard.2022.08.052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/10/2022] [Accepted: 08/24/2022] [Indexed: 01/28/2023]
Abstract
INTRODUCTION The role of erythrocytes in the acute coronary syndrome (ACS) is complex. The aim of this review in terms of PICO (P: patients; I: intervention; C: comparison; O: outcome) was to summarize systematic reviews in patients (P) with acute coronary syndrome, evaluating the effects of (I) 1) iron deficiency, 2) administration of an erythropoiesis-stimulating agent (ESA), 3) anemia on admission, 4) red blood cell transfusion, 5) a restrictive transfusion strategy in comparison (C) to 1) no iron deficiency, 2) no ESA 3) no anemia on admission, 4) no red blood cell transfusion, 5) a liberal transfusion strategy on mortality (O). METHODS We used AMSTAR2 to assess the methodological quality of systematic reviews and grade the available research. The primary endpoint was all-cause mortality. RESULTS Using the data from 2,787,005 patients, the following conditions were associated with worse outcome in patients with ACS: anemia on admission (RR 2.08 95%CI 1.70-2.55) and transfusion (1.93 95%CI 1.12-3.34) of red blood cells. A liberal transfusion (RR 0.86 95%CI 0.70-1-05), administration of ESA (RR 0.55 95%CI 0.22-1.33) and iron deficiency (OR 1.24 95%CI 0.12-13.13) were not associated with altered all-cause mortality. CONCLUSION Patients suffering from ACS and anemia on admission are at particular risk for adverse outcome. There is evidence of associations between adverse outcomes and receiving red blood cell transfusions.
Collapse
Affiliation(s)
- Christian Jung
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Duesseldorf, Germany.
| | - Richard Rezar
- Department of Cardiology and Intensive Care Medicine, Paracelsus Medical University of Salzburg, Müllner Hauptstraße 48, 5020 Salzburg, Austria.
| | - Patricia Wischmann
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Duesseldorf, Germany
| | - Maryna Masyuk
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Duesseldorf, Germany
| | - Christian Datz
- Department of Internal Medicine, Teaching Hospital of the Paracelsus Medical University Salzburg, General Hospital Oberndorf, Paracelsus Medical University Salzburg, Oberndorf, Austria.
| | - Raphael Romano Bruno
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Duesseldorf, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Duesseldorf, Germany
| | - Bernhard Wernly
- Department of Internal Medicine, Teaching Hospital of the Paracelsus Medical University Salzburg, General Hospital Oberndorf, Paracelsus Medical University Salzburg, Oberndorf, Austria; Center for Public Health and Healthcare Research, Paracelsus Medical University of Salzburg, Strubergasse 21, 5020 Salzburg, Austria
| |
Collapse
|
10
|
Zhuang J, Zhang X, Liu Q, Zhu M, Huang X. Targeted delivery of nanomedicines for promoting vascular regeneration in ischemic diseases. Am J Cancer Res 2022; 12:6223-6241. [PMID: 36168632 PMCID: PMC9475455 DOI: 10.7150/thno.73421] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/15/2022] [Indexed: 11/24/2022] Open
Abstract
Ischemic diseases, the leading cause of disability and death, are caused by the restriction or blockage of blood flow in specific tissues, including ischemic cardiac, ischemic cerebrovascular and ischemic peripheral vascular diseases. The regeneration of functional vasculature network in ischemic tissues is essential for treatment of ischemic diseases. Direct delivery of pro-angiogenesis factors, such as VEGF, has demonstrated the effectiveness in ischemic disease therapy but suffering from several obstacles, such as low delivery efficacy in disease sites and uncontrolled modulation. In this review, we summarize the molecular mechanisms of inducing vascular regeneration, providing the guidance for designing the desired nanomedicines. We also introduce the delivery of various nanomedicines to ischemic tissues by passive or active targeting manner. To achieve the efficient delivery of nanomedicines in various ischemic diseases, we highlight targeted delivery of nanomedicines and controllable modulation of disease microenvironment using nanomedicines.
Collapse
Affiliation(s)
- Jie Zhuang
- School of Medicine, Nankai University, Tianjin 300071, China.,Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, China.,Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiangyun Zhang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, China.,Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Qiqi Liu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, China.,Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Mingsheng Zhu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, China.,Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xinglu Huang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, China.,Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
11
|
de Paula LJC, Uchida AH, Rezende PC, Soares P, Scudeler TL. Protective or Inhibitory Effect of Pharmacological Therapy on Cardiac Ischemic Preconditioning: A Literature Review. Curr Vasc Pharmacol 2022; 20:409-428. [PMID: 35986546 DOI: 10.2174/1570161120666220819163025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/23/2022] [Accepted: 05/31/2022] [Indexed: 01/25/2023]
Abstract
Ischemic preconditioning (IP) is an innate phenomenon, triggered by brief, non-lethal cycles of ischemia/reperfusion applied to a tissue or organ that confers tolerance to a subsequent more prolonged ischemic event. Once started, it can reduce the severity of myocardial ischemia associated with some clinical situations, such as percutaneous coronary intervention (PCI) and intermittent aortic clamping during coronary artery bypass graft surgery (CABG). Although the mechanisms underlying IP have not been completely elucidated, several studies have shown that this phenomenon involves the participation of cell triggers, intracellular signaling pathways, and end-effectors. Understanding this mechanism enables the development of preconditioning mimetic agents. It is known that a range of medications that activate the signaling cascades at different cellular levels can interfere with both the stimulation and the blockade of IP. Investigations of signaling pathways underlying ischemic conditioning have identified a number of therapeutic targets for pharmacological manipulation. This review aims to present and discuss the effects of several medications on myocardial IP.
Collapse
Affiliation(s)
| | | | - Paulo Cury Rezende
- Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Paulo Soares
- Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Thiago Luis Scudeler
- Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
12
|
Abdu FA, Mohammed AQ, Liu L, Yin G, Xu S, Mohammed AA, Mareai RM, Xu Y, Che W. Metabolic syndrome and the risk of adverse cardiovascular events in patients with myocardial infarction with non-obstructive coronary arteries. Nutr Metab Cardiovasc Dis 2022; 32:666-674. [PMID: 35140026 DOI: 10.1016/j.numecd.2022.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/13/2021] [Accepted: 01/04/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND AND AIMS Metabolic syndrome (MeS) is recognized as a significant predictor of poor outcomes in coronary artery disease. However, its prognostic implications in myocardial infarction with non-obstructive coronary arteries (MINOCA) have not been examined. We aimed at investigating the role of MeS on the clinical outcomes in MINOCA patients. METHODS AND RESULTS Patients diagnosed with MINOCA between 2015 and 2019 were included. MeS was defined according to modified NCEP-ATPIII criteria. The primary endpoint was major adverse cardiovascular events (MACE). Cox regression analysis was used to evaluate the correlation between MeS and the hazard of MACE. The integrated discrimination improvement (IDI) and net reclassification index (NRI) were performed to assess MeS incremental predictive value. Of 281 MINOCA patients, 83 (29.5%) patients satisfied the MeS criteria. During a median follow-up duration of 28 months, MINOCA patients with MeS had a notably higher rate of MACE than those without MeS (30.1% vs. 17.6%, respectively P = 0.020). Cox regression analysis revealed that MeS was associated with an increased hazard of MACE (adjusted HR 2.126; 95% CI: 1.193-3.787, P = 0.010). When each component of MeS was analyzed as a categorized variable separately, only high fasting blood glucose and BMI ≥25 kg/m2 were associated with an increased hazard of MACE. Moreover, MeS had an incremental predictive ability for MACE when added to a model with clinical risk factors. CONCLUSION MeS is relatively common in patients with MINOCA. The presence of MeS significantly increased the hazard of MACE among the MINOCA population.
Collapse
Affiliation(s)
- Fuad A Abdu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Abdul-Quddus Mohammed
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lu Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guoqing Yin
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Siling Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ayman A Mohammed
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Redhwan M Mareai
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Wenliang Che
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Department of Cardiology, Shanghai Tenth People's Hospital Chongming Branch, Shanghai, China.
| |
Collapse
|
13
|
Yousuf MS, Samad K, Ahmed SS, Siddiqui KM, Ullah H. Cardiac Surgery and Blood-Saving Techniques: An Update. Cureus 2022; 14:e21222. [PMID: 35186524 PMCID: PMC8844256 DOI: 10.7759/cureus.21222] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2022] [Indexed: 11/12/2022] Open
Abstract
Cardiac surgery is typically attributed with a significant risk of intraoperative blood loss and allogeneic blood transfusions. Intraoperative blood loss, allogenic blood transfusions, high dose anticoagulation requirement, and interactions with cardiopulmonary bypass (CPB) have all been linked to cardiac surgeries. To reduce unnecessary transfusions and their negative effects, it is recommended to follow evidence-based multidisciplinary strategies, which are collectively termed patient blood management (PBM). This review highlights the most recent blood conservation strategies in adult cardiac surgery, which can be employed pre-operatively, intra-operatively, and postoperatively, to enhance red cell mass and attenuate the utilization of packed red blood cells (PRBCs) and other blood products.
Collapse
Affiliation(s)
| | - Khalid Samad
- Anaesthesia and Critical Care, The Aga Khan University Hospital, Karachi, PAK
- Anaesthesia and Critical Care, Aga Khan Health Service, Pakistan (AKHS,P), Karachi, PAK
| | | | | | - Hameed Ullah
- Anaesthesiology, The Aga Khan University Hospital, Karachi, PAK
| |
Collapse
|
14
|
Sun P, Kumar N, Tin A, Zhao J, Brown MR, Lin Z, Yang ML, Zheng Q, Jia J, Bielak LF, Yu B, Boerwinkle E, Hunker KL, Coresh J, Chen YE, Huo Y, Kardia SL, Khoriaty R, Zhou X, Morrison AC, Zhang Y, Ganesh SK. Epidemiologic and Genetic Associations of Erythropoietin With Blood Pressure, Hypertension, and Coronary Artery Disease. Hypertension 2021; 78:1555-1566. [PMID: 34488438 PMCID: PMC8516734 DOI: 10.1161/hypertensionaha.121.17597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/05/2021] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Pengfei Sun
- Department of cardiology, Peking University First hospital, Beijing, 100034, China
| | - Nitin Kumar
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Adrienne Tin
- Johns Hopkins Bloomberg School of Public Health, MD, 21205, USA
- University of Mississippi Medical Center, MS, 38677, USA
| | - Jing Zhao
- Department of cardiology, Peking University First hospital, Beijing, 100034, China
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Zesen Lin
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Min-Lee Yang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Qiwen Zheng
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jia Jia
- Department of cardiology, Peking University First hospital, Beijing, 100034, China
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Bing Yu
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Kristina L. Hunker
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Josef Coresh
- Johns Hopkins Bloomberg School of Public Health, MD, 21205, USA
| | - Y. Eugene Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Yong Huo
- Department of cardiology, Peking University First hospital, Beijing, 100034, China
| | - Sharon L.R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Rami Khoriaty
- Division of Hematology, Department of Internal Medicine and Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Xiang Zhou
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Alanna C. Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yan Zhang
- Department of cardiology, Peking University First hospital, Beijing, 100034, China
- Institute of Cardiovascular Disease, Peking University First Hospital, Beijing, 100034, China
| | - Santhi K. Ganesh
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW High mechanical index impulses from a diagnostic transducer are utilized in myocardial perfusion imaging, but can also be utilized therapeutically in three cardiovascular applications: (a) thrombus dissolution (sonothrombolysis), (b) improving microvascular flow in ischemic territories (sonoperfusion), and (c) targeted drug and nucleic acid delivery. The targeted therapeutic effect appears to be based on acoustic cavitation of the intravascular microbubbles which results in endothelial shear and pore formation, as well as mechanical destruction of thrombi. RECENT FINDINGS Within the last 5 years, clinical trials have been performed in acute myocardial infarction demonstrating successful reductions in myocardial infarct size with sonothrombolysis added to current guideline-based treatment. In patients with severe peripheral arterial disease, brief improvements in calf microvascular blood flow have been observed for 1 h after 10 min of sonoperfusion therapy. Targeted ultrasound therapies are developing for prevention of microvascular obstruction in acute coronary syndromes and peripheral vascular disease.
Collapse
|
16
|
Nohara S, Yamamoto M, Yasukawa H, Nagata T, Takahashi J, Shimozono K, Yanai T, Sasaki T, Okabe K, Shibata T, Akagaki D, Mawatari K, Fukumoto Y. SOCS3 deficiency in cardiomyocytes elevates sensitivity of ischemic preconditioning that synergistically ameliorates myocardial ischemia reperfusion injury. PLoS One 2021; 16:e0254712. [PMID: 34292971 PMCID: PMC8297769 DOI: 10.1371/journal.pone.0254712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 07/01/2021] [Indexed: 11/19/2022] Open
Abstract
Ischemic preconditioning (IPC) is the most powerful endogenous cardioprotective form of cellular adaptation. However, the inhibitory or augmenting mechanism underlying cardioprotection via IPC remains largely unknown. Suppressor of cytokine signaling-3 (SOCS3) is a cytokine-inducible potent negative feedback regulator of the signal transducer and activator of transcription-3 (STAT3) signaling pathway. Here, we aimed to determine whether cardiac SOCS3 deficiency and IPC would synergistically reduce infarct size after myocardial ischemia reperfusion injury. We evaluated STAT3 activation and SOCS3 induction after ischemic conditioning (IC) using western blot analysis and real-time PCR, and found that myocardial IC alone transiently activated myocardial STAT3 and correspondingly induced SOCS3 expression in wild-type mice. Compared with wild-type mice, cardiac-specific SOCS3 knockout (SOCS3-CKO) mice showed significantly greater and more sustained IC-induced STAT3 activation. Following ischemia reperfusion, IPC substantially reduced myocardial infarct size and significantly enhanced STAT3 phosphorylation in SOCS3-CKO mice compared to in wild-type mice. Real-time PCR array analysis revealed that SOCS3-CKO mice after IC exhibited significantly increased expressions of several anti-apoptotic genes and SAFE pathway-related genes. Moreover, real-time PCR analysis revealed that myocardial IC alone rapidly induced expression of the STAT3-activating cytokine erythropoietin in the kidney at 1 h post-IC. We also found that the circulating erythropoietin level was promptly increased at 1 h after myocardial IC. Myocardial SOCS3 deficiency and IPC exert synergistic effects in the prevention of myocardial injury after ischemia reperfusion. Our present results suggest that myocardial SOCS3 is a potent inhibitor of IPC-induced cardioprotection, and that myocardial SOCS3 inhibition augment IPC-mediated cardioprotection during ischemia reperfusion injury.
Collapse
Affiliation(s)
- Shoichiro Nohara
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Mai Yamamoto
- Cardiovascular Research Institute, Kurume University, Kurume, Japan
| | - Hideo Yasukawa
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
- * E-mail:
| | - Takanobu Nagata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Jinya Takahashi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Koutatsu Shimozono
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Toshiyuki Yanai
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Tomoko Sasaki
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Kota Okabe
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Tatsuhiro Shibata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Daiki Akagaki
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Kazutoshi Mawatari
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Yoshihiro Fukumoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
- Cardiovascular Research Institute, Kurume University, Kurume, Japan
| |
Collapse
|
17
|
Boyle AJ, Schultz C, Selvanayagam JB, Moir S, Kovacs R, Dib N, Zlotnick D, Al-Omary M, Sugito S, Selvarajah A, Collins N, McLachlan G. Calcium/Calmodulin-Dependent Protein Kinase II Delta Inhibition and Ventricular Remodeling After Myocardial Infarction: A Randomized Clinical Trial. JAMA Cardiol 2021; 6:762-768. [PMID: 33851966 DOI: 10.1001/jamacardio.2021.0676] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Importance After anterior ST-segment elevation myocardial infarction (STEMI), left ventricular (LV) remodeling results in heart failure and death. Calcium/calmodulin-dependent protein kinase II delta (CaMKIId) is a key molecular mediator of adverse LV remodeling. Objective To determine whether NP202, an orally active inhibitor of CaMKIId, prevents LV remodeling in patients after anterior STEMI with early residual LV dysfunction. Design, Setting, and Participants A randomized, double-blind, placebo-controlled multicenter clinical trial of NP202 vs placebo in patients after primary percutaneous coronary intervention (PCI) for anterior STEMI was performed from November 19, 2015, to August 1, 2018. The study was performed at 32 sites across the US, Australia, and New Zealand. Patients presenting with anterior STEMI who underwent PCI within 12 hours of symptom onset and left ventricular ejection fraction (LVEF) less than 45% on screening echocardiogram 48 hours after primary PCI were included in the study. Baseline cardiovascular magnetic resonance (CMR) imaging was performed within 5 days of the STEMI and before administration of the study drug. Follow-up CMR was performed after 3 months. Data were analyzed from November 19, 2015, to August 1, 2018. Interventions Patients were randomly assigned to NP202, 1000 mg, daily for 3 months vs corresponding placebo. Main Outcomes and Measures The primary end point was change in LV end-systolic volume index (LVESVi) on CMR. Secondary end points were change in LV end-diastolic volume index, change in LVEF, change in infarct size, and change in diastolic function. Safety and tolerability were also assessed. Results A total of 147 patients (mean [SD] age, 58 [11] years; 129 men [88%]; 130 White patients [88%]) who experienced anterior STEMI treated with primary PCI were randomized to receive NP202 (73 [49.7%]) or placebo (74 [50.3%]). Baseline LVEF was similar between groups. At baseline, patients randomized to NP202 had greater LVESVi (48.2 mL/m2) than that in the placebo group (41.3 mL/m2; P = .03). However, the groups were otherwise well matched. For the primary end point of change in LVESVi from baseline to 3 months, there was no significant difference between the placebo (median [interquartile range] change, -0.60 [-9.28 to 5.99] mL/m2) and NP202 groups (-3.53 [-9.24 to 4.81] mL/m2) (P = .78). There was also no difference in the secondary efficacy end points assessed by CMR. NP202 was well tolerated and demonstrated an acceptable safety profile. Major adverse cardiac and cerebrovascular event rates were similar between groups. Two deaths occurred in each group during the follow-up period. Conclusions and Relevance Three months of treatment with NP202 after primary PCI for anterior STEMI with residual LV dysfunction did not improve LV remodeling. The drug was safe and well tolerated. Trial Registration ClinicalTrials.gov Identifier: NCT02557217.
Collapse
Affiliation(s)
- Andrew J Boyle
- Department of Cardiovascular Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia.,University of Newcastle, Newcastle, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Carl Schultz
- University of Western Australia School of Medicine, Perth, Australia.,Department of Cardiology, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Joseph B Selvanayagam
- Flinders Medical Centre, Flinders University, Adelaide, South Australia, Australia.,South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Stuart Moir
- MonashHeart, Melbourne, Victoria, Australia.,Monash University, Melbourne, Victoria, Australia
| | | | - Nabil Dib
- Dignity Healthcare, Gilbert, Arizona
| | | | - Mohammed Al-Omary
- Department of Cardiovascular Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia.,University of Newcastle, Newcastle, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Stuart Sugito
- Department of Cardiovascular Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Aravinda Selvarajah
- Department of Cardiovascular Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Nicholas Collins
- Department of Cardiovascular Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia.,University of Newcastle, Newcastle, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | | |
Collapse
|
18
|
Wu Y, Yang B. Erythropoietin Receptor/β Common Receptor: A Shining Light on Acute Kidney Injury Induced by Ischemia-Reperfusion. Front Immunol 2021; 12:697796. [PMID: 34276689 PMCID: PMC8278521 DOI: 10.3389/fimmu.2021.697796] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/17/2021] [Indexed: 12/29/2022] Open
Abstract
Acute kidney injury (AKI) is a health problem worldwide, but there is a lack of early diagnostic biomarkers and target-specific treatments. Ischemia-reperfusion (IR), a major cause of AKI, not only induces kidney injury, but also stimulates the self-defense system including innate immune responses to limit injury. One of these responses is the production of erythropoietin (EPO) by adjacent normal tissue, which is simultaneously triggered, but behind the action of its receptors, either by the homodimer EPO receptor (EPOR)2 mainly involved in erythropoiesis or the heterodimer EPOR/β common receptor (EPOR/βcR) which has a broad range of biological protections. EPOR/βcR is expressed in several cell types including tubular epithelial cells at low levels or absent in normal kidneys, but is swiftly upregulated by hypoxia and inflammation and also translocated to cellular membrane post IR. EPOR/βcR mediates anti-apoptosis, anti-inflammation, pro-regeneration, and remodeling via the PI3K/Akt, STAT3, and MAPK signaling pathways in AKI. However, the precise roles of EPOR/βcR in the pathogenesis and progression of AKI have not been well defined, and its potential as an earlier biomarker for AKI diagnosis and monitoring repair or chronic progression requires further investigation. Here, we review biological functions and mechanistic signaling pathways of EPOR/βcR in AKI, and discuss its potential clinical applications as a biomarker for effective diagnosis and predicting prognosis, as well as directing cell target drug delivery.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Basic Medical Research Centre, Medical School, Nantong University, Nantong, China.,Nantong-Leicester Joint Institute of Kidney Science, Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Bin Yang
- Nantong-Leicester Joint Institute of Kidney Science, Nephrology, Affiliated Hospital of Nantong University, Nantong, China.,Department of Cardiovascular Sciences, College of Life Sciences, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
19
|
Therapies to prevent post-infarction remodelling: From repair to regeneration. Biomaterials 2021; 275:120906. [PMID: 34139506 DOI: 10.1016/j.biomaterials.2021.120906] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 05/02/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022]
Abstract
Myocardial infarction is the first cause of worldwide mortality, with an increasing incidence also reported in developing countries. Over the past decades, preclinical research and clinical trials continually tested the efficacy of cellular and acellular-based treatments. However, none of them resulted in a drug or device currently used in combination with either percutaneous coronary intervention or coronary artery bypass graft. Inflammatory, proliferation and remodelling phases follow the ischaemic event in the myocardial tissue. Only recently, single-cell sequencing analyses provided insights into the specific cell populations which determine the final fibrotic deposition in the affected region. In this review, ischaemia, inflammation, fibrosis, angiogenesis, cellular stress and fundamental cellular and molecular components are evaluated as therapeutic targets. Given the emerging evidence of biomaterial-based systems, the increasing use of injectable hydrogels/scaffolds and epicardial patches is reported both as acellular and cellularised/functionalised treatments. Since several variables influence the outcome of any experimented treatment, we return to the pathological basis with an unbiased view towards any specific process or cellular component. Thus, by evaluating the benefits and limitations of the approaches based on these targets, the reader can weigh the rationale of each of the strategies that reached the clinical trials stage. As recent studies focused on the relevance of the extracellular matrix in modulating ischaemic remodelling and enhancing myocardial regeneration, we aim to portray current trends in the field with this review. Finally, approaches towards feasible translational studies that are as yet unexplored are also suggested.
Collapse
|
20
|
Helix B Surface Peptide Protects Cardiomyocytes From Hypoxia/Reoxygenation-induced Autophagy Through the PI3K/Akt Pathway. J Cardiovasc Pharmacol 2021; 76:181-188. [PMID: 32404595 DOI: 10.1097/fjc.0000000000000849] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Helix B surface peptide (HBSP) is a newly discovered tissue-protective erythropoietin derivative that provides benefits after myocardial ischemia/reperfusion. This study explores the cardioprotective effects of HBSP in myocardial cells in response to hypoxia/reoxygenation injury and its potential mechanism. METHODS In this study, rat ventricular (H9c2) cell cultures were established and pretreated with HBSP. H9c2 cardiomyocytes were randomly assigned to the control, H/R, H/R + LY294002 (a PI3K inhibitor), HBSP + H/R, and HBSP + H/R + LY294002 groups. The pretreated cardiomyocytes underwent H/R, and the cardiomyocytes were monitored for viability through a CCK-8 assay, whereas flow cytometry was used to test cell apoptosis. Orgotein Superoxide Dismutase (SOD) and lactate dehydrogenase (LDH) expression were monitored by SOD and LDH kits, respectively. The expression of LC3 autophagosomes was determined by immunocytochemistry. The expression of LC3II/LC3I, p-Mammalian Target of Rapamycin (mTOR) mTOR, mTOR, Beclin 1, p-PI3K, PI3K p-Akt, and Akt was determined by Western blotting. RESULTS HBSP increased cell viability and reduced SOD and LDH production, and it also reduced H/R-induced cell apoptosis. Moreover, the expression of the autophagy-related proteins (LC3II/LC3I) was inhibited by HBSP, whereas the expression of p-PI3K, p-Akt, and p-mTOR was enhanced. However, the PI3K inhibitor (LY294002) notably abolished these effects in H9c2 cells. CONCLUSIONS HBSP inhibits excessive autophagy and apoptosis induced by H/R by activating the PI3K/Akt pathway. HBSP may potentially be a therapeutic intervention for myocardial ischemia/reperfusion injury.
Collapse
|
21
|
Eswarappa M, Cantarelli C, Cravedi P. Erythropoietin in Lupus: Unanticipated Immune Modulating Effects of a Kidney Hormone. Front Immunol 2021; 12:639370. [PMID: 33796104 PMCID: PMC8007959 DOI: 10.3389/fimmu.2021.639370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/24/2021] [Indexed: 11/24/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multiorgan autoimmune disease with variable clinical presentation, typically characterized by a relapsing-remitting course. SLE has a multifactorial pathogenesis including genetic, environmental, and hormonal factors that lead to loss of tolerance against self-antigens and autoantibody production. Mortality in SLE patients remains significantly higher than in the general population, in part because of the limited efficacy of available treatments and the associated toxicities. Therefore, novel targeted therapies are urgently needed to improve the outcomes of affected individuals. Erythropoietin (EPO), a kidney-produced hormone that promotes red blood cell production in response to hypoxia, has lately been shown to also possess non-erythropoietic properties, including immunomodulatory effects. In various models of autoimmune diseases, EPO limits cell apoptosis and favors cell clearance, while reducing proinflammatory cytokines and promoting the induction of regulatory T cells. Notably, EPO has been shown to reduce autoimmune response and decrease disease severity in mouse models of SLE. Herein, we review EPO's non-erythropoietic effects, with a special focus on immune modulating effects in SLE and its potential clinical utility.
Collapse
Affiliation(s)
- Meghana Eswarappa
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Chiara Cantarelli
- UO Nefrologia, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Paolo Cravedi
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
22
|
Gunata M, Parlakpinar H. A review of myocardial ischaemia/reperfusion injury: Pathophysiology, experimental models, biomarkers, genetics and pharmacological treatment. Cell Biochem Funct 2020; 39:190-217. [PMID: 32892450 DOI: 10.1002/cbf.3587] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/03/2020] [Accepted: 08/14/2020] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases are known to be the most fatal diseases worldwide. Ischaemia/reperfusion (I/R) injury is at the centre of the pathology of the most common cardiovascular diseases. According to the World Health Organization estimates, ischaemic heart disease is the leading global cause of death, causing more than 9 million deaths in 2016. After cardiovascular events, thrombolysis, percutaneous transluminal coronary angioplasty or coronary bypass surgery are applied as treatment. However, after restoring coronary blood flow, myocardial I/R injury may occur. It is known that this damage occurs due to many pathophysiological mechanisms, especially increasing reactive oxygen types. Besides causing cardiomyocyte death through multiple mechanisms, it may be an important reason for affecting other cell types such as platelets, fibroblasts, endothelial and smooth muscle cells and immune cells. Also, polymorphonuclear leukocytes are associated with myocardial I/R damage during reperfusion. This damage may be insufficient in patients with co-morbidity, as it is demonstrated that it can be prevented by various endogenous antioxidant systems. In this context, the resulting data suggest that optimal cardioprotection may require a combination of additional or synergistic multi-target treatments. In this review, we discussed the pathophysiology, experimental models, biomarkers, treatment and its relationship with genetics in myocardial I/R injury. SIGNIFICANCE OF THE STUDY: This review summarized current information on myocardial ischaemia/reperfusion injury (pathophysiology, experimental models, biomarkers, genetics and pharmacological therapy) for researchers and reveals guiding data for researchers, especially in the field of cardiovascular system and pharmacology.
Collapse
Affiliation(s)
- Mehmet Gunata
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Hakan Parlakpinar
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| |
Collapse
|
23
|
Li J, Sun D, Li Y. Novel Findings and Therapeutic Targets on Cardioprotection of Ischemia/ Reperfusion Injury in STEMI. Curr Pharm Des 2020; 25:3726-3739. [PMID: 31692431 DOI: 10.2174/1381612825666191105103417] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/30/2019] [Indexed: 12/19/2022]
Abstract
Acute ST-segment elevation myocardial infarction (STEMI) remains a leading cause of morbidity and mortality around the world. A large number of STEMI patients after the infarction gradually develop heart failure due to the infarcted myocardium. Timely reperfusion is essential to salvage ischemic myocardium from the infarction, but the restoration of coronary blood flow in the infarct-related artery itself induces myocardial injury and cardiomyocyte death, known as ischemia/reperfusion injury (IRI). The factors contributing to IRI in STEMI are complex, and microvascular obstruction, inflammation, release of reactive oxygen species, myocardial stunning, and activation of myocardial cell death are involved. Therefore, additional cardioprotection is required to prevent the heart from IRI. Although many mechanical conditioning procedures and pharmacological agents have been identified as effective cardioprotective approaches in animal studies, their translation into the clinical practice has been relatively disappointing due to a variety of reasons. With new emerging data on cardioprotection in STEMI over the past few years, it is mandatory to reevaluate the effectiveness of "old" cardioprotective interventions and highlight the novel therapeutic targets and new treatment strategies of cardioprotection.
Collapse
Affiliation(s)
- Jianqiang Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Danghui Sun
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Yue Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| |
Collapse
|
24
|
Legallois D, Hodzic A, Alexandre J, Dolladille C, Saloux E, Manrique A, Roule V, Labombarda F, Milliez P, Beygui F. Definition of left ventricular remodelling following ST-elevation myocardial infarction: a systematic review of cardiac magnetic resonance studies in the past decade. Heart Fail Rev 2020; 27:37-48. [DOI: 10.1007/s10741-020-09975-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
25
|
Murali S, Vogrin S, Noaman S, Dinh DT, Brennan AL, Lefkovits J, Reid CM, Cox N, Chan W. Bleeding Severity in Percutaneous Coronary Intervention (PCI) and Its Impact on Short-Term Clinical Outcomes. J Clin Med 2020; 9:jcm9051426. [PMID: 32403442 PMCID: PMC7291133 DOI: 10.3390/jcm9051426] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/04/2020] [Accepted: 05/06/2020] [Indexed: 11/27/2022] Open
Abstract
Bleeding severity in patients undergoing percutaneous coronary intervention (PCI), defined by the Bleeding Academic Research Consortium (BARC), portends adverse prognosis. We analysed data from 37,866 Australian patients undergoing PCI enrolled in the Victorian Cardiac Outcomes Registry (VCOR), and investigated the association between increasing BARC severity and in-hospital and 30-day major adverse cardiac and cerebrovascular events (MACCE) (a composite of mortality, myocardial infarction, stent thrombosis, target vessel revascularisation, or stroke). Independent predictors associated with major bleeding (BARC groups 3&5), and MACCE were also assessed. There was a stepwise increase in in-hospital and 30-day MACCE with greater severity of bleeding. Independent predictors of bleeding included female sex (Odds Ratio (OR) 1.34), age (OR 1.02), fibrinolytic therapy (OR 1.77), femoral access (OR 1.51), and ticagrelor (OR 1.42), all significant at the p < 0.001 level. Following adjustment of clinically important variables, BARC 3&5 bleeds (OR 4.37) were still predictive of cumulative in-hospital and 30-day MACCE. In conclusion, major bleeding is an uncommon but potentially fatal PCI complication and was independently associated with greater MACCE rates. Efforts to mitigate the occurrence of bleeding, including radial access and judicious use of potent antiplatelet therapies, may ameliorate the risk of short-term adverse clinical outcomes.
Collapse
Affiliation(s)
- Shashank Murali
- Department of Medicine, University of Melbourne, Melbourne 3010, Victoria, Australia; (S.M.); (S.N.)
| | - Sara Vogrin
- Department of Medicine-Western Health, Melbourne Medical School, University of Melbourne, Melbourne 3010, Victoria, Australia;
| | - Samer Noaman
- Department of Medicine, University of Melbourne, Melbourne 3010, Victoria, Australia; (S.M.); (S.N.)
- Department of Cardiology, Western Health, St Albans 3021, Victoria, Australia;
- Department of Cardiology, Alfred Health, Melbourne 3004, Victoria, Australia
| | - Diem T. Dinh
- School of Public Health & Preventive Medicine, Monash University, Melbourne 3004, Victoria, Australia; (D.T.D.); (A.L.B.); (J.L.); (C.M.R.)
| | - Angela L. Brennan
- School of Public Health & Preventive Medicine, Monash University, Melbourne 3004, Victoria, Australia; (D.T.D.); (A.L.B.); (J.L.); (C.M.R.)
| | - Jeffrey Lefkovits
- School of Public Health & Preventive Medicine, Monash University, Melbourne 3004, Victoria, Australia; (D.T.D.); (A.L.B.); (J.L.); (C.M.R.)
| | - Christopher M. Reid
- School of Public Health & Preventive Medicine, Monash University, Melbourne 3004, Victoria, Australia; (D.T.D.); (A.L.B.); (J.L.); (C.M.R.)
- School of Public Health, Curtin University, Perth 6102, Western Australia, Australia
| | - Nicholas Cox
- Department of Cardiology, Western Health, St Albans 3021, Victoria, Australia;
| | - William Chan
- Department of Medicine, University of Melbourne, Melbourne 3010, Victoria, Australia; (S.M.); (S.N.)
- Department of Cardiology, Western Health, St Albans 3021, Victoria, Australia;
- Department of Cardiology, Alfred Health, Melbourne 3004, Victoria, Australia
- Correspondence: ; Tel.: +61-(03)-8345-1333
| |
Collapse
|
26
|
Takahashi J, Yamamoto M, Yasukawa H, Nohara S, Nagata T, Shimozono K, Yanai T, Sasaki T, Okabe K, Shibata T, Mawatari K, Kakuma T, Aoki H, Fukumoto Y. Interleukin-22 Directly Activates Myocardial STAT3 (Signal Transducer and Activator of Transcription-3) Signaling Pathway and Prevents Myocardial Ischemia Reperfusion Injury. J Am Heart Assoc 2020; 9:e014814. [PMID: 32301368 PMCID: PMC7428538 DOI: 10.1161/jaha.119.014814] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Interleukin (IL)-22, a member of the IL-10 cytokine family, is the only known cytokine that is secreted by immune cells but does not target immune cells; it mainly targets epithelial cells. In this study, we aimed to determine whether IL-22 administration could activate the myocardial STAT3 (signal transducer and activator of transcription-3) signaling pathway, and thus prevent myocardial injury, in a mouse model of ischemia reperfusion injury. METHODS AND RESULTS We evaluated the STAT3 activation after IL-22 injection by Western blot analysis and immunostaining for phosphorylated STAT3 in the heart and found that STAT3 activation in heart tissue rapidly peaked after IL-22 injection. Coimmunostaining of phosphorylated STAT3 and α-actinin revealed that STAT3 activation occurred in cardiomyocytes after IL-22 administration. In heart tissue from intact mice, real-time PCR demonstrated significant expression of IL-22 receptor subunit 1, and coimmunostaining of IL-22 receptor subunit 1 and α-actinin showed IL-22 receptor subunit 1 expression in cardiomyocytes. In cultured cardiomyocytes, IL-22 activated STAT3, and we detected IL-22 receptor subunit 1 expression. Overall, these results indicated that IL-22 directly activated the myocardial IL-22-receptor subunit 1-STAT3 signaling pathway. Following ischemia reperfusion, compared with PBS-treated mice, IL-22-treated mice exhibited a significantly reduced infarct size, significantly reduced myocardial apoptosis, and significantly enhanced phosphorylated STAT3 expression. Moreover, heart tissue from IL-22-treated mice exhibited a significantly reduced expression ratio of phosphorylated p53 to p53. CONCLUSIONS Our present findings suggest that IL-22 directly activated the myocardial STAT3 signaling pathway and acted as a cardioprotective cytokine to ameliorate acute myocardial infarction after ischemia reperfusion.
Collapse
Affiliation(s)
- Jinya Takahashi
- Division of Cardiovascular MedicineDepartment of Internal MedicineKurume University School of MedicineKurumeJapan
| | - Mai Yamamoto
- Cardiovascular Research InstituteKurume UniversityKurumeJapan
| | - Hideo Yasukawa
- Division of Cardiovascular MedicineDepartment of Internal MedicineKurume University School of MedicineKurumeJapan
| | - Shoichiro Nohara
- Division of Cardiovascular MedicineDepartment of Internal MedicineKurume University School of MedicineKurumeJapan
| | - Takanobu Nagata
- Division of Cardiovascular MedicineDepartment of Internal MedicineKurume University School of MedicineKurumeJapan
| | - Koutatsu Shimozono
- Division of Cardiovascular MedicineDepartment of Internal MedicineKurume University School of MedicineKurumeJapan
| | - Toshiyuki Yanai
- Division of Cardiovascular MedicineDepartment of Internal MedicineKurume University School of MedicineKurumeJapan
| | - Tomoko Sasaki
- Division of Cardiovascular MedicineDepartment of Internal MedicineKurume University School of MedicineKurumeJapan
| | - Kota Okabe
- Division of Cardiovascular MedicineDepartment of Internal MedicineKurume University School of MedicineKurumeJapan
| | - Tatsuhiro Shibata
- Division of Cardiovascular MedicineDepartment of Internal MedicineKurume University School of MedicineKurumeJapan
| | - Kazutoshi Mawatari
- Division of Cardiovascular MedicineDepartment of Internal MedicineKurume University School of MedicineKurumeJapan
| | | | - Hiroki Aoki
- Cardiovascular Research InstituteKurume UniversityKurumeJapan
| | - Yoshihiro Fukumoto
- Division of Cardiovascular MedicineDepartment of Internal MedicineKurume University School of MedicineKurumeJapan
- Cardiovascular Research InstituteKurume UniversityKurumeJapan
| |
Collapse
|
27
|
Bruno RR, Kelm M, Jung C. Spotlight on comorbidities in STEMI patients. Endocrinol Diabetes Metab 2020; 3:e00102. [PMID: 31922029 PMCID: PMC6947710 DOI: 10.1002/edm2.102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 10/27/2019] [Indexed: 01/28/2023] Open
Affiliation(s)
- Raphael Romano Bruno
- Division of Cardiology, Pulmonology, and Vascular Medicine Medical Faculty University Hospital Düsseldorf Dusseldorf Germany
| | - Malte Kelm
- Division of Cardiology, Pulmonology, and Vascular Medicine Medical Faculty University Hospital Düsseldorf Dusseldorf Germany
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine Medical Faculty University Hospital Düsseldorf Dusseldorf Germany
| |
Collapse
|
28
|
Yang C, Zhang C, Jia J, Wang L, Zhang W, Li J, Xu M, Rong R, Zhu T. Cyclic helix B peptide ameliorates acute myocardial infarction in mice by inhibiting apoptosis and inflammatory responses. Cell Death Discov 2019; 5:78. [PMID: 30911412 PMCID: PMC6423043 DOI: 10.1038/s41420-019-0161-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 02/22/2019] [Accepted: 03/04/2019] [Indexed: 12/19/2022] Open
Abstract
Cyclic helix B peptide (CHBP) is a peptide derivant of erythropoietin with powerful tissue-protective efficacies in a variety of organ injuries, but without erythropoietic effect. However, the role of CHBP in acute myocardial infarction (AMI) and related mechanisms are not studied yet. In this study, we found in a murine AMI model that the administration of CHBP could ameliorate cardiac injury, increase the survival rate, inhibit cardiomyocyte apoptosis, improve cardiac function and remodeling, and reduce the expression of inflammatory cytokines in the serum and kidney tissue both at 24 h and 8 weeks following AMI. This study suggests that CHBP has the potential to be used as an effective drug in the treatment of AMI.
Collapse
Affiliation(s)
- Cheng Yang
- 1Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032 China.,2Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032 China.,3Zhangjiang Institute of Fudan University, Shanghai, 201203 China
| | - Chao Zhang
- 1Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032 China.,2Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032 China
| | - Jianguo Jia
- 4Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032 China.,5Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032 China
| | - Lingyan Wang
- 6Biomedical Research Center, Zhongshan Hospital, Fudan University, Shanghai, 200032 China
| | - Weitao Zhang
- 1Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032 China.,2Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032 China
| | - Jiawei Li
- 1Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032 China.,2Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032 China
| | - Ming Xu
- 1Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032 China.,2Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032 China
| | - Ruiming Rong
- 1Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032 China.,2Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032 China.,7Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, 200032 China
| | - Tongyu Zhu
- 1Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032 China.,2Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032 China
| |
Collapse
|
29
|
Seo WW, Suh JW, Oh IY, Yoon CH, Cho YS, Youn TJ, Chae IH, Choi DJ. Efficacy of IntraCoronary Erythropoietin Delivery BEfore Reperfusion-Gauging Infarct Size in Patients with Acute ST-segment Elevation Myocardial Infarction (ICEBERG). Int Heart J 2019; 60:255-263. [PMID: 30799375 DOI: 10.1536/ihj.18-035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Previous clinical studies have shown inconsistent results regarding the effect of erythropoietin in ST-segment elevation myocardial infarction (STEMI). This study investigated whether directed intracoronary infusion of darbepoetin-α into ischemic myocardium before reperfusion would reduce infarct size or post-infarct remodeling in STEMI patients.Eighty STEMI patients received one of the following treatments simultaneously with the first balloon inflation: intracoronary darbepoetin-α 300 μg (n = 40) or saline (n = 40), administered via the over-the-wire balloon system. The primary endpoint was infarct size estimated by serial cardiac enzyme levels after procedure. The secondary endpoints were (1) infarct size and proportion of salvaged myocardium measured with cardiac magnetic resonance (CMR) at baseline; (2) post-infarct remodeling (PIR), defined as an increase in left ventricular end-diastolic volume more than 20% at 4 months compared to the baseline on CMR; and (3) composite cardiovascular endpoints assessed at 4 months.The peak CK-MB [median 270.0 (interquartile range 139.8-356.3) versus 231.5 (131.0-408.5) ng/mL, P = 0.55] and troponin-I [128.5 (63.5-227.8) versus 109.0 (43.8-220.0) ng/mL, P = 0.52) ] did not differ between the darbepoetin-α and control group. Fifty-seven patients completed the baseline and 4-month follow-up CMR. There were no differences in infarct size [30.6 (18.1-49.8) versus 31.5 (22.5-47.3) cm3, P = 0.91), proportion of salvaged myocardium [26.7% (15.9-42.6%) versus 35.8% (22.4-48.8%), P = 0.12) or PIR (8.0% versus 6.7%, P = 0.62) between the two groups. Composite cardiovascular outcomes did not differ between the two groups.In conclusion, administration of intracoronary darbepoetin-α before reperfusion did not reduce infarct size or post-infarct remodeling in STEMI patients.
Collapse
Affiliation(s)
- Won-Woo Seo
- Division of Cardiology, Department of Internal Medicine, Kangdong Sacred Heart Hospital
| | - Jung-Won Suh
- Department of Internal Medicine, Seoul National University College of Medicine, Cardiovascular Center, Seoul National University Bundang Hospital
| | - Il-Young Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Cardiovascular Center, Seoul National University Bundang Hospital
| | - Chang-Hwan Yoon
- Department of Internal Medicine, Seoul National University College of Medicine, Cardiovascular Center, Seoul National University Bundang Hospital
| | - Young-Seok Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Cardiovascular Center, Seoul National University Bundang Hospital
| | - Tae-Jin Youn
- Department of Internal Medicine, Seoul National University College of Medicine, Cardiovascular Center, Seoul National University Bundang Hospital
| | - In-Ho Chae
- Department of Internal Medicine, Seoul National University College of Medicine, Cardiovascular Center, Seoul National University Bundang Hospital
| | - Dong-Ju Choi
- Department of Internal Medicine, Seoul National University College of Medicine, Cardiovascular Center, Seoul National University Bundang Hospital
| |
Collapse
|
30
|
Guedeney P, Sorrentino S, Claessen B, Mehran R. The link between anemia and adverse outcomes in patients with acute coronary syndrome. Expert Rev Cardiovasc Ther 2019; 17:151-159. [DOI: 10.1080/14779072.2019.1575729] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Paul Guedeney
- The Zena and Michael A. Weiner Cardiovascular Institute, The Icahn School of Medicine at Mount Sinai, New York, USA
- Institut de Cardiologie, Sorbonne Université, ACTION Study group, INSERM UMRS 1166, Hôpital Pitié-Salpêtrière (AP-HP), Paris, France
| | - Sabato Sorrentino
- The Zena and Michael A. Weiner Cardiovascular Institute, The Icahn School of Medicine at Mount Sinai, New York, USA
- Division of cardiology, Department of Medical and Surgical Science, Magna Graecia University, Catanzaro, Italy
| | - Bimmer Claessen
- The Zena and Michael A. Weiner Cardiovascular Institute, The Icahn School of Medicine at Mount Sinai, New York, USA
| | - Roxana Mehran
- The Zena and Michael A. Weiner Cardiovascular Institute, The Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
31
|
Effect of Erythropoietin Administration on Myocardial Viability and Coronary Microvascular Dysfunction in Anterior Acute Myocardial Infarction: Randomized Controlled Trial in the Japanese Population. Cardiol Ther 2018; 7:151-162. [PMID: 30353280 PMCID: PMC6251819 DOI: 10.1007/s40119-018-0122-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Indexed: 01/29/2023] Open
Abstract
Introduction Cardioprotective effects of erythropoietin (EPO) on infarcted myocardium in acute myocardial infarction (AMI) patients have been inconclusive. This study aimed to assess the effect of EPO administration on coronary microvascular dysfunction (CMD) and myocardial viability in anterior AMI. We also evaluated the serial changes in CMD and cardiac remodeling in these patients. Methods Patients with a successful percutaneous coronary intervention (PCI) for the first anterior AMI were randomly assigned to two groups (EPO and control groups), and given single-dose intravenous administration of recombinant human EPO (12,000 IU) or saline after PCI. Delayed-enhanced cardiac magnetic resonance imaging was performed at 1 week after AMI to assess the average of transmural extent of infarction and infarct size. Coronary flow velocity reserve (CFVR) of the left anterior descending coronary artery was measured by Doppler echocardiography at 1 week, 1 month, and 8 months after AMI. All patients underwent clinical follow-up for the assessment of cardiac remodeling. Results Sixty-one patients (EPO 32, control 29) were eligible for analysis. EPO group (2.4 ± 1.2) had a tendency of smaller transmural extent of infarction than that of control group (2.9 ± 1.1; p = 0.063). CFVR-8 months improved significantly in EPO group (2.9 ± 0.6) compared to control group (2.6 ± 0.5; p = 0.04). Left atrial (LA) volume − 8 months was significantly lower in EPO group (47 ± 11) than those of control group (65 ± 20; p = 0.004). Conclusions A single medium dose of EPO could have a favorable effect on CMD and LA remodeling in the chronic phase of anterior AMI. Trial Registration The institutional ethics committee of Wakayama Medical University, identifier, 1125. Electronic supplementary material The online version of this article (10.1007/s40119-018-0122-1) contains supplementary material, which is available to authorized users.
Collapse
|
32
|
Kudenchuk PJ. Erythropoietin for Out-of-Hospital Cardiac Arrest: Growing Together or Apart? J Am Coll Cardiol 2018; 68:50-2. [PMID: 27364050 DOI: 10.1016/j.jacc.2016.03.598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 03/02/2016] [Indexed: 11/18/2022]
Affiliation(s)
- Peter J Kudenchuk
- Department of Medicine, Division of Cardiology/Arrhythmia Services and King County Emergency Medical Services, Seattle, Washington.
| |
Collapse
|
33
|
Parviz Y, Waleed M, Vijayan S, Adlam D, Lavi S, Al Nooryani A, Iqbal J, Stone GW. Cellular and molecular approaches to enhance myocardial recovery after myocardial infarction. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2018; 20:351-364. [PMID: 29958820 DOI: 10.1016/j.carrev.2018.05.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/22/2018] [Accepted: 05/29/2018] [Indexed: 10/14/2022]
Abstract
Reperfusion therapy has resulted in significant improvement in post-myocardial infarction morbidity and mortality in over the last 4 decades. Nonetheless, it is well recognized that simply restoring patency of the epicardial artery may not stop or reverse damage at microvascular level, and myocardial salvage is often suboptimal. Numerous efforts have been undertaken to elucidate the mechanisms underlying extensive myonecrosis to facilitate the discovery of therapies to provide additional and incremental benefits over current therapeutic pathways. To date, conclusively effective strategies to promote myocardial recovery have not yet been established. Novel approaches are investigating the foundational cellular and molecular bases of myocardial ischemia and irreversible injury. Herein, we review the emerging concepts and proposed therapies that may improve myocardial protection and reduce infarct size. We examine the preclinical and clinical evidence for reduced infarct size with these strategies, including anti-inflammatory agents, intracellular ion channel modulators, agents affecting the reperfusion injury salvage kinase (RISK) and nitric oxide signaling pathways, modulators of mitochondrial function, anti-apoptotic agents, and stem cell and gene therapy. We review the potential reasons of failures to date and the potential for new strategies to further promote myocardial recovery and improve prognosis.
Collapse
Affiliation(s)
- Yasir Parviz
- New York Presbyterian Hospital, Columbia University Medical Centre and the Cardiovascular Research Foundation, New York, NY, USA.
| | | | | | - David Adlam
- Department of Cardiovascular Sciences, University of Leicester, Cardiovascular Research Centre, UK
| | - Shahar Lavi
- Division of Cardiology, London Health Sciences Centre, Western University, London, Ontario, Canada
| | | | - Javaid Iqbal
- South Yorkshire Cardiothoracic Centre, Northern General Hospital, Sheffield, UK
| | - Gregg W Stone
- New York Presbyterian Hospital, Columbia University Medical Centre and the Cardiovascular Research Foundation, New York, NY, USA
| |
Collapse
|
34
|
Minamino T, Higo S, Araki R, Hikoso S, Nakatani D, Suzuki H, Yamada T, Okutsu M, Yamamoto K, Fujio Y, Ishida Y, Ozawa T, Kato K, Toba K, Aizawa Y, Komuro I. Low-Dose Erythropoietin in Patients With ST-Segment Elevation Myocardial Infarction (EPO-AMI-II) - A Randomized Controlled Clinical Trial. Circ J 2018; 82:1083-1091. [PMID: 29398672 DOI: 10.1253/circj.cj-17-0889] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
BACKGROUND Erythropoietin (EPO) has antiapoptotic and tissue-protective effects, but previous clinical studies using high-dose EPO have not shown cardioprotective effects, probably because of platelet activation and a lack of knowledge regarding the optimal dose. In contrast, a small pilot study using low-dose EPO has shown improvement in left ventricular function without adverse cardiovascular events. METHODS AND RESULTS We performed a multicenter (25 hospitals), prospective, randomized, double-blind, placebo-controlled, dose-finding study to clarify the efficacy and safety of low-dose EPO in patients with ST-segment elevation myocardial infarction (STEMI) under the Evaluation System of Investigational Medical Care of the Ministry of Health, Labor and Welfare of Japan. In total, 198 STEMI patients with low left ventricular ejection fraction (LVEF <50%) were randomly assigned to receive intravenous administration of EPO (6,000 or 12,000 IU) or placebo within 6 h of successful percutaneous coronary intervention. At 6 months, there was no significant dose-response relationship in LVEF improvement among the 3 groups tested (EPO 12,000 IU: 5.4±9.3%, EPO 6,000 IU: 7.3±7.7%, Placebo: 8.1±8.3%, P=0.862). Low-dose EPO also did not improve cardiac function, as evaluated by 99 mTc-MIBI SPECT or NT-proBNP at 6 months and did not increase adverse events. CONCLUSIONS Administration of low-dose EPO did not improve LVEF at 6 months in STEMI patients (UMIN000005721).
Collapse
Affiliation(s)
- Tetsuo Minamino
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University
| | - Shuichiro Higo
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Ryo Araki
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Shungo Hikoso
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Daisaku Nakatani
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Hiroshi Suzuki
- Department of Cardiology, Showa University Fujigaoka Hospital
| | | | - Masaaki Okutsu
- Department of Internal Medicine, Kawasaki Medical School General Medical Center
| | - Kouji Yamamoto
- Department of Medical Statistics, Osaka City University Graduate School of Medicine
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Yoshio Ishida
- Department of Internal Medicine, Kaizuka City Hospital
| | - Takuya Ozawa
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences
| | - Kiminori Kato
- Department of Laboratory Medicine and Clinical Epidemiology for Prevention of Noncommunicable Diseases, Niigata University Graduate School of Medical and Dental Sciences
| | - Ken Toba
- Department of Hematology, Tachikawa Medical Center
| | - Yoshifusa Aizawa
- Department of Research and Development, Tachikawa Medical Center
| | - Issei Komuro
- Department of Cardiovascular Medicine, Tokyo University Graduate School of Medicine
| |
Collapse
|
35
|
Heusch G, Gersh BJ. The pathophysiology of acute myocardial infarction and strategies of protection beyond reperfusion: a continual challenge. Eur Heart J 2018; 38:774-784. [PMID: 27354052 DOI: 10.1093/eurheartj/ehw224] [Citation(s) in RCA: 302] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/12/2016] [Indexed: 12/15/2022] Open
Abstract
The incidence of ST segment elevation myocardial infarction (STEMI) has decreased over the last two decades in developed countries, but mortality from STEMI despite widespread access to reperfusion therapy is still substantial as is the development of heart failure, particularly among an expanding older population. In developing countries, the incidence of STEMI is increasing and interventional reperfusion is often not available. We here review the pathophysiology of acute myocardial infarction and reperfusion, notably the temporal and spatial evolution of ischaemic and reperfusion injury, the different modes of cell death, and the resulting coronary microvascular dysfunction. We then go on to briefly characterize the cardioprotective phenomena of ischaemic preconditioning, ischaemic postconditioning, and remote ischaemic conditioning and their underlying signal transduction pathways. We discuss in detail the attempts to translate conditioning strategies and drug therapy into the clinical setting. Most attempts have failed so far to reduce infarct size and improve clinical outcomes in STEMI patients, and we discuss potential reasons for such failure. Currently, it appears that remote ischaemic conditioning and a few drugs (atrial natriuretic peptide, exenatide, metoprolol, and esmolol) reduce infarct size, but studies with clinical outcome as primary endpoint are still underway.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Hufelandstr. 55, 45122 Essen, Germany
| | - Bernard J Gersh
- Division of Cardiovascular Diseases, Mayo Clinic and Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
36
|
Zhou H, Huang J, Zhu L, Cao Y. Erythropoietin alleviates post-resuscitation myocardial dysfunction in rats potentially through increasing the expression of angiotensin II receptor type 2 in myocardial tissues. Mol Med Rep 2018; 17:5184-5192. [PMID: 29393490 PMCID: PMC5865983 DOI: 10.3892/mmr.2018.8473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 12/01/2017] [Indexed: 02/06/2023] Open
Abstract
Activation of renin-angiotensin system (RAS) is one of the pathological mechanisms associated with myocardial ischemia-reperfusion injury following resuscitation. The present study aimed to determine whether erythropoietin (EPO) improves post‑resuscitation myocardial dysfunction and how it affects the renin‑angiotensin system. Sprague‑Dawley rats were randomly divided into sham, vehicle, epinephrine (EP), EPO and EP + EPO groups. Excluding the sham group, all groups underwent cardiopulmonary resuscitation (CPR) 4 min after asphyxia‑induced cardiac arrest (CA). EP and/or EPO was administrated by intravenous injection when CPR began. The results demonstrated that the vehicle group exhibited lower mean arterial pressure, left ventricular systolic pressure, maximal ascending rate of left ventricular pressure during left ventricular isovolumic contraction and maximal descending rate of left ventricular pressure during left ventricular isovolumic relaxation (+LVdP/dt max and ‑LVdP/dt max, respectively), and higher left ventricular end‑diastolic pressure, compared with the sham group following return of spontaneous circulation (ROSC). Few significant differences were observed concerning the myocardial function between the vehicle and EP groups; however, compared with the vehicle group, EPO reversed myocardial function indices following ROSC, excluding‑LVdP/dt max. Serum renin and angiotensin (Ang) II levels were measured by ELISA. The serum levels of renin and Ang II were significantly increased in the vehicle group compared with the sham group, which was also observed for the myocardial expression of renin and Ang II receptor type 1 (AT1R), as determined by reverse transcription‑quantitative polymerase chain reaction and western blotting. EPO alone did not significantly reduce the high serum levels of renin and Ang II post-resuscitation, but changed the protein levels of renin and AT1R expression in myocardial tissues. However, EPO enhanced the myocardial expression of Ang II receptor type 2 (AT2R) following ROSC. In conclusion, the present study confirmed that CA resuscitation activated the renin‑Ang II‑AT1R signaling pathway, which may contribute to myocardial dysfunction in rats. The present study confirmed that EPO treatment is beneficial for protecting cardiac function post‑resuscitation, and the roles of EPO in alleviating post‑resuscitation myocardial dysfunction may potentially be associated with enhanced myocardial expression of AT2R.
Collapse
Affiliation(s)
- Hourong Zhou
- Department of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jia Huang
- Emergency Department, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Li Zhu
- Institute of Anesthesia, Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Yu Cao
- Department of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
37
|
Chaudhary R, Garg J, Krishnamoorthy P, Bliden K, Shah N, Agarwal N, Gupta R, Sharma A, Kern KB, Patel NC, Gurbel P. Erythropoietin therapy after out-of-hospital cardiac arrest: A systematic review and meta-analysis. World J Cardiol 2017; 9:830-837. [PMID: 29317989 PMCID: PMC5746625 DOI: 10.4330/wjc.v9.i12.830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/28/2017] [Accepted: 11/22/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To assess safety and efficacy of early erythropoietin (Epo) administration in patients with out-of-hospital cardiac arrest (OHCA).
METHODS A systematic literature search was performed using PubMed, MEDLINE, EMBASE, EBSCO, CINAHL, Web of Science and Cochrane databases, of all studies published from the inception through October 10, 2016. Inclusion criteria included: (1) Adult humans with OHCA and successful sustained return of spontaneous circulation; and (2) studies including mortality/brain death, acute thrombotic events as their end points. Primary efficacy outcome was “brain death or Cerebral Performance Category (CPC) score of 5”. Secondary outcomes were “CPC score 1, and 2-4”, “overall thrombotic events” and “acute coronary stent thrombosis”.
RESULTS We analyzed a total of 606 participants (n = 276 received Epo and n = 330 with standard of care alone) who experienced OHCA enrolled in 3 clinical trials. No significant difference was observed between the Epo and no Epo group in brain death or CPC score 5 (OR = 0.77; 95%CI: 0.42-1.39), CPC score 1 (OR = 1.16, 95%CI: 0.82-1.64), and CPC score 2-4 (OR = 0.77, 95%CI: 0.44-1.36). Epo group was associated with increased thrombotic complications (OR = 2.41, 95%CI: 1.26-4.62) and acute coronary stent thrombosis (OR = 8.16, 95%CI: 1.39-47.99). No publication bias was observed.
CONCLUSION Our study demonstrates no improvement in neurological outcomes and increased incidence of thrombotic events and acute coronary stent thrombosis in OHCA patients who were treated with Epo in addition to standard therapy.
Collapse
Affiliation(s)
- Rahul Chaudhary
- Department of Medicine, Sinai Hospital of Baltimore, Baltimore, MD 21215, United States
| | - Jalaj Garg
- Division of Cardiology, Lehigh Valley Health Network, Allentown, PA 18103, United States
| | - Parasuram Krishnamoorthy
- Department of Medicine, Division of Cardiology, Einstein Healthcare Network, Philadelphia, PA 19141, United States
| | - Kevin Bliden
- Inova Heart and Vascular Institute, Inova Medical Center, Fairfax, VA 22042, United States
| | - Neeraj Shah
- Division of Cardiology, Lehigh Valley Health Network, Allentown, PA 18103, United States
| | - Nayan Agarwal
- Division of Cardiovascular Medicine, University of Florida, Gainesville, FL 32611, United States
| | - Rahul Gupta
- Queens Cardiac Care, Queens, NY 11428, United States
| | - Abhishek Sharma
- Division of Cardiovascular Medicine, State University of New York, Brooklyn, NY 12246, United States
| | - Karl B Kern
- Division of Cardiology, University of Arizona College of Medicine, Tucson, AZ 85721, United States
| | - Nainesh C Patel
- Division of Cardiology, Lehigh Valley Health Network, Allentown, PA 18103, United States
| | - Paul Gurbel
- Inova Heart and Vascular Institute, Inova Medical Center, Fairfax, VA 22042, United States
| |
Collapse
|
38
|
Helix B Surface Peptide Protects Cardiomyocytes Against Hypoxia/Reoxygenation-induced Apoptosis Through Mitochondrial Pathways. J Cardiovasc Pharmacol 2017; 67:418-26. [PMID: 26828322 DOI: 10.1097/fjc.0000000000000367] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Helix B surface peptide (HBSP), a newly developed tissue-protective erythropoietin derivative, has beneficial effects on myocardial ischemia. This study aimed to investigate the cardio-protective effects of HBSP against hypoxia/reoxygenation (H/R) injury and its possible mechanism. METHODS A rat-derived cardiomyocyte cell line (H9C2 cells) were established and pretreated with HBSP. The pretreated primary cultures were subjected to H/R and monitored for cell viability using the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide assay. Intracellular reactive oxygen species (ROS) levels, apoptosis, and mitochondrial membrane potential (ΔΨm) were detected by flow cytometry. The expression of cytochrome C and Bcl-2 family proteins, as well as the activities of caspases 3 and 9 were determined by Western blot analysis and a colorimetric method, respectively. RESULTS HBSP reduced apoptotic cells in cardiomyocytes subjected to H/R. In HBSP-treated cardiomyocytes, the H/R-induced mitochondrial ROS production, ΔΨm collapse, and cytochrome C release from mitochondria to the cytosol significantly decreased. Moreover, HBSP inhibited the activation of caspases 9 and 3, as well as the alteration of Bcl-2 family proteins, which were induced by H/R. CONCLUSIONS These results indicated that HBSP has protective effects against H/R-induced apoptosis by regulating the mitochondrial pathway. This mechanism involves inhibiting mitochondrial ROS generation, inhibiting caspase-3 activity, reducing ΔΨm collapse, reducing cytochrome release, and balancing anti and proapoptotic Bcl-2 family proteins.
Collapse
|
39
|
Mesgarpour B, Heidinger BH, Roth D, Schmitz S, Walsh CD, Herkner H. Harms of off-label erythropoiesis-stimulating agents for critically ill people. Cochrane Database Syst Rev 2017; 8:CD010969. [PMID: 28841235 PMCID: PMC6373621 DOI: 10.1002/14651858.cd010969.pub2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Anaemia is a common problem experienced by critically-ill people. Treatment with erythropoiesis-stimulating agents (ESAs) has been used as a pharmacologic strategy when the blunted response of endogenous erythropoietin has been reported in critically-ill people. The use of ESAs becomes more important where adverse clinical outcomes of transfusing blood products is a limitation. However, this indication for ESAs is not licensed by regulatory authorities and is called off-label use. Recent studies concern the harm of ESAs in a critical care setting. OBJECTIVES To focus on harms in assessing the effects of erythropoiesis-stimulating agents (ESAs), alone or in combination, compared with placebo, no treatment or a different active treatment regimen when administered off-label to critically-ill people. SEARCH METHODS We conducted a systematic search of the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, PsycINFO via OvidSP, CINAHL, all evidence-based medicine (EBM) reviews including IPA and SCI-Expanded, Conference Proceedings Citation Index- Science, BIOSIS Previews and TOXLINE up to February 2017. We also searched trials registries, checked reference lists of relevant studies and tracked their citations by using SciVerse Scopus. SELECTION CRITERIA We considered randomized controlled trials (RCTs) and controlled observational studies, which compared scheduled systemic administration of ESAs versus other effective interventions, placebo or no treatment in critically-ill people. DATA COLLECTION AND ANALYSIS Two review authors independently screened and evaluated the eligibility of retrieved records, extracted data and assessed the risks of bias and quality of the included studies. We resolved differences in opinion by consensus or by involving a third review author. We assessed the evidence using GRADE and created a 'Summary of findings' table. We used fixed-effect or random-effects models, depending on the heterogeneity between studies. We fitted three-level hierarchical Bayesian models to calculate overall treatment effect estimates. MAIN RESULTS Of the 27,865 records identified, 39 clinical trials and 14 observational studies, including a total of 945,240 participants, were eligible for inclusion. Five studies are awaiting classification. Overall, we found 114 adverse events in 33 studies (30 RCTs and three observational studies), and mortality was reported in 41 studies (32 RCTs and nine observational studies). Most studies were at low to moderate risk of bias for harms outcomes. However, overall harm assessment and reporting were of moderate to low quality in the RCTs, and of low quality in the observational studies. We downgraded the GRADE quality of evidence for venous thromboembolism and mortality to very low and low, respectively, because of risk of bias, high inconsistency, imprecision and limitations of study design.It is unclear whether there is an increase in the risk of any adverse events (Bayesian risk ratio (RR) 1.05, 95% confidence interval (CI) 0.93 to 1.21; 3099 participants; 9 studies; low-quality evidence) or venous thromboembolism (Bayesian RR 1.04, 95% CI 0.70 to 1.41; 18,917 participants; 18 studies; very low-quality evidence).There was a decreased risk of mortality with off-label use of ESAs in critically-ill people (Bayesian RR 0.76, 95% CI 0.61 to 0.92; 930,470 participants; 34 studies; low-quality evidence). AUTHORS' CONCLUSIONS Low quality of evidence suggests that off-label use of ESAs may reduce mortality in a critical care setting. There was a lack of high-quality evidence about the harm of ESAs in critically-ill people. The information for biosimilar ESAs is less conclusive. Most studies neither evaluated ESAs' harm as a primary outcome nor predefined adverse events. Any further studies of ESA should address the quality of evaluating, recording and reporting of adverse events.
Collapse
Affiliation(s)
| | | | - Dominik Roth
- Medical University of ViennaDepartment of Emergency MedicineAllgemeines Krankenhaus, Währinger Gürtel
18‐20,ViennaAustria1090
| | - Susanne Schmitz
- Luxembourg Institute of HealthDepartment of Population Health1A‐B, rue Thomas EdisonStrassenLuxembourg1445
| | - Cathal D Walsh
- Department of Mathematics and StatisticsHealth Research Institute (HRI) and MACSIUniversity of LimerickIreland
| | - Harald Herkner
- Medical University of ViennaDepartment of Emergency MedicineAllgemeines Krankenhaus, Währinger Gürtel
18‐20,ViennaAustria1090
| |
Collapse
|
40
|
Cahill TJ, Choudhury RP, Riley PR. Heart regeneration and repair after myocardial infarction: translational opportunities for novel therapeutics. Nat Rev Drug Discov 2017; 16:699-717. [DOI: 10.1038/nrd.2017.106] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
41
|
The Erythropoietin System Protects the Heart Upon Injury by Cardiac Progenitor Cell Activation. VITAMINS AND HORMONES 2017. [PMID: 28629520 DOI: 10.1016/bs.vh.2017.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Erythropoietin (EPO) is a growth hormone, widely known for its role in erythropoiesis. The broad expression of erythropoietin receptor (EPOR) in adult organs suggested that EPO may also affect other cells besides late erythroid progenitors. In the embryonic heart, EPOR is expressed in all cells including the immature proliferating cardiomyocytes. In contrast to the embryonic heart in adulthood, EPOR expression is decreased and mainly detected in immature proliferating cells (i.e., resident cardiac progenitor cells) rather than in terminally differentiated cells (i.e., cardiomyocytes). Since cardiac progenitor cells are considered a regenerative cell source upon cardiac injury, the protective action of the EPO system was tested by creating an erythroid-rescued EPOR knockout mouse model. Although these mice appear to have less immature proliferating myocytes during embryogenesis, they reach adulthood without apparent morphological defects. However, upon ischemia reperfusion, these animals show a greater infarct size, suggesting that the EPO/EPOR protects the heart upon injury. Indeed preclinical studies showed that EPO administration postinfarction improves cardiac function via neoangiogenesis, antiapoptotic mechanisms, and/or CPC activation. Despite the promising preclinical data, large cohort clinical studies in humans failed to show a significant amelioration in cardiac function upon systemic injection of EPO in patients with myocardial infarctions. The discrepancy between preclinical and clinical trials may be due to differences between the doses, the way of delivery, the homogeneity of the cohorts, and last but not least the species differences. These data pinpoint the importance of carrying out preclinical studies in human models of disease as engineered human cardiac tissue that will provide a better understanding of the expression pattern of EPOR and the role of its ligand in human cardiac cells. Such studies may be able to bridge the gap between preclinical rodent data and human clinical trials and thus lead to the design of more successful clinical studies.
Collapse
|
42
|
Cambria E, Pasqualini FS, Wolint P, Günter J, Steiger J, Bopp A, Hoerstrup SP, Emmert MY. Translational cardiac stem cell therapy: advancing from first-generation to next-generation cell types. NPJ Regen Med 2017; 2:17. [PMID: 29302353 PMCID: PMC5677990 DOI: 10.1038/s41536-017-0024-1] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 05/16/2017] [Accepted: 05/22/2017] [Indexed: 12/16/2022] Open
Abstract
Acute myocardial infarction and chronic heart failure rank among the major causes of morbidity and mortality worldwide. Except for heart transplantation, current therapy options only treat the symptoms but do not cure the disease. Stem cell-based therapies represent a possible paradigm shift for cardiac repair. However, most of the first-generation approaches displayed heterogeneous clinical outcomes regarding efficacy. Stemming from the desire to closely match the target organ, second-generation cell types were introduced and rapidly moved from bench to bedside. Unfortunately, debates remain around the benefit of stem cell therapy, optimal trial design parameters, and the ideal cell type. Aiming at highlighting controversies, this article provides a critical overview of the translation of first-generation and second-generation cell types. It further emphasizes the importance of understanding the mechanisms of cardiac repair and the lessons learned from first-generation trials, in order to improve cell-based therapies and to potentially finally implement cell-free therapies.
Collapse
Affiliation(s)
- Elena Cambria
- Institute for Regenerative Medicine, University of Zurich, Zurich, 8044 Switzerland.,Division of Surgical Research, University Hospital of Zurich, Zurich, 8091 Switzerland
| | | | - Petra Wolint
- Institute for Regenerative Medicine, University of Zurich, Zurich, 8044 Switzerland.,Division of Surgical Research, University Hospital of Zurich, Zurich, 8091 Switzerland
| | - Julia Günter
- Institute for Regenerative Medicine, University of Zurich, Zurich, 8044 Switzerland.,Division of Surgical Research, University Hospital of Zurich, Zurich, 8091 Switzerland
| | - Julia Steiger
- Institute for Regenerative Medicine, University of Zurich, Zurich, 8044 Switzerland.,Division of Surgical Research, University Hospital of Zurich, Zurich, 8091 Switzerland
| | - Annina Bopp
- Institute for Regenerative Medicine, University of Zurich, Zurich, 8044 Switzerland.,Division of Surgical Research, University Hospital of Zurich, Zurich, 8091 Switzerland
| | - Simon P Hoerstrup
- Institute for Regenerative Medicine, University of Zurich, Zurich, 8044 Switzerland.,Division of Surgical Research, University Hospital of Zurich, Zurich, 8091 Switzerland.,Heart Center Zurich, University Hospital of Zurich, Zurich, Switzerland.,Wyss Translational Center Zurich, Zurich, Switzerland
| | - Maximilian Y Emmert
- Institute for Regenerative Medicine, University of Zurich, Zurich, 8044 Switzerland.,Division of Surgical Research, University Hospital of Zurich, Zurich, 8091 Switzerland.,Heart Center Zurich, University Hospital of Zurich, Zurich, Switzerland.,Wyss Translational Center Zurich, Zurich, Switzerland
| |
Collapse
|
43
|
Özyüncü N, Güleç S, Özdöl Ç, Candemir B, Ongun A, Tulunay Kaya C, Erol Ç. Impact of serum erythropoietin level on collateral vessel development in patients with coronary artery disease. Anatol J Cardiol 2017; 17:386-391. [PMID: 28315563 PMCID: PMC5469086 DOI: 10.14744/anatoljcardiol.2017.7419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2017] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE Experimental data have shown that Erythropoietin (EPO) stimulates angiogenesis and neovascularization which may result in improved collateral development. The aim of this study was to investigate the association between serum EPO levels and the extent of coronary collaterals. Patient characteristics possibly related with coronary collaterals were also sought. METHODS A total of 256 patients with high grade coronary stenosis or occlusion were evaluated for the extent of coronary collaterals using Rentrop classification. Patients with grade 0 or 1 collaterals were grouped as poor collaterals, while grade 2 or 3 collaterals were grouped as good collaterals. RESULTS Mean age of the study population was 63 years, 77% were males. Subjects with good collaterals were significantly more likely to have anemia (p=0.038) and stable angina pectoris as clinical presentation (p=0.40). Serum EPO levels were not different among good and poor collateral groups (10.4±9.4 mU/mL vs. 9.7±11 mU/mL, p=0.397). The prevalence of all other cardiovascular risk factors, medications, and angiographic characteristics were similar between the two groups. After adjusting for age, gender, and clinical presentation with stable angina pectoris, presence of anemia persisted to be a significant correlate of the good collateral formation (OR: 1.95; 95%; CI: 1.07-3.54, p=0.029). CONCLUSION There has been conflicting results from trials studying the effects of serum EPO on coronary collateral development. The present study, with the largest patient population studying this topic, suggests that presence of anemia, but not serum EPO level, is associated with good collateral development.
Collapse
Affiliation(s)
- Nil Özyüncü
- Department of Cardiology, Faculty of Medicine, Ankara University; Ankara-Turkey.
| | | | | | | | | | | | | |
Collapse
|
44
|
Steppich B, Groha P, Ibrahim T, Schunkert H, Laugwitz KL, Hadamitzky M, Kastrati A, Ott I. Effect of Erythropoietin in patients with acute myocardial infarction: five-year results of the REVIVAL-3 trial. BMC Cardiovasc Disord 2017; 17:38. [PMID: 28109258 PMCID: PMC5251242 DOI: 10.1186/s12872-016-0464-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 12/29/2016] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Erythropoietin (EPO) has been suggested to promote cardiac repair after MI. However, the randomized, double-blind, placebo controlled REVIVAL-3 trial showed that short term high dose EPO in timely reperfused myocardium does not improve left ventricular ejection fraction after 6 months. Moreover, the study raised safety concerns due to a trend towards a higher incidence of adverse clinical events as well as a increase in neointima formation after treatment with EPO. The present study therefore aimed to assess the 5-year clinical outcomes. METHODS After successful reperfusion 138 patients with STEMI were randomly assigned to receive epoetin beta (3.33×104 U, n = 68) or placebo (n = 70) immediately, 24 and 48 h after percutaneous coronary intervention. The primary outcome of the present study- the combined incidence of MACE 5 years after randomization - occurred in 25% of the patients assigned to epoetin beta and 17% of the patients assigned to placebo (RR 1.5; 95% CI 0.8-3.5; p = 0.26). Target lesion revascularization was required in 15 patients (22.1%) treated with epoetin-ß and 9 patients (12.9%) treated with placebo (p = 0.15). Analysis of patients in the upper and lower quartile of baseline hemoglobin as an indirect estimate of endogenous erythropoietin levels revealed no significant impact of endogenous erythropoietin on efficiency of exogen administered epoetin-ß in terms of death and MACE. CONCLUSION These long-term follow-up data show that epoetin beta does not improve clinical outcomes of patients with acute myocardial infarction. TRIAL REGISTRATION URL www.clinicaltrials.gov ; Unique identifier NCT00390832; trial registration date October 19th 2006.
Collapse
Affiliation(s)
- Birgit Steppich
- Deutsches Herzzentrum der Technischen Universität München, Lazarettstr. 36, 80636, Munich, Germany.
| | - Philip Groha
- Deutsches Herzzentrum der Technischen Universität München, Lazarettstr. 36, 80636, Munich, Germany
| | - Tareq Ibrahim
- Medizinische Klinik Klinikum rechts der Isar der Technischen Universität München, Ismaningerstr. 22, 81675, Munich, Germany
| | - Heribert Schunkert
- Medizinische Klinik Klinikum rechts der Isar der Technischen Universität München, Ismaningerstr. 22, 81675, Munich, Germany
| | - Karl-Ludwig Laugwitz
- Medizinische Klinik Klinikum rechts der Isar der Technischen Universität München, Ismaningerstr. 22, 81675, Munich, Germany
| | - Martin Hadamitzky
- Deutsches Herzzentrum der Technischen Universität München, Lazarettstr. 36, 80636, Munich, Germany
| | - Adnan Kastrati
- Deutsches Herzzentrum der Technischen Universität München, Lazarettstr. 36, 80636, Munich, Germany
| | - Ilka Ott
- Deutsches Herzzentrum der Technischen Universität München, Lazarettstr. 36, 80636, Munich, Germany
| | | |
Collapse
|
45
|
Grasso G, Alafaci C, Macdonald RL. Management of aneurysmal subarachnoid hemorrhage: State of the art and future perspectives. Surg Neurol Int 2017; 8:11. [PMID: 28217390 PMCID: PMC5288992 DOI: 10.4103/2152-7806.198738] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 12/01/2016] [Indexed: 12/25/2022] Open
Abstract
Background: Aneurysmal subarachnoid hemorrhage (SAH) accounts for 5% of strokes and carries a poor prognosis. It affects around 6 cases per 100,000 patient years occurring at a relatively young age. Methods: Common risk factors are the same as for stroke, and only in a minority of the cases, genetic factors can be found. The overall mortality ranges from 32% to 67%, with 10–20% of patients with long-term dependence due to brain damage. An explosive headache is the most common reported symptom, although a wide spectrum of clinical disturbances can be the presenting symptoms. Brain computed tomography (CT) allow the diagnosis of SAH. The subsequent CT angiography (CTA) or digital subtraction angiography (DSA) can detect vascular malformations such as aneurysms. Non-aneurysmal SAH is observed in 10% of the cases. In patients surviving the initial aneurysmal bleeding, re-hemorrhage and acute hydrocephalus can affect the prognosis. Results: Although occlusion of an aneurysm by surgical clipping or endovascular procedure effectively prevents rebleeding, cerebral vasospasm and the resulting cerebral ischemia occurring after SAH are still responsible for the considerable morbidity and mortality related to such a pathology. A significant amount of experimental and clinical research has been conducted to find ways in preventing these complications without sound results. Conclusions: Even though no single pharmacological agent or treatment protocol has been identified, the main therapeutic interventions remain ineffective and limited to the manipulation of systemic blood pressure, alteration of blood volume or viscosity, and control of arterial dioxide tension.
Collapse
Affiliation(s)
- Giovanni Grasso
- Department of Experimental Biomedicine and Clinical Neurosciences (BIONEC), Section of Neurosurgery, University of Palermo, Palermo, Italy
| | | | - R Loch Macdonald
- Division of Neurosurgery, St. Michael's Hospital, Labatt Family Centre of Excellence in Brain Injury and Trauma Research, Keenan Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
46
|
Hamo CE, Klem I, Rao SV, Songco V, Najjar S, Lakatta EG, Raman SV, Harrington RA, Heitner JF. The Systematic Evaluation of Identifying the Infarct Related Artery Utilizing Cardiac Magnetic Resonance in Patients Presenting with ST-Elevation Myocardial Infarction. PLoS One 2017; 12:e0169108. [PMID: 28060863 PMCID: PMC5218460 DOI: 10.1371/journal.pone.0169108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 12/12/2016] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Identification of the infarct-related artery (IRA) in patients with STEMI using coronary angiography (CA) is often based on the ECG and can be challenging in patients with severe multi-vessel disease. The current study aimed to determine how often percutaneous intervention (PCI) is performed in a coronary artery different from the artery supplying the territory of acute infarction on cardiac magnetic resonance imaging (CMR). METHODS We evaluated 113 patients from the Reduction of infarct Expansion and Ventricular remodeling with Erythropoetin After Large myocardial infarction (REVEAL) trial, who underwent CMR within 4±2 days of revascularization. Blinded reviewers interpreted CA to determine the IRA and CMR to determine the location of infarction on a 17-segment model. In patients with multiple infarcts on CMR, acuity was determined with T2-weighted imaging and/or evidence of microvascular obstruction. RESULTS A total of 5 (4%) patients were found to have a mismatch between the IRA identified on CMR and CA. In 4/5 cases, there were multiple infarcts noted on CMR. Thirteen patients (11.5%) had multiple infarcts in separate territories on CMR with 4 patients (3.5%) having multiple acute infarcts and 9 patients (8%) having both acute and chronic infarcts. CONCLUSIONS In this select population of patients, the identification of the IRA by CA was incorrect in 4% of patients presenting with STEMI. Four patients with a mismatch had an acute infarction in more than one coronary artery territory on CMR. The role of CMR in patients presenting with STEMI with multi-vessel disease on CA deserves further investigation.
Collapse
Affiliation(s)
- Carine E. Hamo
- Department of Medicine, Stony Brook University Hospital, Stony Brook New York, United States of America
| | - Igor Klem
- The Duke Clinical Research Institute, Durham, North Carolina, United States of America
| | - Sunil V. Rao
- The Duke Clinical Research Institute, Durham, North Carolina, United States of America
| | - Vincent Songco
- Division of Cardiology, New York Methodist Hospital, Brooklyn, New York, United States of America
| | - Samer Najjar
- MedStar Health Research Institute, Washington, DC, United States of America
- Intramural Research Program, National Institute of Aging, the National Institute of Health, Baltimore, Maryland, United States of America
| | - Edward G. Lakatta
- Intramural Research Program, National Institute of Aging, the National Institute of Health, Baltimore, Maryland, United States of America
| | - Subha V. Raman
- Division of Cardiovascular Medicine, Ohio State University, Columbus, Ohio, United States of America
| | - Robert A. Harrington
- Department of Medicine, Stanford University, Palo Alto, California, United States of America
| | - John F. Heitner
- Division of Cardiology, New York Methodist Hospital, Brooklyn, New York, United States of America
| |
Collapse
|
47
|
So Many Targets ∗. J Am Coll Cardiol 2017; 69:434-436. [PMID: 28126161 DOI: 10.1016/j.jacc.2016.10.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 10/17/2016] [Indexed: 11/24/2022]
|
48
|
Signaling Pathways in Cardiac Myocyte Apoptosis. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9583268. [PMID: 28101515 PMCID: PMC5215135 DOI: 10.1155/2016/9583268] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/20/2016] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases, the number 1 cause of death worldwide, are frequently associated with apoptotic death of cardiac myocytes. Since cardiomyocyte apoptosis is a highly regulated process, pharmacological intervention of apoptosis pathways may represent a promising therapeutic strategy for a number of cardiovascular diseases and disorders including myocardial infarction, ischemia/reperfusion injury, chemotherapy cardiotoxicity, and end-stage heart failure. Despite rapid growth of our knowledge in apoptosis signaling pathways, a clinically applicable treatment targeting this cellular process is currently unavailable. To help identify potential innovative directions for future research, it is necessary to have a full understanding of the apoptotic pathways currently known to be functional in cardiac myocytes. Here, we summarize recent progress in the regulation of cardiomyocyte apoptosis by multiple signaling molecules and pathways, with a focus on the involvement of these pathways in the pathogenesis of heart disease. In addition, we provide an update regarding bench to bedside translation of this knowledge and discuss unanswered questions that need further investigation.
Collapse
|
49
|
Kang J, Kim TW, Hur J, Kim HS. Strategy to Prime the Host and Cells to Augment Therapeutic Efficacy of Progenitor Cells for Patients with Myocardial Infarction. Front Cardiovasc Med 2016; 3:46. [PMID: 27933299 PMCID: PMC5121226 DOI: 10.3389/fcvm.2016.00046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 11/08/2016] [Indexed: 11/23/2022] Open
Abstract
Cell therapy in myocardial infarction (MI) is an innovative strategy that is regarded as a rescue therapy to repair the damaged myocardium and to promote neovascularization for the ischemic border zone. Among several stem cell sources for this purpose, autologous progenitors from bone marrow or peripheral blood would be the most feasible and safest cell-source. Despite the theoretical benefit of cell therapy, this method is not widely adopted in the actual clinical practice due to its low therapeutic efficacy. Various methods have been used to augment the efficacy of cell therapy in MI, such as using different source of progenitors, genetic manipulation of cells, or priming of the cells or hosts (patients) with agents. Among these methods, the strategy to augment the therapeutic efficacy of the autologous peripheral blood mononuclear cells (PBMCs) by priming agents may be the most feasible and the safest method that can be applied directly to the clinic. In this review, we will discuss the current status and future directions of priming PBMCs or patients, as for cell therapy of MI.
Collapse
Affiliation(s)
- Jeehoon Kang
- Department of Medicine, Seoul National University Hospital, Seoul, South Korea; Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science & Technology, Seoul National University, Seoul, South Korea
| | - Tae-Won Kim
- Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science & Technology, Seoul National University, Seoul, South Korea; National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, South Korea
| | - Jin Hur
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital , Seoul , South Korea
| | - Hyo-Soo Kim
- Department of Medicine, Seoul National University Hospital, Seoul, South Korea; Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science & Technology, Seoul National University, Seoul, South Korea; National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, South Korea
| |
Collapse
|
50
|
Liu CW, Liao PC, Chen KC, Hsu JC, Li AH, Tu CM, Wu YW. Baseline Hemoglobin Levels Associated with One-Year Mortality in ST-Segment Elevation Myocardial Infarction Patients. ACTA CARDIOLOGICA SINICA 2016; 32:656-666. [PMID: 27899852 DOI: 10.6515/acs20160106a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND The association between hemoglobin (Hb) levels and mortality in patients with ST-segment elevation myocardial infarction (STEMI) remains controversial. The purpose of this study was to examine the mortality among STEMI patients with anemia or erythrocytosis, and further establish the relationship between mortality and the increment of Hb level. METHODS Between 2006 and 2012, 951 consecutive patients with STEMI undergoing primary percutaneous coronary intervention in a medical center in Northern Taiwan were enrolled in our study, including 535 patients with normal Hb level, 148 with anemia (male Hb ≤ 13 g/dl, female ≤ 12) and 268 with erythrocytosis (male Hb ≥ 16, female ≥ 15). RESULTS Patients in the anemia group were the oldest, and had higher morbidity than the normal Hb group, followed by the erythrocytosis group. In regression analyses, neither anemia nor erythrocytosis was associated with 30-day and 1-year mortality. Each 1-g/dl increment of Hb level was not associated with 30-day mortality both in patients with anemia or erythrocytosis. However, it was associated with a decreased risk of 1-year mortality in anemic patients [hazard ratio (HR): 0.756, 95% confidence interval (CI): 0.608-0.938, p = 0.011] and an increased risk of 1-year mortality in those with erythrocytosis (HR: 2.086, 95%CI: 1.106-3.937, p = 0.023). In multivariate analysis, each 1-g/dl increment of Hb level was associated with 1-year mortality both in anemic patients and those with erythrocytosis (HR: 0.788, 95%CI: 0.621-0.999, p = 0.049; HR: 2.302, 95%CI: 1.051-5.04, p = 0.037). CONCLUSIONS Higher hemoglobin levels in STEMI patients with anemia were associated with decreased risks of 1-year mortality, whereas higher hemoglobin levels in those with erythrocytosis were associated with increased risks of one-year mortality.
Collapse
Affiliation(s)
- Cheng-Wei Liu
- Cardiology Division of Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City; ; Department of Internal Medicine, Tri-Service General Hospital, Songshan Branch; ; National Defense Medical Center
| | - Pen-Chih Liao
- Cardiology Division of Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City
| | - Kuo-Chin Chen
- Cardiology Division of Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City
| | - Jung-Cheng Hsu
- Cardiology Division of Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City; ; Chihlee Institute of Technology
| | - Ai-Hsien Li
- Cardiology Division of Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City
| | - Chung-Ming Tu
- Cardiology Division of Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City; ; Chihlee Institute of Technology
| | - Yen-Wen Wu
- Cardiology Division of Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City; ; Departments of Internal Medicine; ; Departments of Nuclear Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei; ; Department of Nuclear Medicine, Far Eastern Memorial Hospital, New Taipei City; ; National Yang-Ming University School of Medicine, Taipei, Taiwan
| |
Collapse
|