1
|
Sung PH, Yue Y, Chen YL, Chiang JY, Cheng BC, Yang CC, Chai HT, Yip HK. Combined dapagliflozin and roxadustat effectively protected heart and kidney against cardiorenal syndrome-induced damage in rodent through activation of cell stress-Nfr2/ARE signalings and stabilizing HIF-1α. Biomed Pharmacother 2024; 180:117567. [PMID: 39423754 DOI: 10.1016/j.biopha.2024.117567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND This study tested whether combined dapagliflozin (DAPA) and roxadustat (ROX) therapy was superior to a singular therapy in protecting heart and kidney functions in rats with cardiorenal syndrome (CRS). METHODS AND RESULTS An in vitro study demonstrated that the cell survival (PI3K/Akt/mTOR)/cell stress (ERK1/2, JNK/p-38) signaling was significantly activated by combination therapy with ROX-DAPA (all p<0.001). Additionally, these two signaling pathways further significantly upregulated the hypoxia-induced factor (HIF)-1α which, in turn, significantly upregulated Nrf2/ARE (HO-1/NQO-1) and angiogenesis/cell-growth factors (EPO/SDF-1α/VEGF/FGF/IGF-2) and downregulated hypoxia-inducible factor prolyl-4-hydroxylase-1 (all p<0.001). Adult-male SD rats were categorized into Groups 1 (sham-operated control)/2 (CRS)/3 (CRS+ROX)/4 (CRS+DAPA)/5 (CRS+ROX+DAPA). By Day 60 after rodent CRS induction, the levels of BUN/creatinine and the ratio of urine protein to creatinine were lowest in Group 1, highest in Group 2, and significantly lower in Group 5 than in Groups 3 and 4; however, they were similar in the latter two groups, whereas the left-ventricular-ejection-fraction exhibited the opposite trend of creatinine among the groups (all p<0.0001). The protein expression levels of cell-survival (p-PI3K/p-Akt-p-mTOR)/cell-stress (p-JNK/p-p38/p-ERK1/2)/Nrf2-ARE (HO-1/NQO-1/SIRT1/SIRT3) signaling factors and angiogenesis factors (HIF-1α/VEGF/SDF-1α/FGF/IGF-2/EPO) significantly and progressively increased from Groups 1-5 (all p<0.0001). CONCLUSION Combined DAPA-ROX therapy has a synergistic effect on protecting heart and kidney functions against CRS-induced damage in rodents.
Collapse
Affiliation(s)
- Pei-Hsun Sung
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan, ROC; Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan, ROC; Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan, ROC
| | - Ya Yue
- The First Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Yi-Ling Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan, ROC; Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan, ROC
| | - John Y Chiang
- Department of Computer Science and Engineering, National Sun Yat-Sen University, Kaohsiung 804201, Taiwan, ROC; Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung 807378, Taiwan, ROC
| | - Ben-Chung Cheng
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan, ROC
| | - Chih-Chao Yang
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan, ROC
| | - Han-Tan Chai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan, ROC
| | - Hon-Kan Yip
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan, ROC; Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan, ROC; Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan, ROC; Department of Nursing, Asia University, Taichung 413305, Taiwan, ROC; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404333, Taiwan, ROC; School of Medicine, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan, ROC.
| |
Collapse
|
2
|
Yang P, Zhu L, Wang S, Gong J, Selvaraj JN, Ye L, Chen H, Zhang Y, Wang G, Song W, Li Z, Cai L, Zhang H, Zhang D. Engineered model of heart tissue repair for exploring fibrotic processes and therapeutic interventions. Nat Commun 2024; 15:7996. [PMID: 39266508 PMCID: PMC11393355 DOI: 10.1038/s41467-024-52221-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 08/30/2024] [Indexed: 09/14/2024] Open
Abstract
Advancements in human-engineered heart tissue have enhanced the understanding of cardiac cellular alteration. Nevertheless, a human model simulating pathological remodeling following myocardial infarction for therapeutic development remains essential. Here we develop an engineered model of myocardial repair that replicates the phased remodeling process, including hypoxic stress, fibrosis, and electrophysiological dysfunction. Transcriptomic analysis identifies nine critical signaling pathways related to cellular fate transitions, leading to the evaluation of seventeen modulators for their therapeutic potential in a mini-repair model. A scoring system quantitatively evaluates the restoration of abnormal electrophysiology, demonstrating that the phased combination of TGFβ inhibitor SB431542, Rho kinase inhibitor Y27632, and WNT activator CHIR99021 yields enhanced functional restoration compared to single factor treatments in both engineered and mouse myocardial infarction model. This engineered heart tissue repair model effectively captures the phased remodeling following myocardial infarction, providing a crucial platform for discovering therapeutic targets for ischemic heart disease.
Collapse
Affiliation(s)
- Pengcheng Yang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Lihang Zhu
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Shiya Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Jixing Gong
- Center of Translational Medicine, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangdong, China
| | - Jonathan Nimal Selvaraj
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Lincai Ye
- Shanghai Institute for Congenital Heart Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai, China
| | - Hanxiao Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yaoyao Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Gongxin Wang
- Henan SCOPE Research Institute of Electrophysiology Co. Ltd., Kaifeng, China
| | - Wanjun Song
- Beijing Geek Gene Technology Co. Ltd., Beijing, China
| | - Zilong Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Lin Cai
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China.
| | - Hao Zhang
- Shanghai Institute for Congenital Heart Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai, China.
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China.
- Cardiovascular Research Institute, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Marrow JP, Alshamali R, Edgett BA, Allwood MA, Cochrane KLS, Al-Sabbag S, Ayoub A, Ask K, Hare GMT, Brunt KR, Simpson JA. Cardiomyocyte crosstalk with endothelium modulates cardiac structure, function, and ischemia-reperfusion injury susceptibility through erythropoietin. Front Physiol 2024; 15:1397049. [PMID: 39011088 PMCID: PMC11246973 DOI: 10.3389/fphys.2024.1397049] [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: 03/06/2024] [Accepted: 06/03/2024] [Indexed: 07/17/2024] Open
Abstract
Erythropoietin (EPO) exerts non-canonical roles beyond erythropoiesis that are developmentally, structurally, and physiologically relevant for the heart as a paracrine factor. The role for paracrine EPO signalling and cellular crosstalk in the adult is uncertain. Here, we provided novel evidence showing cardiomyocyte restricted loss of function in Epo in adult mice induced hyper-compensatory increases in Epo expression by adjacent cardiac endothelial cells via HIF-2α independent mechanisms. These hearts showed concentric cellular hypertrophy, elevated contractility and relaxation, and greater resistance to ischemia-reperfusion injury. Voluntary exercise capacity compared to control hearts was improved independent of any changes to whole-body metabolism or blood O2 content or delivery (i.e., hematocrit). Our findings suggest cardiac EPO had a localized effect within the normoxic heart, which was regulated by cell-specific EPO-reciprocity between cardiomyocytes and endothelium. Within the heart, hyper-compensated endothelial Epo expression was accompanied by elevated Vegfr1 and Vegfb RNA, that upon pharmacological pan-inhibition of VEGF-VEGFR signaling, resulted in a paradoxical upregulation in whole-heart Epo. Thus, we provide the first evidence that a novel EPO-EPOR/VEGF-VEGFR axis exists to carefully mediate cardiac homeostasis via cardiomyocyte-endothelial EPO crosstalk.
Collapse
Affiliation(s)
- Jade P Marrow
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
- IMPART Investigator Team Canada, Guelph, ON, Canada
| | - Razan Alshamali
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
- IMPART Investigator Team Canada, Guelph, ON, Canada
| | - Brittany A Edgett
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
- IMPART Investigator Team Canada, Guelph, ON, Canada
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Melissa A Allwood
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
- IMPART Investigator Team Canada, Guelph, ON, Canada
| | - Kyla L S Cochrane
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
- IMPART Investigator Team Canada, Guelph, ON, Canada
| | - Sara Al-Sabbag
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Anmar Ayoub
- Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Kjetil Ask
- Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Gregory M T Hare
- IMPART Investigator Team Canada, Guelph, ON, Canada
- Department of Anesthesiology and Pain Medicine, St Michael's Hospital, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
| | - Keith R Brunt
- IMPART Investigator Team Canada, Guelph, ON, Canada
- Department of Pharmacology, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Jeremy A Simpson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
- IMPART Investigator Team Canada, Guelph, ON, Canada
| |
Collapse
|
4
|
Wang Y, Li Y, Liu D. Erythropoietin promoted intraplaque angiogenesis by PI3K/AKT/mTOR signaling pathway in atherosclerosis. Tissue Cell 2023; 82:102084. [PMID: 37060746 DOI: 10.1016/j.tice.2023.102084] [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: 11/20/2022] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND This study aimed to investigate role of erythropoietin in atherosclerosis and explore whether underlying mechanism is associated with PI3K/AKT/mTOR pathway. METHODS High-fat-diet-induced atherosclerosis model was established in apolipoprotein E knockout mice (C57BL/6 genetic background). Mice were randomly divided into the control group and the EPO group. Hematoxylin-eosin was performed for the determination of atherosclerotic lesions. The expression levels of related proteins were detected by western blot analysis. RESULTS Erythropoietin significantly enhanced the incidence of hemorrhage in atherosclerotic plaques compared with the control group. The proteins' expression signaling pathways (including PI3K, AKT, and mTOR) and angiogenesis-related proteins (VEGF, COX-2, and HIF-1α) were proved to be up-regulated by erythropoietin. Additionally, erythropoietin significantly enhanced the incidence of hemorrhage in the atherosclerotic plaques compared with the control group. The vitro experiments were conducted in macrophages at 21% O2 or 1% O2. The data showed that expression of p-PI3K, p-AKT, p-mTOR, VEGF, COX-2, and HIF-1α related proteins increased in 1% O2 group than 21% O2 group. Moreover, compared with control group, protein expression including p-PI3K, p-AKT, p-mTOR, VEGF, COX-2, and HIF-1α was markedly increased in EPO group, decreased in inhibitors group, and similar results were observed in EPO+ inhibitors group. CONCLUSION The present study demonstrated that erythropoietin might promote angiogenesis in atherosclerotic vulnerable by activating PI3K/AKT/mTOR signaling pathway in atherosclerotic, providing a novel therapeutic target for atherosclerotic targeted therapy.
Collapse
Affiliation(s)
- Ying Wang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, P.R. China.; Department of Cardiology, Hebei General Hospital, Shijiazhuang, P.R. China
| | - Yongjun Li
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, P.R. China.; Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China.
| | - Dongxia Liu
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, P.R. China
| |
Collapse
|
5
|
Association between Anti-Erythropoietin Receptor Antibodies and Cardiac Function in Patients on Hemodialysis: A Multicenter Cross-Sectional Study. Biomedicines 2022; 10:biomedicines10092092. [PMID: 36140193 PMCID: PMC9495431 DOI: 10.3390/biomedicines10092092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiac dysfunction is an important prognostic predictor of cardiovascular mortality in patients on hemodialysis (HD). Erythropoietin (EPO) has been reported to improve cardiac function by binding to the EPO receptor (EPOR) on cardiomyocytes. This study investigated whether anti-EPOR antibodies were associated with left ventricular cardiac function in patients undergoing HD. This multicenter, cross-sectional observational study included 377 patients (median age, 70 years; 267 (70.8%) males) with chronic kidney disease (CKD) undergoing stable maintenance HD. Serum levels of anti-EPOR antibodies were measured, and echocardiography was used to assess the left ventricular mass index (LVMI) and left ventricular ejection fraction (LVEF). Anti-EPOR antibodies were found in 17 patients (4.5%). LVMI was greater (median of 135 g/m2 vs. 115 g/m2, p = 0.042), and the prevalence of LVEF < 50% was higher (35.3% vs. 15.6%, p = 0.032) in patients with anti-EPOR antibodies than in those without. Multivariable linear regression and logistic regression analysis (after adjusting for known risk factors of heart failure) revealed that anti-EPOR antibodies were independently associated with LVMI (coefficient 16.2%; 95% confidence interval (CI) 1.0−35.0%, p = 0.043) and LVEF <50% (odds ratio 3.20; 95% CI 1.05−9.73, p = 0.041). Thus, anti-EPOR antibody positivity was associated with left ventricular dysfunction in patients undergoing HD.
Collapse
|
6
|
Shoeibi S, Mozdziak P, Mohammadi S. Important signals regulating coronary artery angiogenesis. Microvasc Res 2017; 117:1-9. [PMID: 29247718 DOI: 10.1016/j.mvr.2017.12.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 01/03/2023]
Abstract
Angiogenesis is a complex process of budding, the formation of new blood vessels from pre-existing microvessels, via migration, proliferation and survival. Vascular angiogenesis factors include different classes of molecules that have a fundamental role in blood vessel formation. Numerous inducers of angiogenesis, such as the members of the vascular endothelial growth factor (VEGF) family, basic fibroblast growth factor (bFGF), angiopoietin (Ang), hepatocyte growth factor (HGF), and hypoxia inducible factor-1 (HIF-1), have an important role in angiogenesis. However, VEGF, platelet-derived growth factor (PDGF), and transforming growth factor β (TGF-β) expression appear to be important in intraplaque angiogenesis. Interaction and combined effects between growth factors is essential in endothelial cell migration, proliferation, differentiation, and endothelial cell-cell communication that ultimately lead to the microvessel formation. Since VEGF has a key role during angiogenesis; it may be considered as a good therapeutic target in the clinic. The essential function of several angiogenic factors involved in coronary angiogenesis and intraplaque angiogenesis in atherosclerosis are carefully considered along with the use of angiogenic factors in clinical practice.
Collapse
Affiliation(s)
- Sara Shoeibi
- Cellular and Molecular research Center, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Paul Mozdziak
- Graduate Physiology Program, North Carolina State University, Raleigh, NC
| | - Shabnam Mohammadi
- Department of Basic Sciences, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
7
|
Mechanisms and mediators of hypertension induced by erythropoietin and related molecules. Nephrol Dial Transplant 2017; 33:1690-1698. [DOI: 10.1093/ndt/gfx324] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/23/2017] [Indexed: 11/14/2022] Open
|
8
|
van den Hoogenhof MMG, van der Made I, Beqqali A, de Groot NE, Damanafshan A, van Oort RJ, Pinto YM, Creemers EE. The RNA-binding protein Rbm38 is dispensable during pressure overload-induced cardiac remodeling in mice. PLoS One 2017; 12:e0184093. [PMID: 28850611 PMCID: PMC5574583 DOI: 10.1371/journal.pone.0184093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/17/2017] [Indexed: 11/23/2022] Open
Abstract
The importance of tightly controlled alternative pre-mRNA splicing in the heart is emerging. The RNA binding protein Rbm24 has recently been identified as a pivotal cardiac splice factor, which governs sarcomerogenesis in the heart by controlling the expression of alternative protein isoforms. Rbm38, a homolog of Rbm24, has also been implicated in RNA processes such as RNA splicing, RNA stability and RNA translation, but its function in the heart is currently unknown. Here, we investigated the role of Rbm38 in the healthy and diseased adult mouse heart. In contrast to the heart- and skeletal muscle-enriched protein Rbm24, Rbm38 appears to be more broadly expressed. We generated somatic Rbm38 -/- mice and show that global loss of Rbm38 results in hematopoietic defects. Specifically, Rbm38 -/- mice were anemic and displayed enlarged spleens with extramedullary hematopoiesis, as has been shown earlier. The hearts of Rbm38 -/- mice were mildly hypertrophic, but cardiac function was not affected. Furthermore, Rbm38 deficiency did not affect cardiac remodeling (i.e. hypertrophy, LV dilation and fibrosis) or performance (i.e. fractional shortening) after pressure-overload induced by transverse aorta constriction. To further investigate molecular consequences of Rbm38 deficiency, we examined previously identified RNA stability, splicing, and translational targets of Rbm38. We found that stability targets p21 and HuR, splicing targets Mef2d and Fgfr2, and translation target p53 were not altered, suggesting that these Rbm38 targets are tissue-specific or that Rbm38 deficiency may be counteracted by a redundancy mechanism. In this regard, we found a trend towards increased Rbm24 protein expression in Rbm38 -/- hearts. Overall, we conclude that Rbm38 is critical in hematopoiesis, but does not play a critical role in the healthy and diseased heart.
Collapse
Affiliation(s)
| | - Ingeborg van der Made
- Department of Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Abdelaziz Beqqali
- Department of Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Nina E. de Groot
- Department of Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Amin Damanafshan
- Department of Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Ralph J. van Oort
- Department of Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Yigal M. Pinto
- Department of Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Esther E. Creemers
- Department of Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
9
|
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
|
10
|
Hayashi T, Joki N, Tanaka Y, Hase H. Anaemia and early phase cardiovascular events on haemodialysis. Nephrology (Carlton) 2016; 20 Suppl 4:1-6. [PMID: 26456311 DOI: 10.1111/nep.12642] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2015] [Indexed: 01/30/2023]
Abstract
Although the mechanism of the cardio-renal anaemia syndrome (CRAS) has been elucidated in considerable detail over the past decade, cardiovascular disease (CVD) remains a leading cause of death among patients with end-stage kidney disease (ESKD) undergoing haemodialysis, and these patients' cardiovascular mortality is greater than that of the general population. Recent studies have reported that the CVD risk increases with advancing chronic kidney disease (CKD) stage. Furthermore, the incidence of cardiovascular events is highest during the first week after dialysis initiation, with increased risk in incident haemodialysis patients. This accumulated evidence demonstrates that how patients are managed during the pre-dialysis phase may have important implications on long-term outcomes in ESKD. Anaemia, a non-traditional risk factor for CVD, advances exponentially along with declining kidney function due to insufficient erythropoietin production. Anaemia causes functional abnormalities of the heart, as represented by cardiac hypertrophy, which results from increased cardiac workload induced by an increased preload. Left ventricular hypertrophy (LVH), a traditional risk factor for CVD, is especially associated with advanced CKD stage and could be a major risk factor for cardiovascular complications such as ischaemic heart disease, heart failure, and sudden cardiac death. In ESKD, anaemia develops more severely and requires a higher amount of erythropoiesis-stimulating agent (ESA) therapy before dialysis initiation. This suggests that improvement in anaemia management during the pre-dialysis phase may have a beneficial effect on cardiac hypertrophy and contribute to reducing the CVD risk after initiating haemodialysis.
Collapse
Affiliation(s)
- Toshihide Hayashi
- Division of Nephrology, Toho University Ohashi Medical Center, Tokyo, Japan
| | - Nobuhiko Joki
- Division of Nephrology, Toho University Ohashi Medical Center, Tokyo, Japan
| | - Yuri Tanaka
- Division of Nephrology, Toho University Ohashi Medical Center, Tokyo, Japan
| | - Hiroki Hase
- Division of Nephrology, Toho University Ohashi Medical Center, Tokyo, Japan
| |
Collapse
|
11
|
Nagai T, Nishimura K, Honma T, Higashiyama A, Sugano Y, Nakai M, Honda S, Iwakami N, Okada A, Kawakami S, Kanaya T, Asaumi Y, Aiba T, Nishida Y, Kubota Y, Sugiyama D, Okamura T, Noguchi T, Kusano K, Ogawa H, Yasuda S, Anzai T. Prognostic significance of endogenous erythropoietin in long-term outcome of patients with acute decompensated heart failure. Eur J Heart Fail 2016; 18:803-13. [PMID: 27126377 DOI: 10.1002/ejhf.537] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 02/28/2016] [Accepted: 03/05/2016] [Indexed: 12/20/2022] Open
Abstract
AIMS Although previous reports suggest that an elevated endogenous erythropoietin (EPO) level is associated with worse clinical outcomes in chronic heart failure (HF) patients, the prognostic implication of EPO in patients with acute decompensated HF (ADHF) and underlying mechanisms of the high EPO level in severe HF patients who have a poor prognosis remain unclear. METHODS AND RESULTS We examined 539 consecutive ADHF patients with EPO measurement on admission from our registry. During a median follow-up period of 329 days, a higher EPO level on admission was independently associated with worse clinical outcomes [hazard ratio (HR) 1.25, 95% confidence interval (CI) 1.06-1.48, P = 0.008], and haemoglobin level was the strongest determinant of EPO level (P < 0.001), whereas estimated glomerular filtration rate (eGFR) was not significant in multivariate regression analysis. In the anaemic subgroup of 318 patients, a higher EPO level than expected on the basis of their haemoglobin level was related to increased adverse events (HR 1.63, 95% CI 1.05-2.49, P = 0.028). Moreover, estimated plasma volume excess rate was positively associated with EPO level (P = 0.003), and anaemic patients with a higher than expected EPO level tended to have a higher estimated plasma volume excess rate and plasma lactate level, and lower systemic oxygen saturation level with the preservation of the reticulocyte production index than those with a lower than expected EPO level. CONCLUSION A high EPO level predicts long-term worse clinical outcomes in ADHF patients, independent of anaemia and impaired renal function. Anaemia and hypoxia due to severe congestion may synergistically contribute to a high EPO level in high-risk HF patients.
Collapse
Affiliation(s)
- Toshiyuki Nagai
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Kunihiro Nishimura
- Preventive Medicine and Epidemiology Informatics, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Takehiro Honma
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Aya Higashiyama
- Preventive Medicine and Epidemiology Informatics, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yasuo Sugano
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Michikazu Nakai
- Preventive Medicine and Epidemiology Informatics, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Satoshi Honda
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Naotsugu Iwakami
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Atsushi Okada
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Shoji Kawakami
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Tomoaki Kanaya
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yasuhide Asaumi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Takeshi Aiba
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yoko Nishida
- Foundation for Biomedical Research and Innovation, Hyogo, Japan
| | - Yoshimi Kubota
- Department of Environmental and Preventive Medicine, Hyogo College of Medicine, Hyogo, Japan
| | - Daisuke Sugiyama
- Department of Preventive Medicine and Public Health, Keio University, Tokyo, Japan
| | - Tomonori Okamura
- Department of Preventive Medicine and Public Health, Keio University, Tokyo, Japan
| | - Teruo Noguchi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Kengo Kusano
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Hisao Ogawa
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Satoshi Yasuda
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | | |
Collapse
|
12
|
Suzuki K, Satoh K, Ikeda S, Sunamura S, Otsuki T, Satoh T, Kikuchi N, Omura J, Kurosawa R, Nogi M, Numano K, Sugimura K, Aoki T, Tatebe S, Miyata S, Mukherjee R, Spinale FG, Kadomatsu K, Shimokawa H. Basigin Promotes Cardiac Fibrosis and Failure in Response to Chronic Pressure Overload in Mice. Arterioscler Thromb Vasc Biol 2016; 36:636-46. [DOI: 10.1161/atvbaha.115.306686] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/16/2016] [Indexed: 01/08/2023]
Affiliation(s)
- Kota Suzuki
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Kimio Satoh
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Shohei Ikeda
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Shinichiro Sunamura
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Tomohiro Otsuki
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Taijyu Satoh
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Nobuhiro Kikuchi
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Junichi Omura
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Ryo Kurosawa
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Masamichi Nogi
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Kazuhiko Numano
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Koichiro Sugimura
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Tatsuo Aoki
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Shunsuke Tatebe
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Satoshi Miyata
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Rupak Mukherjee
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Francis G. Spinale
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Kenji Kadomatsu
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| | - Hiroaki Shimokawa
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.S., K.S., S.I., S.S., T.O., T.S., N.K., J.O., R.K., M.N., K.N., K.S., T.A., S.T., H.S.); Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston (R.M.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia (F.G.S.); and Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan (K.K.)
| |
Collapse
|
13
|
Chen YH, Chou CY, Sun CY. Impairment of the Cellular Distribution and Stability of the Erythropoietin Receptor Through the Direct Targeting of Aristolochic Acid. Toxicol Sci 2015; 147:246-54. [PMID: 26136230 DOI: 10.1093/toxsci/kfv125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Aristolochic acid (AA) nephropathy is complicated with early onset and severe anemia. The molecular pathological mechanism of AA-induced anemia remains unclear. The aim of this study was to evaluate the putative pathological roles of the erythropoietin receptor (EPOR) in AA-induced anemia in both AA nephropathy zebrafish and cultured human renal tubular cells (HK2). Immunofluorescence staining experiments revealed that AA colocalizes with the EPOR in zebrafish embryos as well as in the cytoplasm of HK2 cells. After exogenous EPO stimulation, the EPOR was detected in the plasma membrane of HK cells. However, cotreatment with AA and EPO inhibited EPOR signaling and its membrane localization upon EPO stimulation. The results of studies with a protein synthesis inhibitor and a lysosome inhibitor revealed that AA accelerates the lysosomal degradation of EPOR. The molecular docking results suggest that AA may interact with the N-terminus of EPOR. Together with the results of light absorption and in vitro competition assays, we concluded that AA treatment impairs EPOR membrane localization, accelerates its lysosomal degradation, and consequently downregulates EPOR signaling by direct targeting. The results of this study may further detail the pathological mechanism of severe anemia complicated with AA nephropathy.
Collapse
Affiliation(s)
- Yau-Hung Chen
- *Department of Chemistry, Tamkang University, Tamsui, New Taipei City 251, Taiwan;
| | - Chi-Yuan Chou
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Chiao-Yin Sun
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung, Taiwan; and Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
| |
Collapse
|
14
|
|
15
|
Hsu PL, Horng LY, Peng KY, Wu CL, Sung HC, Wu RT. Activation of mitochondrial function and Hb expression in non-haematopoietic cells by an EPO inducer ameliorates ischaemic diseases in mice. Br J Pharmacol 2014; 169:1461-76. [PMID: 23530756 DOI: 10.1111/bph.12197] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 01/31/2013] [Accepted: 02/04/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Many organs suffer from ischaemic injuries that reduce their ability to generate sufficient energy, which is required for functional maintenance and repair. Erythropoietin (EPO) ameliorates ischaemic injuries by pleiotropic effects. The aim of this study was to investigate the effect and mechanism of a small molecule EH-201, and found it as a potent EPO inducer and its effect in non-haematopoietic cells for therapeutic potential in ischemic disorders. EXPERIMENTAL APPROACH Mice kidney slices, primary hepatocytes, primary cardiomyocytes and C2C12 myoblasts were treated with EH-201. The effects of this treatment on EPO, Hb expression and mitochondrial biogenesis were analysed. In vivo, doxorubicin-induced cardiomyopathic mice were treated with EH-201. The mice were subjected to an endurance test, electrocardiography and echocardiography, and a histological examination of the isolated hearts was performed. EH-201 was also administered to cisplatin-induced nephropathic mice. KEY RESULTS In non-haematopoietic cells, EH-201 was potent at inducing EPO. EH-201 also stimulated mitochondrial biogenesis and enhanced the expression of Hb by a mechanism dependent on EPO-mediated signalling. In mechanistic studies, using EPO and EPO receptor-neutralizing antibodies, we confirmed that EH-201 enhances EPO-EPOR autocrine activity. EH-201 robustly increased the endurance performance activity of healthy and cardiomyopathic mice during hypoxic stress, enhanced myocardial mitochondrial biogenesis and Hb expression, and also improved cardiac function. EH-201 ameliorated anaemia and renal dysfunction in nephropathic mice. CONCLUSIONS AND IMPLICATIONS The enhancement and recovery of cellular functions through the stimulation of mitochondrial activity and Hb production in non-haematopoietic cells by an inducer of endogenous EPO has potential as a therapeutic strategy for ischaemic diseases.
Collapse
Affiliation(s)
- Pei-Lun Hsu
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
16
|
Jun JH, Jun NH, Shim JK, Shin EJ, Kwak YL. Erythropoietin protects myocardium against ischemia-reperfusion injury under moderate hyperglycemia. Eur J Pharmacol 2014; 745:1-9. [PMID: 25446919 DOI: 10.1016/j.ejphar.2014.09.038] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 09/23/2014] [Accepted: 09/23/2014] [Indexed: 01/26/2023]
Abstract
Erythropoietin (EPO), an essential hormone for erythropoiesis, provides protection against myocardial ischemia/reperfusion (I/R) injury. Hyperglycemia during acute myocardial infarction aggravates organ damage and attenuates the efficacies of various protective measures. This study aimed to investigate the protective role of EPO against myocardial I/R injury under a clinically relevant moderate hyperglycemic condition and its associated mechanisms. Eighty-two Sprague-Dawley rats were randomly assigned to six groups: normoglycemia-Sham, normoglycemia-I/R-control-saline (IRC), normoglycemia-I/R-EPO (IRE), hyperglycemia-Sham, hyperglycemia-IRC, and hyperglycemia-IRE. The rats received 1.2 g/kg dextrose or same volume of normal saline depending on the group. I/R was induced by a 30 min period of ischemia followed by reperfusion for 4 h. For 1 h before I/R injury, intravenous 4000 IU/kg of EPO was administered. EPO pretreatment significantly reduced the number of apoptotic cells and the infarct size compared with those of the control groups. EPO increased GATA-4 phosphorylation and acetylation against I/R in hyperglycemic myocardium. It also enhanced ERK induced GATA-4 post-translational modifications such as increased GATA-4 phosphorylation and acetylation, and decreased GATA-4 ubiquitination following hypoxia-reoxygenation in H9c2 cells in hyperglycemic medium. Increased GATA-4 stability by EPO diminished I/R-related down-regulation of Bcl-2 and reduction of caspase-3 activities in hyperglycemic myocardium. In conclusion, EPO pretreatment before I/R injury conveyed significant myocardial protection under moderate hyperglycemic condition through mechanisms involved in reduction of caspase-3 activity and up-regulation of Bcl-2 in association with enhanced ERK-induced GATA-4 stability.
Collapse
Affiliation(s)
- Ji Hae Jun
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Na-Hyung Jun
- Department of Anesthesiology and Pain Medicine, National Health Insurance Corporation Ilsan Hospital, Goyang, Republic of Korea
| | - Jae-Kwang Shim
- Department of Anesthesiology and Pain Medicine, Yonsei Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Jung Shin
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Lan Kwak
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
17
|
Pourrajab F, Babaei Zarch M, Baghi Yazdi M, Rahimi Zarchi A, Vakili Zarch A. Application of stem cell/growth factor system, as a multimodal therapy approach in regenerative medicine to improve cell therapy yields. Int J Cardiol 2014; 173:12-9. [PMID: 24612559 DOI: 10.1016/j.ijcard.2014.02.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 12/26/2013] [Accepted: 02/08/2014] [Indexed: 12/13/2022]
Abstract
Stem cells hold a great promise for regenerative medicine, especially for replacing cells in infarcted organ that hardly have any intrinsic renewal capacity, including heart and brain. Signaling pathways that regulate pluripotency or lineage-specific gene and protein expression have been the major focus of stem cell research. Between them, there are some well known signaling pathways such as GF/GFR systems, SDF-1α/CXC4 ligand receptor interaction and PI3K/Akt signaling, and cytokines may regulate cell fate decisions, and can be utilized to positively influence cell therapy outcomes or accentuate synergistic compliance. For example, contributing factors in the progression of heart failure are both the loss of cardiomyocytes after myocardial infarction, and the absence of an adequate endogenous repair signaling. Combining cell engraftment with therapeutic signaling factor delivery is more exciting in terms of host progenitor/donor stem cell survival and proliferation. Thus stem cell-based therapy, besides triggering signaling pathways through GF/GFR systems can become a realistic option in regenerative processes for replacing lost cells and reconstituting the damaged organ, as before.
Collapse
Affiliation(s)
- Fatemeh Pourrajab
- School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Clinical Biochemistry and Molecular Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | | | | | - Abolfazl Rahimi Zarchi
- School of Nursing, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Abbas Vakili Zarch
- School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
18
|
The Role of Treatment for Anemia as a Therapeutic Target in the Management of Chronic Heart Failure: Insights After RED-HF. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2013; 16:279. [DOI: 10.1007/s11936-013-0279-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
19
|
Erythropoietin and the heart: physiological effects and the therapeutic perspective. Int J Cardiol 2013; 171:116-25. [PMID: 24377712 DOI: 10.1016/j.ijcard.2013.12.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 10/08/2013] [Accepted: 12/10/2013] [Indexed: 01/22/2023]
Abstract
Erythropoietin (Epo) has been thought to act exclusively on erythroid progenitor cells. The identification of Epo receptor (EpoR) in non-haematopoietic cells and tissues including neurons, astrocytes, microglia, immune cells, cancer cell lines, endothelial cells, bone marrow stromal cells, as well as cells of myocardium, reproductive system, gastrointestinal tract, kidney, pancreas and skeletal muscle indicates that Epo has pleiotropic actions. Epo shows signals through protein kinases, anti-apoptotic proteins and transcription factors. In light of interest of administering recombinant human erythropoietin (rhEpo) and its analogues for limiting infarct size and left ventricular (LV) remodelling after acute myocardial infarction (AMI) in humans, the foremost studies utilising rhEpo are reviewed. The putative mechanisms involved in Epo-induced cardioprotection are related to the antiapoptotic, anti-inflammatory and angiogenic effects of Epo. Thus, cardioprotective potentials of rhEpo are reviewed in this article by focusing on clinical applicability. An overview of non-haematopoietic Epo analogues, which are a reliable alternative to the classic EpoR agonists and may prevent undesired side effects, is also provided.
Collapse
|
20
|
Wen Y, Xu J, Ma X, Gao Q. High-dose erythropoietin in acute ST-segment elevation myocardial infarction: a meta-analysis of randomized controlled trials. Am J Cardiovasc Drugs 2013; 13:435-42. [PMID: 24097294 DOI: 10.1007/s40256-013-0042-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE We sought to perform a meta-analysis to evaluate the potential influence of high-dose erythropoietin (EPO) on cardiac function parameters in patients with acute ST-segment elevation myocardial infarction (STEMI). METHODS AND RESULTS By searching PubMed, EMBASE, and the Cochrane Library (up to December 2012), seven randomized controlled trials (RCTs) reporting cardiac functional parameters with a total of 1,250 acute STEMI patients were identified. When applied to patients with acute STEMI, high-dose EPO was relatively safe and no increase in all-caused death and severe adverse effects were indicated. Estimates were pooled from fixed or random effects models. Compared with controls, high-dose EPO resulted in a slight but significant improvement in left ventricular ejection fraction of 1.02 % [95 % confidence interval (CI) 0.17-1.88, P = 0.019, I (2) = 0 %] and an improvement in left ventricular end-systolic volume of -4.61 ml (95 % CI -7.64 to -1.58, P = 0.003, I (2) = 27.7 %). CONCLUSIONS Available evidence suggested that high-dose EPO has limited cardio-protective effects in patients with STEMI. However, considering the relatively short follow-up durations and small patient populations in the current RCTs, the effects of high-dose EPO on clinical outcomes in patients with STEMI need to be evaluated in larger prospective RCTs of longer duration.
Collapse
Affiliation(s)
- Yanting Wen
- Center for Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing, 210093, China
| | | | | | | |
Collapse
|
21
|
Nakayama M, Takeda M, Asaumi Y, Shimokawa H. Identification and visualization of stimulus-specific transcriptional activity in cardiac hypertrophy in mice. Int J Cardiovasc Imaging 2013; 30:211-9. [PMID: 24162179 DOI: 10.1007/s10554-013-0314-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/14/2013] [Indexed: 10/26/2022]
Abstract
Identification of specific signaling pathways for cardiac hypertrophy in living animals is challenging because no methods have been established to directly observe sequential molecular signaling events at the transcriptional level during pathogenesis. Here, our aim was to develop a useful method for monitoring the specific signaling pathways involved in the development of cardiac hypertrophy in vivo. Expression profiling of the left ventricle by microarray was performed in 2 different mouse models of cardiac hypertrophy: mechanical pressure overload by transverse aortic constriction (TAC) and neurohumoral activation by angiotensin II (Ang II) infusion. To annotate the information on transcription factor-binding sites, we collected promoter sequences and identified significantly frequent transcription factor-binding sites in the promoter regions of coregulated genes from both models (P < 0.05, binomial probability). Finally, we injected a firefly luciferase vector plasmid containing each transcription factor-binding site into the left ventricle in both models. In the TAC and Ang II models, we selected 379 and 12 upregulated genes, respectively. Twenty binding sites for transcription factors, including activator protein 4, were identified in the TAC model, and 4 sites for transcription factors, including ecotropic viral integration 1, were identified in the Ang II model. GATA-binding sites were noted in both models of cardiac hypertrophy. Using the firefly luciferase reporter, we demonstrated the enhancement of transcriptional activity during the progression of cardiac hypertrophy using in vivo imaging in live mice. These results suggested that our approach was useful for the identification of unique transcription factors that characterize different models of cardiac hypertrophy in vivo.
Collapse
Affiliation(s)
- Masaharu Nakayama
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan,
| | | | | | | |
Collapse
|
22
|
Plasma vascular endothelial growth factor and angiogenin are positively related to erythropoietin dose in hemodialysis patients. Adv Med Sci 2013; 58:143-9. [PMID: 23640951 DOI: 10.2478/v10039-012-0071-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE Experimental data confirmed that erythropoietin (EPO) administration alters the course of various pathological situations such as heart failure and tumor growth by inducing vascular endothelial growth factor-A (VEGF-A) expression. The effect of EPO dose on plasma VEGF-A level in hemodialysis (HD) patients was evaluated. The effect of EPO dose on plasma angiogenin level in HD patients was also evaluated, since angiogenin is necessary for angiogenesis induced by VEGF-A. METHODS Thirty two HD patients (10 diabetics) enrolled into the study. Patients were iron replete and did not suffer from infections, autoimmune diseases or malignancies. Plasma VEGF-A and angiogenin, as well as serum interleukin-6 and tumor necrosis factor-α were measured by means of ELISA. RESULTS Weekly EPO dose per kg of dry body weight was positively related to both VEGF-A and angiogenin, whereas no relation was detected among VEGF-A or angiogenin and hemoglobin, inflammation or presence of diabetes mellitus. These relations among EPO dose and VEGF-A or angiogenin remained after adjustment for hemoglobin concentration or inflammation or presence of diabetes mellitus. CONCLUSIONS EPO dose may affect plasma VEGF-A and angiogenin concentrations in HD patients.
Collapse
|
23
|
Roubille F, Prunier F, Barrère-Lemaire S, Leclercq F, Piot C, Kritikou EA, Rhéaume E, Busseuil D, Tardif JC. What is the Role of Erythropoietin in Acute Myocardial Infarct? Bridging the Gap Between Experimental Models and Clinical Trials. Cardiovasc Drugs Ther 2013; 27:315-31. [DOI: 10.1007/s10557-013-6461-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
24
|
Dame C. GATA4: the missing link between Epo and cardioprotection?! Nutr Metab Cardiovasc Dis 2013; 23:e19-e20. [PMID: 23541168 DOI: 10.1016/j.numecd.2013.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 01/03/2013] [Accepted: 01/08/2013] [Indexed: 11/16/2022]
|
25
|
Roubille F, Micheau A, Combes S, Thibaut S, Souteyrand G, Cayla G, Bonello L, Lesavre N, Sportouch-Dukhan C, Klein F, Berboucha S, Cade S, Cung TT, Raczka F, Macia JC, Gervasoni R, Cransac F, Leclercq F, Barrère-Lemaire S, Paganelli F, Mottref P, Vernhet Kovacsik H, Ovize M, Piot C. Intracoronary administration of darbepoetin-alpha at onset of reperfusion in acute myocardial infarction: Results of the randomized Intra-Co-EpoMI trial. Arch Cardiovasc Dis 2013; 106:135-45. [DOI: 10.1016/j.acvd.2012.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 11/13/2012] [Accepted: 12/04/2012] [Indexed: 01/29/2023]
|
26
|
Abstract
The hematopoietic growth factor erythropoietin (Epo) circulates in plasma and controls the oxygen carrying capacity of the blood (Fisher. Exp Biol Med (Maywood) 228:1-14, 2003). Epo is produced primarily in the adult kidney and fetal liver and was originally believed to play a role restricted to stimulation of early erythroid precursor proliferation, inhibition of apoptosis, and differentiation of the erythroid lineage. Early studies showed that mice with targeted deletion of Epo or the Epo receptor (EpoR) show impaired erythropoiesis, lack mature erythrocytes, and die in utero around embryonic day 13.5 (Wu et al. Cell 83:59-67, 1995; Lin et al. Genes Dev. 10:154-164, 1996). These animals also exhibited heart defects, abnormal vascular development as well as increased apoptosis in the brain suggesting additional functions for Epo signaling in normal development of the central nervous system and heart. Now, in addition to its well-known role in erythropoiesis, a diverse array of cells have been identified that produce Epo and/or express the Epo-R including endothelial cells, smooth muscle cells, and cells of the central nervous system (Masuda et al. J Biol Chem. 269:19488-19493, 1994; Marti et al. Eur J Neurosci. 8:666-676, 1996; Bernaudin et al. J Cereb Blood Flow Metab. 19:643-651, 1999; Li et al. Neurochem Res. 32:2132-2141, 2007). Endogenously produced Epo and/or expression of the EpoR gives rise to autocrine and paracrine signaling in different organs particularly during hypoxia, toxicity, and injury conditions. Epo has been shown to regulate a variety of cell functions such as calcium flux (Korbel et al. J Comp Physiol B. 174:121-128, 2004) neurotransmitter synthesis and cell survival (Velly et al. Pharmacol Ther. 128:445-459, 2010; Vogel et al. Blood. 102:2278-2284, 2003). Furthermore Epo has neurotrophic effects (Grimm et al. Nat Med. 8:718-724, 2002; Junk et al. Proc Natl Acad Sci U S A. 99:10659-10664, 2002), can induce an angiogenic phenotype in cultured endothelial cells and is a potent angiogenic factor in vivo (Ribatti et al. Eur J Clin Invest. 33:891-896, 2003) and might enhance ventilation in hypoxic conditions (Soliz et al. J Physiol. 568:559-571, 2005; Soliz et al. J Physiol. 583, 329-336, 2007). Thus multiple functions have been identified breathing new life and exciting possibilities into what is really an old growth factor.This review will address the function of Epo in non-hematopoietic tissues with significant emphasis on the brain and heart.
Collapse
Affiliation(s)
- Omolara O Ogunshola
- Institute of Veterinary Physiology, Vetsuisse Faculty and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
27
|
Prognostic significance of left ventricular hypertrophy observed at dialysis initiation depends on the pre-dialysis use of erythropoiesis-stimulating agents. Clin Exp Nephrol 2012; 17:294-303. [PMID: 23100176 DOI: 10.1007/s10157-012-0705-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 09/28/2012] [Indexed: 01/20/2023]
Abstract
BACKGROUND Recent experimental studies suggest that erythropoietin promotes beneficial myocardial remodeling during left ventricular hypertrophy (LVH); however, such compensatory capacity may be limited due to insufficient erythropoietin production in chronic kidney disease patients. Thus, this study aimed to explore the effect of pre-dialysis erythropoiesis-stimulating agent (ESA) use on the prognostic significance of LVH in dialyzed patients. METHODS This retrospective study included 404 consecutive patients who started dialysis between 2001 and 2009. The interaction of ESA with the association between left ventricular mass index (LVMI) observed at dialysis initiation and all-cause and cardiovascular mortality was analyzed at the end of 2010 using the Cox model. RESULTS During a median follow-up of 36.5 months, 164 patients died, 31 of them from heart failure. The frequency of pre-dialysis ESA use was 58.7 % and median LVMI was 160.3 g/m(2). Of interest, patients with the lowest tertile of LVMI had worse survival compared with those with each subsequent tertile. LVMI was inversely associated with all-cause mortality [hazard ratio (HR) 0.991, 95 % confidence interval (CI) 0.988-0.995, P = 0.000] after extensive adjustment including ejection fraction, whereas the prognostic value of LVMI for cardiovascular mortality was dependent on pre-dialysis ESA use [adjusted HR 1.010, 95 % CI 0.999-1.020, P = 0.065 for pre-dialysis ESA(+) and 0.978, 95 % CI 0.967-0.989, P = 0.000 for pre-dialysis ESA(-), respectively]. CONCLUSIONS Our results suggest that reverse epidemiology may exist between LVH and mortality and that pre-dialysis ESA use may modify the prognostic significance of LVH observed at dialysis initiation for cardiovascular mortality in dialyzed patients.
Collapse
|
28
|
Jun JH, Shim JK, Ryoo HM, Kwak YL. Erythropoietin-activated ERK/MAP kinase enhances GATA-4 acetylation via phosphorylation of serine 261 of GATA-4. J Cell Physiol 2012; 228:190-7. [DOI: 10.1002/jcp.24121] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
29
|
|
30
|
Abstract
Erythropoietin (Epo) is an essential hormone that binds and activates the Epo receptor (EpoR) resident on the surface of erythroid progenitor cells, thereby promoting erythropoiesis. Recombinant human erythropoietin has been used successfully for over 20 years to treat anemia in millions of patients. In addition to erythropoiesis, Epo has also been reported to have other effects, such as tissue protection and promotion of tumor cell growth or survival. This became of significant concern in 2003, when some clinical trials in cancer patients reported increased tumor progression and worse survival outcomes in patients treated with erythropoiesis-stimulating agents (ESAs). One of the potential mechanisms proffered to explain the observed safety issues was that functional EpoR was expressed in tumors and/or endothelial cells, and that ESAs directly stimulated tumor growth and/or antagonized tumor ablative therapies. Since then, numerous groups have performed further research evaluating this potential mechanism with conflicting data and conclusions. Here, we review the biology of endogenous Epo and EpoR expression and function in erythropoiesis, and evaluate the evidence pertaining to the expression of EpoR on normal nonhematopoietic and tumor cells.
Collapse
|
31
|
Hoch M, Fischer P, Stapel B, Missol-Kolka E, Sekkali B, Scherr M, Favret F, Braun T, Eder M, Schuster-Gossler K, Gossler A, Hilfiker A, Balligand JL, Drexler H, Hilfiker-Kleiner D. Erythropoietin preserves the endothelial differentiation capacity of cardiac progenitor cells and reduces heart failure during anticancer therapies. Cell Stem Cell 2012; 9:131-43. [PMID: 21816364 DOI: 10.1016/j.stem.2011.07.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 05/31/2011] [Accepted: 07/05/2011] [Indexed: 11/17/2022]
Abstract
Anticancer therapies, such as targeting of STAT3 or the use of anthracyclins (doxorubicin), can induce cardiomyopathy. In mice prone to developing heart failure as a result of reduced cardiac STAT3 expression (cardiomyocyte-restricted deficiency of STAT3) or treatment with doxorubicin, we observed impaired endothelial differentiation capacity of Sca-1(+) cardiac progenitor cells (CPCs) in conjunction with attenuated CCL2/CCR2 activation. Mice in both models also displayed reduced erythropoietin (EPO) levels in the cardiac microenvironment. EPO binds to CPCs and seems to be responsible for maintaining an active CCL2/CCR2 system. Supplementation with the EPO derivative CERA in a hematocrit-inactive low dose was sufficient to upregulate CCL2, restore endothelial differentiation of CPCs, and preserve the cardiac microvasculature and cardiac function in both mouse models. Thus, low-dose EPO treatment could potentially be exploited as a therapeutic strategy to reduce the risk of heart failure in certain treatment regimens.
Collapse
Affiliation(s)
- Melanie Hoch
- Department of Cardiology and Angiology, Medical School Hannover, 30625 Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
It has been well established that blood and bone share a unique, regulatory relationship with one another, though the specifics of this relationship still remain unanswered. Erythropoietin (Epo) is known primarily for its role as a hematopoietic hormone. However, after the discovery of Epo receptor outside the hematopoietic tissues, Epo has been avidly studied for its possible nonhematopoietic effects. It has been proposed that Epo interacts with bone both directly, by activating bone marrow stromal cells, and indirectly, through signaling pathways on hematopoietic stem cells. Yet, the role of Epo in regulating skeletal maintenance and regeneration remains controversial. Here, we review the current state of knowledge pertaining to the effects of Epo on the skeleton.
Collapse
Affiliation(s)
- S J McGee
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109-1078, USA
| | | | | | | | | |
Collapse
|
33
|
Salisch SV, Klar M, Thurisch B, Bungert J, Dame C. Gata4 and Sp1 regulate expression of the erythropoietin receptor in cardiomyocytes. J Cell Mol Med 2012; 15:1963-72. [PMID: 21029371 PMCID: PMC3918051 DOI: 10.1111/j.1582-4934.2010.01193.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Experimental studies indicate significant cardioprotective effects of recombinant erythropoietin (Epo) by binding to the Epo receptor (EpoR) and by inducing various molecular mechanisms, including activation of Gata4, a transcription factor that induces anti-apoptotic genes. However, specific molecular mechanisms of EpoR regulation in cardiomyocytes are unknown. We identified a 774 bp regulatory domain in the EpoR 5' flanking region by reporter gene assays in murine HL-1 cardiomyocytes. The binding sites for Gata and Sp transcription factors both significantly contributed to EpoR promoter activity. DNA-binding studies (EMSA and ChIP assays) identified Gata4 and Sp1 as EpoR promoter-binding proteins in HL1 cardiomyocytes. Although Sp1 alone stimulates EpoR only slightly, forced expression of Gata4 significantly induced EpoR mRNA expression. In addition, knockdown of Gata4 (but also of Sp1) resulted in a significant decrease of EpoR transcript levels in HL-1 cardiomyocytes. Cumulative in vitro data suggest that function of the Sp1 site is essential for the Gata4-mediated transcription. In vivo, analysis of transgenic mice expressing an inducible small-hairpin RNA against Gata4 confirmed suppression of EpoR expression in the heart. Treating mice with high-dose doxorubicin not only resulted in Gata4 protein depletion, but also down-regulated EpoR, followed by up-regulation of EpoR transcripts when Gata4 levels recovered. In conclusion, we identified Gata4 as novel regulator of EpoR transcription in cardiomyocytes. In models of cardiac injury, down-regulation of Gata4 or Sp1 may limit the accessibility of the EpoR for binding of erythropoiesis-stimulating agents (ESA). Thereby our data underline the essential role of Gata4 in mediating cardioprotective effects.
Collapse
Affiliation(s)
- Sandy von Salisch
- Department of Neonatology, Charité- Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | |
Collapse
|
34
|
Kagaya Y, Asaumi Y, Wang W, Takeda M, Nakano M, Satoh K, Fukumoto Y, Shimokawa H. Current Perspectives on Protective Roles of Erythropoietin in Cardiovascular System: Erythropoietin Receptor as a Novel Therapeutic Target. TOHOKU J EXP MED 2012; 227:83-91. [DOI: 10.1620/tjem.227.83] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Yutaka Kagaya
- Comprehensive Education Center for Community Medicine, Tohoku University Graduate School of Medicine
| | - Yasuhide Asaumi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Wanting Wang
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Morihiko Takeda
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Makoto Nakano
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Kimio Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Yoshihiro Fukumoto
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| |
Collapse
|
35
|
Yang J, Xiao Z, Li T, Gu X, Fan B. Erythropoietin promotes the growth of pituitary adenomas by enhancing angiogenesis. Int J Oncol 2011; 40:1230-7. [PMID: 22086127 PMCID: PMC3584615 DOI: 10.3892/ijo.2011.1261] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Accepted: 10/25/2011] [Indexed: 11/06/2022] Open
Abstract
rhEPO is frequently used in clinical practice to treat anemia. However, recently rhEPO has been reported to accelerate tumor growth, progression and metastasis. Many pituitary adenoma patients, particularly those with macroprolactinomas, tend to have anemia and may need rhEPO therapy. To date, whether rhEPO has deleterious effects on pituitary adenomas has not been defined. Here we demonstrated for the first time that human pituitary adenomas are EPOR negative tumors and rhEPO accelerated the tumor growth of MMQ pituitary adenoma xenografts via enhancement of angiogenesis in vivo, whereas rhEPO displayed no direct effect on MMQ cells in vitro. Our mechanistic study showed that rhEPO administration increased phosphorylation of JAK2, STAT3 and VEGF expression in human umbilical vein endothelial cells (HUVECs) in vitro and in MMQ cell xenografts in vivo. Furthermore, VEGF inhibitor attenuated rhEPO induced angiogenesis and delayed tumor growth in MMQ pituitary adenoma xenografts in vivo. JAK2 inhibitor AG490 attenuated EPO induced HUVECs proliferation, phosphorylation of JAK2, STAT3 and VEGF upregulation in vitro and inhibited EPO induced vessel formation in Chicken chorioallantoic membrane (CAM) angiogenesis model in vivo. These results suggest that rhEPO administration may promote the growth of pituitary adenomas by enhancing angiogenesis through EPO-JAK2-STAT3-VEGF signaling pathway. rhEPO should be used with caution in anemia patients bearing pituitary adenoma due to its potential deleterious effects.
Collapse
Affiliation(s)
- Jinsheng Yang
- Department of Neurosurgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, PR China
| | | | | | | | | |
Collapse
|
36
|
Ammar HI, Saba S, Ammar RI, Elsayed LA, Ghaly WBAA, Dhingra S. Erythropoietin protects against doxorubicin-induced heart failure. Am J Physiol Heart Circ Physiol 2011; 301:H2413-21. [PMID: 21984540 DOI: 10.1152/ajpheart.01096.2010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The hormone erythropoietin (EPO) has been demonstrated to have cardioprotective properties. The present study investigates the role of EPO to prevent heart failure following cancer treatment with doxorubicin [adriamycin (AD)]. Male Wistar rats (150 ± 10 g) were treated with saline (vehicle control group); with EPO, subcutaneously at 1,000 IU/kg body wt, three times per week for 4 wk (EPO group); with adriamycin, intraperitoneally at 2.5 mg/kg body wt, three times per week for 2 wk (AD group); and with adriamycin and EPO (EPO-AD group). Echocardiographic measurements showed that EPO-AD treatment prevented the AD-induced decline in cardiac function. Each of the hearts was then exposed to ischemia and reperfusion during Langendorff perfusion. The percentage of recovery after ischemia-reperfusion was significantly greater in EPO-AD than the AD-treated group for left ventricular developed pressure, maximal increase in pressure, and rate pressure product. The level of oxidative stress was significantly higher in AD (5 μM for 24 h)-exposed isolated cardiomyocytes; EPO (5 U/ml for 48 h) treatment prevented this. EPO treatment also decreased AD-induced cardiomyocyte apoptosis, which was associated with the decrease in the Bax-to-Bcl2 ratio and caspase-3 activation. Immunostaining of myocardial tissue for CD31 showed a significant decrease in the number of capillaries in AD-treated animals. EPO-AD treatment restored the number of capillaries. In conclusion, EPO treatment effectively prevented AD-induced heart failure. The protective effect of EPO was associated with a decreased level of oxidative stress and apoptosis in cardiomyocytes as well as improved myocardial angiogenesis.
Collapse
|
37
|
|
38
|
High glucose stimulates the expression of erythropoietin in rat glomerular epithelial cells. Lab Anim Res 2011; 27:245-50. [PMID: 21998614 PMCID: PMC3188732 DOI: 10.5625/lar.2011.27.3.245] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 09/15/2011] [Accepted: 09/15/2011] [Indexed: 02/04/2023] Open
Abstract
It has been reported that the levels of erythropoietin are associated with diabetes mellitus. Glomerular epithelial cells, located in the renal cortex, play an important role in the regulation of kidney function and hyperglycemia-induced cell loss of glomerular epithelial cells is implicated in the onset of diabetic nephropathy. This study investigated the effect of high glucose on erythropoietin and erythropoietin receptor expression in rat glomerular epithelial cells. We found that 25 mM D-glucose, but not mannitol or L-glucose, stimulated erythropoietin mRNA and protein expression in a time dependent manner (>4 h) in rat glomerular epithelial cells. In addition, 25 mM glucose, but not mannitol or L-glucose, also increased the phosphorylation of erythropoietin receptor, suggesting a role for erythropoietin receptor phosphorylation in erythropoietin synthesis. We conclude that high glucose stimulates erythropoietin production and erythropoietin receptor phosphorylation in rat glomerular epithelial cells.
Collapse
|
39
|
Wang W, Kagaya Y, Asaumi Y, Fukui S, Takeda M, Shimokawa H. Protective Effects of Recombinant Human Erythropoietin against Pressure Overload-Induced Left Ventricular Remodeling and Premature Death in Mice. TOHOKU J EXP MED 2011; 225:131-43. [DOI: 10.1620/tjem.225.131] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Wanting Wang
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Yutaka Kagaya
- Graduate Medical Education Center, Tohoku University Hospital
| | - Yasuhide Asaumi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Shigefumi Fukui
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Morihiko Takeda
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW In 1985-1989, erythropoietin (EPO), its receptor (EPOR), and janus kinase 2 were cloned; established to be essential for definitive erythropoiesis; and initially intensely studied. Recently, new impetus, tools, and model systems have emerged to re-examine EPO/EPOR actions, and are addressed in this review. Impetus includes indications that EPO affects significantly more than standard erythroblast survival pathways, the development of novel erythropoiesis-stimulating agents, increasing evidence for EPO/EPOR cytoprotection of ischemically injured tissues, and potential EPO-mediated worsening of tumorigenesis. RECENT FINDINGS New findings are reviewed in four functional contexts: (pro)erythroblast survival mechanisms, new candidate EPO/EPOR effects on erythroid cell development and new EPOR responses, EPOR downmodulation and trafficking, and novel erythropoiesis-stimulating agents. SUMMARY As Current Opinion, this monograph seeks to summarize, and provoke, new EPO/EPOR action concepts. Specific problems addressed include: beyond (and before) BCL-XL, what key survival factors are deployed in early-stage proerythroblasts? Are distinct EPO/EPOR signals transduced in stage-selective fashions? Is erythroblast proliferation also modulated by EPO/EPOR signals? What functions are subserved by new noncanonical EPO/EPOR response factors (e.g. podocalyxin like-1, tribbles 3, reactive oxygen species, and nuclear factor kappa B)? What key regulators mediate EPOR inhibition and trafficking? And for emerging erythropoiesis-stimulating agents, to what extent do activities parallel EPOs (or differ in advantageous, potentially complicating ways, or both)?
Collapse
|
41
|
Ruifrok WPT, Lipsic E, de Boer RA, van Gilst WH, van Veldhuisen DJ. Erythropoiesis stimulation in acute ischemic syndromes. Heart Fail Clin 2010; 6:313-21. [PMID: 20630406 DOI: 10.1016/j.hfc.2009.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Erythropoietin (EPO) is a hematopoietic hormone with extensive nonhematopoietic properties. The discovery of an EPO receptor outside the hematopoietic system has fuelled research into the beneficial effects of EPO for various conditions, predominantly in cardiovascular disease. Experimental evidence has revealed the cytoprotective properties of EPO, and it seems that the EPO-EPO receptor system provides a powerful backbone against acute myocardial ischemia, gaining from the different properties of EPO. There is an ongoing discussion about possible discrepancy between preclinical and clinical effects of EPO on the cardiovascular system. Large, randomized, placebo-controlled clinical trials are underway to give a final verdict on EPO treatment for acute coronary syndromes.
Collapse
Affiliation(s)
- Willem-Peter T Ruifrok
- Department of Cardiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands.
| | | | | | | | | |
Collapse
|
42
|
Ueda K, Takano H, Niitsuma Y, Hasegawa H, Uchiyama R, Oka T, Miyazaki M, Nakaya H, Komuro I. Sonic hedgehog is a critical mediator of erythropoietin-induced cardiac protection in mice. J Clin Invest 2010; 120:2016-29. [PMID: 20484812 DOI: 10.1172/jci39896] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Accepted: 03/24/2010] [Indexed: 12/24/2022] Open
Abstract
Erythropoietin reportedly has beneficial effects on the heart after myocardial infarction, but the underlying mechanisms of these effects are unknown. We here demonstrate that sonic hedgehog is a critical mediator of erythropoietin-induced cardioprotection in mice. Treatment of mice with erythropoietin inhibited left ventricular remodeling and improved cardiac function after myocardial infarction, independent of erythropoiesis and the mobilization of bone marrow-derived cells. Erythropoietin prevented cardiomyocyte apoptosis and increased the number of capillaries and mature vessels in infarcted hearts by upregulating the expression of angiogenic cytokines such as VEGF and angiopoietin-1 in cardiomyocytes. Erythropoietin also increased the expression of sonic hedgehog in cardiomyocytes, and inhibition of sonic hedgehog signaling suppressed the erythropoietin-induced increase in angiogenic cytokine expression. Furthermore, the beneficial effects of erythropoietin on infarcted hearts were abolished by cardiomyocyte-specific deletion of sonic hedgehog. These results suggest that erythropoietin protects the heart after myocardial infarction by inducing angiogenesis through sonic hedgehog signaling.
Collapse
Affiliation(s)
- Kazutaka Ueda
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Westenbrink BD, Ruifrok WPT, Voors AA, Tilton RG, van Veldhuisen DJ, Schoemaker RG, van Gilst WH, de Boer RA. Vascular endothelial growth factor is crucial for erythropoietin-induced improvement of cardiac function in heart failure. Cardiovasc Res 2010; 87:30-9. [DOI: 10.1093/cvr/cvq041] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
44
|
Santhanam AVR, d'Uscio LV, Katusic ZS. Cardiovascular effects of erythropoietin an update. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2010; 60:257-85. [PMID: 21081221 DOI: 10.1016/b978-0-12-385061-4.00009-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Erythropoietin (EPO) is a therapeutic product of recombinant DNA technology and it has been in clinical use as stimulator of erythropoiesis over the last two decades. Identification of EPO and its receptor (EPOR) in the cardiovascular system expanded understanding of physiological and pathophysiological role of EPO. In experimental models of cardiovascular and cerebrovascular disorders, EPO exerts protection either by preventing apoptosis of cardiac myocytes, smooth muscle cells, and endothelial cells, or by increasing endothelial production of nitric oxide. In addition, EPO stimulates mobilization of progenitor cells from bone marrow thereby accelerating repair of injured endothelium and neovascularization. A novel signal transduction pathway involving EPOR--β-common heteroreceptor is postulated to enhance EPO-mediated tissue protection. A better understanding of the role of β-common receptor signaling as well as development of novel analogs of EPO with enhanced nonhematopoietic protective effects may expand clinical application of EPO in prevention and treatment of cardiovascular and cerebrovascular disorders.
Collapse
|
45
|
Taniguchi N, Nakamura T, Sawada T, Matsubara K, Furukawa K, Hadase M, Nakahara Y, Nakamura T, Matsubara H. Erythropoietin Prevention Trial of Coronary Restenosis and Cardiac Remodeling After ST-Elevated Acute Myocardial Infarction (EPOC-AMI) - A Pilot, Randomized, Placebo-Controlled Study -. Circ J 2010; 74:2365-71. [DOI: 10.1253/circj.cj-10-0267] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | - Takeshi Nakamura
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine
| | - Takahisa Sawada
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine
| | | | | | | | | | | | - Hiroaki Matsubara
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine
| |
Collapse
|
46
|
Vogiatzi G, Briasoulis A, Tousoulis D, Papageorgiou N, Stefanadis C. Is there a role for erythropoietin in cardiovascular disease? Expert Opin Biol Ther 2009; 10:251-64. [PMID: 20028188 DOI: 10.1517/14712590903547819] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
47
|
Abstract
Anemia is frequently observed in patients with chronic heart failure and is increasingly recognized as an important and treatable condition. The present review will summarize the current knowledge of the prevalence, causative factors, and pathophysiologic correlates of anemia in chronic heart failure. Despite increasing knowledge of anemia there are very few evidence-based recommendations for treatment. Potential benefits of therapy with supplemental hematinics such as iron, subcutaneous erythropoiesis-stimulating agents, or both will also be discussed.
Collapse
Affiliation(s)
- Stavros G Drakos
- 3rd Cardiology Department, University of Athens School of Medicine, Athens, Greece
| | | | | | | |
Collapse
|
48
|
Klopsch C, Furlani D, Gäbel R, Li W, Pittermann E, Ugurlucan M, Kundt G, Zingler C, Titze U, Wang W, Ong LL, Wagner K, Li RK, Ma N, Steinhoff G. Intracardiac injection of erythropoietin induces stem cell recruitment and improves cardiac functions in a rat myocardial infarction model. J Cell Mol Med 2009; 13:664-79. [PMID: 19449462 PMCID: PMC3822874 DOI: 10.1111/j.1582-4934.2008.00546.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Erythropoietin (EPO) protects the myocardium from ischaemic injury and promotes beneficial remodelling. We assessed the therapeutic efficacy of intracardiac EPO injection and EPO-mediated stem cell homing in a rat myocardial infarction (MI) model. Following MI, EPO (3000 U/kg) or saline was delivered by intracardiac injection. Compared to myocardial infarction control group (MIC), EPO significantly improved left ventricular function (n =11-14, P < 0.05) and decreased right ventricular wall stress (n = 8, P < 0.05) assessed by pressure-volume loops after 6 weeks. MI-EPO hearts exhibited smaller infarction size (20.1 +/- 1.1% versus 27.8 +/- 1.2%; n = 6-8, P < 0.001) and greater capillary density (338.5 +/- 14.7 versus 259.8 +/- 9.2 vessels per mm2; n = 6-8, P < 0.001) than MIC hearts. Direct EPO injection reduced post-MI myocardial apoptosis by approximately 41% (0.27 +/- 0.03% versus 0.42 +/- 0.03%; n = 6, P= 0.005). The chemoattractant SDF-1 was up-regulated significantly assessed by quantitative realtime PCR and immunohistology. c-Kit(+) and CD34(+) stem cells were significantly more numerous in MI-EPO than in MIC at 24 hrs in peripheral blood (n = 7, P < 0.05) and 48 hrs in the infarcted hearts (n = 6, P < 0.001). Further, the mRNAs of Akt, eNOS and EPO receptor were significantly enhanced in MI-EPO hearts (n = 7, P < 0.05). Intracardiac EPO injection restores myocardial functions following MI, which may attribute to the improved early recruitment of c-Kit(+) and CD34(+) stem cells via the enhanced expression of chemoattractant SDF-1.
Collapse
|
49
|
Burger DE, Xiang FL, Hammoud L, Jones DL, Feng Q. Erythropoietin protects the heart from ventricular arrhythmia during ischemia and reperfusion via neuronal nitric-oxide synthase. J Pharmacol Exp Ther 2009; 329:900-7. [PMID: 19307451 DOI: 10.1124/jpet.109.150896] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Erythropoietin (EPO) is a potent cardioprotective agent in models of myocardial ischemia and reperfusion (I/R). It has been suggested recently that EPO may also reduce ventricular arrhythmia after I/R. The present study investigated the role of neuronal nitric oxide synthase (nNOS) on the antiarrhythmic effects of EPO. EPO treatment increased nNOS expression in isolated neonatal mouse ventricular myocytes. Cotreatment with the phosphatidylinositol 3 (PI3)-kinase inhibitor, LY294002 [2-(4-morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one hydrochloride], or treatment of cardiomyocytes infected with a dominant negative adenovirus targeted to Akt1 (ADV-dnAkt1) blocked the effects of EPO on nNOS expression, suggesting that EPO regulates nNOS expression via PI3-kinase and Akt. To examine the in vivo antiarrhythmic effects of EPO, wild-type (WT) and nNOS(-/-) mice were anesthetized and, after a baseline measurement, subjected to myocardial I/R to provoke ventricular arrhythmias. Pretreatment with EPO 24 h before ischemia increased nNOS expression and significantly reduced the number of premature ventricular contractions (PVCs) and the incidence of ventricular tachycardia (VT) in WT mice. In contrast, treatment with EPO had no effect on PVCs or the incidence of VT in nNOS(-/-) mice. Furthermore, EPO treatment after ischemia significantly reduced the threshold dose of cesium chloride (CsCl) to induce VT. We conclude that EPO via nNOS protects the heart from spontaneous and CsCl-induced ventricular arrhythmia during myocardial I/R.
Collapse
MESH Headings
- Animals
- Arrhythmias, Cardiac/chemically induced
- Arrhythmias, Cardiac/etiology
- Arrhythmias, Cardiac/prevention & control
- Cells, Cultured
- Cesium/pharmacology
- Chlorides/pharmacology
- Electrocardiography
- Enzyme Inhibitors/pharmacology
- Erythropoietin/pharmacology
- Erythropoietin/therapeutic use
- Gene Expression/drug effects
- Gene Expression/genetics
- Heart/drug effects
- Heart/physiopathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardial Infarction/etiology
- Myocardial Infarction/pathology
- Myocardial Infarction/prevention & control
- Myocardial Reperfusion Injury/complications
- Myocardial Reperfusion Injury/pathology
- Myocardium/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Nitric Oxide Synthase Type I/physiology
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphoinositide-3 Kinase Inhibitors
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Tachycardia, Ventricular/chemically induced
- Tachycardia, Ventricular/physiopathology
- Tachycardia, Ventricular/prevention & control
- Ventricular Premature Complexes/physiopathology
- Ventricular Premature Complexes/prevention & control
Collapse
Affiliation(s)
- Dylan E Burger
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | | | | | | | | |
Collapse
|
50
|
Fukui S, Fukumoto Y, Suzuki J, Saji K, Nawata J, Shinozaki T, Kagaya Y, Watanabe J, Shimokawa H. Diabetes mellitus accelerates left ventricular diastolic dysfunction through activation of the renin–angiotensin system in hypertensive rats. Hypertens Res 2009; 32:472-80. [DOI: 10.1038/hr.2009.43] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|