1
|
Guo M, Watanabe T, Shinoka T. Injectable Stem Cell-Based Therapies for Myocardial Regeneration: A Review of the Literature. J Funct Biomater 2025; 16:152. [PMID: 40422817 DOI: 10.3390/jfb16050152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/28/2025] Open
Abstract
Stem cell-based therapies are an emerging treatment modality aimed at replenishing lost cardiomyocytes and improving myocardial function after cardiac injury. This review examines the current state of research on injectable stem cell therapies in the setting of cardiovascular disease given their relative simplicity and ability for deep myocardial tissue penetration. Various methods of cell delivery, ranging in level of invasiveness and procedural complexity, have been developed, and numerous cell types have been studied as potential sources of stem cells, each with distinct advantages and disadvantages. We discuss key challenges associated with this approach, including low stem cell retention after transplantation and the innovative biomolecular strategies that have been explored to address this issue. Overall, investigations into the application of stem cells toward cardiac regeneration remain predominantly in the preclinical stage with a number of small, early-phase clinical trials. However, continued scientific advancements in stem cell technology may provide transformative treatment options for patients with heart failure, offering improved survival and quality of life.
Collapse
Affiliation(s)
- Marissa Guo
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Tatsuya Watanabe
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Toshiharu Shinoka
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
- Department of Cardiothoracic Surgery, Nationwide Children's Hospital, Columbus, OH 43205, USA
| |
Collapse
|
2
|
Li W, Shi X, Zhang D, Hu J, Zhao S, Ye S, Wang J, Liu X, Zhang Q, Wang Z, Zhang Y, Yan L. Adipose derived mesenchymal stem cell-seeded regenerated silk fibroin scaffolds reverse liver fibrosis in mice. J Mater Chem B 2025; 13:4201-4213. [PMID: 40059659 DOI: 10.1039/d5tb00275c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Liver fibrosis (LF) is an important process in the progression of chronic liver disease to cirrhosis. We have previously demonstrated that a regenerated silk fibroin scaffold loaded with adipose-derived stem cells (RSF + ADSCs) can repair acute liver injury. In this study, we established a chronic LF animal model using carbon tetrachloride (CCl4) and a high-fat diet. We then investigated the liver repair capacity after transplanting RSF + ADSC scaffolds and RSF scaffolds onto the liver surface of mice. Compared with the control group, the concentrations of ALT and AST in the serum were significantly reduced in the RSF and RSF + ADSC groups. HE staining and Masson trichrome staining revealed a decrease in the SAF score in both the RSF and RSF + ADSC groups. Meanwhile, the biomarkers of blood vessels and bile ducts, such as CD34, ERG, muc1, and CK19, were significantly elevated in the RSF + ADSC group. Finally, transcriptome analysis showed that the PPAR signaling pathway, which inhibits liver fibrosis, was significantly upregulated in both the RSF and RSF + ADSC groups. Our study suggests that, compared with RSF scaffolds alone, RSF + ADSCs have a significant repair effect on chronic LF in mice.
Collapse
Affiliation(s)
- Weilong Li
- The Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, PR China.
| | - Xiaonan Shi
- The Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, PR China.
| | - Daxu Zhang
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, PR China
| | - Jingjing Hu
- The Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, PR China.
| | - Shuo Zhao
- Department of Critical Care Medicine,Aerospace Central Hospital,Beijing,, PR China
| | - Shujun Ye
- The Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, PR China.
| | - Jingyi Wang
- The Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, PR China.
| | - Xiaojiao Liu
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai, 201620, PR China.
| | - Qian Zhang
- School of nursing, Lanzhou University, Gansu 730000, PR China
| | - Zhanbo Wang
- Department of Pathology, Chinese PLA General Hospital, Beijing 100853, P. R. China.
| | - Yaopeng Zhang
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai, 201620, PR China.
| | - Li Yan
- The Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, PR China.
| |
Collapse
|
3
|
Tang XL, Wysoczynski M, Gumpert AM, Solanki M, Li Y, Wu WJ, Zheng S, Ruble H, Li H, Stowers H, Zheng S, Ou Q, Tanveer N, Slezak J, Kalra DK, Bolli R. Intravenous infusions of mesenchymal stromal cells have cumulative beneficial effects in a porcine model of chronic ischaemic cardiomyopathy. Cardiovasc Res 2024; 120:1939-1952. [PMID: 39163570 PMCID: PMC11630033 DOI: 10.1093/cvr/cvae173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 07/08/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024] Open
Abstract
AIMS The development of cell therapy as a widely available clinical option for ischaemic cardiomyopathy is hindered by the invasive nature of current cell delivery methods. Furthermore, the rapid disappearance of cells after transplantation provides a cogent rationale for using repeated cell doses, which, however, has not been done thus far in clinical trials because it is not feasible with invasive approaches. The goal of this translational study was to test the therapeutic utility of the intravenous route for cell delivery. METHODS AND RESULTS Pigs with chronic ischaemic cardiomyopathy induced by myocardial infarction received one or three intravenous doses of allogeneic bone marrow mesenchymal stromal cells (MSCs) or placebo 35 days apart. Rigour guidelines, including blinding and randomization, were strictly followed. A comprehensive assessment of left ventricular (LV) function was conducted with three independent methods (echocardiography, magnetic resonance imaging, and haemodynamic studies). The results demonstrate that three doses of MSCs improved both load-dependent and independent indices of LV function and reduced myocardial hypertrophy and fibrosis; in contrast, one dose failed to produce most of these benefits. CONCLUSIONS To our knowledge, this is the first study to show that intravenous infusion of a cell product improves LV function and structure in a large animal model of chronic ischaemic cardiomyopathy and that repeated infusions are necessary to produce robust effects. This study, conducted in a clinically relevant model, supports a new therapeutic strategy based on repeated intravenous infusions of allogeneic MSCs and provides a foundation for a first-in-human trial testing this strategy in patients with chronic ischaemic cardiomyopathy.
Collapse
Affiliation(s)
- Xian-Liang Tang
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Marcin Wysoczynski
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Anna M Gumpert
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Mitesh Solanki
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Yan Li
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Wen-Jian Wu
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Shirong Zheng
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Halina Ruble
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Hong Li
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Heather Stowers
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Shengnan Zheng
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Qinghui Ou
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Nida Tanveer
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Jan Slezak
- Centre of Experimental Medicine, Institute for Heart Research, Bratislava, Slovakia
| | - Dinesh K Kalra
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| |
Collapse
|
4
|
Takejima AL, Machado-Júnior PAB, Blume GG, Simeoni RB, Francisco JC, Tonial MS, Marqueze LFB, Noronha L, Olandoski M, Abdelwahid E, Carvalho KATDE, Pinho RA, Guarita-Souza LC. Bone-marrow mononuclear cells and acellular human amniotic membrane improve global cardiac function without inhibition of the NLRP3 Inflammasome in a rat model of heart failure. AN ACAD BRAS CIENC 2024; 96:e20230053. [PMID: 38451595 DOI: 10.1590/0001-3765202420230053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/19/2023] [Indexed: 03/08/2024] Open
Abstract
Recent studies have suggested that therapies with stem cells and amniotic membrane can modulate the inflammation following an ischemic injury in the heart. This study evaluated the effects of bone-marrow mononuclear cells (BMMC) and acellular human amniotic membrane (AHAM) on cardiac function and NLRP3 complex in a rat model of heart failure.On the 30th day,the echocardiographic showed improvements on ejection fraction and decreased pathological ventricular remodeling on BMMC and AHAM groups.Oxidative stress analysis was similar between the three groups,and the NLRP3 inflammasome activity were not decreased with the therapeutic use of both BMMC and AHAM,in comparison to the control group.
Collapse
Affiliation(s)
- Aline L Takejima
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Paulo André B Machado-Júnior
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Gustavo G Blume
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Rossana Baggio Simeoni
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Julio Cesar Francisco
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Murilo S Tonial
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Luis Felipe B Marqueze
- Pontifícia Universidade Católica do Paraná (PUCPR), Laboratory of Exercise Biochemistry in Health, School of Medicine, 1555 Imaculada Conceição Street, Prado Velho, 80215-901 Curitiba, PR, Brazil
| | - Lucia Noronha
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Marcia Olandoski
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Eltyeb Abdelwahid
- Northwestern University, Feinberg School of Medicine, Feinberg Cardiovascular Research Institute, 303 E. Chicago Ave., Tarry 14-725, 60611 Chicago, IL, USA
| | - Katherine A T DE Carvalho
- The Pelé Pequeno Príncipe Institute, Cell Therapy and Biotechnology in Regenerative Medicine Department, Child and Adolescent Health Research & Pequeno Príncipe Faculties, 1632 Silva Jardim Ave., Água Verde, 80240-020 Curitiba, PR, Brazil
| | - Ricardo A Pinho
- Pontifícia Universidade Católica do Paraná (PUCPR), Laboratory of Exercise Biochemistry in Health, School of Medicine, 1555 Imaculada Conceição Street, Prado Velho, 80215-901 Curitiba, PR, Brazil
| | - Luiz César Guarita-Souza
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| |
Collapse
|
5
|
Tan FH, Bronner ME. Regenerative loss in the animal kingdom as viewed from the mouse digit tip and heart. Dev Biol 2024; 507:44-63. [PMID: 38145727 PMCID: PMC10922877 DOI: 10.1016/j.ydbio.2023.12.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/30/2023] [Accepted: 12/19/2023] [Indexed: 12/27/2023]
Abstract
The myriad regenerative abilities across the animal kingdom have fascinated us for centuries. Recent advances in developmental, molecular, and cellular biology have allowed us to unearth a surprising diversity of mechanisms through which these processes occur. Developing an all-encompassing theory of animal regeneration has thus proved a complex endeavor. In this chapter, we frame the evolution and loss of animal regeneration within the broad developmental constraints that may physiologically inhibit regenerative ability across animal phylogeny. We then examine the mouse as a model of regeneration loss, specifically the experimental systems of the digit tip and heart. We discuss the digit tip and heart as a positionally-limited system of regeneration and a temporally-limited system of regeneration, respectively. We delve into the physiological processes involved in both forms of regeneration, and how each phase of the healing and regenerative process may be affected by various molecular signals, systemic changes, or microenvironmental cues. Lastly, we also discuss the various approaches and interventions used to induce or improve the regenerative response in both contexts, and the implications they have for our understanding regenerative ability more broadly.
Collapse
Affiliation(s)
- Fayth Hui Tan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
6
|
Shen Y, Kim IM, Tang Y. Re: Single-cell transcriptome analysis for characterizing primary Sca-1 positive, non-endothelial cardiac cells. J Mol Cell Cardiol 2023; 182:28-29. [PMID: 37454412 PMCID: PMC11987645 DOI: 10.1016/j.yjmcc.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/06/2023] [Accepted: 07/01/2023] [Indexed: 07/18/2023]
Affiliation(s)
- Yan Shen
- Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Il-Man Kim
- Anatomy, Cell Biology & Physiology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Yaoliang Tang
- Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
7
|
Luis J, Eastlake K, Lamb WDB, Limb GA, Jayaram H, Khaw PT. Cell-Based Therapies for Glaucoma. Transl Vis Sci Technol 2023; 12:23. [PMID: 37494052 PMCID: PMC10383000 DOI: 10.1167/tvst.12.7.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 07/27/2023] Open
Abstract
Glaucomatous optic neuropathy (GON) is the major cause of irreversible visual loss worldwide and can result from a range of disease etiologies. The defining features of GON are retinal ganglion cell (RGC) degeneration and characteristic cupping of the optic nerve head (ONH) due to tissue remodeling, while intraocular pressure remains the only modifiable GON risk factor currently targeted by approved clinical treatment strategies. Efforts to understand the mechanisms that allow species such as the zebrafish to regenerate their retinal cells have greatly increased our understanding of regenerative signaling pathways. However, proper integration within the retina and projection to the brain by the newly regenerated neuronal cells remain major hurdles. Meanwhile, a range of methods for in vitro differentiation have been developed to derive retinal cells from a variety of cell sources, including embryonic and induced pluripotent stem cells. More recently, there has been growing interest in the implantation of glial cells as well as cell-derived products, including neurotrophins, microRNA, and extracellular vesicles, to provide functional support to vulnerable structures such as RGC axons and the ONH. These approaches offer the advantage of not relying upon the replacement of degenerated cells and potentially targeting earlier stages of disease pathogenesis. In order to translate these techniques into clinical practice, appropriate cell sourcing, robust differentiation protocols, and accurate implantation methods are crucial to the success of cell-based therapy in glaucoma. Translational Relevance: Cell-based therapies for glaucoma currently under active development include the induction of endogenous regeneration, implantation of exogenously derived retinal cells, and utilization of cell-derived products to provide functional support.
Collapse
Affiliation(s)
- Joshua Luis
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - Karen Eastlake
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - William D. B. Lamb
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - G. Astrid Limb
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - Hari Jayaram
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - Peng T. Khaw
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| |
Collapse
|
8
|
Clavellina D, Balkan W, Hare JM. Stem cell therapy for acute myocardial infarction: Mesenchymal Stem Cells and induced Pluripotent Stem Cells. Expert Opin Biol Ther 2023; 23:951-967. [PMID: 37542462 PMCID: PMC10837765 DOI: 10.1080/14712598.2023.2245329] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/07/2023]
Abstract
INTRODUCTION Acute myocardial infarction (AMI) remains a leading cause of death in the United States. The limited capacity of cardiomyocytes to regenerate and the restricted contractility of scar tissue after AMI are not addressed by current pharmacologic interventions. Mesenchymal stem/stromal cells (MSCs) have emerged as a promising therapeutic approach due to their low antigenicity, ease of harvesting, and efficacy and safety in preclinical and clinical studies, despite their low survival and engraftment rates. Other stem cell types, such as induced pluripotent stem cells (iPSCs) also show promise, and optimizing cardiac repair requires integrating emerging technologies and strategies. AREAS COVERED This review offers insights into advancing cell-based therapies for AMI, emphasizing meticulously planned trials with a standardized definition of AMI, for a bench-to-bedside approach. We critically evaluate fundamental studies and clinical trials to provide a comprehensive overview of the advances, limitations and prospects for cell-based therapy in AMI. EXPERT OPINION MSCs continue to show potential promise for treating AMI and its sequelae, but addressing their low survival and engraftment rates is crucial for clinical success. Integrating emerging technologies such as pluripotent stem cells and conducting well-designed trials will harness the full potential of cell-based therapy in AMI management. Collaborative efforts are vital to developing effective stem cell therapies for AMI patients.
Collapse
Affiliation(s)
- Diana Clavellina
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
9
|
Kahrizi MS, Mousavi E, Khosravi A, Rahnama S, Salehi A, Nasrabadi N, Ebrahimzadeh F, Jamali S. Recent advances in pre-conditioned mesenchymal stem/stromal cell (MSCs) therapy in organ failure; a comprehensive review of preclinical studies. Stem Cell Res Ther 2023; 14:155. [PMID: 37287066 DOI: 10.1186/s13287-023-03374-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 05/10/2023] [Indexed: 06/09/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs)-based therapy brings the reassuring capability to regenerative medicine through their self-renewal and multilineage potency. Also, they secret a diversity of mediators, which are complicated in moderation of deregulated immune responses, and yielding angiogenesis in vivo. Nonetheless, MSCs may lose biological performance after procurement and prolonged expansion in vitro. Also, following transplantation and migration to target tissue, they encounter a harsh milieu accompanied by death signals because of the lack of proper tensegrity structure between the cells and matrix. Accordingly, pre-conditioning of MSCs is strongly suggested to upgrade their performances in vivo, leading to more favored transplantation efficacy in regenerative medicine. Indeed, MSCs ex vivo pre-conditioning by hypoxia, inflammatory stimulus, or other factors/conditions may stimulate their survival, proliferation, migration, exosome secretion, and pro-angiogenic and anti-inflammatory characteristics in vivo. In this review, we deliver an overview of the pre-conditioning methods that are considered a strategy for improving the therapeutic efficacy of MSCs in organ failures, in particular, renal, heart, lung, and liver.
Collapse
Affiliation(s)
| | - Elnaz Mousavi
- Department of Endodontics, School of Dentistry, Guilan University of Medical Sciences, Rasht, Iran
| | - Armin Khosravi
- Department of Periodontics, Dental School, Islamic Azad University, Isfahan (Khorasgan) Branch, Isfahan, Iran
| | - Sara Rahnama
- Department of Pediatric Dentistry, School of Dentistry, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Salehi
- Department of Oral and Maxillofacial Radiology, School of Dentistry, Islamic Azad University, Isfahan (Khorasgan) Branch, Isfahan, Iran
| | - Navid Nasrabadi
- Department of Endodontics, School of Dentistry, Birjand University of Medical Sciences, Birjand, Iran
| | - Farnoosh Ebrahimzadeh
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Samira Jamali
- Department of Endodontics, Stomatological Hospital, College of Stomatology, Xi'an Jiaotong University, Shaanxi, People's Republic of China.
| |
Collapse
|
10
|
Prakash N, Kim J, Jeon J, Kim S, Arai Y, Bello AB, Park H, Lee SH. Progress and emerging techniques for biomaterial-based derivation of mesenchymal stem cells (MSCs) from pluripotent stem cells (PSCs). Biomater Res 2023; 27:31. [PMID: 37072836 PMCID: PMC10114339 DOI: 10.1186/s40824-023-00371-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/26/2023] [Indexed: 04/20/2023] Open
Abstract
The use of mesenchymal stem cells (MSCs) for clinical purposes has skyrocketed in the past decade. Their multilineage differentiation potentials and immunomodulatory properties have facilitated the discovery of therapies for various illnesses. MSCs can be isolated from infant and adult tissue sources, which means they are easily available. However, this raises concerns because of the heterogeneity among the various MSC sources, which limits their effective use. Variabilities arise from donor- and tissue-specific differences, such as age, sex, and tissue source. Moreover, adult-sourced MSCs have limited proliferation potentials, which hinders their long-term therapeutic efficacy. These limitations of adult MSCs have prompted researchers to develop a new method for generating MSCs. Pluripotent stem cells (PSCs), such as embryonic stem cells and induced PSCs (iPSCs), can differentiate into various types of cells. Herein, a thorough review of the characteristics, functions, and clinical importance of MSCs is presented. The existing sources of MSCs, including adult- and infant-based sources, are compared. The most recent techniques for deriving MSCs from iPSCs, with a focus on biomaterial-assisted methods in both two- and three-dimensional culture systems, are listed and elaborated. Finally, several opportunities to develop improved methods for efficiently producing MSCs with the aim of advancing their various clinical applications are described.
Collapse
Affiliation(s)
- Nityanand Prakash
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Jiseong Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Jieun Jeon
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Siyeon Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Alvin Bacero Bello
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, 06911, Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea.
| |
Collapse
|
11
|
Shaik R, Xu J, Wang Y, Hong Y, Zhang G. Fibrin-Enriched Cardiac Extracellular Matrix Hydrogel Promotes In Vitro Angiogenesis. ACS Biomater Sci Eng 2023; 9:877-888. [PMID: 36630688 PMCID: PMC10064974 DOI: 10.1021/acsbiomaterials.2c01148] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Angiogenesis is essential for cardiac repair after myocardial infarction. Promoting angiogenesis has been demonstrated as an effective approach for myocardial infarction treatment. Several different strategies for inducing myocardial angiogenesis have been explored, including exogenous delivery of angiogenic genes, proteins, microRNAs, cells, and extracellular vesicles. Various types of injectable hydrogels have been investigated for cardiac tissue repair. One of the most promising injectable hydrogels in cardiac regeneration is a cardiac extracellular matrix hydrogel that is derived from decellularized porcine myocardium. It can be delivered minimally invasively via transendocardial delivery. The safety and efficacy of cardiac extracellular matrix hydrogels have been shown in small and large animal myocardial infarction models as well as clinical trials. The main mechanisms underlying the therapeutic benefits of cardiac extracellular matrix hydrogels have been elucidated and involved in the modulation of the immune response, downregulation of pathways related to heart failure progression and fibrosis, upregulation of genes important for cardiac muscle contraction, and enhancing cardiomyocyte differentiation and maturation from stem cells. However, no potent capillary network formation induced by cardiac extracellular matrix hydrogels has been reported. In this study, we tested the feasibility of incorporating a fibrin matrix into cardiac extracellular matrix hydrogels to improve the angiogenic properties of the hydrogel. Our in vitro results demonstrate that fibrin-enriched cardiac extracellular matrix hydrogels can induce robust endothelial cell tube formation from human umbilical vein endothelial cells and promote the sprouting of human mesenchymal stem cell spheroids. The obtained information from this study is very critical toward the future in vivo evaluation of fibrin-enriched cardiac extracellular matrix hydrogels in promoting myocardial angiogenesis.
Collapse
Affiliation(s)
- Rubia Shaik
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Jiazhu Xu
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Yong Wang
- Department of Biomedical Engineering, Pennsylvania State University, State College, University Park, Pennsylvania 16801, United States
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Ge Zhang
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
12
|
Mechanotransduction of mesenchymal stem cells (MSCs) during cardiomyocytes differentiation. Heliyon 2022; 8:e11624. [DOI: 10.1016/j.heliyon.2022.e11624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 09/15/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
|
13
|
Handley EL, Callanan A. Modulation of Tissue Microenvironment Following Myocardial Infarction. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Ella Louise Handley
- Institute for Bioengineering School of Engineering University of Edinburgh Edinburgh EH9 3DW UK
| | - Anthony Callanan
- Institute for Bioengineering School of Engineering University of Edinburgh Edinburgh EH9 3DW UK
| |
Collapse
|
14
|
Lee H, Lee TW, Chandrasekharan A, Sung SE, Yim SG, Kim S, Seong KY, Seo MS, Yang SY. Injectable Self-Crosslinkable Thiolated Hyaluronic Acid for Stem Cell Therapy of Atopic Dermatitis. ACS Biomater Sci Eng 2022; 8:1613-1622. [PMID: 35245045 DOI: 10.1021/acsbiomaterials.1c01374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Stem cell therapies offer great promise in regenerative medicine to reinstate the normal function of diseased tissue, thereby avoiding the need for replacement. In stem cell therapies, damaged cells are replaced or restored by regulating inflammation and the immune system. However, the low survival rate and local retention of transplanted cells pose a significant challenge. In this study, injectable self-crosslinkable hydrogels using thiol-functionalized hyaluronic acid (HA-SH) were developed to improve the efficacy of mesenchymal stem cells (MSCs) for treating atopic dermatitis (AD)-related inflammatory lesions. The gelation kinetics and mechanical properties of HA-SH hydrogels were easily tuned by varying the concentration of the polymer in the precursor solution before injection. The MSC-laden HA-SH hydrogels exhibited high cell viability (>80%) for 1 week and good in vivo biocompatibility after implantation beneath the mouse skin. Moreover, the MSC-laden HA-SH hydrogel showed increased expression of anti-inflammatory cytokines, which can alleviate the immune response. In an AD animal model, a reduction in epidermal thickness and mast cell infiltration was achieved by applying a self-crosslinkable HA-SH solution including MSCs. This HA-based injectable hydrogel represents a potential carrier of stem cells, and its strong immunomodulation capabilities can be utilized for treating inflammation-related diseases.
Collapse
Affiliation(s)
- Hyeseon Lee
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea
| | - Tae Wook Lee
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea
| | - Ajeesh Chandrasekharan
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea
| | - Soo-Eun Sung
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea.,Department of Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea
| | - Sang-Gu Yim
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea
| | - Sodam Kim
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea
| | - Keum-Yong Seong
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea
| | - Min-Soo Seo
- Department of Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea
| | - Seung Yun Yang
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea
| |
Collapse
|
15
|
Tracy EP, Stielberg V, Rowe G, Benson D, Nunes SS, Hoying JB, Murfee WL, LeBlanc AJ. State of the field: cellular and exosomal therapeutic approaches in vascular regeneration. Am J Physiol Heart Circ Physiol 2022; 322:H647-H680. [PMID: 35179976 PMCID: PMC8957327 DOI: 10.1152/ajpheart.00674.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 01/19/2023]
Abstract
Pathologies of the vasculature including the microvasculature are often complex in nature, leading to loss of physiological homeostatic regulation of patency and adequate perfusion to match tissue metabolic demands. Microvascular dysfunction is a key underlying element in the majority of pathologies of failing organs and tissues. Contributing pathological factors to this dysfunction include oxidative stress, mitochondrial dysfunction, endoplasmic reticular (ER) stress, endothelial dysfunction, loss of angiogenic potential and vascular density, and greater senescence and apoptosis. In many clinical settings, current pharmacologic strategies use a single or narrow targeted approach to address symptoms of pathology rather than a comprehensive and multifaceted approach to address their root cause. To address this, efforts have been heavily focused on cellular therapies and cell-free therapies (e.g., exosomes) that can tackle the multifaceted etiology of vascular and microvascular dysfunction. In this review, we discuss 1) the state of the field in terms of common therapeutic cell population isolation techniques, their unique characteristics, and their advantages and disadvantages, 2) common molecular mechanisms of cell therapies to restore vascularization and/or vascular function, 3) arguments for and against allogeneic versus autologous applications of cell therapies, 4) emerging strategies to optimize and enhance cell therapies through priming and preconditioning, and, finally, 5) emerging strategies to bolster therapeutic effect. Relevant and recent clinical and animal studies using cellular therapies to restore vascular function or pathologic tissue health by way of improved vascularization are highlighted throughout these sections.
Collapse
Affiliation(s)
- Evan Paul Tracy
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Virginia Stielberg
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Gabrielle Rowe
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Daniel Benson
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
- Department of Bioengineering, University of Louisville, Louisville, Kentucky
| | - Sara S Nunes
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Heart & Stroke/Richard Lewar Centre of Excellence, University of Toronto, Toronto, Ontario, Canada
| | - James B Hoying
- Advanced Solutions Life Sciences, Manchester, New Hampshire
| | - Walter Lee Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida
| | - Amanda Jo LeBlanc
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
16
|
Sharma A, Gupta S, Archana S, Verma RS. Emerging Trends in Mesenchymal Stem Cells Applications for Cardiac Regenerative Therapy: Current Status and Advances. Stem Cell Rev Rep 2022; 18:1546-1602. [PMID: 35122226 DOI: 10.1007/s12015-021-10314-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2021] [Indexed: 12/29/2022]
Abstract
Irreversible myocardium infarction is one of the leading causes of cardiovascular disease (CVD) related death and its quantum is expected to grow in coming years. Pharmacological intervention has been at the forefront to ameliorate injury-related morbidity and mortality. However, its outcomes are highly skewed. As an alternative, stem cell-based tissue engineering/regenerative medicine has been explored quite extensively to regenerate the damaged myocardium. The therapeutic modality that has been most widely studied both preclinically and clinically is based on adult multipotent mesenchymal stem cells (MSC) delivered to the injured heart. However, there is debate over the mechanistic therapeutic role of MSC in generating functional beating cardiomyocytes. This review intends to emphasize the role and use of MSC in cardiac regenerative therapy (CRT). We have elucidated in detail, the various aspects related to the history and progress of MSC use in cardiac tissue engineering and its multiple strategies to drive cardiomyogenesis. We have further discussed with a focus on the various therapeutic mechanism uncovered in recent times that has a significant role in ameliorating heart-related problems. We reviewed recent and advanced technologies using MSC to develop/create tissue construct for use in cardiac regenerative therapy. Finally, we have provided the latest update on the usage of MSC in clinical trials and discussed the outcome of such studies in realizing the full potential of MSC use in clinical management of cardiac injury as a cellular therapy module.
Collapse
Affiliation(s)
- Akriti Sharma
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai, 600036, Tamil Nadu, India
| | - Santosh Gupta
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai, 600036, Tamil Nadu, India
| | - S Archana
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai, 600036, Tamil Nadu, India
| | - Rama Shanker Verma
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai, 600036, Tamil Nadu, India.
| |
Collapse
|
17
|
Bagno LL, Salerno AG, Balkan W, Hare JM. Mechanism of Action of Mesenchymal Stem Cells (MSCs): impact of delivery method. Expert Opin Biol Ther 2021; 22:449-463. [PMID: 34882517 DOI: 10.1080/14712598.2022.2016695] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Mesenchymal stromal cells (MSCs; AKA mesenchymal stem cells) stimulate healing and reduce inflammation. Promising therapeutic responses are seen in many late-phase clinical trials, but others have not satisfied their primary endpoints, making translation of MSCs into clinical practice difficult. These inconsistencies may be related to the route of MSC delivery, lack of product optimization, or varying background therapies received in clinical trials over time. AREAS COVERED Here we discuss the different routes of MSC delivery, highlighting the proposed mechanism(s) of therapeutic action as well as potential safety concerns. PubMed search criteria used: MSC plus: local administration; routes of administration; delivery methods; mechanism of action; therapy in different diseases. EXPERT OPINION Direct injection of MSCs using a controlled local delivery approach appears to have benefits in certain disease states, but further studies are required to make definitive conclusions regarding the superiority of one delivery method over another.
Collapse
Affiliation(s)
- Luiza L Bagno
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alessandro G Salerno
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami
| |
Collapse
|
18
|
Hopes and Hurdles of Employing Mesenchymal Stromal Cells in the Treatment of Cardiac Fibrosis. Int J Mol Sci 2021; 22:ijms222313000. [PMID: 34884805 PMCID: PMC8657815 DOI: 10.3390/ijms222313000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 12/04/2022] Open
Abstract
Excessive cardiac fibrosis plays a crucial role in almost all types of heart disease. Generally, cardiac fibrosis is a scarring process triggered in response to stress, injury, or aging and is characterized by the accumulation of activated myofibroblasts that deposit high levels of extracellular matrix proteins in the myocardium. While it is beneficial for cardiac repair in the short term, it can also result in pathological remodeling, tissue stiffening, and cardiac dysfunction, contributing to the progression of heart failure, arrhythmia, and sudden cardiac death. Despite its high prevalence, there is a lack of effective and safe therapies that specifically target myofibroblasts to inhibit or even reverse pathological cardiac fibrosis. In the past few decades, cell therapy has been under continuous evaluation as a potential treatment strategy, and several studies have shown that transplantation of mesenchymal stromal cells (MSCs) can reduce cardiac fibrosis and improve heart function. Mechanistically, it is believed that the heart benefits from MSC therapy by stimulating innate anti-fibrotic and regenerative reactions. The mechanisms of action include paracrine signaling and cell-to-cell interactions. In this review, we provide an overview of the anti-fibrotic properties of MSCs and approaches to enhance them and discuss future directions of MSCs for the treatment of cardiac fibrosis.
Collapse
|
19
|
Functions of Mesenchymal Stem Cells in Cardiac Repair. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1312:39-50. [PMID: 33330961 DOI: 10.1007/5584_2020_598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Myocardial infarction (MI) and heart failure (HF) are significant contributors of mortality worldwide. Mesenchymal stem cells (MSCs) hold a great potential for cardiac regenerative medicine-based therapies. Their therapeutic potential has been widely investigated in various in-vitro and in-vivo preclinical models. Besides, they have been tested in clinical trials of MI and HF with various outcomes. Differentiation to lineages of cardiac cells, neovascularization, anti-fibrotic, anti-inflammatory, anti-apoptotic and immune modulatory effects are the main drivers of MSC functions during cardiac repair. However, the main mechanisms regulating these functions and cross-talk between cells are not fully known yet. Increasing line of evidence also suggests that secretomes of MSCs and/or their extracellular vesicles play significant roles in a paracrine manner while mediating these functions. This chapter aims to summarize and highlight cardiac repair functions of MSCs during cardiac repair.
Collapse
|
20
|
Petryk N, Shevchenko O. Anti-inflammatory Activity of Mesenchymal Stem Cells in λ-Carrageenan-Induced Chronic Inflammation in Rats: Reactions of the Blood System, Leukocyte-Monocyte Ratio. Inflammation 2021; 43:1893-1901. [PMID: 32462547 DOI: 10.1007/s10753-020-01262-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The variety of chronic diseases caused by a chronic inflammatory process is an unresolved problem in developed countries. Due to this, modern medicine lacks effective, pathogenetic mechanisms of treatment or at least improvement of the quality of life of people with so-called diseases of civilization. The purpose of this study was to investigate the anti-inflammatory and immunomodulatory ability of mesenchymal stem cells (MSCs) in a model of λ-carrageenan secondary chronic inflammation in rats. MSCs derived from rat femoral epiphysis were used. At the current level of medicine, many highly specific markers of chronic inflammation are available that will also be studied later (α-TNF, IL 6, C-reactive protein); however, this article will consider the study of the most accessible but at the same time very informative indicators of the inflammatory process-a common total blood count-leukocytes, leukocyte formula, and LMR. The study was performed on 132 male Wistar rats weighing 180-200 g, which were divided into 12 groups. The inflammation was caused by the introduction of 10 mg λ-carrageenan intramuscular in right hip. After induction of edema, the experimental groups of rats were administered MSCs into the inflamed site, in the amount of 1-2 million cells once. Blood sampling was performed from 6 h to 28 days. With one-way ANOVA followed by Tukey-Kramer multiple comparisons test, p < 0.05, we compared our groups of animals. In the detailed dynamics of inflammation, from the 6th hour to the 28th day (ten terms), the reactions of the blood system and their mechanisms were investigated. There were intact rats-6 animals without any interventions, as well as rats administered MSCs without causing inflammation (6 animals) in experiment. In this experiment, the lymphocyte-monocyte ratio in rats is described for the first time, demonstrating the suppression of chronic inflammation by means of MSCs.
Collapse
Affiliation(s)
- Nataliia Petryk
- Department of Pathology, Kharkiv National Medical University, Kharkiv, Ukraine.
| | | |
Collapse
|
21
|
Shi H, Xue T, Yang Y, Jiang C, Huang S, Yang Q, Lei D, You Z, Jin T, Wu F, Zhao Q, Ye X. Microneedle-mediated gene delivery for the treatment of ischemic myocardial disease. SCIENCE ADVANCES 2020; 6:eaaz3621. [PMID: 32596444 PMCID: PMC7299628 DOI: 10.1126/sciadv.aaz3621] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 05/04/2020] [Indexed: 06/01/2023]
Abstract
Cardiovascular disorders are still the primary cause of mortality worldwide. Although intramyocardial injection can effectively deliver agents to the myocardium, this approach is limited because of its restriction to needle-mediated injection and the minor retention of agents in the myocardium. Here, we engineered phase-transition microneedles (MNs) coated with adeno-associated virus (AAV) and achieved homogeneous distribution of AAV delivery. Bioluminescence imaging revealed the successful delivery and transfection of AAV-luciferase. AAV-green fluorescent protein-transfected cardiomyocytes were homogeneously distributed on postoperative day 28. AAV-vascular endothelial growth factor (VEGF)-loaded MNs improved heart function by enhancing VEGF expression, promoting functional angiogenesis, and activating the Akt signaling pathway. The results indicated the superiority of MNs over direct muscle injection. Consequently, MNs might emerge as a promising tool with great versatility for delivering various agents to treat ischemic myocardial disease.
Collapse
Affiliation(s)
- Hongpeng Shi
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Tong Xue
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Yang Yang
- Department of Cardiothoracic Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P. R. China
| | - Chenyu Jiang
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Shixing Huang
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Qi Yang
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Dong Lei
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, International Joint Laboratory for Advanced Fiber and Low-Dimension Materials, College of Materials Science and Engineering Donghua University, Shanghai 201620, P. R. China
| | - Zhengwei You
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, International Joint Laboratory for Advanced Fiber and Low-Dimension Materials, College of Materials Science and Engineering Donghua University, Shanghai 201620, P. R. China
| | - Tuo Jin
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Fei Wu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Qiang Zhao
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Xiaofeng Ye
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| |
Collapse
|
22
|
Guo Y, Yu Y, Hu S, Chen Y, Shen Z. The therapeutic potential of mesenchymal stem cells for cardiovascular diseases. Cell Death Dis 2020; 11:349. [PMID: 32393744 PMCID: PMC7214402 DOI: 10.1038/s41419-020-2542-9] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are derived from a wide range of sources and easily isolated and cultured. MSCs have the capacity for in vitro amplification and self-renewal, low immunogenicity and immunomodulatory properties, and under certain conditions, MSCs can be differentiated into a variety of cells. In the cardiovascular system, MSCs can protect the myocardium by reducing the level of inflammation, promoting the differentiation of myocardial cells around infarct areas and angiogenesis, increasing apoptosis resistance, and inhibiting fibrosis, which are ideal qualities for cardiovascular repair. Preclinical studies have shown that MSCs can be transplanted and improve cardiac repair, but challenges, such as their low rate of migration to the ischemic myocardium, low tissue retention, and low survival rate after transplantation, remain. This article reviews the potential and methods of MSC transplantation in the treatment of cardiovascular diseases (CVDs) and the challenges of the clinical use of MSCs.
Collapse
Affiliation(s)
- Yajun Guo
- Institute for Cardiovascular Science, Soochow University, Suzhou 215006, China.,Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, Suzhou 215006, China
| | - Yunsheng Yu
- Institute for Cardiovascular Science, Soochow University, Suzhou 215006, China.,Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, Suzhou 215006, China
| | - Shijun Hu
- Institute for Cardiovascular Science, Soochow University, Suzhou 215006, China. .,Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, Suzhou 215006, China. .,State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou 215123, China. .,Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, China.
| | - Yueqiu Chen
- Institute for Cardiovascular Science, Soochow University, Suzhou 215006, China. .,Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, Suzhou 215006, China.
| | - Zhenya Shen
- Institute for Cardiovascular Science, Soochow University, Suzhou 215006, China. .,Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, Suzhou 215006, China.
| |
Collapse
|
23
|
Zhang J, Cheng T, Chen Y, Gao F, Guan F, Yao M. A chitosan-based thermosensitive scaffold loaded with bone marrow-derived mesenchymal stem cells promotes motor function recovery in spinal cord injured mice. Biomed Mater 2020; 15:035020. [DOI: 10.1088/1748-605x/ab785f] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
24
|
Effect of resveratrol combined with atorvastatin on re-endothelialization after drug-eluting stents implantation and the underlying mechanism. Life Sci 2020; 245:117349. [PMID: 31981632 DOI: 10.1016/j.lfs.2020.117349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 11/20/2022]
Abstract
AIMS To explore whether the combination of atorvastatins and resveratrol is superior to each individual drug alone regarding re-endothelialization after drug-eluting stents (DESs) implantation. MATERIALS AND METHODS Ninety-four rabbits were randomized into control, atorvastatin, resveratrol, and combined medication groups. Abdominal aorta injury was induced via ballooning, followed by DES implantation. Neointimal formation and re-endothelialization after stent implantation were assessed via optical coherence tomography and scanning electron microscopy. The effects of resveratrol and atorvastatin on bone marrow-derived mesenchymal derived stem cells (BMSCs) were assessed. KEY FINDINGS Compared with the findings in the resveratrol and atorvastatin groups, the neointimal area and mean neointimal thickness were greater in the combined medication group, which also exhibited improved re-endothelialization. Compared with the effects of monotherapy, combined treatment further protected BMSCs against rapamycin-induced apoptosis and improved cell migration. Combined medication significantly upregulated Akt, p-Akt, eNOS, p-eNOS, and CXCR4 expression in BMSCs compared with the effects of monotherapy, and these effects were abolished by the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002. SIGNIFICANCE The combination of atorvastatin and resveratrol has the potential of accelerating re-endothelialization after stent implantation, reducing the risk of thrombosis and improving the safety of DESs.
Collapse
|
25
|
von Bahr V, Millar JE, Malfertheiner MV, Ki KK, Passmore MR, Bartnikowski N, Redd MA, Cavaye M, Suen JY, McAuley DF, Fraser JF. Mesenchymal stem cells may ameliorate inflammation in an ex vivo model of extracorporeal membrane oxygenation. Perfusion 2020; 34:15-21. [PMID: 30966907 DOI: 10.1177/0267659119830857] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Mesenchymal stem cells exhibit immunomodulatory properties which are currently being investigated as a novel treatment option for Acute Respiratory Distress Syndrome. However, the feasibility and efficacy of mesenchymal stem cell therapy in the setting of extracorporeal membrane oxygenation is poorly understood. This study aimed to characterise markers of innate immune activation in response to mesenchymal stem cells during an ex vivo simulation of extracorporeal membrane oxygenation. METHODS Ex vivo extracorporeal membrane oxygenation simulations (n = 10) were conducted using a commercial extracorporeal circuit with a CO2-enhanced fresh gas supply and donor human whole blood. Heparinised circuits (n = 4) were injected with 40 × 106-induced pluripotent stem cell-derived human mesenchymal stem cells, while the remainder (n = 6) acted as controls. Simulations were maintained, under physiological conditions, for 240 minutes. Circuits were sampled at 15, 30, 60, 120 and 240 minutes and assessed for levels of interleukin-1β, interleukin-6, interleukin-8, interleukin-10, tumour necrosis factor-α, transforming growth factor-β1, myeloperoxidase and α-Defensin-1. In addition, haemoglobin, platelet and leukocyte counts were performed. RESULTS There was a trend towards reduced levels of pro-inflammatory cytokines in mesenchymal stem cell-treated circuits and a significant increase in transforming growth factor-β1. Blood cells and markers of neutrophil activation were reduced in mesenchymal stem cell circuits during the length of the simulation. As previously reported, the addition of mesenchymal stem cells resulted in a reduction of flow and increased trans-oxygenator pressures in comparison to controls. CONCLUSIONS The addition of mesenchymal stem cells during extracorporeal membrane oxygenation may cause an increase in transforming growth factor-β1. This is despite their ability to adhere to the membrane oxygenator. Further studies are required to confirm these findings.
Collapse
Affiliation(s)
- Viktor von Bahr
- 1 Critical Care Research Group, The Prince Charles Hospital, The University of Queensland, Brisbane, QLD, Australia
- 2 Section for Anesthesiology and Intensive Care Medicine, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jonathan E Millar
- 1 Critical Care Research Group, The Prince Charles Hospital, The University of Queensland, Brisbane, QLD, Australia
- 3 Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Maximillian V Malfertheiner
- 1 Critical Care Research Group, The Prince Charles Hospital, The University of Queensland, Brisbane, QLD, Australia
- 4 Department of Internal Medicine II, Cardiology and Pneumology, University Medical Center Regensburg, Regensburg, Germany
| | - Katrina K Ki
- 1 Critical Care Research Group, The Prince Charles Hospital, The University of Queensland, Brisbane, QLD, Australia
| | - Margaret R Passmore
- 1 Critical Care Research Group, The Prince Charles Hospital, The University of Queensland, Brisbane, QLD, Australia
| | - Nicole Bartnikowski
- 1 Critical Care Research Group, The Prince Charles Hospital, The University of Queensland, Brisbane, QLD, Australia
| | - Meredith A Redd
- 5 Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Michael Cavaye
- 1 Critical Care Research Group, The Prince Charles Hospital, The University of Queensland, Brisbane, QLD, Australia
| | - Jacky Y Suen
- 1 Critical Care Research Group, The Prince Charles Hospital, The University of Queensland, Brisbane, QLD, Australia
| | - Danny F McAuley
- 3 Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - John F Fraser
- 1 Critical Care Research Group, The Prince Charles Hospital, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
26
|
Comparative Analysis of the Healing Effects of the Transplanted Cell Sheets to the Experimentally Injured Maxillary Sinuses. J Craniofac Surg 2019; 30:2285-2292. [PMID: 31232985 DOI: 10.1097/scs.0000000000005638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND In our study, the authors aimed to obtain a live and functional sinus epithelium with mesenchymal stem cells and nasal mucosa epithelial cells from rabbits which are cultured in temperature-responsive culture plates to get a single-layer. METHODOLOGY/PRINCIPAL Twenty-two female New Zealand rabbits were included in the study. Two of them were used to obtain mesenchymal stem cells. A total of 40 maxillary sinuses were randomly divided into 5 groups: 1) control group which is used to investigate normal rabbit maxillary mucosa, 2) secondary healing group, 3) mesenchymal stem cell graft group, 4) differentiated mesenchymal stem cell group, and 5) nasal mucosal graft group. The animals were sacrificed at the 28th day after the surgery.Scanning electron microscopy, transmission electron microscopy, and immunohistochemical investigations were performed. RESULTS With these investigations, it was shown that; all graft groups were histologically better than secondary healing group and when the authors compared the graft groups, differentiated mesenchymal stem cell group were the best. CONCLUSION Our study results showed that endoscopic sinus surgery and treatment with cell sheets, which were generated in temperature-responsive culture dishes, had more functional respiratory epithelium.
Collapse
|
27
|
Zhao L, Cheng G, Choksi K, Samanta A, Girgis M, Soder R, Vincent RJ, Wulser M, De Ruyter M, McEnulty P, Hauptman J, Yang Y, Weiner CP, Dawn B. Transplantation of Human Umbilical Cord Blood-Derived Cellular Fraction Improves Left Ventricular Function and Remodeling After Myocardial Ischemia/Reperfusion. Circ Res 2019; 125:759-772. [PMID: 31462157 DOI: 10.1161/circresaha.119.315216] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Rationale: Human umbilical cord blood (hUCB) contains diverse populations of stem/progenitor cells. Whether hUCB-derived nonhematopoietic cells would induce cardiac repair remains unknown. Objective: To examine whether intramyocardial transplantation of hUCB-derived CD45-Lin- nonhematopoietic cellular fraction after a reperfused myocardial infarction in nonimmunosuppressed rats would improve cardiac function and ameliorate ventricular remodeling. Methods and Results: Nonhematopoietic CD45-Lin- cells were isolated from hUCB. Flow cytometry and quantitative polymerase chain reaction were used to characterize this subpopulation. Age-matched male Fischer 344 rats underwent a 30-minute coronary occlusion followed by reperfusion and 48 hours later received intramyocardial injection of vehicle or hUCB CD45-Lin- cells. After 35 days, compared with vehicle-treated rats, CD45-Lin- cell-treated rats exhibited improved left ventricular function, blunted left ventricular hypertrophy, greater preservation of viable myocardium in the infarct zone, and superior left ventricular remodeling. Mechanistically, hUCB CD45-Lin- cell injection favorably modulated molecular pathways regulating myocardial fibrosis, cardiomyocyte apoptosis, angiogenesis, and inflammation in postinfarct ventricular myocardium. Rare persistent transplanted human cells could be detected at both 4 and 35 days after myocardial infarction. Conclusions: Transplantation of hUCB-derived CD45-Lin- nonhematopoietic cellular subfraction after a reperfused myocardial infarction in nonimmunosuppressed rats ameliorates left ventricular dysfunction and improves remodeling via favorable paracrine modulation of molecular pathways. These findings with human cells in a clinically relevant model of myocardial ischemia/reperfusion in immunocompetent animals may have significant translational implications.Visual Overview: An online visual overview is available for this article.
Collapse
Affiliation(s)
- Lin Zhao
- From the Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine (L.Z., G.C., M.G., J.H., Y.Y., B.D.)
| | - Guangming Cheng
- From the Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine (L.Z., G.C., M.G., J.H., Y.Y., B.D.)
| | - Kashyap Choksi
- Cardiology Consultants of South Georgia, Thomasville (K.C.)
| | - Anweshan Samanta
- Department of Internal Medicine (A.S.), University of Missouri-Kansas City
| | - Magdy Girgis
- From the Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine (L.Z., G.C., M.G., J.H., Y.Y., B.D.)
| | - Rupal Soder
- Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (R.S., R.J.V., M.W., C.P.W.)
| | - Robert J Vincent
- Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (R.S., R.J.V., M.W., C.P.W.)
| | - Michael Wulser
- Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (R.S., R.J.V., M.W., C.P.W.)
| | - Matt De Ruyter
- Department of Orthopedic Surgery (M.D.R.), University of Missouri-Kansas City
| | - Patrick McEnulty
- Department of Radiology, University of Kansas School of Medicine-Wichita (P.M.)
| | - Jeryl Hauptman
- From the Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine (L.Z., G.C., M.G., J.H., Y.Y., B.D.)
| | - Yanjuan Yang
- From the Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine (L.Z., G.C., M.G., J.H., Y.Y., B.D.)
| | - Carl P Weiner
- Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (R.S., R.J.V., M.W., C.P.W.)
| | - Buddhadeb Dawn
- From the Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine (L.Z., G.C., M.G., J.H., Y.Y., B.D.)
| |
Collapse
|
28
|
Yan W, Abu-El-Rub E, Saravanan S, Kirshenbaum LA, Arora RC, Dhingra S. Inflammation in myocardial injury: mesenchymal stem cells as potential immunomodulators. Am J Physiol Heart Circ Physiol 2019; 317:H213-H225. [PMID: 31125258 PMCID: PMC6732476 DOI: 10.1152/ajpheart.00065.2019] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/13/2019] [Accepted: 05/20/2019] [Indexed: 02/08/2023]
Abstract
Ischemic heart disease is a growing worldwide epidemic. Improvements in medical and surgical therapies have reduced early mortality after acute myocardial infarction and increased the number of patients living with chronic heart failure. The irreversible loss of functional cardiomyocytes puts these patients at significant risk of ongoing morbidity and mortality after their index event. Recent evidence suggests that inflammation is a key mediator of postinfarction adverse remodeling in the heart. In this review, we discuss the cardioprotective and deleterious effects of inflammation and its mediators during acute myocardial infarction. We also explore the role of mesenchymal stem cell therapy to limit secondary injury and promote myocardial healing after myocardial infarction.
Collapse
Affiliation(s)
- Weiang Yan
- Institute of Cardiovascular Sciences, Saint Boniface Hospital Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba , Winnipeg , Canada
- Section of Cardiac Surgery, Department of Surgery, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba , Winnipeg , Canada
| | - Ejlal Abu-El-Rub
- Institute of Cardiovascular Sciences, Saint Boniface Hospital Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba , Winnipeg , Canada
| | - Sekaran Saravanan
- Centre for Nanotechnology and Advanced Biomaterials, Department of Bioengineering, SASTRA University , Thanjavur, Tamil Nadu , India
| | - Lorrie A Kirshenbaum
- Institute of Cardiovascular Sciences, Saint Boniface Hospital Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba , Winnipeg , Canada
| | - Rakesh C Arora
- Institute of Cardiovascular Sciences, Saint Boniface Hospital Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba , Winnipeg , Canada
- Section of Cardiac Surgery, Department of Surgery, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba , Winnipeg , Canada
| | - Sanjiv Dhingra
- Institute of Cardiovascular Sciences, Saint Boniface Hospital Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba , Winnipeg , Canada
| |
Collapse
|
29
|
Lu L, Liu Y, Zhang X, Lin J. The therapeutic role of bone marrow stem cell local injection in rat experimental periodontitis. J Oral Rehabil 2019; 47 Suppl 1:73-82. [PMID: 31220354 DOI: 10.1111/joor.12843] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/26/2019] [Accepted: 06/15/2019] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem cell therapy brings hope for regenerating damaged periodontal tissues. The present study aimed to investigate the therapeutic role of local bone marrow stem cell (BMSC) injection in ligation-induced periodontitis and the underlying mechanisms. Alveolar bone lesion was induced by placing ligatures subgingivally around the bilateral maxillary second molars for 28 days. The alveolar bone lesion was confirmed by micro-CT analysis and bone histomorphometry. Allogeneic BMSC transplantation was carried out at 28 day after ligation. The survival state of the transplanted BMSC was observed by bioluminescent imaging. The implantation of the BMSC into the gingival tissues and periodontal ligament was confirmed by green fluorescent protein (GFP) immunohistochemical staining. The expression level of pro-inflammatory, tumour necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), and receptor activator of nuclear factor-κ B ligand (RANKL) and osteoprotegerin (OPG) in periodontal tissues were evaluated by immunohistochemical staining and real-time PCR. Significant reverse of alveolar bone lesion was observed after BMSC transplantation. The expression of TNF-α and IL-1β was down-regulated by BMSC transplantation. The number of tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts in the periodontal ligament was reduced, and the increased RANKL expression and decreased OPG expression were also reversed after BMSC transplantation. It is concluded that allogeneic BMSC local injection could inhibit the inflammation of the periodontitis tissue and promote periodontal tissue regeneration.
Collapse
Affiliation(s)
- Lei Lu
- Department of Oral Anatomy and Physiology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Yang Liu
- Department of Stomatology, Technology Innovation Park, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Xu Zhang
- Department of Oral Anatomy and Physiology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China.,Department of Stomatology, the Fourth Hospital of Harbin Medical University, Harbin, China
| | - Jiang Lin
- Department of Periodontology, the Fourth Hospital of Harbin Medical University, Harbin, China.,Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
30
|
Ahmed ESA, Ahmed NH, Medhat AM, Said UZ, Rashed LA, Abdel Ghaffar ARB. Mesenchymal stem cells targeting PI3K/AKT pathway in leukemic model. Tumour Biol 2019; 41:1010428319846803. [PMID: 31018830 DOI: 10.1177/1010428319846803] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells have therapeutic properties that are related to their potentials for trans-differentiation, immunomodulation, anti-inflammatory, inhibitory effect on tumor proliferation, and induction of apoptosis. This study was performed to analyze the role of mesenchymal stem cells as an alternative for cellular signaling growth factors involved in the pathogenesis of leukemogenesis in rats. Treatment of rats with 7,12-dimethyl benz [a] anthracene induced leukemogenesis appeared as a significant decrease in hematological parameters with concomitant significant increase in bone marrow oxidative and inflammatory indices (transforming growth factor beta and interleukin-6) in comparison with normal groups. On the contrary, Western immunoblotting showed a significant increase in the signaling growth factors: PI3K, AKT, mTOR proteins and a significant decrease in PTEN in 7,12-dimethyl benz [a] anthracene-treated group. In addition, a significant increase in the transcript levels of B cell lymphoma-2 protein gene in the 7,12-dimethyl benz [a] anthracene group, while that of C-X-C motif chemokine receptor-4 and B cell lymphoma-2 protein associated x-protein were significantly downregulated compared to controls. Meanwhile, therapeutic mesenchymal stem cells treatment predict a significant improvement versus 7,12-dimethyl benz [a] anthracene group through the modulation of growth factors that confront bone marrow dysplasia. In the same direction treatment of 7,12-dimethyl benz [a] anthracene group with mesenchymal stem cells, it induced apoptosis and increased the homing efficacy to bone marrow. In conclusion, mesenchymal stem cells improve hematopoiesis and alleviate inflammation, and modulated PI3K/AKT signaling pathway contributed to experimental leukemogenesis.
Collapse
Affiliation(s)
- Esraa S A Ahmed
- 1 National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Neamat H Ahmed
- 1 National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Amina M Medhat
- 2 Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Ussama Z Said
- 1 National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Laila A Rashed
- 3 Biochemistry & Molecular Biology Department, Medicine Faculty, Cairo University, Cairo, Egypt
| | | |
Collapse
|
31
|
Huang K, Hu S, Cheng K. A New Era of Cardiac Cell Therapy: Opportunities and Challenges. Adv Healthc Mater 2019; 8:e1801011. [PMID: 30548836 PMCID: PMC6368830 DOI: 10.1002/adhm.201801011] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/05/2018] [Indexed: 12/15/2022]
Abstract
Myocardial infarction (MI), caused by coronary heart disease (CHD), remains one of the most common causes of death in the United States. Over the last few decades, scientists have invested considerable resources on the study and development of cell therapies for myocardial regeneration after MI. However, due to a number of limitations, they are not yet readily available for clinical applications. Mounting evidence supports the theory that paracrine products are the main contributors to the regenerative effects attributed to these cell therapies. The next generation of cell-based MI therapies will identify and isolate cell products and derivatives, integrate them with biocompatible materials and technologies, and use them for the regeneration of damaged myocardial tissue. This review discusses the progress made thus far in pursuit of this new generation of cell therapies. Their fundamental regenerative mechanisms, their potential to combine with other therapeutic products, and their role in shaping new clinical approaches for heart tissue engineering, are addressed.
Collapse
Affiliation(s)
- Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27607, USA
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27607, USA
- Pharmacoengineeirng and Molecular Pharmaceutics Division, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
32
|
Miklíková M, Jarkovská D, Čedíková M, Švíglerová J, Kuncová J, Nalos L, Kubíková T, Liška V, Holubová M, Lysák D, Králíčková M, Vištejnová L, Štengl M. Beneficial effects of mesenchymal stem cells on adult porcine cardiomyocytes in non-contact co-culture. Physiol Res 2018; 67:S619-S631. [PMID: 30607969 DOI: 10.33549/physiolres.934051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been reported to improve survival of cardiomyocytes (CMCs) and overall regeneration of cardiac tissue. Despite promising preclinical results, interactions of MSCs and CMCs, both direct and indirect, remain unclear. In this study, porcine bone marrow MSCs and freshly isolated porcine primary adult CMCs were used for non-contact co-culture experiments. Morphology, viability and functional parameters of CMCs were measured over time and compared between CMCs cultured alone and CMCs co-cultured with MSCs. In non-contact co-culture, MSCs improved survival of CMCs. CMCs co-cultured with MSCs maintained CMCs morphology and viability in significantly higher percentage than CMCs cultured alone. In viable CMCs, mitochondrial respiration was preserved in both CMCs cultured alone and in CMCs co-cultured with MSCs. Comparison of cellular contractility and calcium handling, measured in single CMCs, revealed no significant differences between viable CMCs from co-culture and CMCs cultured alone. In conclusion, non-contact co-culture of porcine MSCs and CMCs improved survival of CMCs with a sufficient preservation of functional and mitochondrial parameters.
Collapse
Affiliation(s)
- M Miklíková
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Bak DH, Choi MJ, Kim SR, Lee BC, Kim JM, Jeon ES, Oh W, Lim ES, Park BC, Kim MJ, Na J, Kim BJ. Human umbilical cord blood mesenchymal stem cells engineered to overexpress growth factors accelerate outcomes in hair growth. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 22:555-566. [PMID: 30181702 PMCID: PMC6115345 DOI: 10.4196/kjpp.2018.22.5.555] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/04/2018] [Accepted: 07/19/2018] [Indexed: 12/22/2022]
Abstract
Human umbilical cord blood mesenchymal stem cells (hUCB-MSCs) are used in tissue repair and regeneration; however, the mechanisms involved are not well understood. We investigated the hair growth-promoting effects of hUCB-MSCs treatment to determine whether hUCB-MSCs enhance the promotion of hair growth. Furthermore, we attempted to identify the factors responsible for hair growth. The effects of hUCB-MSCs on hair growth were investigated in vivo, and hUCB-MSCs advanced anagen onset and hair follicle neogeneration. We found that hUCB-MSCs co-culture increased the viability and up-regulated hair induction-related proteins of human dermal papilla cells (hDPCs) in vitro. A growth factor antibody array revealed that secretory factors from hUCB-MSCs are related to hair growth. Insulin-like growth factor binding protein-1 (IGFBP-1) and vascular endothelial growth factor (VEGF) were increased in co-culture medium. Finally, we found that IGFBP-1, through the co-localization of an IGF-1 and IGFBP-1, had positive effects on cell viability; VEGF secretion; expression of alkaline phosphatase (ALP), CD133, and β-catenin; and formation of hDPCs 3D spheroids. Taken together, these data suggest that hUCB-MSCs promote hair growth via a paracrine mechanism.
Collapse
Affiliation(s)
- Dong Ho Bak
- Department of Dermatology, College of Medicine, Chung-Ang University, Seoul 06973, Korea.,Department of Medicine, Graduate School, Chung-Ang University, Seoul 06973, Korea
| | - Mi Ji Choi
- Department of Dermatology, College of Medicine, Chung-Ang University, Seoul 06973, Korea.,Department of Medicine, Graduate School, Chung-Ang University, Seoul 06973, Korea
| | - Soon Re Kim
- Department of Dermatology, College of Medicine, Chung-Ang University, Seoul 06973, Korea
| | - Byung Chul Lee
- Department of Dermatology, College of Medicine, Chung-Ang University, Seoul 06973, Korea
| | - Jae Min Kim
- Department of Medicine, Graduate School, Chung-Ang University, Seoul 06973, Korea
| | - Eun Su Jeon
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Seongnam 13494, Korea
| | - Wonil Oh
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Seongnam 13494, Korea
| | - Ee Seok Lim
- Thema Dermatologic Clinic, Seoul 06524, Korea
| | - Byung Cheol Park
- Department of Dermatology, Dankook Medical College, Cheonan 31116, Korea
| | | | - Jungtae Na
- Department of Dermatology, College of Medicine, Chung-Ang University, Seoul 06973, Korea
| | - Beom Joon Kim
- Department of Dermatology, College of Medicine, Chung-Ang University, Seoul 06973, Korea.,Department of Medicine, Graduate School, Chung-Ang University, Seoul 06973, Korea
| |
Collapse
|
34
|
Wysoczynski M, Khan A, Bolli R. New Paradigms in Cell Therapy: Repeated Dosing, Intravenous Delivery, Immunomodulatory Actions, and New Cell Types. Circ Res 2018; 123:138-158. [PMID: 29976684 PMCID: PMC6050028 DOI: 10.1161/circresaha.118.313251] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Perhaps the most important advance in the field of cell therapy for heart disease has been the recognition that all stem/progenitor cells (both adult and embryonic) fail to engraft in the heart to a significant extent and thus work via paracrine mechanisms. This fundamental advance has led to 4 new paradigms that are discussed in this review and that may importantly shape, or even revolutionize, the future of the field: (1) repeated cell therapy, (2) intravenous cell therapy, (3) immunomodulatory actions of cell therapy, and (4) new cell types. Because virtually all of our current knowledge of cell therapy is predicated on the effects of a single cell dose, the idea that the full therapeutic effects of a cell product require repeated doses is disruptive and has far-reaching implications. For example, inadequate dosing (single-dose protocols) may be responsible, at least in part, for the borderline or disappointing results obtained to date in clinical trials; furthermore, future studies (both preclinical and clinical) may need to incorporate repeated cell administrations. Another disruptive idea, supported by emerging preclinical and clinical evidence, is that intravenously injected cells can produce beneficial effects on the heart, presumably via release of paracrine factors in extracardiac organs or endocrine factors into the systemic circulation. Intravenous administration would obviate the need for direct delivery of cells to the heart, making cell therapy simpler, cheaper, safer, more scalable, and more broadly available, even on an outpatient basis. Although the mechanism of action of cell therapy remains elusive, there is compelling in vitro evidence that transplanted cells modulate the function of various immune cell types via release of paracrine factors, such as extracellular vesicles, although in vivo evidence is still limited. Investigation of the new paradigms reviewed herein should be a top priority because it may profoundly transform cell therapy and finally make it a reality.
Collapse
Affiliation(s)
- Marcin Wysoczynski
- From the Institute of Molecular Cardiology, University of Louisville, KY
| | - Abdur Khan
- From the Institute of Molecular Cardiology, University of Louisville, KY
| | - Roberto Bolli
- From the Institute of Molecular Cardiology, University of Louisville, KY.
| |
Collapse
|
35
|
Baek SM, Son Y, Hong HS. Substance P blocks the impairment of paracrine potential of MSC due to long term culture. Mol Cell Toxicol 2018. [DOI: 10.1007/s13273-018-0031-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
36
|
Wang S, Hu S, Wang J, Liu Y, Zhao R, Tong M, Cui H, Wu N, Chen X. Conditioned medium from bone marrow-derived mesenchymal stem cells inhibits vascular calcification through blockade of the BMP2-Smad1/5/8 signaling pathway. Stem Cell Res Ther 2018; 9:160. [PMID: 29895327 PMCID: PMC5998505 DOI: 10.1186/s13287-018-0894-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 04/17/2018] [Accepted: 05/02/2018] [Indexed: 12/13/2022] Open
Abstract
Background Arterial calcification is associated with cardiovascular disease as a complication of advanced atherosclerosis and is a significant contributor to cardiovascular morbidity and mortality. Osteoblastic differentiation of vascular smooth muscle cells (VSMCs) plays an important role in arterial calcification and is characterized by cellular necrosis, inflammation, and lipoprotein and phospholipid complexes, especially in atherosclerotic calcification. The conditioned medium from bone marrow-derived mesenchymal stem cells (MSC-CM) is well known as a rich source of autologous cytokines and is universally used for tissue regeneration in current clinical medicine. Here, we demonstrate that MSC-CM inhibits beta-glycerophosphate (β-GP)-induced vascular calcification through blockade of the bone morphogenetic protein-2 (BMP2)–Smad1/5/8 signaling pathway. Methods VSMC calcification was induced by β-GP followed by treatment with MSC-CM. Mineral deposition was assessed by Alizarin Red S staining. Intracellular calcium content was determined colorimetrically by the o-cresolphthalein complexone method and alkaline phosphatase (ALP) activity was measured by the para-nitrophenyl phosphate method. Expression of BMP2, BMPR1A, BMPR1B, BMPR2, msh homeobox 2 (Msx2), Runt-related transcription factor 2 (Runx2), and osteocalcin (OC), representative osteoblastic markers, was assessed using real-time polymerase chain reaction analysis while the protein expression of BMP2, Runx2, and phosphorylated Smad1/5/8 was detected by western blot analysis. Results Our data demonstrated that MSC-CM inhibits osteoblastic differentiation and mineralization of VSMCs as evidenced by decreased calcium content, ALP activity, and decreased expression of BMP-2, Runx2, Msx2, and OC. MSC-CM suppressed the expression of phosphorylated Smad1/5/8 and the β-GP-induced translocation from the cytoplasm to the nucleus. Further study demonstrated that human recombinant BMP-2 overcame the suppression of VSMC calcification by MSC-CM. Conclusion MSC-CM may act as a novel therapy for VSMC calcification by mediating the BMP2–Smad1/5/8 signaling pathway
Collapse
Affiliation(s)
- Shuangshuang Wang
- Department of Cardiology, Ningbo First Hospital, Ningbo, 315000, China
| | - Siwang Hu
- Spine Tumor Center, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Jian Wang
- Department of Cardiology, Ningbo First Hospital, Ningbo, 315000, China
| | - Yahui Liu
- Department of Cardiology, Ningbo First Hospital, Ningbo, 315000, China
| | - Ruochi Zhao
- Department of Cardiology, Ningbo First Hospital, Ningbo, 315000, China
| | - Maoqing Tong
- Department of Cardiology, Ningbo First Hospital, Ningbo, 315000, China
| | - Hanbin Cui
- Department of Cardiology, Ningbo First Hospital, Ningbo, 315000, China
| | - Nan Wu
- Department of Cardiology, Ningbo First Hospital, Ningbo, 315000, China
| | - Xiaomin Chen
- Department of Cardiology, Ningbo First Hospital, Ningbo, 315000, China.
| |
Collapse
|
37
|
Devetzi M, Goulielmaki M, Khoury N, Spandidos DA, Sotiropoulou G, Christodoulou I, Zoumpourlis V. Genetically‑modified stem cells in treatment of human diseases: Tissue kallikrein (KLK1)‑based targeted therapy (Review). Int J Mol Med 2018; 41:1177-1186. [PMID: 29328364 PMCID: PMC5819898 DOI: 10.3892/ijmm.2018.3361] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/02/2018] [Indexed: 12/12/2022] Open
Abstract
The tissue kallikrein-kinin system (KKS) is an endogenous multiprotein metabolic cascade which is implicated in the homeostasis of the cardiovascular, renal and central nervous system. Human tissue kallikrein (KLK1) is a serine protease, component of the KKS that has been demonstrated to exert pleiotropic beneficial effects in protection from tissue injury through its anti-inflammatory, anti-apoptotic, anti-fibrotic and anti-oxidative actions. Mesenchymal stem cells (MSCs) or endothelial progenitor cells (EPCs) constitute populations of well-characterized, readily obtainable multipotent cells with special immunomodulatory, migratory and paracrine properties rendering them appealing potential therapeutics in experimental animal models of various diseases. Genetic modification enhances their inherent properties. MSCs or EPCs are competent cellular vehicles for drug and/or gene delivery in the targeted treatment of diseases. KLK1 gene delivery using adenoviral vectors or KLK1 protein infusion into injured tissues of animal models has provided particularly encouraging results in attenuating or reversing myocardial, renal and cerebrovascular ischemic phenotype and tissue damage, thus paving the way for the administration of genetically modified MSCs or EPCs with the human tissue KLK1 gene. Engraftment of KLK1-modified MSCs and/or KLK1-modified EPCs resulted in advanced beneficial outcome regarding heart and kidney protection and recovery from ischemic insults. Collectively, findings from pre-clinical studies raise the possibility that tissue KLK1 may be a novel future therapeutic target in the treatment of a wide range of cardiovascular, cerebrovascular and renal disorders.
Collapse
Affiliation(s)
- Marina Devetzi
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Maria Goulielmaki
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Nicolas Khoury
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | | | - Ioannis Christodoulou
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| |
Collapse
|
38
|
Miao C, Lei M, Hu W, Han S, Wang Q. A brief review: the therapeutic potential of bone marrow mesenchymal stem cells in myocardial infarction. Stem Cell Res Ther 2017; 8:242. [PMID: 29096705 PMCID: PMC5667518 DOI: 10.1186/s13287-017-0697-9] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Myocardial infarction (MI) results in dysfunction and irreversible loss of cardiomyocytes and is among the most serious health threats today. Bone marrow mesenchymal stem cells (BMSCs), with their capacity for multidirectional differentiation, low immunogenicity, and high portability, can serve as ideal seed cells in cardiovascular disease therapy. In this review, we examine recent literature concerning the application of BMSCs for the treatment of MI and consider the following aspects: activity of transplanted cells, migration and homing of BMSCs, immunomodulatory and anti-inflammatory effects of BMSCs, anti-fibrotic activity of BMSCs, the role of BMSCs in angiogenesis, and differentiation of BMSCs into cardiomyocyte-like cells and endothelial cells. Each aspect is complementary to the others and together they promote the repair of cardiomyocytes by BMSCs after MI. Although transplantation of BMSCs has enabled new options for MI treatment, the critical issue we must now address is the reduced viability of transplanted BMSCs due to inadequate blood supply, poor nourishment of cells, and generation of free radicals. More clinical trials are needed to prove the therapeutic potential of BMSCs in MI.
Collapse
Affiliation(s)
- Chi Miao
- Cardiovascular Department Of Internal Medicine, The Fourth Affiliated Hospital of China Medical University, Chongshandong Street No.4, Shenyang, 110032, China
| | - Mingming Lei
- Cardiovascular Department Of Internal Medicine, The Fourth Affiliated Hospital of China Medical University, Chongshandong Street No.4, Shenyang, 110032, China
| | - Weina Hu
- Cardiovascular Department Of Internal Medicine, The Fourth Affiliated Hospital of China Medical University, Chongshandong Street No.4, Shenyang, 110032, China
| | - Shuo Han
- Cardiovascular Department Of Internal Medicine, The Fourth Affiliated Hospital of China Medical University, Chongshandong Street No.4, Shenyang, 110032, China
| | - Qi Wang
- Cardiovascular Department Of Internal Medicine, The Fourth Affiliated Hospital of China Medical University, Chongshandong Street No.4, Shenyang, 110032, China.
| |
Collapse
|
39
|
Epstein SE, Luger D, Lipinski MJ. Paracrine-Mediated Systemic Anti-Inflammatory Activity of Intravenously Administered Mesenchymal Stem Cells. Circ Res 2017; 121:1044-1046. [DOI: 10.1161/circresaha.117.311925] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Stephen E. Epstein
- From the MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, DC
| | - Dror Luger
- From the MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, DC
| | - Michael J. Lipinski
- From the MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, DC
| |
Collapse
|
40
|
Liang Y, Li X, Zhang Y, Yeung SC, Zhen Z, Ip MSM, Tse HF, Lian Q, Mak JCW. Induced Pluripotent Stem Cells-Derived Mesenchymal Stem Cells Attenuate Cigarette Smoke-Induced Cardiac Remodeling and Dysfunction. Front Pharmacol 2017; 8:501. [PMID: 28804458 PMCID: PMC5532447 DOI: 10.3389/fphar.2017.00501] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/14/2017] [Indexed: 12/11/2022] Open
Abstract
The strong relationship between cigarette smoking and cardiovascular disease (CVD) has been well-documented, but the mechanisms by which smoking increases CVD risk appear to be multifactorial and incompletely understood. Mesenchymal stem cells (MSCs) are regarded as an important candidate for cell-based therapy in CVD. We hypothesized that MSCs derived from induced pluripotent stem cell (iPSC-MSCs) or bone marrow (BM-MSCs) might alleviate cigarette smoke (CS)-induced cardiac injury. This study aimed to investigate the effects of BM-MSCs or iPSC-MSCs on CS-induced changes in serum and cardiac lipid profiles, oxidative stress and inflammation as well as cardiac function in a rat model of passive smoking. Male Sprague-Dawley rats were randomly selected for exposure to either sham air (SA) as control or 4% CS for 1 h per day for 56 days. On day 29 and 43, human adult BM-MSCs, iPSC-MSCs or PBS were administered intravenously to CS-exposed rats. Results from echocardiography, serum and cardiac lipid profiles, cardiac antioxidant capacity, cardiac pro- and anti-inflammatory cytokines and cardiac morphological changes were evaluated at the end of treatment. iPSC-MSC-treated group showed a greater effect in the improvement of CS-induced cardiac dysfunction over BM-MSCs-treated group as shown by increased percentage left ventricular ejection fraction and percentage fractional shortening, in line with the greater reversal of cardiac lipid abnormality. In addition, iPSC-MSCs administration attenuated CS-induced elevation of cardiac pro-inflammatory cytokines as well as restoration of anti-inflammatory cytokines and anti-oxidative markers, leading to ameliorate cardiac morphological abnormalities. These data suggest that iPSC-MSCs on one hand may restore CS-induced cardiac lipid abnormality and on the other hand may attenuate cardiac oxidative stress and inflammation via inhibition of CS-induced NF-κB activation, leading to improvement of cardiac remodeling and dysfunction. Thus, iPSC-MSCs may be a promising candidate in cell-based therapy to prevent cardiac complications in smokers.
Collapse
Affiliation(s)
- Yingmin Liang
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong KongPok Fu Lam, Hong Kong
| | - Xiang Li
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong KongPok Fu Lam, Hong Kong
| | - Yuelin Zhang
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong
| | - Sze Chun Yeung
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong KongPok Fu Lam, Hong Kong
| | - Zhe Zhen
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong
| | - Mary S M Ip
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong KongPok Fu Lam, Hong Kong.,Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong KongPok Fu Lam, Hong Kong
| | - Hung Fat Tse
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong
| | - Qizhou Lian
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong KongPok Fu Lam, Hong Kong.,Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong KongPok Fu Lam, Hong Kong.,Department of Ophthalmology, The University of Hong KongPok Fu Lam, Hong Kong
| | - Judith C W Mak
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong KongPok Fu Lam, Hong Kong.,Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong KongPok Fu Lam, Hong Kong.,Department of Pharmacology and Pharmacy, The University of Hong KongPok Fu Lam, Hong Kong
| |
Collapse
|
41
|
Meng X, Li J, Yu M, Yang J, Zheng M, Zhang J, Sun C, Liang H, Liu L. Transplantation of mesenchymal stem cells overexpressing IL10 attenuates cardiac impairments in rats with myocardial infarction. J Cell Physiol 2017; 233:587-595. [DOI: 10.1002/jcp.25919] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 03/17/2017] [Indexed: 01/14/2023]
Affiliation(s)
- Xin Meng
- Department of UltrasonographyXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Jianping Li
- Department of Radiation OncologyXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Ming Yu
- Department of UltrasonographyXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Jian Yang
- Department of Cardiovascular SurgeryXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Minjuan Zheng
- Department of UltrasonographyXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Jinzhou Zhang
- Department of Cardiovascular SurgeryXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Chao Sun
- Department of UltrasonographyXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Hongliang Liang
- Department of Cardiovascular SurgeryXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Liwen Liu
- Department of UltrasonographyXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| |
Collapse
|
42
|
Der Sarkissian S, Lévesque T, Noiseux N. Optimizing stem cells for cardiac repair: Current status and new frontiers in regenerative cardiology. World J Stem Cells 2017; 9:9-25. [PMID: 28154736 PMCID: PMC5253186 DOI: 10.4252/wjsc.v9.i1.9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/20/2016] [Accepted: 10/24/2016] [Indexed: 02/06/2023] Open
Abstract
Cell therapy has the potential to improve healing of ischemic heart, repopulate injured myocardium and restore cardiac function. The tremendous hope and potential of stem cell therapy is well understood, yet recent trials involving cell therapy for cardiovascular diseases have yielded mixed results with inconsistent data thereby readdressing controversies and unresolved questions regarding stem cell efficacy for ischemic cardiac disease treatment. These controversies are believed to arise by the lack of uniformity of the clinical trial methodologies, uncertainty regarding the underlying reparative mechanisms of stem cells, questions concerning the most appropriate cell population to use, the proper delivery method and timing in relation to the moment of infarction, as well as the poor stem cell survival and engraftment especially in a diseased microenvironment which is collectively acknowledged as a major hindrance to any form of cell therapy. Indeed, the microenvironment of the failing heart exhibits pathological hypoxic, oxidative and inflammatory stressors impairing the survival of transplanted cells. Therefore, in order to observe any significant therapeutic benefit there is a need to increase resilience of stem cells to death in the transplant microenvironment while preserving or better yet improving their reparative functionality. Although stem cell differentiation into cardiomyocytes has been observed in some instance, the prevailing reparative benefits are afforded through paracrine mechanisms that promote angiogenesis, cell survival, transdifferentiate host cells and modulate immune responses. Therefore, to maximize their reparative functionality, ex vivo manipulation of stem cells through physical, genetic and pharmacological means have shown promise to enable cells to thrive in the post-ischemic transplant microenvironment. In the present work, we will overview the current status of stem cell therapy for ischemic heart disease, discuss the most recurring cell populations employed, the mechanisms by which stem cells deliver a therapeutic benefit and strategies that have been used to optimize and increase survival and functionality of stem cells including ex vivo preconditioning with drugs and a novel “pharmaco-optimizer” as well as genetic modifications.
Collapse
|
43
|
Quan L, Wang Y, Liang J, Qiu T, Wang H, Zhang Y, Zhang Y, Hui Q, Tao K. Screening for genes, transcription factors and miRNAs associated with the myogenic and osteogenic differentiation of human adipose tissue-derived stem cells. Int J Mol Med 2016; 38:1839-1849. [PMID: 27779643 DOI: 10.3892/ijmm.2016.2788] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/09/2016] [Indexed: 11/05/2022] Open
Abstract
In the present study, we aimed to reveal the molecular mechanisms responsible for the differentiation of human adipose tissue-derived stem cells (hASCs) into myocytes and osteoblasts. Microarray data GSE37329 were obtained from the Gene Expression Omnibus database, including three hASC cell lines from healthy donors, two osteogenic lineages and two myogenic lineages from the in vitro‑induction of hASCs. Differentially expressed genes (DEGs) in the two lineages were firstly screened. Subsequently, the underlying functions of the two sets of DEGs were investigated by Gene Ontology function and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, followed by protein-protein interaction (PPI) network construction. Regulatory relationships between transcription factors (TFs) and microRNAs (miRNAs or miRs) with target genes were finally explored using different algorithms. A total of 665 and 485 DEGs were identified from the hASC‑derived myogenic and osteogenic lineages, respectively. The shared upregulated genes (n=205) in the two sets of DEGs were mainly involved in metabolism-related pathways, whereas the shared downregulated genes (n=128) were significantly enriched in the transforming growth factor-β (TGF-β) signaling pathway. Four genes, vascular endothelial growth factor A (VEGFA), fibroblast growth factor 2 (FGF2), nerve growth factor (NGF) and interleukin 1B (IL1B), presented with relatively higher degrees in both PPI networks. The transcription factor RAD21 was predicted to target shared upregulated and downregulated genes as well as specific downregulated genes in the myogenic and the osteogenic lineages. In addition, miRNA-DEG interaction analysis revealed that hsa-miR-1 regulated the most shared DEGs in the two lineages. There may be a correlation between the four genes, VEGFA, FGF2, IL1B and NGF, and the differentiation of hASCs into myocytes and osteoblasts. The TF RAD21 and hsa-miR-1 may play important roles in regulating the expression of differentiation-associated genes.
Collapse
Affiliation(s)
- Liangliang Quan
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Yang Wang
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Jiulong Liang
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Tao Qiu
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Hongyi Wang
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Ye Zhang
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Yu Zhang
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Qiang Hui
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Kai Tao
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| |
Collapse
|
44
|
Could stem cells be the future therapy for sepsis? Blood Rev 2016; 30:439-452. [PMID: 27297212 DOI: 10.1016/j.blre.2016.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 12/15/2022]
Abstract
The severity and threat of sepsis is well known, and despite several decades of research, the mortality continues to be high. Stem cells have great potential to be used in various clinical disorders. The innate ability of stem cells such as pluripotency, self-renewal makes them potential agents for therapeutic intervention. The pathophysiology of sepsis is a plethora of complex mechanisms which include the initial microbial infection, followed by "cytokine storm," endothelial dysfunction, coagulation cascade, and the late phase of apoptosis and immune paralysis which ultimately results in multiple organ dysfunction. Stem cells could potentially alter each step of this complex pathophysiology of sepsis. Multiple organ dysfunction associated with sepsis most often leads to death and stem cells have shown their ability to prevent the organ damage and improve the organ function. The possible mechanisms of therapeutic potential of stem cells in sepsis have been discussed in detail. The route of administration, dose level, and timing also play vital role in the overall effect of stem cells in sepsis.
Collapse
|
45
|
Yu M, Wang J, Fang Q, Liu P, Chen S, Zhe N, Lin X, Zhang Y, Zhao J, Zhou Z. High expression of heme oxygenase-1 in target organs may attenuate acute graft-versus-host disease through regulation of immune balance of TH17/Treg. Transpl Immunol 2016; 37:10-17. [PMID: 27168057 DOI: 10.1016/j.trim.2016.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 05/02/2016] [Accepted: 05/03/2016] [Indexed: 12/31/2022]
Abstract
The high incidence of acute graft-versus-host disease (aGVHD) is a serious complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Grades III and IV aGVHD are the leading causes of death in allo-HSCT recipients. Heme oxygenase-1(HO-1) has anti-inflammatory and immune-regulatory functions. In this study, we evaluated the none GVHD and grade I-IV patients samples which were collected at the first re-examination after successful allo-HSCT, we found that expressions of HO-1 mRNA in the bone marrow and peripheral blood mononuclear cells of allo-HSCT recipients who had subsequent non-GVHD and grade I aGVHD were significantly higher than those in patients with Grade III-IV aGVHD. We then demonstrated that enhanced expression of HO-1 in target organs by infusing HO-1-gene-modified Mesenchymal stem cells (MSCs) alleviated the clinical and histopathological severity of aGVHD in experimental mice. Flow cytometry revealed a higher expression of Treg cells and a lower expression of TH17 cells in splenic and lymph node tissues of mice with enhanced HO-1 expression, as compared to that in the aGVHD mice. This was further substantiated by lower expression levels of ROR-Υt and IL-17A mRNA, and higher levels of Foxp3 mRNA in the splenic tissue of mice with enhanced HO-1 expression. Our results indicate that high expression of HO-1 may reduce the severity of aGVHD by regulation of the TH17/Treg balance.
Collapse
Affiliation(s)
- Meisheng Yu
- Graduate School, Guizhou Medical University, PR China
| | - Jishi Wang
- Guizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, PR China; Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China.
| | - Qin Fang
- Department of Pharmacy, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, Guizhou 550058, PR China
| | - Ping Liu
- Graduate School, Guizhou Medical University, PR China
| | - Shuya Chen
- Graduate School, Guizhou Medical University, PR China
| | - Nana Zhe
- Graduate School, Guizhou Medical University, PR China
| | - Xiaojing Lin
- Graduate School, Guizhou Medical University, PR China
| | - Yaming Zhang
- Guizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, PR China
| | - Jiangyuan Zhao
- Guizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, PR China
| | - Zhen Zhou
- Department of Pharmacy, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, Guizhou 550058, PR China
| |
Collapse
|
46
|
Alestalo K, Miettinen JA, Vuolteenaho O, Huikuri H, Lehenkari P. Bone Marrow Mononuclear Cell Transplantation Restores Inflammatory Balance of Cytokines after ST Segment Elevation Myocardial Infarction. PLoS One 2015; 10:e0145094. [PMID: 26690350 PMCID: PMC4687062 DOI: 10.1371/journal.pone.0145094] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/29/2015] [Indexed: 01/10/2023] Open
Abstract
Background Acute myocardial infarction (AMI) launches an inflammatory response and a repair process to compensate cardiac function. During this process, the balance between proinflammatory and anti-inflammatory cytokines is important for optimal cardiac repair. Stem cell transplantation after AMI improves tissue repair and increases the ventricular ejection fraction. Here, we studied in detail the acute effect of bone marrow mononuclear cell (BMMNC) transplantation on proinflammatory and anti-inflammatory cytokines in patients with ST segment elevation myocardial infarction (STEMI). Methods Patients with STEMI treated with thrombolysis followed by percutaneous coronary intervention (PCI) were randomly assigned to receive either BMMNC or saline as an intracoronary injection. Cardiac function was evaluated by left ventricle angiogram during the PCI and again after 6 months. The concentrations of 27 cytokines were measured from plasma samples up to 4 days after the PCI and the intracoronary injection. Results Twenty-six patients (control group, n = 12; BMMNC group, n = 14) from the previously reported FINCELL study (n = 80) were included to this study. At day 2, the change in the proinflammatory cytokines correlated with the change in the anti-inflammatory cytokines in both groups (Kendall’s tau, control 0.6; BMMNC 0.7). At day 4, the correlation had completely disappeared in the control group but was preserved in the BMMNC group (Kendall’s tau, control 0.3; BMMNC 0.7). Conclusions BMMNC transplantation is associated with preserved balance between pro- and anti-inflammatory cytokines after STEMI in PCI-treated patients. This may partly explain the favorable effect of stem cell transplantation after AMI.
Collapse
Affiliation(s)
- Kirsi Alestalo
- Surgery Clinic, Medical Research Center, Oulu University Hospital, Oulu, Finland
- Department of Anatomy and Cell Biology, Medical Research Center, University of Oulu, Oulu, Finland
- * E-mail:
| | - Johanna A. Miettinen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Olli Vuolteenaho
- Department of Physiology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Heikki Huikuri
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Petri Lehenkari
- Surgery Clinic, Medical Research Center, Oulu University Hospital, Oulu, Finland
- Department of Anatomy and Cell Biology, Medical Research Center, University of Oulu, Oulu, Finland
| |
Collapse
|
47
|
van Zuylen VL, den Haan MC, Geutskens SB, Roelofs H, Fibbe WE, Schalij MJ, Atsma DE. Post-myocardial infarct inflammation and the potential role of cell therapy. Cardiovasc Drugs Ther 2015; 29:59-73. [PMID: 25583678 DOI: 10.1007/s10557-014-6568-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Myocardial infarction triggers reparative inflammatory processes programmed to repair damaged tissue. However, often additional injury to the myocardium occurs through the course of this inflammatory process, which ultimately can lead to heart failure. The potential beneficial effects of cell therapy in treating cardiac ischemic disease, the number one cause of death worldwide, are being studied extensively, both in clinical trials using adult stem cells as well as in fundamental research on cardiac stem cells and regenerative biology. This review summarizes the current knowledge on molecular and cellular processes implicated in post-infarction inflammation and discusses the potential beneficial role cell therapy might play in this process. Due to its immunomodulatory properties, the mesenchymal stromal cell is a candidate to reverse the disease progression of the infarcted heart towards heart failure, and therefore is emphasized in this review.
Collapse
Affiliation(s)
- Vanessa-leigh van Zuylen
- Department of Cardiology, Leiden University Medical Center, P.O. Box 9600, 2300, RC, Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
48
|
Transplantation of human mesenchymal stem cells into the cisterna magna and its neuroprotective effects in a parkinsonian animal model. Mol Cell Toxicol 2015. [DOI: 10.1007/s13273-015-0038-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
49
|
Damous LL, Nakamuta JS, Carvalho AETSD, Carvalho KC, Soares JM, Simões MDJ, Krieger JE, Baracat EC. Does adipose tissue-derived stem cell therapy improve graft quality in freshly grafted ovaries? Reprod Biol Endocrinol 2015; 13:108. [PMID: 26394676 PMCID: PMC4580300 DOI: 10.1186/s12958-015-0104-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/11/2015] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND A major concern in ovarian transplants is substantial follicle loss during the initial period of hypoxia. Adipose tissue-derived stem cells (ASCs) have been employed to improve angiogenesis when injected into ischemic tissue. This study evaluated the safety and efficacy of adipose tissue-derived stem cells (ASCs) therapy in the freshly grafted ovaries 30 days after injection. METHODS Rat ASCs (rASCs) obtained from transgenic rats expressing green fluorescent protein (GFP)-(5 × 10(4) cells/ovary) were injected in topic (intact) or freshly grafted ovaries of 30 twelve-week-old adult female Wistar rats. The whole ovary was grafted in the retroperitoneum without vascular anastomosis, immediately after oophorectomy. Vaginal smears were performed daily to assess the resumption of the estrous cycle. Estradiol levels, grafts morphology and follicular viability and density were analyzed. Immunohistochemistry assays were conducted to identify and quantify rASC-GFP(+), VEGF tissue expression, apoptosis (cleaved caspase-3 and TUNEL), and cell proliferation (Ki-67). Quantitative gene expression (qPCR) for VEGF-A, Bcl2, EGF and TGF-β1 was evaluated using RT-PCR and a double labeling immunofluorescence assay for GFP and Von Willebrand Factor (VWF) was performed. RESULTS Grafted ovaries treated with rASC-GFP(+) exhibited earlier resumption of the estrous phase (p < 0.05), increased VEGF-A expression (11-fold in grafted ovaries and 5-fold in topic ovaries vs. control) and an increased number of blood vessels (p < 0.05) in ovarian tissue without leading to apoptosis or cellular proliferation (p > 0.05). Estradiol levels were similar among groups (p > 0.05). rASC-GFP(+) were observed in similar quantities in the topic and grafted ovaries (p > 0.05), and double-labeling for GFP and vWF was observed in both injected groups. CONCLUSION rASC therapy in autologous freshly ovarian grafts could be feasible and safe, induces earlier resumption of the estrous phase and enhances blood vessels in rats. This pilot study may be useful in the future for new researches on frozen-thawed ovarian tissue.
Collapse
Affiliation(s)
- Luciana L Damous
- Laboratório de Ginecologia Estrutural e Molecular (LIM-58), Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 2nd floor, room 2113, Pacaembu, 01246-903, São Paulo, Brazil.
| | - Juliana S Nakamuta
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, 05403-000, São Paulo, Brazil.
| | - Ana E T Saturi de Carvalho
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, 05403-000, São Paulo, Brazil.
| | - Katia Candido Carvalho
- Laboratório de Ginecologia Estrutural e Molecular (LIM-58), Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 2nd floor, room 2113, Pacaembu, 01246-903, São Paulo, Brazil.
| | - José Maria Soares
- Laboratório de Ginecologia Estrutural e Molecular (LIM-58), Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 2nd floor, room 2113, Pacaembu, 01246-903, São Paulo, Brazil.
| | - Manuel de Jesus Simões
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), Botucatu St 740. Ed. Lemos Torres, 2nd floor, Vila Clementino, 04023-009, São Paulo, Brazil.
| | - José Eduardo Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, 05403-000, São Paulo, Brazil.
| | - Edmund Chada Baracat
- Laboratório de Ginecologia Estrutural e Molecular (LIM-58), Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 2nd floor, room 2113, Pacaembu, 01246-903, São Paulo, Brazil.
| |
Collapse
|
50
|
Abstract
Heart failure remains a major cause of death and disability, requiring rapid development of new therapies. Bone marrow-derived mesenchymal stem cell (MSC)-based therapy is an emerging approach for the treatment of both acute and chronic heart failure. Following successful experimental studies in a range of models, more than 40 clinical trials of MSC-based therapy for heart failure have now been registered, and the results of completed clinical trials so far have shown feasibility and safety of this approach with therapeutic potential suggested (though preliminarily). However, there appear to be several critical issues to be solved before this treatment could become a widespread standard therapy for heart failure. In this review, we comprehensively and systemically summarize a total of 73 preclinical studies and 11 clinical trial reports published to date. By analyzing the data in these reports, (1) improvement in the cell delivery method to the heart in order to enhance donor cell engraftment, (2) elucidation of mechanisms underpinning the therapeutic effects of the treatment differentiation and/or treatment secretion, and (3) validation of the utility of allogeneic MSCs which could enhance the efficacy and expand the application/indication of this therapeutic approach are highlighted as future perspectives. These important respects are further discussed in this review article with referencing latest scientific and clinical information.
Collapse
Affiliation(s)
- Takuya Narita
- Cardiothoracic Surgery, National Heart Centre, Singapore, Singapore
| | | |
Collapse
|