1
|
Barrère-Lemaire S, Vincent A, Jorgensen C, Piot C, Nargeot J, Djouad F. Mesenchymal stromal cells for improvement of cardiac function following acute myocardial infarction: a matter of timing. Physiol Rev 2024; 104:659-725. [PMID: 37589393 DOI: 10.1152/physrev.00009.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/05/2023] [Accepted: 08/16/2023] [Indexed: 08/18/2023] Open
Abstract
Acute myocardial infarction (AMI) is the leading cause of cardiovascular death and remains the most common cause of heart failure. Reopening of the occluded artery, i.e., reperfusion, is the only way to save the myocardium. However, the expected benefits of reducing infarct size are disappointing due to the reperfusion paradox, which also induces specific cell death. These ischemia-reperfusion (I/R) lesions can account for up to 50% of final infarct size, a major determinant for both mortality and the risk of heart failure (morbidity). In this review, we provide a detailed description of the cell death and inflammation mechanisms as features of I/R injury and cardioprotective strategies such as ischemic postconditioning as well as their underlying mechanisms. Due to their biological properties, the use of mesenchymal stromal/stem cells (MSCs) has been considered a potential therapeutic approach in AMI. Despite promising results and evidence of safety in preclinical studies using MSCs, the effects reported in clinical trials are not conclusive and even inconsistent. These discrepancies were attributed to many parameters such as donor age, in vitro culture, and storage time as well as injection time window after AMI, which alter MSC therapeutic properties. In the context of AMI, future directions will be to generate MSCs with enhanced properties to limit cell death in myocardial tissue and thereby reduce infarct size and improve the healing phase to increase postinfarct myocardial performance.
Collapse
Affiliation(s)
- Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Anne Vincent
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Christian Jorgensen
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Christophe Piot
- Département de Cardiologie Interventionnelle, Clinique du Millénaire, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Farida Djouad
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| |
Collapse
|
2
|
Asserson DB. Allogeneic Mesenchymal Stem Cells After In Vivo Transplantation: A Review. Cell Reprogram 2023; 25:264-276. [PMID: 37971885 DOI: 10.1089/cell.2023.0084] [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] [Indexed: 11/19/2023] Open
Abstract
Autologous mesenchymal stem cells (MSCs) are ideal for tissue regeneration because of their ability to circumvent host rejection, but their procurement and processing present logistical and time-sensitive challenges. Allogeneic MSCs provide an alternative cell-based therapy capable of positively affecting all human organ systems, and can be readily available. Extensive research has been conducted in the treatment of autoimmune, degenerative, and inflammatory diseases with such stem cells, and has demonstrated predominantly safe outcomes with minimal complications. Nevertheless, continued clinical trials are necessary to ascertain optimal harvest and transplant techniques.
Collapse
Affiliation(s)
- Derek B Asserson
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
3
|
Wang W, Tayier B, Guan L, Yan F, Mu Y. Optimization of the cotransfection of SERCA2a and Cx43 genes for myocardial infarction complications. Life Sci 2023; 331:122067. [PMID: 37659592 DOI: 10.1016/j.lfs.2023.122067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/27/2023] [Accepted: 08/30/2023] [Indexed: 09/04/2023]
Abstract
As our previous study showed, the therapeutic effect of two genes (SERCA2a and Cx43) on heart failure after myocardial infarction (MI) was greater than that of single gene (SERCA2a or Cx43) therapy for bone marrow stem cell (BMSC) transplantation. Based on previous research, the aim of this study was to investigate the optimal ratio of codelivery of SERCA2a and Cx43 genes for MI therapy after biotinylated microbubble (BMB) transplantation via ultrasonic-targeted microbubble destruction (UTMD). Forty rats underwent left anterior descending (LAD) ligation and BMSC injection into the infarct and border zones. Four weeks later, the genes SERCA2a and Cx43 were codelivered at different ratios (1:1, 1:2 and 2:1) into the infarcted heart via UTMD. Cardiac mechanoelectrical function was determined at 4 wks after gene delivery, and the hearts of the rats were harvested for measurement of MI size and detection of SERCA2a and Cx43 expression. Q-PCR analysis of the expression of Nkx2.5 and GATA4 in the myocardial infarct zone and measurement of neovascularization in infarcted hearts. After comparing the therapeutic effects of different cogene ratios, the SERCA2a/Cx43-1:2 group showed remarkable cardiac electrical stability and strengthened the role of anti-arrhythmia. In conclusion, the optimum ratio of the SERCA2a/Cx43 gene is 1:2, which is advantageous for maintaining cardiac electrophysiological stability.
Collapse
Affiliation(s)
- Wei Wang
- Department of Echocardiography, Xinjiang Medical University Affiliated First Hospital, Urumqi, China; Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, China; Department of Ultrasound, Urumqi Friendship Hospital, Urumqi, China
| | - Baihetiya Tayier
- Department of Echocardiography, Xinjiang Medical University Affiliated First Hospital, Urumqi, China; Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, China
| | - Lina Guan
- Department of Echocardiography, Xinjiang Medical University Affiliated First Hospital, Urumqi, China; Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, China
| | - Fei Yan
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Yuming Mu
- Department of Echocardiography, Xinjiang Medical University Affiliated First Hospital, Urumqi, China; Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, China.
| |
Collapse
|
4
|
Tan YL, Eng SP, Hafez P, Abdul Karim N, Law JX, Ng MH. Mesenchymal Stromal Cell Mitochondrial Transfer as a Cell Rescue Strategy in Regenerative Medicine: A Review of Evidence in Preclinical Models. Stem Cells Transl Med 2022; 11:814-827. [PMID: 35851922 PMCID: PMC9397650 DOI: 10.1093/stcltm/szac044] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/14/2022] [Indexed: 12/02/2022] Open
Abstract
Mesenchymal stromal cells (MSC) have excellent clinical potential and numerous properties that ease its clinical translation. Mitochondria play a crucial role in energy metabolism, essential for cellular activities, such as proliferation, differentiation, and migration. However, mitochondrial dysfunction can occur due to diseases and pathological conditions. Research on mitochondrial transfer from MSCs to recipient cells has gained prominence. Numerous studies have demonstrated that mitochondrial transfer led to increased adenosine triphosphate (ATP) production, recovered mitochondrial bioenergetics, and rescued injured cells from apoptosis. However, the complex mechanisms that lead to mitochondrial transfer from healthy MSCs to damaged cells remain under investigation, and the factors contributing to mitochondrial bioenergetics recovery in recipient cells remain largely ambiguous. Therefore, this review demonstrates an overview of recent findings in preclinical studies reporting MSC mitochondrial transfer, comprised of information on cell sources, recipient cells, dosage, route of administration, mechanism of transfer, pathological conditions, and therapeutic effects. Further to the above, this research discusses the potential challenges of this therapy in its clinical settings and suggestions to overcome its challenges.
Collapse
Affiliation(s)
- Yu Ling Tan
- Center for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia , Kuala Lumpur , Malaysia
| | | | - Pezhman Hafez
- Yakin Splendour Global Holdings Berhad , Kuala Lumpur , Malaysia
| | - Norwahidah Abdul Karim
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia , Kuala Lumpur , Malaysia
| | - Jia Xian Law
- Center for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia , Kuala Lumpur , Malaysia
| | - Min Hwei Ng
- Center for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia , Kuala Lumpur , Malaysia
| |
Collapse
|
5
|
Shin HS, Shin HH, Shudo Y. Current Status and Limitations of Myocardial Infarction Large Animal Models in Cardiovascular Translational Research. Front Bioeng Biotechnol 2021; 9:673683. [PMID: 33996785 PMCID: PMC8116580 DOI: 10.3389/fbioe.2021.673683] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/06/2021] [Indexed: 01/16/2023] Open
Abstract
Establishing an appropriate disease model that mimics the complexities of human cardiovascular disease is critical for evaluating the clinical efficacy and translation success. The multifaceted and complex nature of human ischemic heart disease is difficult to recapitulate in animal models. This difficulty is often compounded by the methodological biases introduced in animal studies. Considerable variations across animal species, modifications made in surgical procedures, and inadequate randomization, sample size calculation, blinding, and heterogeneity of animal models used often produce preclinical cardiovascular research that looks promising but is irreproducible and not translatable. Moreover, many published papers are not transparent enough for other investigators to verify the feasibility of the studies and the therapeutics' efficacy. Unfortunately, successful translation of these innovative therapies in such a closed and biased research is difficult. This review discusses some challenges in current preclinical myocardial infarction research, focusing on the following three major inhibitors for its successful translation: Inappropriate disease model, frequent modifications to surgical procedures, and insufficient reporting transparency.
Collapse
Affiliation(s)
- Hye Sook Shin
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Heather Hyeyoon Shin
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Yasuhiro Shudo
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
6
|
Malik R, Darche FA, Rivinius R, Seckinger A, Krause U, Koenen M, Thomas D, Katus HA, Schweizer PA. Quantitative Efficacy and Fate of Mesenchymal Stromal Cells Targeted to Cardiac Sites by Radiofrequency Catheter Ablation. Cell Transplant 2020; 29:963689720914236. [PMID: 32207339 PMCID: PMC7444233 DOI: 10.1177/0963689720914236] [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] [Indexed: 11/29/2022] Open
Abstract
Engraftment and functional integration of stem cells or stem cell-derived cells within cardiac tissue is an important prerequisite for cell replacement therapy aiming at the treatment of heart disease. Recently, a novel intravenous approach for application of mesenchymal stromal cells (MSCs) to cardiac sites has been established using radiofrequency catheter ablation (RFCA)-guided targeting, bypassing the need for open chest surgery or direct myocardial cell injection. However, little is known about the quantitative efficacy and longevity of this strategy. We performed selective power-controlled RFCA with eight ablation pulses (30 W, 60 s each) to induce heat-mediated lesions at the right atrial appendices (RAAs) of pigs. Different concentrations of human bone marrow-derived MSCs (105 to 1.6 × 106 cells/kg bodyweight) labeled with superparamagnetic iron oxide (SPIO) particles were infused intravenously in nine pigs one d after RFCA treatment and hearts were explanted 8 d later to quantify the number of engrafted cells. Prussian blue staining revealed high numbers of SPIO-labeled cells in areas surrounding the RFCA-induced lesions. Cell numbers were evaluated by quantitative real-time polymerase chain reaction using specific primers for human MSCs (hMSCs), which indicated that up to 106 hMSCs, corresponding to ∼3.9% of the systemically applied human cells, engrafted within the RAAs of RFCA-treated pigs. Of note, infused hMSCs were observed in nontargeted organs, as well, but appeared at very low concentrations. To assess long-term deposition of MSCs, RAAs of three pigs were analyzed after 6 months, which revealed few persisting hMSCs at targeted sites. RFCA-mediated targeting of MSCs provides a novel minimal invasive strategy for cardiac stem cell engraftment. Qualitative and quantitative results of our large animal experiments indicate an efficient guidance of MSCs to selected cardiac regions, although only few cells remained at targeted sites 6 mo after cell transplantation.
Collapse
Affiliation(s)
- Rizwan Malik
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany
| | - Fabrice A Darche
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Rasmus Rivinius
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Anja Seckinger
- Department of Hematology, Oncology and Rheumatology, Medical University Hospital Heidelberg, Heidelberg, Germany
| | - Ulf Krause
- Department of Hematology, Oncology and Rheumatology, Medical University Hospital Heidelberg, Heidelberg, Germany.,Institute for Transfusion Medicine and Cellular Therapy, University Hospital Muenster, Domagstrasse, Muenster, Germany
| | - Michael Koenen
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,Department of Molecular Neurobiology, Max-Planck-Institute for Medical Research, Jahnstrasse, Heidelberg, Germany
| | - Dierk Thomas
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Patrick A Schweizer
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
7
|
Liu Z, Mikrani R, Zubair HM, Taleb A, Naveed M, Baig MMFA, Zhang Q, Li C, Habib M, Cui X, Sembatya KR, Lei H, Zhou X. Systemic and local delivery of mesenchymal stem cells for heart renovation: Challenges and innovations. Eur J Pharmacol 2020; 876:173049. [PMID: 32142771 DOI: 10.1016/j.ejphar.2020.173049] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 02/20/2020] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
In the beginning stage of heart disease, the blockage of blood flow frequently occurs due to the persistent damage and even death of myocardium. Cicatricial tissue developed after the death of myocardium can affect heart function, which ultimately leads to heart failure. In recent years, several studies carried out about the use of stem cells such as embryonic, pluripotent, cardiac and bone marrow-derived stem cells as well as myoblasts to repair injured myocardium. Current studies focus more on finding appropriate measures to enhance cell homing and survival in order to increase paracrine function. Until now, there is no universal delivery route for mesenchymal stem cells (MSCs) for different diseases. In this review, we summarize the advantages and challenges of the systemic and local pathways of MSC delivery. In addition, we also describe some advanced measures of cell delivery to improve the efficiency of transplantation. The combination of cells and therapeutic substances could be the most reliable method, which allows donor cells to deliver sufficient amounts of paracrine factors and provide long-lasting effects. The cardiac support devices or tissue engineering techniques have the potential to facilitate the controlled release of stem cells on local tissue for a sustained period. A novel promising epicardial drug delivery system is highlighted here, which not only provides MSCs with a favorable environment to promote retention but also increases the contact area and a number of cells recruited in the heart muscle.
Collapse
Affiliation(s)
- Ziwei Liu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Reyaj Mikrani
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | | | - Abdoh Taleb
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China
| | - Muhammad Naveed
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China
| | - Mirza Muhammad Faran Asraf Baig
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu, 210023, PR China
| | - Qin Zhang
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Cuican Li
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Murad Habib
- Department of Surgery, Ayub Teaching Hospital, Abbottabad, Pakistan
| | - Xingxing Cui
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Kiganda Raymond Sembatya
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Han Lei
- Department of Pharmacy, Jiangsu Worker Medical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Xiaohui Zhou
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China; Department of Surgery, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu Province, 210017, PR China; Department of Surgery, Nanjing Shuiximen Hospital, Nanjing, Jiangsu Province, 210017, PR China.
| |
Collapse
|
8
|
Hsu MF, Yu SH, Chuang SJ, Kuo TKC, Singal PK, Huang CY, Kao CL, Kuo CH. Can mesenchymal stem cell lysate reverse aging? Aging (Albany NY) 2019; 10:2900-2910. [PMID: 30362957 PMCID: PMC6224235 DOI: 10.18632/aging.101595] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/12/2018] [Indexed: 12/16/2022]
Abstract
Recent findings regarding uses of adipose-derived mesenchymal stem cell (MSC)-lysate on weight loss and improved glucose tolerance in mice on a high-fat diet suggest an encouraging possibility of using MSC lysate for an anti-aging intervention in humans. However, weight loss and lipopenia during late life can be as life-threatening as hyperglycemia during early adulthood. For this 3-year lifelong experiment, a total of 92 rats were randomized into the vehicle-injected group (F=22; M=24) and the MSC lysate injected group (F=22, M=24). We examined longevity, spontaneous locomotor activity, and body composition in rats maintained on a normal diet and received an intermittent treatment of human adipose-derived MSC lysate (3 times a week, 11 times a month given every second month), starting at 12 months of age until natural death. In substantiating previous knowledge regarding the effects of long-term MSC lysate treatments on fat loss and insulin resistance, the present findings also highlighted a shortened average lifespan, a longer inactive time, and a greater bone loss with a relative increase of lean mass in MSC lysate rats with respect to controls. Conclusion: Our data suggest that MSC lysate treatments stimulate disparity in tissue development and produce a cachexia-like effect to decrease longevity.
Collapse
Affiliation(s)
- Ming-Fen Hsu
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Szu-Hsien Yu
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Sheng-Ju Chuang
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan.,Université Catholique de Louvain and de Duve Institute, Brussels, Belgium
| | - Tom Kwang-Chun Kuo
- Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Pawan K Singal
- Institute of Cardiovascular Sciences, St Boniface General Hospital Research Centre and Department of Physiology and Pathophysiology, Faculty of Medicine, University of Manitoba, Winnipeg, Canada
| | - Chih-Yang Huang
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan.,Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chung-Lan Kao
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital and National Yang Ming University, Taipei, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| |
Collapse
|
9
|
Lim M, Wang W, Liang L, Han ZB, Li Z, Geng J, Zhao M, Jia H, Feng J, Wei Z, Song B, Zhang J, Li J, Liu T, Wang F, Li T, Li J, Fang Y, Gao J, Han Z. Intravenous injection of allogeneic umbilical cord-derived multipotent mesenchymal stromal cells reduces the infarct area and ameliorates cardiac function in a porcine model of acute myocardial infarction. Stem Cell Res Ther 2018; 9:129. [PMID: 29751831 PMCID: PMC5948807 DOI: 10.1186/s13287-018-0888-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 04/13/2018] [Accepted: 04/25/2018] [Indexed: 12/31/2022] Open
Abstract
Background Multipotent mesenchymal stromal cell (MSC) therapy has been widely recognized as a feasible strategy for regenerating injured myocardial tissue. However, little is known about the efficacy of intravenous injection of allogeneic umbilical cord (UC) MSCs in preclinical models of porcine myocardial infarction. Methods Different dosages of allogeneic UC-MSCs or the vehicle [phosphate-buffered saline (PBS)] were delivered intravenously into an acute myocardial infarction (AMI) porcine model twice after coronary ligation. Echocardiography was performed to examine the cardiac function and single photon emission computed tomography (SPECT) and positron emission tomography (PET)/computed tomography (CT) was performed to detect cardiac perfusion and nonviable myocardium. At the end of the experiment, 2,3,5-triphenyl-tetrazolium chloride (TTC) staining and Masson T staining were performed to determine the infarct area. The protein and gene expression levels associated with cardiac function, inflammation, and angiogenesis were examined by Western blot and real time polymerase chain reaction (PCR). In vivo trafficking of intravenous injection of allogeneic UC-MSCs enhanced green fluorescent protein (eGFP) was detected by real time PCR and immunofluorescence. Results After systemic delivery, allogeneic UC-MSCs were largely distributed in the lungs and some in the infracted myocardium. At week 8 following AMI, echocardiography demonstrated significantly improved fractional shortening in the high-dose (1.5 × 106 cells/kg) group. SPECT-PET/CT showed that UC-MSC treatment in both high and low doses markedly ameliorated the left ventricle (LV) infarct area but did not significantly improve the myocardial perfusion defect. LV remodeling was inhibited by UC-MSC therapy, as reflected by a marked reduction in rthe fibrosis area at basal, middle, and apical levels and reduced extracellular matrix deposition in the total myocardial area. Inflammatory biomarkers (tumor necrosis factor alpha and interleukin-6) were reduced and pro-angiogenesis factors (vascular endothelial growth factor and platelet/endothelial cell adhesion molecule 1) were augmented in the myocardial infarct and border area. High-dose UC-MSCs increased the connexin 43 (Cx43) (myocardium preservation) expression in remote area of the LV myocardium after AMI. Conclusions Intravenous injection of UC-MSCs is a feasible and effective way to preserve LV function and ameliorate myocardial remodeling in porcine AMI. The cardioprotective effects of UC-MSCs were attributed to paracrine factors that appear to augment angiogenesis, limit inflammation, and preserve Cx43 gap junction. Electronic supplementary material The online version of this article (10.1186/s13287-018-0888-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Meikuang Lim
- National Engineering Research Center of Cell Products, Tianjin AmCellGene Engineering Co., Ltd, Tianjin, People's Republic of China.,Beijing Institute of Stem Cells, Health & Biotech Co., Ltd, Beijing, People's Republic of China
| | - Weiqiang Wang
- Beijing Institute of Stem Cells, Health & Biotech Co., Ltd, Beijing, People's Republic of China
| | - Lu Liang
- National Engineering Research Center of Cell Products, Tianjin AmCellGene Engineering Co., Ltd, Tianjin, People's Republic of China
| | - Zhi-Bo Han
- National Engineering Research Center of Cell Products, Tianjin AmCellGene Engineering Co., Ltd, Tianjin, People's Republic of China.,State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, People's Republic of China.,JiangXi Engineering Research Center for Stem Cell, ShangRao, Jiangxi, People's Republic of China
| | - Zongjin Li
- Beijing Institute of Stem Cells, Health & Biotech Co., Ltd, Beijing, People's Republic of China
| | - Jie Geng
- National Engineering Research Center of Cell Products, Tianjin AmCellGene Engineering Co., Ltd, Tianjin, People's Republic of China.,Beijing Institute of Stem Cells, Health & Biotech Co., Ltd, Beijing, People's Republic of China
| | - Meng Zhao
- National Engineering Research Center of Cell Products, Tianjin AmCellGene Engineering Co., Ltd, Tianjin, People's Republic of China
| | - Honghong Jia
- National Engineering Research Center of Cell Products, Tianjin AmCellGene Engineering Co., Ltd, Tianjin, People's Republic of China
| | - Jie Feng
- National Engineering Research Center of Cell Products, Tianjin AmCellGene Engineering Co., Ltd, Tianjin, People's Republic of China
| | - Zhe Wei
- National Engineering Research Center of Cell Products, Tianjin AmCellGene Engineering Co., Ltd, Tianjin, People's Republic of China
| | - Baoquan Song
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, People's Republic of China
| | - Jiemin Zhang
- Animal Medical Experiment Center, TEDA International Cardiovascular Hospital, Tianjin, People's Republic of China
| | - Jun Li
- Animal Medical Experiment Center, TEDA International Cardiovascular Hospital, Tianjin, People's Republic of China
| | - Tianwen Liu
- Animal Medical Experiment Center, TEDA International Cardiovascular Hospital, Tianjin, People's Republic of China
| | - Fan Wang
- Animal Medical Experiment Center, TEDA International Cardiovascular Hospital, Tianjin, People's Republic of China
| | - Ting Li
- Nuclear Medicine Department, TEDA International Cardiovascular Hospital, Tianjin, People's Republic of China
| | - Jianming Li
- Nuclear Medicine Department, TEDA International Cardiovascular Hospital, Tianjin, People's Republic of China
| | - Yihu Fang
- Institute of Stem Cell, Jiangxi Medical College, ShangRao, Jiangxi, People's Republic of China
| | - Jianhua Gao
- Institute of Stem Cell, Jiangxi Medical College, ShangRao, Jiangxi, People's Republic of China
| | - Zhongchao Han
- National Engineering Research Center of Cell Products, Tianjin AmCellGene Engineering Co., Ltd, Tianjin, People's Republic of China. .,Beijing Institute of Stem Cells, Health & Biotech Co., Ltd, Beijing, People's Republic of China. .,State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, People's Republic of China.
| |
Collapse
|
10
|
Diverging Concepts and Novel Perspectives in Regenerative Medicine. Int J Mol Sci 2017; 18:ijms18051021. [PMID: 28486410 PMCID: PMC5454934 DOI: 10.3390/ijms18051021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/28/2017] [Accepted: 05/03/2017] [Indexed: 12/31/2022] Open
Abstract
Regenerative medicine has rapidly evolved, due to progress in cell and molecular biology allowing the isolation, characterization, expansion, and engineering of cells as therapeutic tools. Despite past limited success in the clinical translation of several promising preclinical results, this novel field is now entering a phase of renewed confidence and productivity, marked by the commercialization of the first cell therapy products. Ongoing issues in the field include the use of pluripotent vs. somatic and of allogenic vs. autologous stem cells. Moreover, the recognition that several of the observed beneficial effects of cell therapy are not due to integration of the transplanted cells, but rather to paracrine signals released by the exogenous cells, is generating new therapeutic perspectives in the field. Somatic stem cells are outperforming embryonic and induced pluripotent stem cells in clinical applications, mainly because of their more favorable safety profile. Presently, both autologous and allogeneic somatic stem cells seem to be equally safe and effective under several different conditions. Recognition that a number of therapeutic effects of transplanted cells are mediated by paracrine signals, and that such signals can be found in extracellular vesicles isolated from culture media, opens novel therapeutic perspectives in the field of regenerative medicine.
Collapse
|
11
|
Tao ZW, Favreau JT, Guyette JP, Hansen KJ, Lessard J, Burford E, Pins GD, Gaudette GR. Delivering stem cells to the healthy heart on biological sutures: effects on regional mechanical function. J Tissue Eng Regen Med 2017; 11:220-230. [PMID: 24753390 PMCID: PMC4664584 DOI: 10.1002/term.1904] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 11/04/2013] [Accepted: 03/17/2014] [Indexed: 12/11/2022]
Abstract
Current cardiac cell therapies cannot effectively target and retain cells in a specific area of the heart. Cell-seeded biological sutures were previously developed to overcome this limitation, demonstrating targeted delivery with > 60% cell retention. In this study, both cell-seeded and non-seeded fibrin-based biological sutures were implanted into normal functioning rat hearts to determine the effects on mechanical function and fibrotic response. Human mesenchymal stem cells (hMSCs) were used based on previous work and established cardioprotective effects. Non-seeded or hMSC-seeded sutures were implanted into healthy athymic rat hearts. Before cell seeding, hMSCs were passively loaded with quantum dot nanoparticles. One week after implantation, regional stroke work index and systolic area of contraction (SAC) were evaluated on the epicardial surface above the suture. Cell delivery and retention were confirmed by quantum dot tracking, and the fibrotic tissue area was evaluated. Non-seeded biological sutures decreased SAC near the suture from 0.20 ± 0.01 measured in sham hearts to 0.08 ± 0.02, whereas hMSC-seeded biological sutures dampened the decrease in SAC (0.15 ± 0.02). Non-seeded sutures also displayed a small amount of fibrosis around the sutures (1.0 ± 0.1 mm2 ). Sutures seeded with hMSCs displayed a significant reduction in fibrosis (0.5 ± 0.1 mm2 , p < 0.001), with quantum dot-labelled hMSCs found along the suture track. These results show that the addition of hMSCs attenuates the fibrotic response observed with non-seeded sutures, leading to improved regional mechanics of the implantation region. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Ze-Wei Tao
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA
| | - John T Favreau
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Jacques P Guyette
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Katrina J Hansen
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Jeffrey Lessard
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Evans Burford
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA
| | - George D Pins
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Glenn R Gaudette
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA
| |
Collapse
|
12
|
Maria ATJ, Maumus M, Le Quellec A, Jorgensen C, Noël D, Guilpain P. Adipose-Derived Mesenchymal Stem Cells in Autoimmune Disorders: State of the Art and Perspectives for Systemic Sclerosis. Clin Rev Allergy Immunol 2016; 52:234-259. [DOI: 10.1007/s12016-016-8552-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
13
|
Human adipose mesenchymal stem cells as potent anti-fibrosis therapy for systemic sclerosis. J Autoimmun 2016; 70:31-9. [PMID: 27052182 DOI: 10.1016/j.jaut.2016.03.013] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/21/2016] [Accepted: 03/23/2016] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Displaying immunosuppressive and trophic properties, mesenchymal stem/stromal cells (MSC) are being evaluated as promising therapeutic options in a variety of autoimmune and degenerative diseases. Although benefits may be expected in systemic sclerosis (SSc), a rare autoimmune disease with fibrosis-related mortality, MSC have yet to be evaluated in this specific condition. While autologous approaches could be inappropriate because of functional alterations in MSC from patients, the objective of the present study was to evaluate allogeneic and xenogeneic MSC in the HOCl-induced model of diffuse SSc. We also questioned the source of human MSC and compared bone marrow- (hBM-MSC) and adipose-derived MSC (hASC). METHODS HOCl-challenged BALB/c mice received intravenous injection of BM-MSC from syngeneic BALB/c or allogeneic C57BL/6 mice, and xenogeneic hBM-MSC or hASC (3 donors each). Skin thickness was measured during the experiment. At euthanasia, histology, immunostaining, collagen determination and RT-qPCR were performed in skin and lungs. RESULTS Xenogeneic hBM-MSC were as effective as allogeneic or syngeneic BM-MSC in decreasing skin thickness, expression of Col1, Col3, α-Sma transcripts, and collagen content in skin and lungs. This anti-fibrotic effect was not associated with MSC migration to injured skin or with long-term MSC survival. Interestingly, compared with hBM-MSC, hASC were significantly more efficient in reducing skin fibrosis, which was related to a stronger reduction of TNFα, IL1β, and enhanced ratio of Mmp1/Timp1 in skin and lung tissues. CONCLUSIONS Using primary cells isolated from 3 murine and 6 human individuals, this preclinical study demonstrated similar therapeutic effects using allogeneic or xenogeneic BM-MSC while ASC exerted potent anti-inflammatory and remodeling properties. This sets the proof-of-concept prompting to evaluate the therapeutic efficacy of allogeneic ASC in SSc patients.
Collapse
|
14
|
The challenges and promises of allogeneic mesenchymal stem cells for use as a cell-based therapy. Stem Cell Res Ther 2015; 6:234. [PMID: 26620426 PMCID: PMC4665863 DOI: 10.1186/s13287-015-0240-9] [Citation(s) in RCA: 217] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are ideal for cell-based therapy in various inflammatory diseases because of their immunosuppressive and tissue repair properties. Moreover, their immunosuppressive properties and low immunogenicity contribute to a reduced or weakened immune response elicited by the implantation of allogeneic MSCs compared with other cell types. Therefore, implantation of allogeneic MSCs may be a promising cell-based therapy. In this review, we first summarize the unique advantages of allogeneic MSCs for therapeutic applications. Second, we critically analyze the factors influencing their therapeutic effects, including administration routes, detection time-points, disease models, differentiation of MSCs in vivo, and timing and dosage of MSC administration. Finally, current approaches to allogeneic MSC application are discussed. In conclusion, allogeneic MSCs are a promising option because of their low immunogenicity and immunosuppressive and tissue repair capabilities. Further investigations are needed to enhance the consistency and efficacy of MSCs when used as a cell-based therapy in inflammatory diseases as well as for tissue repair.
Collapse
|
15
|
Tang XL, Rokosh G, Sanganalmath SK, Tokita Y, Keith MCL, Shirk G, Stowers H, Hunt GN, Wu W, Dawn B, Bolli R. Effects of Intracoronary Infusion of Escalating Doses of Cardiac Stem Cells in Rats With Acute Myocardial Infarction. Circ Heart Fail 2015; 8:757-65. [PMID: 25995227 DOI: 10.1161/circheartfailure.115.002210] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 05/15/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Although c-kit(pos) cardiac stem cells (CSCs) preserve left ventricular (LV) function and structure after myocardial infarction, CSC doses have been chosen arbitrarily, and the dose-effect relationship is unknown. METHODS AND RESULTS Rats underwent a 90-minute coronary occlusion followed by 35 days of reperfusion. Vehicle or CSCs at 5 escalating doses (0.3×10(6), 0.75×10(6), 1.5×10(6), 3.0×10(6), and 6.0×10(6) cells/heart) were given intracoronarily 4 h after reperfusion. The lowest dose (0.3×10(6)) had no effect on LV function and morphology, whereas 0.75, 1.5, and 3.0×10(6) significantly improved regional and global LV function (echocardiography and hemodynamic studies). These 3 doses had similar effects on echocardiographic parameters (infarct wall thickening fraction, LV end-systolic and end-diastolic volumes, LV ejection fraction) and hemodynamic variables (LV end-diastolic pressure, LV dP/dtmax, preload adjusted maximal power, end-systolic elastance, preload recruitable stroke work) and produced similar reductions in apoptosis, scar size, infarct wall thinning, and LV expansion index and similar increases in viable myocardium in the risk region (morphometry). Infusion of 6.0×10(6) CSCs markedly increased postprocedural mortality. Green fluorescent protein and 5-bromo-2'-deoxyuridine staining indicated that persistence of donor cells and formation of new myocytes were negligible with all doses. CONCLUSIONS Surprisingly, in this rat model of acute myocardial infarction, the dose-response relationship for intracoronary CSCs is flat. A minimal dose between 0.3 and 0.75×10(6) is necessary for efficacy; above this threshold, a 4-fold increase in cell number does not produce greater improvement in LV function or structure. Further increases in cell dose are harmful.
Collapse
Affiliation(s)
- Xian-Liang Tang
- From the Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville, KY (X.-L.T., G.R., S.K.S., Y.T., M.C.L.K., G.S., H.S., G.N.H., W.W., R.B.); and Division of Cardiovascular Diseases and the Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (B.D.)
| | - Gregg Rokosh
- From the Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville, KY (X.-L.T., G.R., S.K.S., Y.T., M.C.L.K., G.S., H.S., G.N.H., W.W., R.B.); and Division of Cardiovascular Diseases and the Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (B.D.)
| | - Santosh K Sanganalmath
- From the Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville, KY (X.-L.T., G.R., S.K.S., Y.T., M.C.L.K., G.S., H.S., G.N.H., W.W., R.B.); and Division of Cardiovascular Diseases and the Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (B.D.)
| | - Yukichi Tokita
- From the Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville, KY (X.-L.T., G.R., S.K.S., Y.T., M.C.L.K., G.S., H.S., G.N.H., W.W., R.B.); and Division of Cardiovascular Diseases and the Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (B.D.)
| | - Matthew C L Keith
- From the Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville, KY (X.-L.T., G.R., S.K.S., Y.T., M.C.L.K., G.S., H.S., G.N.H., W.W., R.B.); and Division of Cardiovascular Diseases and the Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (B.D.)
| | - Gregg Shirk
- From the Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville, KY (X.-L.T., G.R., S.K.S., Y.T., M.C.L.K., G.S., H.S., G.N.H., W.W., R.B.); and Division of Cardiovascular Diseases and the Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (B.D.)
| | - Heather Stowers
- From the Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville, KY (X.-L.T., G.R., S.K.S., Y.T., M.C.L.K., G.S., H.S., G.N.H., W.W., R.B.); and Division of Cardiovascular Diseases and the Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (B.D.)
| | - Gregory N Hunt
- From the Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville, KY (X.-L.T., G.R., S.K.S., Y.T., M.C.L.K., G.S., H.S., G.N.H., W.W., R.B.); and Division of Cardiovascular Diseases and the Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (B.D.)
| | - Wenjian Wu
- From the Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville, KY (X.-L.T., G.R., S.K.S., Y.T., M.C.L.K., G.S., H.S., G.N.H., W.W., R.B.); and Division of Cardiovascular Diseases and the Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (B.D.)
| | - Buddhadeb Dawn
- From the Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville, KY (X.-L.T., G.R., S.K.S., Y.T., M.C.L.K., G.S., H.S., G.N.H., W.W., R.B.); and Division of Cardiovascular Diseases and the Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (B.D.)
| | - Roberto Bolli
- From the Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville, KY (X.-L.T., G.R., S.K.S., Y.T., M.C.L.K., G.S., H.S., G.N.H., W.W., R.B.); and Division of Cardiovascular Diseases and the Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (B.D.).
| |
Collapse
|
16
|
Mäkelä T, Takalo R, Arvola O, Haapanen H, Yannopoulos F, Blanco R, Ahvenjärvi L, Kiviluoma K, Kerkelä E, Nystedt J, Juvonen T, Lehenkari P. Safety and biodistribution study of bone marrow-derived mesenchymal stromal cells and mononuclear cells and the impact of the administration route in an intact porcine model. Cytotherapy 2015; 17:392-402. [PMID: 25601140 DOI: 10.1016/j.jcyt.2014.12.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 12/02/2014] [Accepted: 12/10/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND AIMS Bone marrow mononuclear cells (BM-MNCs) and bone marrow-derived mesenchymal stem stromal cells (BM-MSCs) could have therapeutic potential for numerous conditions, including ischemia-related injury. Cells transplanted intravascularly may become entrapped in the lungs, which potentially decreases their therapeutic effect and increases the risk for embolism. METHODS Twelve pigs were divided into groups of 3 and received (99m)Tc- hydroxymethyl-propylene-amine-oxime-labeled autologous BM-MNCs or allogeneic BM-MSCs by either intravenous (IV) or intra-arterial (IA) transplantation. A whole body scan and single photon emission computed tomography/computed tomography (SPECT/CT) were performed 8 h later, and tissue biopsies were collected for gamma counting. A helical CT scan was also performed on 4 pigs to detect possible pulmonary embolism, 2 after IV BM-MSC injection and 2 after saline injection. RESULTS The transplantation route had a greater impact on the biodistribution of the BM-MSCs than the BM-MNCs. The BM-MNCs accumulated in the spleen and bones, irrespective of the administration route. The BM-MSCs had relatively higher uptake in the kidneys. The IA transplantation decreased the deposition of BM-MSCs in the lungs and increased uptake in other organs, especially in the liver. Lung atelectases were frequent due to mechanical ventilation and attracted transplanted cells. CT did not reveal any pulmonary embolism. CONCLUSIONS Both administration routes were found to be safe, but iatrogenic atelectasis might be an issue when cells accumulate in the lungs. The IA administration is effective in avoiding pulmonary entrapment of BM-MSCs. The cell type and administration method both have a major impact on the acute homing.
Collapse
Affiliation(s)
- Tuomas Mäkelä
- Department of Surgery, Oulu University Hospital, Oulu, Finland.
| | - Reijo Takalo
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Oiva Arvola
- Department of Surgery, Oulu University Hospital, Oulu, Finland
| | - Henri Haapanen
- Department of Surgery, Oulu University Hospital, Oulu, Finland
| | | | - Roberto Blanco
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Lauri Ahvenjärvi
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Kai Kiviluoma
- Department of Anaesthesiology, Oulu University Hospital, Oulu, Finland
| | - Erja Kerkelä
- Finnish Red Cross Blood Service, Research and Cell Therapy Services, Helsinki, Finland
| | - Johanna Nystedt
- Finnish Red Cross Blood Service, Research and Cell Therapy Services, Helsinki, Finland
| | - Tatu Juvonen
- Department of Surgery, Oulu University Hospital, Oulu, Finland
| | - Petri Lehenkari
- Department of Anatomy and Cell Biology, Medical Research Center, University of Oulu, Oulu, Finland
| |
Collapse
|
17
|
Therapeutic application of adipose derived stem cells in acute myocardial infarction: lessons from animal models. Stem Cell Rev Rep 2014; 10:389-98. [PMID: 24577790 DOI: 10.1007/s12015-014-9502-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The majority of patients survive an acute myocardial infarction (AMI). Their outcome is negatively influenced by post-AMI events, such as loss of viable cardiomyocytes due to a post-AMI inflammatory response, eventually resulting in heart failure and/or death. Recent pre-clinical animal studies indicate that mesenchymal stem cells derived from adipose tissue (ASC) are new promising candidates that may facilitate cardiovascular regeneration in the infarcted myocardium. In this review we have compared all animal studies in which ASC were used as a therapy post-AMI and have focused on aspects that might be important for future successful clinical application of ASC.
Collapse
|
18
|
Recent advancements in tissue engineering for stem cell-based cardiac therapies. Ther Deliv 2013; 4:503-16. [PMID: 23557290 DOI: 10.4155/tde.13.13] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Advances in cardiac tissue engineering have recently focused on utilizing stem cells to regenerate infarcted and scarred myocardium. Due to their proliferative nature and tremendous potential for differentiation, stem cells are presently being investigated for clinical applications. Unfortunately, limiting factors such as massive cell death and poor retention have hampered clinical outcomes. Consequently, the development of an efficient delivery system for stem cells to the target site is essential. The use of innovative tissue engineering techniques has opened up new horizons within the field of cellular cardiomyoplasty. This paper will present a comprehensive overview of the recent advancements in stem cell technology destined for myocardial tissue repair. In addition, the multidisciplinary approach to tissue engineering presented here will provide the reader with insight into the clinical realization of cellular cardiomyoplasty.
Collapse
|
19
|
Song L, Yang YJ, Dong QT, Qian HY, Gao RL, Qiao SB, Shen R, He ZX, Lu MJ, Zhao SH, Geng YJ, Gersh BJ. Atorvastatin enhance efficacy of mesenchymal stem cells treatment for swine myocardial infarction via activation of nitric oxide synthase. PLoS One 2013; 8:e65702. [PMID: 23741509 PMCID: PMC3669282 DOI: 10.1371/journal.pone.0065702] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Accepted: 04/26/2013] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND In a swine model of acute myocardial infarction (AMI), Statins can enhance the therapeutic efficacy of mesenchymal stem cell (MSCs) transplantation. However, the mechanisms remain unclear. This study aims at assessing whether atorvastatin (Ator) facilitates the effects of MSCs through activation of nitric oxide synthase (NOS), especially endothelial nitric oxide synthase (eNOS), which is known to protect against ischemic injury. METHODS AND RESULTS 42 miniswines were randomized into six groups (n = 7/group): Sham operation; AMI control; Ator only; MSC only, Ator+MSCs and Ator+MSCs+NG-nitrol-L-arginine (L-NNA), an inhibitor of NOS. In an open-heart surgery, swine coronary artery ligation and reperfusion model were established, and autologous bone-marrow MSCs were injected intramyocardium. Four weeks after transplantation, compared with the control group, Ator+MSCs animals exhibited decreased defect areas of both "perfusion" defined by Single-Photon Emission Computed Tomography (-6.2±1.8% vs. 2.0±5.1%, P = 0.0001) and "metabolism" defined by Positron Emission Tomography (-3.00±1.41% vs. 4.20±4.09%, P = 0.0004); Ejection fraction by Magnetic Resonance Imaging increased substantially (14.22±12.8% vs. 1.64±2.64%, P = 0.019). In addition, indices of inflammation, fibrosis, and apoptosis were reduced and survivals of MSCs or MSC-derived cells were increased in Ator+MSCs animals. In Ator or MSCs alone group, perfusion, metabolism, inflammation, fibrosis or apoptosis were reduced but there were no benefits in terms of heart function and cell survival. Furthermore, the above benefits of Ator+MSCs treatment could be partially blocked by L-NNA. CONCLUSIONS Atorvastatin facilitates survival of implanted MSCs, improves function and morphology of infarcted hearts, mediated by activation of eNOS and alleviated by NOS inhibitor. The data reveal the cellular and molecular mechanism for anti-AMI therapy with a combination of statin and stem cells.
Collapse
Affiliation(s)
- Lei Song
- Coronary Heart Disease Center, Department of Cardiology, Fuwai Hospital and Cardiovascular Institute, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Suhr F, Delhasse Y, Bungartz G, Schmidt A, Pfannkuche K, Bloch W. Cell biological effects of mechanical stimulations generated by focused extracorporeal shock wave applications on cultured human bone marrow stromal cells. Stem Cell Res 2013; 11:951-64. [PMID: 23880536 DOI: 10.1016/j.scr.2013.05.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 03/20/2013] [Accepted: 05/18/2013] [Indexed: 12/13/2022] Open
Abstract
Human bone marrow stromal cells (hBMSCs) bear tremendous clinical potential due to their immunomodulatory properties in transplantation settings and their contribution to tissue regeneration. In fact, they are among the most promising types of stem-like cells for therapeutic applications and are the subject of intense research. However, the clinical use of hBMSCs has been confounded by limitations in their availability; they are scarce cells cumbersome to isolate and purify. Additionally, they are difficult to target to the site of injury in regeneration experiments. In order to combat these limitations, focused extracorporeal shock waves (fESW, 0.2/0.3mJ∗mm(-2)) were applied to purified, cultured hBMSCs. fESW (0.2mJ∗mm(-2)) stimulations were found to increase hBMSCs' growth rate (p<0.05), proliferation (p<0.05), migration, cell tracking and wound healing (p<0.05, respectively), as well as to reduce the rate of apoptosis activation (p<0.05). The increase in hBMSC migration behavior was found to be mediated by active remodeling of the actin cytoskeleton as indicated by increased directed stress fiber formations (p<0.05). Furthermore, hBMSCs maintain their differentiation potentials after fESW treatment, whereas 0.2mJ∗mm(-2) is the most effective application. In conclusion, our results establish first-timely that hBMSCs' behavior can be modified and optimized in response to defined mechanical stimulation. These findings appear particularly promising as they suggest that mechanical stress preconditions hBMSCs for improved therapeutic performance without genetic manipulations and that mechanically preconditioned hBMSCs will be advantageous for hBMSC-based tissue regeneration. Therefore, this approach opens the door for exploiting the full potential of these cells in regenerative medicine.
Collapse
Affiliation(s)
- Frank Suhr
- Institute of Cardiovascular Research and Sport Medicine, Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Cologne, Germany.
| | | | | | | | | | | |
Collapse
|
21
|
Huu AL, Prakash S, Shum-Tim D. Cellular cardiomyoplasty: current state of the field. Regen Med 2013; 7:571-82. [PMID: 22817629 DOI: 10.2217/rme.12.28] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cellular cardiomyoplasty employs stem cell therapy to regenerate myocardium. Characterized by their potential for proliferation, differentiation and capacity for self-renewal, stem cells are ideally suited for use in regenerative medicine. Supplementing traditional therapeutic modalities aimed at the palliation of congestive heart failure, cellular cardiomyoplasty is an innovative approach aimed at producing functional, viable myocardium following an acute infarction. The primary focus is to prevent the onset of congestive heart failure; however, potential applications aimed at reversing ischemic heart disease are concurrently in development. After decades of research, cellular cardiomyoplasty has moved beyond traditional in vitro and animal models; it is currently being implemented in clinical trials. Despite this monumental advance, certain limitations remain inherent in this process, preventing stem cell therapy from reaching its full potential. On a cellular level, stem cell retention and viability postimplantation continues to be problematic. Solutions under investigation include pioneering advances in cell delivery, in vitro pretreatment, and tissue engineering. Moreover, questions surrounding optimal cell type and cellular mechanisms concerning cellular cardiomyoplasty remain unanswered. Clarification of these issues is essential to ensure continued progression of this new technology. Stem cell therapy has been highly successful within the in vitro and in vivo environment. However, as clinical trials abound, cellular cardiomyoplasty must transition from an experimental concept to an effective therapeutic treatment. This process is hindered by discordance between scientific accrue and practical applicability. This review will provide a comprehensive summary of current innovations on cellular cardiomyoplasty, and future prospects. There will be a particular emphasis on the clinical aspects of stem cell therapy in an attempt to bridge the gap between science and medicine. Overcoming this barrier will render cellular cardiomyoplasty accessible to patients on a global basis.
Collapse
Affiliation(s)
- Alice Le Huu
- Division of Cardiac Surgery & Surgical Research, Department of Surgery, McGill University Health Center, Montreal, QC, Canada
| | | | | |
Collapse
|
22
|
Yang S, Piao J, Jin L, Zhou Y. Does pretreatment of bone marrow mesenchymal stem cells with 5-azacytidine or double intravenous infusion improve their therapeutic potential for dilated cardiomyopathy? Med Sci Monit Basic Res 2013; 19:20-31. [PMID: 23314418 PMCID: PMC3638670 DOI: 10.12659/msmbr.883737] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 08/27/2012] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND This study was designed to investigate whether pretreatment of bone marrow mesenchymal stem cells (BMSCs) with 5-azacytidine (5-aza) or double intravenous infusion could enhance their therapeutic potential for dilated cardiomyopathy (DCM). MATERIAL/METHODS BMSCs were cultured for 2 weeks in the presence or absence of 5-aza and DCM serum. The cultured BMSCs (Groups 1 and 2), 5-aza-induced BMSCs (Groups 3 and 4), and medium alone (model control) were transplanted into 80 female Wistar rats by intravenous tail vein injection. Double infusion of BMSCs with 1-day time-interval was carried out in Groups 2 and 4. Postmortem histological analysis and evaluation of heart function were performed at 4 weeks post-transplantation. RESULTS Some transplanted BMSCs engrafted into myocardial tissue and were positive for cardiac marker troponin T. The hearts containing transplanted BMSCs secreted a larger amount of vascular endothelial growth factor. Cardiac function parameters and serum level of brain natriuretic peptide (BNP) did not differ among Groups 1, 3, and the model control. As compared with model control, BMSC transplantation in Groups 2 and 4 significantly decreased the serum level of BNP and improved cardiac contractile function, as evidenced by reduced left ventricular end-diastolic and end-systolic diameter, elevated ejection fraction, and fractional shortening. CONCLUSIONS BMSC transplantation is a promising strategy for the treatment of DCM. Pretreatment of BMSCs with 5-aza and DCM serum does not enhance their therapeutic efficacy, and the double intravenous BMSC infusion method is superior to single infusion for preserving cardiac contractile function in a rat model of DCM.
Collapse
|
23
|
Effects of bone marrow mesenchymal stem cells in a rat model of myocardial infarction. Resuscitation 2012; 83:1391-6. [DOI: 10.1016/j.resuscitation.2012.02.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 02/15/2012] [Accepted: 02/21/2012] [Indexed: 01/14/2023]
|
24
|
Guyette JP, Fakharzadeh M, Burford EJ, Tao ZW, Pins GD, Rolle MW, Gaudette GR. A novel suture-based method for efficient transplantation of stem cells. J Biomed Mater Res A 2012; 101:809-18. [PMID: 22961975 DOI: 10.1002/jbm.a.34386] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 06/29/2012] [Accepted: 07/09/2012] [Indexed: 12/13/2022]
Abstract
Advances in regenerative medicine have improved the potential of using cellular therapy for treating several diseases. However, the effectiveness of new cellular therapies is largely limited by low cell engraftment and inadequate localization. To improve on these limitations, we developed a novel delivery mechanism using cell-seeded biological sutures. We demonstrate the ability of cell-seeded biological sutures to efficiently implant human mesenchymal stem cells (hMSCs) to specific regions within the beating heart; a tissue known to have low cell retention and engraftment shortly after delivery. Cell-seeded biological sutures were developed by bundling discrete microthreads extruded from extracellular matrix proteins, attaching a surgical needle to the bundle and seeding the bundle with hMSCs. During cell preparation, hMSCs were loaded with quantum dot nanoparticles for cell tracking within the myocardium. Each biological suture contained an average of 5903 ± 1966 hMSCs/cm suture length. Delivery efficiency was evaluated by comparing cell-seeded biological suture implantation with intramyocardial (IM) cell injections (10,000 hMSCs in 35 μL) into the left ventricle of normal, noninfarcted rat hearts after 1 h. Delivery efficiency of hMSCs by biological sutures (63.6 ± 10.6%) was significantly higher than IM injection (11.8 ± 6.2%; p < 0.05). Cell-tracking analysis indicated suture-delivered hMSCs were found throughout the thickness of the ventricular myocardium: along the entire length of the biological suture track, localizing closely with native myocardium. These results suggest cell-seeded biological sutures can deliver cells to the heart more efficiently than conventional methods, demonstrating an effective delivery method for implanting cells in soft tissue.
Collapse
Affiliation(s)
- Jacques P Guyette
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
de Paula S, Greggio S, Marinowic DR, Machado DC, DaCosta JC. The dose-response effect of acute intravenous transplantation of human umbilical cord blood cells on brain damage and spatial memory deficits in neonatal hypoxia-ischemia. Neuroscience 2012; 210:431-41. [PMID: 22441035 DOI: 10.1016/j.neuroscience.2012.03.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 02/28/2012] [Accepted: 03/02/2012] [Indexed: 01/14/2023]
Abstract
Despite the beneficial effects of cell-based therapies on brain repair shown in most studies, there has not been a consensus regarding the optimal dose of human umbilical cord blood cells (HUCBC) for neonatal hypoxia-ischemia (HI). In this study, we compared the long-term effects of intravenous administration of HUCBC at three different doses on spatial memory and brain morphological changes after HI in newborn Wistar rats. In addition, we tested whether the transplanted HUCBC migrate to the injured brain after transplantation. Seven-day-old animals underwent right carotid artery occlusion and were exposed to 8% O(2) inhalation for 2 h. After 24 h, randomly selected animals were assigned to four different experimental groups: HI rats administered with vehicle (HI+vehicle), HI rats treated with 1×10(6) (HI+low-dose), 1×10(7) (HI+medium-dose), and 1×10(8) (HI+high-dose) HUCBC into the jugular vein. A control group (sham-operated) was also included in this study. After 8 weeks of transplantation, spatial memory performance was assessed using the Morris water maze (MWM), and subsequently, the animals were euthanized for brain morphological analysis using stereological methods. In addition, we performed immunofluorescence and polymerase chain reaction (PCR) analyses to identify HUCBC in the rat brain 7 days after transplantation. The MWM test showed a significant spatial memory recovery at the highest HUCBC dose compared with HI+vehicle rats (P<0.05). Furthermore, the brain atrophy was also significantly lower in the HI+medium- and high-dose groups compared with the HI+vehicle animals (P<0.01; 0.001, respectively). In addition, HUCBC were demonstrated to be localized in host brains by immunohistochemistry and PCR analyses 7 days after intravenous administration. These results revealed that HUCBC transplantation has the dose-dependent potential to promote robust tissue repair and stable cognitive improvement after HI brain injury.
Collapse
Affiliation(s)
- S de Paula
- Laboratório de Neurociências e de Sinalização Celular, Instituto do Cérebro, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | | | | | | | | |
Collapse
|
26
|
Abstract
The contribution of stem cells to cure damaged hearts has finally been unraveled. A large number of preclinical and clinical studies have showed beneficial outcomes after myocardial infarction. In this review, the current understanding of stem cell therapy in preclinical and clinical experiences is summarized. Stem cells from bone marrow have shown a potential to improve cardiac performance after myocardial infarction in animal and early clinical studies. Clinical trials from all over the world have provided safety assessments of stem cell therapy with marginal improvement of clinical outcomes. Thus, further investigations should be encouraged to resolve the discrepancies between studies, clinical issues, and unclear translational findings. This review provides information and commentary on key trials for stem cell-based treat-ment of cardiovascular disease.
Collapse
Affiliation(s)
- Yong Sook Kim
- Heart Research Center, Chonnam National University Hospital, Gwangju, Korea
| | | |
Collapse
|
27
|
Patel AN, Genovese J. Potential clinical applications of adult human mesenchymal stem cell (Prochymal®) therapy. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2011; 4:61-72. [PMID: 24198531 PMCID: PMC3781758 DOI: 10.2147/sccaa.s11991] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In vitro, in vivo animal, and human clinical data show a broad field of application for mesenchymal stem cells (MSCs). There is overwhelming evidence of the usefulness of MSCs in regenerative medicine, tissue engineering, and immune therapy. At present, there are a significant number of clinical trials exploring the use of MSCs for the treatment of various diseases, including myocardial infarction and stroke, in which oxygen suppression causes widespread cell death, and others with clear involvement of the immune system, such as graft-versus-host disease, Crohn’s disease, and diabetes. With no less impact, MSCs have been used as cell therapy to treat defects in bone and cartilage and to help in wound healing, or in combination with biomaterials in tissue engineering development. Among the MSCs, allogeneic MSCs have been associated with a regenerative capacity due to their unique immune modulatory properties. Their immunosuppressive capability without evidence of immunosuppressive toxicity at a global level define their application in the treatment of diseases with a pathogenesis involving uncontrolled activity of the immune system. Until now, the limitation in the number of totally characterized autologous MSCs available represents a major obstacle to their use for adult stem cell therapy. The use of premanufactured allogeneic MSCs from controlled donors under optimal conditions and their application in highly standardized clinical trials would lead to a better understanding of their real applications and reduce the time to clinical translation.
Collapse
Affiliation(s)
- Amit N Patel
- University of Utah School of Medicine, Salt Lake City, UT, USA
| | | |
Collapse
|
28
|
van Dijk A, Naaijkens BA, Jurgens WJFM, Nalliah K, Sairras S, van der Pijl RJ, Vo K, Vonk ABA, van Rossum AC, Paulus WJ, van Milligen FJ, Niessen HWM. Reduction of infarct size by intravenous injection of uncultured adipose derived stromal cells in a rat model is dependent on the time point of application. Stem Cell Res 2011; 7:219-29. [PMID: 21907165 DOI: 10.1016/j.scr.2011.06.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 06/14/2011] [Accepted: 06/16/2011] [Indexed: 01/08/2023] Open
Abstract
Stem cell therapy is a promising tool to improve outcome after acute myocardial infarction (AMI), but needs to be optimized since results from clinical applications remain ambiguous. A potent source of stem cells is the stromal vascular fraction of adipose tissue (SVF), which contains high numbers of adipose derived stem cells (ASC). We hypothesized that: 1) intravenous injection can be used to apply stem cells to the heart. 2) Uncultured SVF cells are easier and safer when cultured ASCs. 3) Transplantation after the acute inflammation period of AMI is favorable over early injection. For this, AMI was induced in rats by 40min of coronary occlusion. One or seven days after AMI, rats were intravenously injected with vehicle, 5×10(6) uncultured rat SVF cells or 1×10(6) rat ASCs. Rats were analyzed 35 days after AMI. Intravenous delivery of both fresh SVF cells and cultured ASCs 7 days after AMI significantly reduced infarct size compared to vehicle. Similar numbers of stem cells were found in the heart, after treatment with fresh SVF cells and cultured ASCs. Importantly, no adverse effects were found after injection of SVF cells. Using cultured ASCs, however, 3 animals had shortness of breath, and one animal died during injection. In contrast to application at 7 days post AMI, injection of SVF cells 1 day post AMI resulted in a small but non-significant infarct reduction (p=0.35). Taken together, intravenous injection of uncultured SVF cells subsequent to the acute inflammation period, is a promising stem cell therapy for AMI.
Collapse
Affiliation(s)
- A van Dijk
- Department of Pathology, VU University Medical Centre, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Simioniuc A, Campan M, Lionetti V, Marinelli M, Aquaro GD, Cavallini C, Valente S, Di Silvestre D, Cantoni S, Bernini F, Simi C, Pardini S, Mauri P, Neglia D, Ventura C, Pasquinelli G, Recchia FA. Placental stem cells pre-treated with a hyaluronan mixed ester of butyric and retinoic acid to cure infarcted pig hearts: a multimodal study. Cardiovasc Res 2011; 90:546-556. [PMID: 21257613 DOI: 10.1093/cvr/cvr018] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/01/2023] Open
|
30
|
Coronary vein infusion of multipotent stromal cells from bone marrow preserves cardiac function in swine ischemic cardiomyopathy via enhanced neovascularization. J Transl Med 2011; 91:553-64. [PMID: 21283079 DOI: 10.1038/labinvest.2010.202] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Few reports have examined the effects of adult bone marrow multipotent stromal cells (MSCs) on large animals, and no useful method has been established for MSC implantation. In this study, we investigate the effects of MSC infusion from the coronary vein in a swine model of chronic myocardial infarction (MI). MI was induced in domestic swine by placing beads in the left coronary artery. Bone marrow cells were aspirated and then cultured to isolate the MSCs. At 4 weeks after MI, MSCs labeled with dye (n=8) or vehicle (n=5) were infused retrogradely from the anterior interventricular vein without any complications. Left ventriculography (LVG) was performed just before and at 4 weeks after cell infusion. The ejection fraction (EF) assessed by LVG significantly decreased from baseline up to a follow-up at 4 weeks in the control group (P<0.05), whereas the cardiac function was preserved in the MSC group. The difference in the EF between baseline and follow-up was significantly greater in the MSC group than in the control group (P<0.05). The MSC administration significantly promoted neovascularization in the border areas compared with the controls (P<0.0005), though it had no affect on cardiac fibrosis. A few MSCs expressed von Willebrand factor in a differentiation assay, but none of them expressed troponin T. In quantitative gene expression analysis, basic fibroblast growth factor and vascular endothelial growth factor (VEGF) levels were significantly higher in the MSC-treated hearts than in the controls (P<0.05, respectively). Immunohistochemical staining revealed VEGF production in the engrafted MSCs. In vitro experiment demonstrated that MSCs significantly stimulated endothelial capillary network formation compared with the VEGF protein (P<0.0001). MSC infusion via the coronary vein prevented the progression of cardiac dysfunction in chronic MI. This favorable effect appeared to derive not from cell differentiation, but from enhanced neovascularization by angiogenic factors secreted from the MSCs.
Collapse
|
31
|
Carrade DD, Owens SD, Galuppo LD, Vidal MA, Ferraro GL, Librach F, Buerchler S, Friedman MS, Walker NJ, Borjesson DL. Clinicopathologic findings following intra-articular injection of autologous and allogeneic placentally derived equine mesenchymal stem cells in horses. Cytotherapy 2010; 13:419-30. [PMID: 21105841 DOI: 10.3109/14653249.2010.536213] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND AIMS The development of an allogeneic mesenchymal stem cell (MSC) product to treat equine disorders would be useful; however, there are limited in vivo safety data for horses. We hypothesized that the injection of self (autologous) and non-self (related allogeneic or allogeneic) MSC would not elicit significant alterations in physical examination, gait or synovial fluid parameters when injected into the joints of healthy horses. METHODS Sixteen healthy horses were used in this study. Group 1 consisted of foals (n = 6), group 2 consisted of their dams (n = 5) and group 3 consisted of half-siblings (n = 5) to group 1 foals. Prior to injection, MSC were phenotyped. Placentally derived MSC were injected into contralateral joints and MSC diluent was injected into a separate joint (control). An examination, including lameness evaluation and synovial fluid analysis, was performed at 0, 24, 48 and 72 h post-injection. RESULTS MSC were major histocompatibility complex (MHC) I positive, MHC II negative and CD86 negative. Injection of allogeneic MSC did not elicit a systemic response. Local responses such as joint swelling or lameness were minimal and variable. Intra-articular MSC injection elicited marked inflammation within the synovial fluid (as measured by nucleated cell count, neutrophil number and total protein concentration). However, there were no significant differences between the degree and type of inflammation elicited by self and non-self-MSC. CONCLUSIONS The healthy equine joint responds similarly to a single intra-articular injection of autologous and allogeneic MSC. This pre-clinical safety study is an important first step in the development of equine allogeneic stem cell therapies.
Collapse
Affiliation(s)
- Danielle D Carrade
- Department of Pathology, Microbiology and Immunology, University of California, Davis, California 95616, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Dubois C, Liu X, Claus P, Marsboom G, Pokreisz P, Vandenwijngaert S, Dépelteau H, Streb W, Chaothawee L, Maes F, Gheysens O, Debyser Z, Gillijns H, Pellens M, Vandendriessche T, Chuah M, Collen D, Verbeken E, Belmans A, Van de Werf F, Bogaert J, Janssens S. Differential Effects of Progenitor Cell Populations on Left Ventricular Remodeling and Myocardial Neovascularization After Myocardial Infarction. J Am Coll Cardiol 2010; 55:2232-43. [DOI: 10.1016/j.jacc.2009.10.081] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 10/05/2009] [Accepted: 10/05/2009] [Indexed: 12/16/2022]
|
33
|
Nakamuta JS, Danoviz ME, Marques FLN, dos Santos L, Becker C, Gonçalves GA, Vassallo PF, Schettert IT, Tucci PJF, Krieger JE. Cell therapy attenuates cardiac dysfunction post myocardial infarction: effect of timing, routes of injection and a fibrin scaffold. PLoS One 2009; 4:e6005. [PMID: 19547700 PMCID: PMC2695782 DOI: 10.1371/journal.pone.0006005] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2009] [Accepted: 05/06/2009] [Indexed: 01/09/2023] Open
Abstract
Background Cell therapy approaches for biologic cardiac repair hold great promises, although basic fundamental issues remain poorly understood. In the present study we examined the effects of timing and routes of administration of bone marrow cells (BMC) post-myocardial infarction (MI) and the efficacy of an injectable biopolymer scaffold to improve cardiac cell retention and function. Methodology/Principal Findings 99mTc-labeled BMC (6×106 cells) were injected by 4 different routes in adult rats: intravenous (IV), left ventricular cavity (LV), left ventricular cavity with temporal aorta occlusion (LV+) to mimic coronary injection, and intramyocardial (IM). The injections were performed 1, 2, 3, or 7 days post-MI and cell retention was estimated by γ-emission counting of the organs excised 24 hs after cell injection. IM injection improved cell retention and attenuated cardiac dysfunction, whereas IV, LV or LV* routes were somewhat inefficient (<1%). Cardiac BMC retention was not influenced by timing except for the IM injection that showed greater cell retention at 7 (16%) vs. 1, 2 or 3 (average of 7%) days post-MI. Cardiac cell retention was further improved by an injectable fibrin scaffold at day 3 post-MI (17 vs. 7%), even though morphometric and function parameters evaluated 4 weeks later displayed similar improvements. Conclusions/Significance These results show that cells injected post-MI display comparable tissue distribution profile regardless of the route of injection and that there is no time effect for cardiac cell accumulation for injections performed 1 to 3 days post-MI. As expected the IM injection is the most efficient for cardiac cell retention, it can be further improved by co-injection with a fibrin scaffold and it significantly attenuates cardiac dysfunction evaluated 4 weeks post myocardial infarction. These pharmacokinetic data obtained under similar experimental conditions are essential for further development of these novel approaches.
Collapse
Affiliation(s)
- Juliana S. Nakamuta
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Maria E. Danoviz
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Fabio L. N. Marques
- Radiopharmacy Laboratory, Nuclear Medicine Center, University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Leonardo dos Santos
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
- Cardiac Physiology and Pathophysiology Laboratory, Cardiology Division, Federal University of Sao Paulo, São Paulo, São Paulo, Brazil
| | - Claudia Becker
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Giovana A. Gonçalves
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Paula F. Vassallo
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Isolmar T. Schettert
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Paulo J. F. Tucci
- Cardiac Physiology and Pathophysiology Laboratory, Cardiology Division, Federal University of Sao Paulo, São Paulo, São Paulo, Brazil
| | - Jose E. Krieger
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|