1
|
Hussain MA, Colicchia M, Veerapen J, Weeraman D, Podaru MN, Jones D, Suzuki K, Mathur A. Circulatory support and stem cell therapy in the management of advanced heart failure: a concise review of available evidence. Regen Med 2019; 14:585-593. [PMID: 31115248 DOI: 10.2217/rme-2018-0121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Stem cell therapy utilizing bone marrow mononuclear cells (BMC's) is a potential strategy to treat heart failure patients with improvement in symptom profile and cardiac function. We describe a rationale for concurrent BMC and left ventricular assist device therapy in selected heart failure patients. This combination therapy has demonstrated improved myocardial perfusion and cardiac function in patients with advanced ischemic cardiomyopathy. Moreover, preclinical data support improved cell retention with left ventricular unloading. The beneficial effects of BMC's are likely through a paracrine mechanism initiating a 'cardiac-repair' process. Combination therapy of BMC's and a left ventricular assist device may exhibit a synergistic effect with improved engraftment of BMC's through left ventricular unloading.
Collapse
Affiliation(s)
- Mohsin A Hussain
- William Harvey Research Institute, Queen Mary University of London, St Bartholomew's Hospital NHS Foundation Trust, London, UK
| | - Martina Colicchia
- William Harvey Research Institute, Queen Mary University of London, St Bartholomew's Hospital NHS Foundation Trust, London, UK
| | - Jessry Veerapen
- William Harvey Research Institute, Queen Mary University of London, St Bartholomew's Hospital NHS Foundation Trust, London, UK
| | - Deshan Weeraman
- William Harvey Research Institute, Queen Mary University of London, St Bartholomew's Hospital NHS Foundation Trust, London, UK
| | - Mihai-Nicolae Podaru
- William Harvey Research Institute, Queen Mary University of London, St Bartholomew's Hospital NHS Foundation Trust, London, UK
| | - Daniel Jones
- William Harvey Research Institute, Queen Mary University of London, St Bartholomew's Hospital NHS Foundation Trust, London, UK
| | - Ken Suzuki
- William Harvey Research Institute, Queen Mary University of London, St Bartholomew's Hospital NHS Foundation Trust, London, UK
| | - Anthony Mathur
- William Harvey Research Institute, Queen Mary University of London, St Bartholomew's Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
2
|
Umemura Y, Ogura H, Matsuura H, Ebihara T, Shimizu K, Shimazu T. Bone marrow-derived mononuclear cell therapy can attenuate systemic inflammation in rat heatstroke. Scand J Trauma Resusc Emerg Med 2018; 26:97. [PMID: 30445981 PMCID: PMC6240199 DOI: 10.1186/s13049-018-0566-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/02/2018] [Indexed: 12/30/2022] Open
Abstract
Background This study was performed to gain insights into novel therapeutic approaches for acute systemic inflammation in heatstroke. Bone marrow-derived mononuclear cells (BMMNCs) secrete anti-inflammatory proteins and have protective effects against acute inflammation. Recent evidence suggested that transplantation of BMMNCs can reduce the acute tissue injury caused by regional myocardial reperfusion and the lung dysfunction induced by lipopolysaccharides. We evaluated whether BMMNCs attenuate systemic inflammatory response induced by severe heatstroke. Material and methods Anesthetized 12-week-old male Wistar rats were subjected to heat stress (41.8 °C for 30 min) with/without transplantation of BMMNCs. Bone marrow cells were harvested from the femur and tibia of other Wistar rats. BMMNCs were separated by density centrifugation, dissolved in phosphate-buffered saline (PBS), and injected intravenously immediately after heat stress (HS-BMMNCs group). The control group was administered an equal volume of PBS, and the sham group underwent the same procedure without heat stress. Results Seven-day survival improved significantly in the HS-BMMNCs group versus control group (83.3% vs 41.7%). Transplantation of BMMNCs significantly suppressed serum levels of pro-inflammatory mediators, such as tumor necrosis factor-alpha, interleukin-6 and histone H3 at 3, 6, and 12 h after heat stress. Besides, the elevation of serum syndecan-1, a main component of the vascular endothelial glycocalyx layer, in the BMMNCs group was significantly suppressed compared to that in the control group at 6 and 12 h after heat stress. Histological analysis revealed that edema of the alveolar septum and vascular endothelial injury in the lung were evident in the control group 6 h after heat stress, whereas the morphological alteration was ameliorated in the HS-BMMNCs group. Also, histological analysis using BMMNCs derived from green fluorescent protein transgenic rats showed that the transplanted BMMNCs migrated into lung, kidney, and spleen at 24 h after heat stress but did not engraft to host tissues. Conclusion Transplantation of BMMNCs attenuated acute systemic inflammation and vascular endothelial injury, reduced organ dysfunction, and improved survival in a rat heatstroke model. These findings provide a possible therapeutic strategy against critical heatstroke.
Collapse
Affiliation(s)
- Yutaka Umemura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroshi Matsuura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takeshi Ebihara
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kentaro Shimizu
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takeshi Shimazu
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
3
|
Pi Z, Lin H, Yang J. Isoflurane reduces pain and inhibits apoptosis of myocardial cells through the phosphoinositide 3-kinase/protein kinase B signaling pathway in mice during cardiac surgery. Mol Med Rep 2018; 17:6497-6505. [PMID: 29488606 PMCID: PMC5928630 DOI: 10.3892/mmr.2018.8642] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/28/2017] [Indexed: 01/27/2023] Open
Abstract
Heart bypass surgery is the most common treatment for myocardial ischemia. Clinical investigations have revealed that isoflurane anesthesia is efficient to alleviate pain during cardiac surgery, including heart bypass surgery. Previous studies have revealed the protective effects of isoflurane on myocardial cells of patients with myocardial ischemia during the perioperative period. The present study aimed to investigate the mechanism underlying the protective effects of isoflurane on myocardial cells in mice with myocardial ischemia. ELISA, flow cytometry, immunofluorescence and western blotting were used to analyze the effects of isoflurane anesthesia on myocardial cells. Briefly, myocardial cell apoptosis and viability, pain, phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) signaling pathway expression and the pharmacodynamics of isoflurane were studied in mice treated with isoflurane for heart bypass surgery. The results demonstrated that isoflurane anesthesia efficiently attenuated pain in mice during surgery. Viability and apoptosis of myocardial cells was also improved by isoflurane in vitro and in vivo. The PI3K/AKT pathway was upregulated in myocardial cells on day 3 post-operation. Mechanistically, isoflurane promoted PI3K/AKT activation, upregulated B-cell lymphoma 2 (Bcl-2)-associated X protein and Bcl-2 expression levels, and reduced the expression levels of caspase-3 and caspase-8 in myocardial cells. In conclusion, the findings indicated that isoflurane is beneficial for pain attenuation and inhibits apoptosis of myocardial cells via the PI3K/AKT signaling pathway in mice during cardiac surgery.
Collapse
Affiliation(s)
- Zhibing Pi
- Department of Anesthesiology of The First Affiliated Hospital of Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Hai Lin
- Department of Anesthesiology of The First Affiliated Hospital of Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jianping Yang
- Department of Anesthesiology of The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
4
|
Wang L, Ma H, Xue Y, Shi H, Ma T, Cui X. Berberine inhibits the ischemia-reperfusion injury induced inflammatory response and apoptosis of myocardial cells through the phosphoinositide 3-kinase/RAC-α serine/threonine-protein kinase and nuclear factor-κB signaling pathways. Exp Ther Med 2017; 15:1225-1232. [PMID: 29403554 PMCID: PMC5780743 DOI: 10.3892/etm.2017.5575] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 07/14/2017] [Indexed: 12/24/2022] Open
Abstract
Myocardial ischemia-reperfusion injury is one of the most common cardiovascular diseases, and can lead to serious damage and dysfunction of the myocardial tissue. Previous studies have demonstrated that berberine exhibits ameliorative effects on cardiovascular disease. The present study further investigated the efficacy and potential mechanism underlying the effects of berberine on ischemia-reperfusion injury in a mouse model. Inflammatory markers were measured in the serum and levels of inflammatory proteins in myocardial cells were investigated after treatment with berberine. In addition, the apoptosis of myocardial cells was investigated after berberine treatment. Apoptosis-associated gene expression levels and apoptotic signaling pathways were analyzed in myocardial cells after treatment with berberine. The phosphoinositide 3-kinase (PI3K)/RAC-α serine/threonine-protein kinase (AKT) and nuclear factor (NF)-κB signaling pathways were also analyzed in myocardial cells after treatment with berberine. Histological analysis was used to analyze the potential benefits of berberine in ischemia-reperfusion injury. The present study identified that inflammatory responses and inflammatory factors were decreased in the myocardial cells of the mouse model of ischemia-reperfusion injury. Mechanism analysis demonstrated that berberine inhibited apoptotic protease-activating factor 1, caspase-3 and caspase-9 expression in myocardial cells. The expression of Bcl2-associated agonist of cell death, Bcl-2-like protein 1 and cellular tumor antigen p53 was upregulated. Expression of NF-κB p65, inhibitor of NF-κB kinase subunit β (IKK-β), NF-κB inhibitor α (IκBα), and NF-κB activity, were inhibited in myocardial cells in the mouse model of ischemia-reperfusion injury. In conclusion, the results of the present study indicate that berberine inhibits inflammatory responses through the NF-κB signaling pathway and suppresses the apoptosis of myocardial cells via the PI3K/AKT signaling pathway in a mouse model of ischemia-reperfusion injury. These results suggest that berberine is a potential drug for the treatment of patients with ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Lixin Wang
- Department of Cardiovascular Surgery, The General Hospital of The Chinese People's Armed Police Forces, Beijing 100039, P.R. China
| | - Hao Ma
- Department of Cardiovascular Surgery, The General Hospital of The Chinese People's Armed Police Forces, Beijing 100039, P.R. China
| | - Yan Xue
- Department of Cardiovascular Surgery, The General Hospital of The Chinese People's Armed Police Forces, Beijing 100039, P.R. China
| | - Haiyan Shi
- Department of Cardiovascular Surgery, The General Hospital of The Chinese People's Armed Police Forces, Beijing 100039, P.R. China
| | - Teng Ma
- Department of Cardiovascular Surgery, The General Hospital of The Chinese People's Armed Police Forces, Beijing 100039, P.R. China
| | - Xiaozheng Cui
- Department of Cardiovascular Surgery, The General Hospital of The Chinese People's Armed Police Forces, Beijing 100039, P.R. China
| |
Collapse
|
5
|
Jeyaraman MM, Rabbani R, Copstein L, Sulaiman W, Farshidfar F, Kashani HH, Qadar SMZ, Guan Q, Skidmore B, Kardami E, Ducas J, Mansour S, Zarychanski R, Abou-Setta AM. Autologous Bone Marrow Stem Cell Therapy in Patients With ST-Elevation Myocardial Infarction: A Systematic Review and Meta-analysis. Can J Cardiol 2017; 33:1611-1623. [PMID: 29173601 DOI: 10.1016/j.cjca.2017.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/02/2017] [Accepted: 10/02/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Randomized controlled trials (RCTs) on bone marrow stem cell (BMSC) therapy in ST-elevation myocardial infarction (STEMI) patients have reported conflicting results. Our main objective was to critically appraise and meta-analyze best-available evidence on efficacy and safety of intracoronary administration of autologous BMSC therapy in STEMI patients after primary percutaneous coronary intervention. METHODS We conducted a search of MEDLINE, PubMed, EMBASE, CENTRAL, Global Health, CINAHL, and conference proceedings in February 2017. Our primary outcome was all-cause mortality. Secondary and safety outcomes included cardiac death, heart failure, arrhythmias, repeat myocardial infarction, or target vessel revascularizations; or improved health-related quality of life, left ventricular ejection fraction, or infarct size. Summary relative and absolute risks were obtained using random effects models. We also evaluated the strength of evidence. RESULTS A comprehensive database search identified 42 RCTs (3365 STEMI patients). BMSC therapy did not significantly decrease mortality (risk ratio, 0.71; 95% confidence interval, 0.45-1.11; I2, 0%; absolute risk reduction, 0.1%; 95% confidence interval, -0.71 to 0.91; 40 trials; 3289 participants; I2, 0%; low strength of evidence). BMSC therapy had no effect on secondary or adverse outcomes. Trial sequential analysis for all-cause mortality showed no evidence of a clinically important difference, with a very low probability that future studies can change the current conclusion. CONCLUSIONS On the basis of evidence from 42 RCTs published in the past 15 years, we provide conclusive evidence for a lack of beneficial effect for autologous BMSC therapy in patients with STEMI.
Collapse
Affiliation(s)
- Maya M Jeyaraman
- The George and Fay Yee Center for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Rasheda Rabbani
- The George and Fay Yee Center for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Leslie Copstein
- The George and Fay Yee Center for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Wasan Sulaiman
- The George and Fay Yee Center for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Farnaz Farshidfar
- The George and Fay Yee Center for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hessam H Kashani
- The George and Fay Yee Center for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sheikh M Z Qadar
- The George and Fay Yee Center for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Qingdong Guan
- Cellular Therapy Laboratory, CancerCare Manitoba, Winnipeg, Manitoba, Canada; Manitoba Center for Advanced Cell and Tissue Therapy, Winnipeg, Manitoba, Canada
| | - Becky Skidmore
- Information Specialist Consultant, Ottawa, Ontario, Canada
| | - Elissavet Kardami
- Department of Human Anatomy and Cell Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - John Ducas
- Section of Cardiology, Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Samer Mansour
- Centre Hospitalier de l'Université de Montreal, Montreal, Quebec, Canada; Faculty of Medicine, Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Ryan Zarychanski
- The George and Fay Yee Center for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Haematology and Medical Oncology, Cancer Care Manitoba, Winnipeg, Manitoba, Canada
| | - Ahmed M Abou-Setta
- The George and Fay Yee Center for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
6
|
Feyen DA, Gaetani R, Doevendans PA, Sluijter JP. Stem cell-based therapy: Improving myocardial cell delivery. Adv Drug Deliv Rev 2016; 106:104-115. [PMID: 27133386 DOI: 10.1016/j.addr.2016.04.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/19/2016] [Accepted: 04/20/2016] [Indexed: 12/15/2022]
Abstract
Stem cell-based therapies form an exciting new class of medicine that attempt to provide the body with the building blocks required for the reconstruction of damaged organs. However, delivering cells to the correct location, while preserving their integrity and functional properties, is a complex undertaking. These challenges have led to the development of a highly dynamic interdisciplinary research field, wherein medical, biological, and chemical sciences have collaborated to develop strategies to overcome the physiological barriers imposed on the cellular therapeutics. In this respect, improving the acute retention and subsequent survival of stem cells is key to effectively increase the effect of the therapy, while proper tissue integration is imperative for stem cells to functionally replace lost cells in damaged organs. In this review, we will use the heart as an example to highlight the current knowledge of therapeutic stem cell utilization, the existing pitfalls and limitations, and the approaches that have been developed to overcome them.
Collapse
|
7
|
Nadlacki B, Suuronen EJ. Biomaterial strategies to improve the efficacy of bone marrow cell therapy for myocardial infarction. Expert Opin Biol Ther 2016; 16:1501-1516. [DOI: 10.1080/14712598.2016.1235149] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
8
|
Salidroside Mitigates Sepsis-Induced Myocarditis in Rats by Regulating IGF-1/PI3K/Akt/GSK-3β Signaling. Inflammation 2015; 38:2178-84. [DOI: 10.1007/s10753-015-0200-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
9
|
Choudhury TR, Mathur A. The birth of 'regenerative pharmacology': a clinical perspective. Br J Pharmacol 2014; 169:239-46. [PMID: 23425309 DOI: 10.1111/bph.12128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
10
|
Guo J, Jie W, Shen Z, Li M, Lan Y, Kong Y, Guo S, Li T, Zheng S. SCF increases cardiac stem cell migration through PI3K/AKT and MMP‑2/‑9 signaling. Int J Mol Med 2014; 34:112-8. [PMID: 24804928 PMCID: PMC4072340 DOI: 10.3892/ijmm.2014.1773] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 04/29/2014] [Indexed: 12/15/2022] Open
Abstract
The transplantation of cardiac stem cells (CSCs) is thought to be responsible for improving the performance of injured heart induced by myocardial infarction (MI). However, the mechanisms involved in the migration of activated CSCs post-MI remain to be clarified. In this study, CSCs were isolated from rat hearts and a cellular migration assay was performed using a 24-well Transwell system. Stem cell factor (SCF) induced CSC migration in a concentration-dependent manner, which could be blocked with an SCF antibody as well as a PI3K/AKT inhibitor, LY294002. Moreover, SCF induced the expression and activity of matrix metalloproteinase (MMP)-2 and MMP-9 in a concentration- and time-dependent manner, as measured by quantitative RT-PCR, western blot analysis and gelatin zymography. Results of western blot analysis revealed phosphorylated AKT was markedly increased in SCF-treated CSCs and that inhibition of SCF/c-Kit signaling or phospho-AKT activity significantly attenuated the SCF-induced expression of MMP-2 and MMP-9. Thus, our results showed that SCF partially mediated CSC migration via the activation of PI3K/AKT/MMP-2/-9 signaling.
Collapse
Affiliation(s)
- Junli Guo
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| | - Wei Jie
- Department of Pathology, School of Basic Medicine Science, Guangdong Medical College, Zhanjiang 524023, P.R. China
| | - Zhihua Shen
- Department of Pathology, School of Basic Medicine Science, Guangdong Medical College, Zhanjiang 524023, P.R. China
| | - Mengsen Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, P.R. China
| | - Youling Lan
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| | - Yueqiong Kong
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| | - Shaoli Guo
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| | - Tianfa Li
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| | - Shaojiang Zheng
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| |
Collapse
|
11
|
Bone marrow mesenchymal stem cell-derived microvesicles protect rat pheochromocytoma PC12 cells from glutamate-induced injury via a PI3K/Akt dependent pathway. Neurochem Res 2014; 39:922-31. [PMID: 24706151 DOI: 10.1007/s11064-014-1288-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 03/10/2014] [Accepted: 03/18/2014] [Indexed: 12/28/2022]
Abstract
Studies have suggested that mesenchymal stem cells (MSCs) can protect neuronal cells from excitotoxicity, but the underlying mechanisms are still remaining elusive. In the study, we show that microvesicles released by rat bone marrow-derived MSCs (rBMSC-MVs) protect rat pheochromocytoma PC12 cells from glutamate-induced excitotoxicity. BMSC-MVs upregulate Akt phosphorylation and Bcl-2 expression, downregulate Bax expression, and reduce the cleavage of caspase-3 in glutamate-treated PC12 cells. Such protective effects are partially abrogated by inhibiting PI3K, indicating that rBMSC-MVs act via the PI3K/Akt pathway. Transplantation of rBMSC-MVs may, therefore, be a promising strategy to treat cerebral injury or some other neuronal diseases involving excitotoxicity.
Collapse
|
12
|
Hamshere S, Choudhury T, Jones DA, Locca D, Mills P, Rothman M, Knight C, Parmar M, Agrawal S, Martin J, Mathur A. A randomised double-blind control study of early intracoronary autologous bone marrow cell infusion in acute myocardial infarction (REGENERATE-AMI). BMJ Open 2014; 4:e004258. [PMID: 24549163 PMCID: PMC3931996 DOI: 10.1136/bmjopen-2013-004258] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
INTRODUCTION Acute myocardial infarction (AMI) remains a major cause of mortality and morbidity worldwide despite the latest therapeutic advances designed to decrease myocardial injury. Preclinical and emerging clinical evidence show that the intracoronary injection of autologous bone marrow mononuclear cells (BMCs) following AMI leads to improvement in left ventricular ejection function (LVEF). In this clinical trial we will for the first time assess the effect of early (<24 h) infusion of autologous BMCs following AMI on cardiac function. METHODS AND ANALYSIS REGENERATE-AMI is a double-blind, randomised, multicentre, placebo-controlled trial to determine whether early (<24 h) intracoronary infusion of BMCs improves LVEF after AMI. The study will enrol 100 patients presenting with an anterior AMI demonstrating anterior regional wall motion abnormality. Patients will be randomised to receive intracoronary infusion of BMCs or placebo (0.9% saline). Primary endpoint will be change in LVEF at 1 year compared to baseline, measured by cardiac MRI. Secondary endpoints at 6 months include the change in global LVEF relative to baseline measured by quantitative left ventriculography and echocardiography, as well as major adverse cardiac events which is also measured at 1 year. ETHICS AND DISSEMINATION The study will be performed in agreement with the Declaration of Helsinki and is approved by local ethics committee (NRES Committee London West London: 07/Q0603/76). TRIAL REGISTRATION http://clincialtrials.gov (NCT00765453). The results of the trial will be published according to the CONSORT statement and will be presented at conferences and reported in peer-reviewed journals.
Collapse
Affiliation(s)
- Stephen Hamshere
- Department of Cardiology, London Chest Hospital, Barts Health NHS Trust, London, UK
- Queen Mary University of London, Cardiology, London Chest Hospital, NIHR Cardiovascular BRU, London, UK
| | - Tawfiq Choudhury
- Department of Cardiology, London Chest Hospital, Barts Health NHS Trust, London, UK
- Queen Mary University of London, Cardiology, London Chest Hospital, NIHR Cardiovascular BRU, London, UK
| | - Daniel A Jones
- Department of Cardiology, London Chest Hospital, Barts Health NHS Trust, London, UK
- Queen Mary University of London, Cardiology, London Chest Hospital, NIHR Cardiovascular BRU, London, UK
| | - Didier Locca
- Department of Cardiology, London Chest Hospital, Barts Health NHS Trust, London, UK
| | - Peter Mills
- Department of Cardiology, London Chest Hospital, Barts Health NHS Trust, London, UK
| | - Martin Rothman
- Department of Cardiology, London Chest Hospital, Barts Health NHS Trust, London, UK
| | - Charles Knight
- Department of Cardiology, London Chest Hospital, Barts Health NHS Trust, London, UK
| | | | - Samir Agrawal
- Stem Cell Laboratory, Barts Health NHS Trust and Blizard Institute, Queen Mary University of London, London, UK
| | - John Martin
- British Heart Foundation Laboratories, Department of Medicine, University College London, London, UK
| | - Anthony Mathur
- Department of Cardiology, London Chest Hospital, Barts Health NHS Trust, London, UK
- Queen Mary University of London, Cardiology, London Chest Hospital, NIHR Cardiovascular BRU, London, UK
| |
Collapse
|
13
|
Xu Y, Hu X, Wang L, Jiang Z, Liu X, Yu H, Zhang Z, Chen H, Chen H, Steinhoff G, Li J. Preconditioning via angiotensin type 2 receptor activation improves therapeutic efficacy of bone marrow mononuclear cells for cardiac repair. PLoS One 2013; 8:e82997. [PMID: 24340072 PMCID: PMC3858344 DOI: 10.1371/journal.pone.0082997] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 11/07/2013] [Indexed: 11/29/2022] Open
Abstract
Background The therapeutic efficiency of bone marrow mononuclear cells (BMMNCs) autologous transplantation for myocardial infarction (MI) remains low. Here we developed a novel strategy to improve cardiac repair by preconditioning BMMNCs via angiotensin II type 2 receptor (AT2R) stimulation. Methods and Results Acute MI in rats led to a significant increase of AT2R expression in BMMNCs. Preconditioning of BMMNCs via AT2R stimulation directly with an AT2R agonist CGP42112A or indirectly with angiotensin II plus AT1R antagonist valsartan led to ERK activation and increased eNOS expression as well as subsequent nitric oxide generation, ultimately improved cardiomyocyte protection invitro as measured by co-culture approach. Intramyocardial transplantation of BMMNCs preconditioned via AT2R stimulation improved survival of transplanted cells in ischemic region of heart tissue and reduced cardiomyocyte apoptosis and inflammation at 3 days after MI. At 4 weeks after transplantation, compared to DMEM and non-preconditioned BMMNCs group, AT2R stimulated BMMNCs group showed enhanced vessel density in peri-infarct region and attenuated infarct size, leading to global heart function improvement. Conclusions Preconditioning of BMMNCs via AT2R stimulation exerts protective effect against MI. Stimulation of AT2R in BMMNCs may provide a new strategy to improving therapeutic efficiency of stem cells for post MI cardiac repair.
Collapse
Affiliation(s)
- Yinchuan Xu
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Cardiovascular key lab of Zhejiang Province, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinyang Hu
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Cardiovascular key lab of Zhejiang Province, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lihan Wang
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Cardiovascular key lab of Zhejiang Province, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhi Jiang
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Cardiovascular key lab of Zhejiang Province, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xianbao Liu
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Cardiovascular key lab of Zhejiang Province, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hong Yu
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Cardiovascular key lab of Zhejiang Province, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhaocai Zhang
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Cardiovascular key lab of Zhejiang Province, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Huiqiang Chen
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Cardiovascular key lab of Zhejiang Province, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Han Chen
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Cardiovascular key lab of Zhejiang Province, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Gustav Steinhoff
- Reference and Translation Center for Cardiac Stem Cell Therapy, University of Rostock, Rostock, Germany
| | - Jun Li
- Clinical Stem Cell Research Center and Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (JW); (JL)
| |
Collapse
|
14
|
Hughey CC, James FD, Ma L, Bracy DP, Wang Z, Wasserman DH, Rottman JN, Shearer J. Diminishing impairments in glucose uptake, mitochondrial content, and ADP-stimulated oxygen flux by mesenchymal stem cell therapy in the infarcted heart. Am J Physiol Cell Physiol 2013; 306:C19-27. [PMID: 24196528 DOI: 10.1152/ajpcell.00156.2013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A constant provision of ATP is of necessity for cardiac contraction. As the heart progresses toward failure following a myocardial infarction (MI), it undergoes metabolic alterations that have the potential to compromise the ability to meet energetic demands. This study evaluated the efficacy of mesenchymal stem cell (MSC) transplantation into the infarcted heart to minimize impairments in the metabolic processes that contribute to energy provision. Seven and twenty-eight days following the MI and MSC transplantation, MSC administration minimized cardiac systolic dysfunction. Hyperinsulinemic-euglycemic clamps, coupled with 2-[(14)C]deoxyglucose administration, were employed to assess systemic insulin sensitivity and tissue-specific, insulin-mediated glucose uptake 36 days following the MI in the conscious, unrestrained, C57BL/6 mouse. The improved systolic performance in MSC-treated mice was associated with a preservation of in vivo insulin-stimulated cardiac glucose uptake. Conserved glucose uptake in the heart was linked to the ability of the MSC treatment to diminish the decline in insulin signaling as assessed by Akt phosphorylation. The MSC treatment also sustained mitochondrial content, ADP-stimulated oxygen flux, and mitochondrial oxidative phosphorylation efficiency in the heart. Maintenance of mitochondrial function and density was accompanied by preserved peroxisome proliferator-activated receptor-γ coactivator-1α, a master regulator of mitochondrial biogenesis. These studies provide insight into mechanisms of action that lead to an enhanced energetic state in the infarcted heart following MSC transplantation that may assist in energy provision and dampen cardiac dysfunction.
Collapse
Affiliation(s)
- Curtis C Hughey
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Jiang H, Qu L, Dou R, Lu L, Bian S, Zhu W. Potential role of mesenchymal stem cells in alleviating intestinal ischemia/reperfusion impairment. PLoS One 2013; 8:e74468. [PMID: 24058571 PMCID: PMC3772852 DOI: 10.1371/journal.pone.0074468] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 07/31/2013] [Indexed: 01/01/2023] Open
Abstract
Background Transplantation of bone marrow mesenchymal stem cells (MSCs) provides a promising therapeutic efficiency for a variety of disorders caused by ischemia or reperfusion impairment. We have previously demonstrated the efficacy of MSCs in mitigating intestinal ischemia/reperfusion (I/R) injuries in rats, but the mechanism by which MSCs engraft ameliorates I/R injuries has largely been unknown. The present study aimed at investigating probable mechanisms by which MSCs exert their function. Methods Male donor derived rat MSCs were implanted into intestine of female recipient rat by direct submucosal injection after superior mesenteric artery clamping and unclamping. The homed MSCs were detected by Y chromosome insitu hybridization probe, and the tumor necrosis factor-α (TNF-α) content in intestinal mucosa was determined by ELISA. Expression of proliferative cell nuclear antigen (PCNA) in bowel mucosa was assayed by real-time PCR and intestinal mucosa expression of phosphorylation extracellular signal-regulated kinase (pERK1/2) and nuclear factor-κB (NF-κB) were evaluated by western blot. Results Four and seven days after MSCs transplantation, the TNF-α content of bowel mucosa in MSCs group was significantly lower than that in saline group. The PCNA in bowel mucosa showed higher expression in MSCs treated group than the saline group, both at 4 and 7 days after cell transplantation. The expression of intestinal mucosal pERK1/2 in MSCs treated group was markedly higher than that in saline group, and the expression of NF-κB in MSCs treated group was noticeably decreased than that in saline group at 4 and 7 days post MSCs transplantation. Conclusion The present investigation provides novel evidence that MSCs have the potential to reduce intestinal I/R injuries probably due to their ability to accelerate cell proliferation and decrease the inflammatory response within intestinal mucosa after ischemia and reperfusion.
Collapse
Affiliation(s)
- Haitao Jiang
- Department of General Surgery Ⅱ, Affiliated Hospital of Qingdao University Medical College, Qingdao, China
- * E-mail:
| | - Linlin Qu
- Department of General Surgery Ⅱ, Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Rongrong Dou
- Department of General Surgery Ⅱ, Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Lianfang Lu
- Department of General Surgery Ⅱ, Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Sishan Bian
- Department of General Surgery, Shandong Provincial Hospital of Traditional Chinese Medicine, Jinan, China
| | - Weiming Zhu
- Research Institute of General Surgery, Jinling Hospital, Nanjing University, Nanjing, China
| |
Collapse
|
16
|
Timmerhaus G, Krasnov A, Takle H, Afanasyev S, Nilsen P, Rode M, Jørgensen SM. Comparison of Atlantic salmon individuals with different outcomes of cardiomyopathy syndrome (CMS). BMC Genomics 2012; 13:205. [PMID: 22646522 PMCID: PMC3443006 DOI: 10.1186/1471-2164-13-205] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 05/30/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cardiomyopathy syndrome (CMS) is a severe disease of Atlantic salmon (Salmo salar L.) associated with significant economic losses in the aquaculture industry. CMS is diagnosed with a severe inflammation and degradation of myocardial tissue caused by a double-stranded RNA virus named piscine myocarditis virus (PMCV), with structural similarities to the Totiviridae family. In the present study we characterized individual host responses and genomic determinants of different disease outcomes. RESULTS From time course studies of experimentally infected Atlantic salmon post-smolts, fish exhibited different outcomes of infection and disease. High responder (HR) fish were characterized with sustained and increased viral load and pathology in heart tissue. Low responder (LR) fish showed declining viral load from 6-10 weeks post infection (wpi) and absence of pathology. Global gene expression (SIQ2.0 oligonucleotide microarray) in HR and LR hearts during infection was compared, in order to characterize differences in the host response and to identify genes with expression patterns that could explain or predict the different outcomes of disease. Virus-responsive genes involved in early antiviral and innate immune responses were upregulated equally in LR and HR at the first stage (2-4 wpi), reflecting the initial increase in virus replication. Repression of heart muscle development was identified by gene ontology enrichment analyses, indicating the early onset of pathology. By six weeks both responder groups had comparable viral load, while increased pathology was observed in HR fish. This was reflected by induced expression of genes implicated in apoptosis and cell death mechanisms, presumably related to lymphocyte regulation and survival. In contrast, LR fish showed earlier activation of NK cell-mediated cytotoxicity and NOD-like receptor signaling pathways. At the late stage of infection, increased pathology and viral load in HR was accompanied by a broad activation of genes involved in adaptive immunity and particularly T cell responses, probably reflecting the increased infiltration and homing of virus-specific T cells to the infected heart. This was in sharp contrast to LR fish, where recovery and reduced viral load was associated with a significantly reduced transcription of adaptive immunity genes and activation of genes involved in energy metabolism. CONCLUSIONS In contrast to LR, a stronger and sustained expression of genes involved in adaptive immune responses in heart tissue of HR at the late stage of disease probably reflected the increased lymphocyte infiltration and pathological outcome. In addition to controlled adaptive immunity and activation of genes involved in cardiac energy metabolism in LR at the late stage, recovery of this group could also be related to an earlier activation of NOD-like receptor signaling and NK cell-mediated cytotoxicity pathways.
Collapse
|
17
|
Zhang JY, Chen ZW, Yao H. Protective effect of urantide against ischemia-reperfusion injury via protein kinase C and phosphtidylinositol 3'-kinase - Akt pathway. Can J Physiol Pharmacol 2012; 90:637-45. [PMID: 22537485 DOI: 10.1139/y2012-048] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Urantide is the most potent UT receptor antagonist compound found to date. Our previous studies have shown that it has cardioprotective effect against ischemia-reperfusion injury. However, it is unclear which signal transduction pathways are involved in the urantide-induced cardioprotective effect. This study was designed to investigate whether the effect of urantide on myocardial ischemia-reperfusion injury in rats via the protein kinase C (PKC) and phosphatidylinositol 3-kinase (PI3K)-Akt signaling pathway. The results showed that urantide at 10 and 30 µg/kg markedly inhibited the increases in serum creatine kinase fraction and lactate dehydrogenase activities and the level of cardiac troponin I, reduced the ratio of myocardial infarct size to area at risk. Urantide significantly decreased the histological damage to the myocardium and modified the ultrastructural damage in cardiac myocytes. In the presence of chelerythrine (an inhibitor of PKC, 1 mg/kg) or LY294002 (an inhibitor of PI3K-Akt, 0.3 mg/kg), the protective effect of urantide was almost completely abolished. Urantide (30 µg/kg) markedly enhanced the expression of p-Akt protein during myocardial ischemia-reperfusion injury, and this enhancement was significantly attenuated by LY294002. Therefore, our results demonstrate that urantide has a potent protective effect against myocardial ischemia-reperfusion injury in rats that may be involved with the PKC and PI3K-Akt signaling pathways.
Collapse
Affiliation(s)
- Jun-Yan Zhang
- Department of Pharmacology, Anhui Medical University, Hefei, PR China
| | | | | |
Collapse
|
18
|
Dai W, Kloner RA. Bone marrow-derived cell transplantation therapy for myocardial infarction: lessons learned and future questions. Am J Transplant 2011; 11:2297-301. [PMID: 21929617 DOI: 10.1111/j.1600-6143.2011.03750.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Over the last decade, many investigators have utilized bone marrow-derived cells for cell transplantation therapy in animal studies and in patients with acute myocardial infarction and chronic heart failure. In those experimental and clinical studies, various doses and types of bone marrow-derived cells have been transplanted to the injured myocardium using a variety of approaches, such as intracoronary infusion or catheter-based direct endomyocardial injection, and at different time points after successful coronary reperfusion. The reported treatment effects are variable, which may be related to differences in cell type and quantity of transplanted cells, timing and approach of cell transplantation and patient selection. In this review, we summarize and discuss the controversies and questions related to the clinical use of bone marrow-derived cells.
Collapse
Affiliation(s)
- W Dai
- The Heart Institute of Good Samaritan Hospital, Los Angeles, CA, USA.
| | | |
Collapse
|
19
|
Bollini S, Cheung KK, Riegler J, Dong X, Smart N, Ghionzoli M, Loukogeorgakis SP, Maghsoudlou P, Dubé KN, Riley PR, Lythgoe MF, De Coppi P. Amniotic fluid stem cells are cardioprotective following acute myocardial infarction. Stem Cells Dev 2011; 20:1985-94. [PMID: 21534857 DOI: 10.1089/scd.2010.0424] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In recent years, various types of stem cells have been characterized and their potential for cardiac regeneration has been investigated. We have previously described the isolation of broadly multipotent cells from amniotic fluid, defined as amniotic fluid stem (AFS) cells. The aim of this study was to investigate the therapeutic potential of human AFS cells (hAFS) in a model of acute myocardial infarction. Wistar rats underwent 30 min of ischemia by ligation of the left anterior descending coronary artery, followed by administration of hAFS cells and 2 h of reperfusion. Infarct size was assessed by 2,3,5-triphenyltetrazolium chloride staining and planimetry. hAFS cells were also analyzed by enzyme-linked immunosorbent assay to detect secretion of putative paracrine factors, such as the actin monomer-binding protein thymosin β4 (Tβ4). The systemic injection of hAFS cells and their conditioned medium (hAFS-CM) was cardioprotective, improving myocardial cell survival and decreasing the infarct size from 53.9%±2.3% (control animals receiving phosphate-buffered saline injection) to 40.0%±3.0% (hAFS cells) and 39.7%±2.5% (hAFS-CM, P<0.01). In addition, hAFS cells were demonstrated to secrete Tβ4, previously shown to be both cardioprotective and proangiogenic. Our results suggest that AFS cells have therapeutic potential in the setting of acute myocardial infarction, which may be mediated through paracrine effectors such as Tβ4. Therefore, AFS cells might represent a novel source for cell therapy and cell transplantation strategies in repair following ischemic heart disease, with a possible paracrine mechanism of action and a potential molecular candidate for acute cardioprotection.
Collapse
Affiliation(s)
- Sveva Bollini
- Surgery Unit, Institute of Child Health and Great Ormond Street Hospital, University College London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|