1
|
Jiang Y, Cai Y, Han R, Xu Y, Xia Z, Xia W. Salvianolic acids and its potential for cardio-protection against myocardial ischemic reperfusion injury in diabetes. Front Endocrinol (Lausanne) 2024; 14:1322474. [PMID: 38283744 PMCID: PMC10811029 DOI: 10.3389/fendo.2023.1322474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/21/2023] [Indexed: 01/30/2024] Open
Abstract
The incidence of diabetes and related mortality rate increase yearly in modern cities. Additionally, elevated glucose levels can result in an increase of reactive oxygen species (ROS), ferroptosis, and the disruption of protective pathways in the heart. These factors collectively heighten the vulnerability of diabetic individuals to myocardial ischemia. Reperfusion therapies have been effectively used in clinical practice. There are limitations to the current clinical methods used to treat myocardial ischemia-reperfusion injury. As a result, reducing post-treatment ischemia/reperfusion injury remains a challenge. Therefore, efforts are underway to provide more efficient therapy. Salvia miltiorrhiza Bunge (Danshen) has been used for centuries in ancient China to treat cardiovascular diseases (CVD) with rare side effects. Salvianolic acid is a water-soluble phenolic compound with potent antioxidant properties and has the greatest hydrophilic property in Danshen. It has recently been discovered that salvianolic acids A (SAA) and B (SAB) are capable of inhibiting apoptosis by targeting the JNK/Akt pathway and the NF-κB pathway, respectively. This review delves into the most recent discoveries regarding the therapeutic and cardioprotective benefits of salvianolic acid for individuals with diabetes. Salvianolic acid shows great potential in myocardial protection in diabetes mellitus. A thorough understanding of the protective mechanism of salvianolic acid could expand its potential uses in developing medicines for treating diabetes mellitus related myocardial ischemia-reperfusion.
Collapse
Affiliation(s)
- Yuxin Jiang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Yin Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Ronghui Han
- Faculty of Chinese Medicine State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao SAR, China
| | - Youhua Xu
- Faculty of Chinese Medicine State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao SAR, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Doctoral Training Platform for Research and Translation, BoShiWan, GuanChong Village, Shuanghe Town, ZhongXiang City, Hubei, China
| | - Weiyi Xia
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Doctoral Training Platform for Research and Translation, BoShiWan, GuanChong Village, Shuanghe Town, ZhongXiang City, Hubei, China
| |
Collapse
|
2
|
Benjanuwattra J, Apaijai N, Chunchai T, Singhanat K, Arunsak B, Intachai K, Chattipakorn SC, Chattipakorn N. The temporal impact of erythropoietin administration on mitochondrial function and dynamics in cardiac ischemia/reperfusion injury. Exp Mol Pathol 2022; 127:104802. [DOI: 10.1016/j.yexmp.2022.104802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/24/2022] [Accepted: 06/01/2022] [Indexed: 11/28/2022]
|
3
|
Gawish MF, Selim SA, Abd El-Star AA, Ahmed SM. Histological and immunohistochemical study of the effect of ozone versus erythropoietin on induced skeletal muscle ischemia-reperfusion injury in adult male rats. Ultrastruct Pathol 2022; 46:96-109. [PMID: 35130793 DOI: 10.1080/01913123.2022.2035874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Ischemia reperfusion (IR) injury of skeletal muscles is a serious problem because of its local and systemic complications. Previous studies reported that ozone and erythropoietin could alleviate IR effect on several organs. The current research is established to evaluate the possible protective role of ozone versus erythropoietin following IR injury of the gastrocnemius muscle. Fifty rats were equally divided into five groups: I control, II ischemia reperfusion (IR), III post-reperfusion ozone treated, IV post-reperfusion erythropoietin-treated, and V recovering post-reperfusion without treatment groups. The right femoral arteries of all rats were clamped for three hours to induce ischemia then clamps were released to allow reperfusion for two hours. Rats of group II were scarified immediately after reperfusion period. Rats of group III were injected with ozone just after reperfusion for 14 days. Animals of group IV were injected with erythropoietin just after reperfusion for 14 days. Rats of group V rats were kept for 2 weeks following reperfusion without treatment. Blood samples were obtained to estimate lactate dehydrogenase (LDH) and creatine kinase (CK) enzymes. Gastrocnemius muscle was processed for measurement of tissue malondialdehyde (MDA), as well as examination by light and electron microscopes. iNOS and PCNA immunohistochemistry and statistical analysis were applied. The current results indicated that both ozone and erythropoietin could be used as protective agents reducing the muscular damage induced by IR injury.
Collapse
Affiliation(s)
- Magdy F Gawish
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, Egypt
| | - Sally A Selim
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, Egypt
| | - Alyaa A Abd El-Star
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, Egypt
| | - Samah M Ahmed
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, Egypt
| |
Collapse
|
4
|
Kaur D, Behl T, Sehgal A, Singh S, Sharma N, Badavath VN, Ul Hassan SS, Hasan MM, Bhatia S, Al-Harassi A, Khan H, Bungau S. Unravelling the potential neuroprotective facets of erythropoietin for the treatment of Alzheimer's disease. Metab Brain Dis 2022; 37:1-16. [PMID: 34436747 DOI: 10.1007/s11011-021-00820-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/09/2021] [Indexed: 02/06/2023]
Abstract
During the last three decades, recombinant DNA technology has produced a wide range of hematopoietic and neurotrophic growth factors, including erythropoietin (EPO), which has emerged as a promising protein drug in the treatment of several diseases. Cumulative studies have recently indicated the neuroprotective role of EPO in preclinical models of acute and chronic neurodegenerative disorders, including Alzheimer's disease (AD). AD is one of the most prevalent neurodegenerative illnesses in the elderly, characterized by the accumulation of extracellular amyloid-ß (Aß) plaques and intracellular neurofibrillary tangles (NFTs), which serve as the disease's two hallmarks. Unfortunately, AD lacks a successful treatment strategy due to its multifaceted and complex pathology. Various clinical studies, both in vitro and in vivo, have been conducted to identify the various mechanisms by which erythropoietin exerts its neuroprotective effects. The results of clinical trials in patients with AD are also promising. Herein, it is summarized and reviews all such studies demonstrating erythropoietin's potential therapeutic benefits as a pleiotropic neuroprotective agent in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Dapinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | | - Syed Shams Ul Hassan
- School of Medicine and Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Mohammad Mehedi Hasan
- Department of Biochemistry and Molecular Biology, Faculty of Life Science, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
- Amity Institute of Pharmacy, Amity University, Noida, Haryana, India
| | - Ahmed Al-Harassi
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
5
|
Cytoprotective effects of erythropoietin: What about the lung? Biomed Pharmacother 2021; 139:111547. [PMID: 33831836 DOI: 10.1016/j.biopha.2021.111547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/17/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
Erythropoietin (Epo) is a pleiotropic cytokine, essential for erythropoiesis. Epo and its receptor (Epo-R) are produced by several tissues and it is now admitted that Epo displays other physiological functions than red blood cell synthesis. Indeed, Epo provides cytoprotective effects, which consist in prevention or fight against pathological processes. This perspective article reviews the various protective effects of Epo in several organs and tries to give a proof of concept about its effects in the lung. The tissue-protective effects of Epo could be a promising approach to limit the symptoms of acute and chronic lung diseases.
Collapse
|
6
|
Zouein FA, Booz GW, Altara R. STAT3 and Endothelial Cell-Cardiomyocyte Dialog in Cardiac Remodeling. Front Cardiovasc Med 2019; 6:50. [PMID: 31069236 PMCID: PMC6491672 DOI: 10.3389/fcvm.2019.00050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/08/2019] [Indexed: 12/18/2022] Open
Abstract
This article presents an overview of the central role of STAT3 in the crosstalk between endothelial cells and cardiac myocytes in the heart. Endothelial cell STAT3 has a key role in inflammation that underlies cardiovascular disease and impacts on cardiac structure and function. STAT3 in endothelial cells contributes to adverse cardiomyocyte genetic reprograming, for instance, during peripartum cardiomyopathy. Conversely, cardiomyocyte STAT3 is important for maintaining endothelial cell function and capillary integrity with aging and hypertension. In addition, STAT3 serves as a sentinel for stress in the heart. Recent evidence has revealed that the redox nature of STAT3 is regulated, and STAT3 is responsive to oxidative stress (ischemia-reperfusion) so as to induce protective genes. At the level of the mitochondrion, STAT3 is important in regulating reactive oxygen species (ROS) formation, metabolism, and mitochondrial integrity. STAT3 may also control calcium release from the ER so as to limit its subsequent uptake by mitochondria and the induction of cell death. Under normal conditions, some STAT3 localizes to intercalated discs of cardiomyocytes and serves to transmit pro-fibrotic gene induction signals in the nucleus with increased blood pressure. Further research is needed to understand how the sentinel role of STAT3 in both endothelial cells and cardiomyocytes is integrated in order to coordinate the response of the heart to both physiological and pathological demands.
Collapse
Affiliation(s)
- Fouad A Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Raffaele Altara
- Department of Pathology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States.,Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, Oslo, Norway
| |
Collapse
|
7
|
Zhang J, Zhao D, Na N, Li H, Miao B, Hong L, Huang Z. Renoprotective effect of erythropoietin via modulation of the STAT6/MAPK/NF-κB pathway in ischemia/reperfusion injury after renal transplantation. Int J Mol Med 2017; 41:25-32. [PMID: 29115389 PMCID: PMC5746301 DOI: 10.3892/ijmm.2017.3204] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/02/2017] [Indexed: 12/23/2022] Open
Abstract
Ischemia/reperfusion injury (IRI) commonly occurs in renal transplantation. Erythropoietin (EPO) exerts a protective effect in IRI. To investigate the underlying molecular mechanism, rat models of renal IRI were established and treated with EPO and/or lentivirus-mediated EPO-siRNA, the signal transducer and activator of transcription 6 (STAT6) inhibitor AS1517499, the JNK inhibitor SP600125, the p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580, and the nuclear factor (NF)-κB inhibitor lactacystin. Histological examination revealed that EPO protected the kidney from IRI, through decreasing the extent of tissue congestion and inflammatory cell infiltration; however, EPO siRNA did not exert the same protective effect. In addition, the EPO level was inversely associated with renal IRI. EPO downregulated the expression of interferon-γ, interleukin (IL)-4, creatinine and caspase-3, and upregulated the expression of IL-10, thymic stromal lymphopoietin, STAT6, p-JNK and p-p38, while the opposite effects were observed with the administration of EPO-siRNA and the specific respective inhibitors. Further results revealed that MAPK (p-JNK and p-p38) acted upstream of NF-κB, and that NF-κB signaling regulated the expression of caspase-1 and -3, which may be responsible for the cytotoxicity associated with IRI. Taken together, the results of the present study demonstrated that EPO exerted a protective effect in renal IRI via the STAT6/MAPK/NF-κB pathway. This protective effect of EPO may improve reperfusion tolerance in ischemic kidneys and benefit transplant recipients.
Collapse
Affiliation(s)
- Jinhua Zhang
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Daqiang Zhao
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Ning Na
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Heng Li
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Bin Miao
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Liangqing Hong
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Zhengyu Huang
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
8
|
Yang Y, Hu W, Di S, Ma Z, Fan C, Wang D, Jiang S, Li Y, Zhou Q, Li T, Luo E. Tackling myocardial ischemic injury: the signal transducer and activator of transcription 3 (STAT3) at a good site. Expert Opin Ther Targets 2016; 21:215-228. [PMID: 28001439 DOI: 10.1080/14728222.2017.1275566] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yang Yang
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi’an, China
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi’an, China
| | - Shouyin Di
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Chongxi Fan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, Xi’an, China
| | - Yue Li
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi’an, China
| | - Qing Zhou
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi’an, China
| | - Erping Luo
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
9
|
Serum Compounds of Energy Metabolism Impairment Are Related to Disability, Disease Course and Neuroimaging in Multiple Sclerosis. Mol Neurobiol 2016; 54:7520-7533. [DOI: 10.1007/s12035-016-0257-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 10/24/2016] [Indexed: 12/15/2022]
|
10
|
Kumarasinghe G, Gao L, Hicks M, Villanueva J, Doyle A, Rao P, Ru Qiu M, Jabbour A, Iyer A, Chew HC, Hayward CS, Macdonald P. Improved heart function from older donors using pharmacologic conditioning strategies. J Heart Lung Transplant 2016; 35:636-46. [DOI: 10.1016/j.healun.2015.12.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 10/22/2015] [Accepted: 12/21/2015] [Indexed: 10/22/2022] Open
|
11
|
Lin CS, Lee SH, Huang HS, Chen YS, Ma MC. H2O2 generated by NADPH oxidase 4 contributes to transient receptor potential vanilloid 1 channel-mediated mechanosensation in the rat kidney. Am J Physiol Renal Physiol 2015; 309:F369-76. [PMID: 26136558 DOI: 10.1152/ajprenal.00462.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 06/17/2015] [Indexed: 11/22/2022] Open
Abstract
The presence of NADPH oxidase (Nox) in the kidney, especially Nox4, results in H2O2 production, which regulates Na(+) excretion and urine formation. Redox-sensitive transient receptor potential vanilloid 1 channels (TRPV1s) are distributed in mechanosensory fibers of the renal pelvis and monitor changes in intrapelvic pressure (IPP) during urine formation. The present study tested whether H2O2 derived from Nox4 affects TRPV1 function in renal sensory responses. Perfusion of H2O2 into the renal pelvis dose dependently increased afferent renal nerve activity and substance P (SP) release. These responses were attenuated by cotreatment with catalase or TRPV1 blockers. In single unit recordings, H2O2 activated afferent renal nerve activity in response to rising IPP but not high salt. Western blots revealed that Nox2 (gp91(phox)) and Nox4 are both present in the rat kidney, but Nox4 is abundant in the renal pelvis and originates from dorsal root ganglia. This distribution was associated with expression of the Nox4 regulators p22(phox) and polymerase δ-interacting protein 2. Coimmunoprecipitation experiments showed that IPP increases polymerase δ-interacting protein 2 association with Nox4 or p22(phox) in the renal pelvis. Interestingly, immunofluorescence labeling demonstrated that Nox4 colocalizes with TRPV1 in sensory fibers of the renal pelvis, indicating that H2O2 generated from Nox4 may affect TRPV1 activity. Stepwise increases in IPP and saline loading resulted in H2O2 and SP release, sensory activation, diuresis, and natriuresis. These effects, however, were remarkably attenuated by Nox inhibition. Overall, these results suggest that Nox4-positive fibers liberate H2O2 after mechanostimulation, thereby contributing to a renal sensory nerve-mediated diuretic/natriuretic response.
Collapse
Affiliation(s)
- Chian-Shiung Lin
- Department of Surgery, Liou-Ying Hospital, Chi-Mei Medical Center, Tainan Hsien, Taiwan
| | - Shang-Hsing Lee
- Department of Urology, Cardinal Tien Hospital, New Taipei, Taiwan
| | - Ho-Shiang Huang
- Department of Urology, National Cheng Kung University, Tainan, Taiwan
| | - Yih-Sharng Chen
- Department of Cardiovascular Surgery, National Taiwan University Hospital, Taipei, Taiwan; and
| | - Ming-Chieh Ma
- School of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan
| |
Collapse
|
12
|
Rong R, Xijun X. Erythropoietin pretreatment suppresses inflammation by activating the PI3K/Akt signaling pathway in myocardial ischemia-reperfusion injury. Exp Ther Med 2015; 10:413-418. [PMID: 26622330 DOI: 10.3892/etm.2015.2534] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 03/24/2015] [Indexed: 02/05/2023] Open
Abstract
Erythropoietin (EPO), a glycoprotein originally known for its important role in the stimulation of erythropoiesis, has recently been shown to have significant protective effects in animal models of kidney and intestinal ischemia-reperfusion injury (IRI). However, the mechanism underlying these protective effects remains unclear. The aim of the current study was to evaluate the effects of EPO on myocardial IRI and to investigate the mechanism underlying these effects. A total of 18 male Sprague Dawley rats were randomly divided into three groups, namely the sham, IRI-saline and IRI-EPO groups. Rats in the IRI-EPO group were administered 5,000 U/kg EPO intraperitoneally 24 h prior to the induction of IRI. IRI was induced by ligating the left descending coronary artery for 30 min, followed by reperfusion for 3 h. Pathological changes in the myocardial tissue were observed and scored. The levels of the proinflammatory cytokines, interleukin (IL)-6, IL-1β and tumor necrosis factor (TNF)-α, were evaluated in the serum and myocardial tissue. Furthermore, the effects of EPO on phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) signaling and EPO receptor (EPOR) phosphorylation were measured. Pathological changes in the myocardial tissue, increased expression levels of TNF-α, IL-6 and IL-1β in the myocardium, and increased serum levels of these mediators, as a result of IRI, were significantly decreased by EPO pretreatment. The effects of EPO were found to be associated with the activation of PI3K/Akt signaling, which suppressed the inflammatory responses, following the initiation of EPOR activation by EPO. Therefore, EPO pretreatment was demonstrated to decrease myocardial IRI, which was associated with activation of EPOR, subsequently increasing PI3K/Akt signaling to inhibit the production and release of inflammatory mediators. Thus, the results of the present study indicated that EPO may be useful for preventing myocardial IRI.
Collapse
Affiliation(s)
- Ren Rong
- Department of Cardiac Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiao Xijun
- Department of Cardiac Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
13
|
Schlüter KD, Schulz R, Schreckenberg R. Arginase induction and activation during ischemia and reperfusion and functional consequences for the heart. Front Physiol 2015. [DOI: 10.3389/fphys.2015.00054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
14
|
Schlüter KD, Schulz R, Schreckenberg R. Arginase induction and activation during ischemia and reperfusion and functional consequences for the heart. Front Physiol 2015; 6:65. [PMID: 25814956 PMCID: PMC4356066 DOI: 10.3389/fphys.2015.00065] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 02/07/2015] [Indexed: 12/17/2022] Open
Abstract
Induction and activation of arginase is among the fastest responses of the heart to ischemic events. Induction of arginase expression and enzyme activation under ischemic conditions shifts arginine consumption from nitric oxide formation (NO) to the formation of ornithine and urea. In the heart such a switch in substrate utilization reduces the impact of the NO/cGMP-pathway on cardiac function that requires intact electromechanical coupling but at the same time it induces ornithine-dependent pathways such as the polyamine metabolism. Both effects significantly reduce the recovery of heart function during reperfusion and thereby limits the success of reperfusion strategies. In this context, changes in arginine consumption trigger cardiac remodeling in an unfavorable way and increases the risk of arrhythmia, specifically in the initial post-ischemic period in which arginase activity is dominating. However, during the entire ischemic period arginase activation might be a meaningful adaptation that is specifically relevant for reperfusion following prolonged ischemic periods. Therefore, a precise understanding about the underlying mechanism that leads to arginase induction as well as of it's mechanistic impact on post-ischemic hearts is required for optimizing reperfusion strategies. In this review we will summarize our current understanding of these processes and give an outlook about possible treatment options for the future.
Collapse
Affiliation(s)
| | - Rainer Schulz
- Physiologisches Institut, Justus-Liebig-Univiersität Giessen Giessen, Germany
| | - Rolf Schreckenberg
- Physiologisches Institut, Justus-Liebig-Univiersität Giessen Giessen, Germany
| |
Collapse
|
15
|
Bond WS, Rex TS. Evidence That Erythropoietin Modulates Neuroinflammation through Differential Action on Neurons, Astrocytes, and Microglia. Front Immunol 2014; 5:523. [PMID: 25374571 PMCID: PMC4205853 DOI: 10.3389/fimmu.2014.00523] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 10/06/2014] [Indexed: 12/15/2022] Open
Abstract
Neuroinflammation is a normal and healthy response to neuronal damage. However, excessive or chronic neuroinflammation exacerbates neurodegeneration after trauma and in progressive diseases such as Alzheimer’s, Parkinson’s, age-related macular degeneration, and glaucoma. Therefore, molecules that modulate neuroinflammation are candidates as neuroprotective agents. Erythropoietin (EPO) is a known neuroprotective agent that indirectly attenuates neuroinflammation, in part, by inhibiting neuronal apoptosis. In this review, we provide evidence that EPO also modulates neuroinflammation upstream of apoptosis by acting directly on glia. Further, the signaling induced by EPO may differ depending on cell type and context possibly as a result of activation of different receptors. While significant progress has been made in our understanding of EPO signaling, this review also identifies areas for future study in terms of the role of EPO in modulating neuroinflammation.
Collapse
Affiliation(s)
- Wesley S Bond
- Vanderbilt Eye Institute, Vanderbilt University Medical Center , Nashville, TN , USA ; Vanderbilt Brain Institute, Vanderbilt University Medical Center , Nashville, TN , USA
| | - Tonia S Rex
- Vanderbilt Eye Institute, Vanderbilt University Medical Center , Nashville, TN , USA ; Vanderbilt Brain Institute, Vanderbilt University Medical Center , Nashville, TN , USA
| |
Collapse
|
16
|
Hypoxic preconditioning protects rat hearts against ischemia–reperfusion injury via the arachidonate12-lipoxygenase/transient receptor potential vanilloid 1 pathway. Basic Res Cardiol 2014; 109:414. [DOI: 10.1007/s00395-014-0414-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 04/22/2014] [Accepted: 05/02/2014] [Indexed: 02/07/2023]
|
17
|
Gobe GC, Morais C, Vesey DA, Johnson DW. Use of high-dose erythropoietin for repair after injury: A comparison of outcomes in heart and kidney. J Nephropathol 2013; 2:154-65. [PMID: 24475445 DOI: 10.12860/jnp.2013.27] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 12/25/2012] [Indexed: 11/20/2022] Open
Abstract
CONTEXT There is a need to define the exact benefits and contraindications of use of high-dose recombinant human erythropoietin (EPO) for its non-hematopoietic function as a cytokine that enhances tissue repair after injury. This review compares the outcomes from use of EPO in the injured heart and kidney, two organs that are thought, traditionally, to have intrinsically-different repair mechanisms. EVIDENCE ACQUISITIONS Directory of Open Access Journals (DOAJ), Google Scholar, Pubmed (NLM), LISTA (EBSCO) and Web of Science have been searched. RESULTS Ongoing work by us on EPO protection of ischemia-reperfusion-injured kidneys indicated, first, that EPO acutely enhanced kidney repair via anti-apoptotic, pro-regenerative mechanisms, and second, that EPO may promote chronic fibrosis in the long term. Work by others on the ischaemia-injured heart has also indicated that EPO promotes repair. Although myocardial infarcts are made up mostly of necrotic tissue, many publications state EPO is anti-apoptotic in the heart, as well as promoting healing via cell differentiation and stimulation of granulation tissue. In the case of the heart, promotion of fibrosis may be advantageous where an infarct has destroyed a zone of cardiomyocytes, but if EPO stimulates progressive fibrosis in the heart, this may promote cardiac failure. CONCLUSIONS A major concern in relation to the use of EPO in a cytoprotective role is its stimulation of long-term inflammation and fibrosis. EPO usage for cytoprotection is undoubtedly advantageous, but it may need to be offset with an anti-inflammatory agent in some organs, like kidney and heart, where progression to chronic fibrosis after acute injury is often recorded.
Collapse
Affiliation(s)
- Glenda C Gobe
- Centre for Kidney Disease Research and ; Discipline of Medicine, School of Medicine. The University of Queensland, Brisbane, Australia
| | - Christudas Morais
- Centre for Kidney Disease Research and ; Discipline of Medicine, School of Medicine. The University of Queensland, Brisbane, Australia
| | - David A Vesey
- Centre for Kidney Disease Research and ; Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
| | - David W Johnson
- Centre for Kidney Disease Research and ; Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
| |
Collapse
|
18
|
Watson AJ, Gao L, Sun L, Tsun J, Doyle A, Faddy SC, Jabbour A, Orr Y, Dhital K, Hicks M, Jansz PC, Macdonald PS. Enhanced preservation of pig cardiac allografts by combining erythropoietin with glyceryl trinitrate and zoniporide. Am J Transplant 2013; 13:1676-87. [PMID: 23668842 DOI: 10.1111/ajt.12249] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 02/25/2013] [Accepted: 03/14/2013] [Indexed: 01/25/2023]
Abstract
Erythropoietin has a tissue-protective effect independent of its erythropoietic effect that may be enhanced by combining it with the nitric oxide donor glyceryl trinitrate (GTN) and the sodium-hydrogen exchange inhibitor zoniporide in rat hearts stored with an extracellular-based preservation solution (EBPS). We thus sought to test this combination of agents in a porcine model of orthotopic heart transplantation incorporating donor brain death and total ischaemic time of approximately 260 min. Pig hearts were stored in one of four storage solutions: unmodified EBPS (CON), EBPS supplemented with GTN and zoniporide (GZ), EBPS supplemented with erythropoietin and zoniporide (EZ), or EBPS supplemented with all three agents (EGZ). A total of 4/5 EGZ hearts were successfully weaned from cardiopulmonary bypass compared with only 2/5 GZ hearts, 0/5 CON hearts and 0/5 EG hearts (p = 0.017). Following weaning from bypass EGZ hearts demonstrated superior contractility and haemodynamics than GZ hearts. All weaned hearts displayed impaired diastolic function. Release of troponin I from EGZ hearts was lower than all other groups. In conclusion, supplementation of EBPS with erythropoietin, glyceryl trinitrate and zoniporide provided superior donor heart preservation than all other strategies tested.
Collapse
Affiliation(s)
- A J Watson
- Transplant Program, The Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Chong ZZ, Shang YC, Wang S, Maiese K. A Critical Kinase Cascade in Neurological Disorders: PI 3-K, Akt, and mTOR. FUTURE NEUROLOGY 2012; 7:733-748. [PMID: 23144589 DOI: 10.2217/fnl.12.72] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neurodegenerative disorders lead to disability and death in a significant proportion of the world's population. However, many disorders of the nervous system remain with limited effective treatments. Kinase pathways in the nervous system that involve phosphoinositide 3-kinase (PI 3-K), protein kinase B (Akt), and the mammalian target of rapamycin (mTOR) offer exciting prospects for the understanding of neurodegenerative pathways and the development of new avenues of treatment. PI 3-K, Akt, and mTOR pathways are vital cellular components that determine cell fate during acute and chronic disorders, such as Huntington's disease, Alzheimer's disease, Parkinson's disease, epilepsy, stroke, and trauma. Yet, the elaborate relationship among these kinases and the variable control of apoptosis and autophagy can lead to unanticipated biological and clinical outcomes. Crucial for the successful translation of PI 3-K, Akt, and mTOR into robust and safe clinical strategies will be the further elucidation of the complex roles that these kinase pathways hold in the nervous system.
Collapse
Affiliation(s)
- Zhao Zhong Chong
- Laboratory of Cellular and Molecular Signaling, Newark, New Jersey 07101 ; New Jersey Health Sciences University, Newark, New Jersey 07101
| | | | | | | |
Collapse
|
20
|
Maiese K, Chong ZZ, Wang S, Shang YC. Oxidant stress and signal transduction in the nervous system with the PI 3-K, Akt, and mTOR cascade. Int J Mol Sci 2012. [PMID: 23203037 PMCID: PMC3509553 DOI: 10.3390/ijms131113830] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress impacts multiple systems of the body and can lead to some of the most devastating consequences in the nervous system especially during aging. Both acute and chronic neurodegenerative disorders such as diabetes mellitus, cerebral ischemia, trauma, Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and tuberous sclerosis through programmed cell death pathways of apoptosis and autophagy can be the result of oxidant stress. Novel therapeutic avenues that focus upon the phosphoinositide 3-kinase (PI 3-K), Akt (protein kinase B), and the mammalian target of rapamycin (mTOR) cascade and related pathways offer exciting prospects to address the onset and potential reversal of neurodegenerative disorders. Effective clinical translation of these pathways into robust therapeutic strategies requires intimate knowledge of the complexity of these pathways and the ability of this cascade to influence biological outcome that can vary among disorders of the nervous system.
Collapse
Affiliation(s)
- Kenneth Maiese
- Laboratory of Cellular and Molecular Signaling, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (S.W.); (Y.C.S.)
- Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
- New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA
- Author to whom correspondence should be addressed: E-Mail:
| | - Zhao Zhong Chong
- Laboratory of Cellular and Molecular Signaling, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (S.W.); (Y.C.S.)
- New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA
| | - Shaohui Wang
- Laboratory of Cellular and Molecular Signaling, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (S.W.); (Y.C.S.)
- New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA
| | - Yan Chen Shang
- Laboratory of Cellular and Molecular Signaling, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (S.W.); (Y.C.S.)
- New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA
| |
Collapse
|
21
|
Maiese K, Chong ZZ, Shang YC, Wang S. Erythropoietin: new directions for the nervous system. Int J Mol Sci 2012; 13:11102-11129. [PMID: 23109841 PMCID: PMC3472733 DOI: 10.3390/ijms130911102] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/16/2012] [Accepted: 08/30/2012] [Indexed: 12/14/2022] Open
Abstract
New treatment strategies with erythropoietin (EPO) offer exciting opportunities to prevent the onset and progression of neurodegenerative disorders that currently lack effective therapy and can progress to devastating disability in patients. EPO and its receptor are present in multiple systems of the body and can impact disease progression in the nervous, vascular, and immune systems that ultimately affect disorders such as Alzheimer's disease, Parkinson's disease, retinal injury, stroke, and demyelinating disease. EPO relies upon wingless signaling with Wnt1 and an intimate relationship with the pathways of phosphoinositide 3-kinase (PI 3-K), protein kinase B (Akt), and mammalian target of rapamycin (mTOR). Modulation of these pathways by EPO can govern the apoptotic cascade to control β-catenin, glycogen synthase kinase-3β, mitochondrial permeability, cytochrome c release, and caspase activation. Yet, EPO and each of these downstream pathways require precise biological modulation to avert complications associated with the vascular system, tumorigenesis, and progression of nervous system disorders. Further understanding of the intimate and complex relationship of EPO and the signaling pathways of Wnt, PI 3-K, Akt, and mTOR are critical for the effective clinical translation of these cell pathways into robust treatments for neurodegenerative disorders.
Collapse
Affiliation(s)
- Kenneth Maiese
- Laboratory of Cellular and Molecular Signaling, Cancer Center, F 1220, New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (Y.C.S.); (S.W.)
- Cancer Institute of New Jersey, New Brunswick, New Jersey 08901, USA
- New Jersey Health Sciences University, Newark, New Jersey 07101, USA
| | - Zhao Zhong Chong
- Laboratory of Cellular and Molecular Signaling, Cancer Center, F 1220, New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (Y.C.S.); (S.W.)
- New Jersey Health Sciences University, Newark, New Jersey 07101, USA
| | - Yan Chen Shang
- Laboratory of Cellular and Molecular Signaling, Cancer Center, F 1220, New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (Y.C.S.); (S.W.)
- New Jersey Health Sciences University, Newark, New Jersey 07101, USA
| | - Shaohui Wang
- Laboratory of Cellular and Molecular Signaling, Cancer Center, F 1220, New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (Y.C.S.); (S.W.)
- New Jersey Health Sciences University, Newark, New Jersey 07101, USA
| |
Collapse
|