1
|
Strecanska M, Sekelova T, Csobonyeiova M, Danisovic L, Cehakova M. Therapeutic applications of mesenchymal/medicinal stem/signaling cells preconditioned with external factors: Are there more efficient approaches to utilize their regenerative potential? Life Sci 2024; 346:122647. [PMID: 38614298 DOI: 10.1016/j.lfs.2024.122647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/25/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Mesenchymal/medicinal stem/signaling cells (MSCs) have emerged as a promising treatment option for various disorders. However, the donor's age, advanced stage of disease, and prolonged in vitro expansion often diminish the innate regenerative potential of MSCs. Besides that, the absence of MSCs' comprehensive "pre-admission testing" can result in the injection of cells with reduced viability and function, which may negatively affect the overall outcome of MSC-based therapies. It is, therefore, essential to develop effective strategies to improve the impaired biological performance of MSCs. This review focuses on the comprehensive characterization of various methods of external MSCs stimulation (hypoxia, heat shock, caloric restriction, acidosis, 3D culture, and application of extracellular matrix) that augment their medicinal potential. To emphasize the significance of MSCs priming, we summarize the effects of individual and combined preconditioning approaches, highlighting their impact on MSCs' response to either physiological or pathological conditions. We further investigate the synergic action of exogenous factors to maximize MSCs' therapeutic potential. Not to omit the field of tissue engineering, the application of pretreated MSCs seeded on scaffolds is discussed as well.
Collapse
Affiliation(s)
- Magdalena Strecanska
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia.
| | - Tatiana Sekelova
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia.
| | - Maria Csobonyeiova
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia.
| | - Lubos Danisovic
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia.
| | - Michaela Cehakova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia.
| |
Collapse
|
2
|
Aouabdi S, Nedjadi T, Alsiary R, Mouffouk F, Ansari HR. Transcriptomics Demonstrates Significant Biological Effect of Growing Stem Cells on RGD-Cotton Scaffold. Tissue Eng Part A 2024. [PMID: 38666698 DOI: 10.1089/ten.tea.2023.0333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024] Open
Abstract
Stem cell therapy provides a viable alternative treatment for degenerated or damaged tissue. Stem cells have been used either alone or in conjunction with an artificial scaffold. The latter provides a structural advantage by enabling the cells to thrive in three-dimensional (3D) settings, closely resembling the natural in vivo environments. Previously, we disclosed the development of a 3D scaffold made from cotton, which was conjugated with arginyl-glycyl-aspartic acid (RGD), to facilitate the growth and proliferation of mesenchymal stem cells (MSCs). This scaffold allowed the MSCs to adhere and proliferate without compromising their viability or their stem cell markers. A comprehensive analysis investigation of the molecular changes occurring in MSCs adhering to the cotton fibers will contribute to the advancement of therapy. The objective of this study is to analyze the molecular processes occurring in the growth of MSCs on a cotton-RGD conjugated-based scaffold by examining their gene expression profiles. To achieve this, we conducted an experiment where MSCs were seeded with and without the scaffold for a duration of 48 h. Subsequently, cells were collected for RNA extraction, cDNA synthesis, and whole-transcriptomic analysis performed on both populations. Our analysis revealed several upregulated and downregulated differently expressed genes in the MSCs adhering to the scaffold compared with the control cells. Through gene ontology analysis, we were able to identify enriched biological processes, molecular functions, pathways, and protein-protein interactions in these differentially expressed genes. Our data suggest that the scaffold may have the potential to enhance osteogenesis in the MSCs. Furthermore, our results indicate that the scaffold does not induce oxidative stress, inflammation, or aging in the MSCs. These findings provide valuable insights for the application of MSCs in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Sihem Aouabdi
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Taoufik Nedjadi
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Rawiah Alsiary
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Fouzi Mouffouk
- Department of Chemistry, Kuwait University, Kuwait, Kuwait
| | - Hifzur Rahman Ansari
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| |
Collapse
|
3
|
Yalçın MB, Bora ES, Erdoğan MA, Çakır A, Erbaş O. The Effect of Adipose-Derived Mesenchymal Stem Cells on Peripheral Nerve Damage in a Rodent Model. J Clin Med 2023; 12:6411. [PMID: 37835055 PMCID: PMC10573691 DOI: 10.3390/jcm12196411] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/27/2023] [Accepted: 10/07/2023] [Indexed: 10/15/2023] Open
Abstract
Peripheral nerve damage is a significant clinical problem with limited therapeutic options. Adipose-derived mesenchymal stem cells (ADSCs) have emerged as a promising therapeutic approach due to their regenerative potential. However, the underlying mechanisms by which ADSCs promote peripheral nerve regeneration remain unclear. In this study, we investigated the role of syndecan-1 and heat shock protein 70 (HSP-70) in mediating the regenerative effects of ADSCs on peripheral nerves. ADSCs were characterized and isolated from the adipose tissue of rats. In vitro experiments were conducted to evaluate the ability of ADSCs to secrete syndecan-1 and HSP-70 in response to stress conditions. To evaluate the therapeutic potential of ADSCs, rats with sciatic nerve injuries were treated with ADSCs and assessed for functional recovery, nerve regeneration, and changes in syndecan-1 and HSP-70 levels. Regeneration was evaluated with Electromyography (EMG) histology. The results showed that ADSCs could secrete syndecan-1 and HSP-70 in response to stress conditions. Furthermore, ADSC treatment significantly improved functional recovery and nerve regeneration and increased syndecan-1 and HSP-70 levels in the injured nerve. On the other hand, ADSCs make improvements histologically through the influence of Nerve growth factor (NGF), Malondialdehyde (MDA), and EMG.
Collapse
Affiliation(s)
- Mehmet Burak Yalçın
- Department of Orthopedics and Traumatology, Bahcelievler Memorial Hospital, Istanbul 34180, Turkey;
| | - Ejder Saylav Bora
- Department of Emergency Medicine, Izmir Atatürk Research and Training Hospital, Izmir 35360, Turkey
| | - Mümin Alper Erdoğan
- Department of Physiology, Faculty of Medicine, Izmir Kâtip Çelebi University, Izmir 35620, Turkey;
| | - Adem Çakır
- Department of Emergency Medicine, Çanakkale Mehmet Akif Ersoy State Hospital, Çanakkale 17100, Turkey;
| | - Oytun Erbaş
- Department of Physiology, Demiroğlu Bilim University, Istanbul 34394, Turkey;
| |
Collapse
|
4
|
Sadeghian I, Heidari R, Raee MJ, Negahdaripour M. Cell-penetrating peptide-mediated delivery of therapeutic peptides/proteins to manage the diseases involving oxidative stress, inflammatory response and apoptosis. J Pharm Pharmacol 2022; 74:1085-1116. [PMID: 35728949 DOI: 10.1093/jpp/rgac038] [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: 12/10/2021] [Accepted: 05/22/2022] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Peptides and proteins represent great potential for modulating various cellular processes including oxidative stress, inflammatory response, apoptosis and consequently the treatment of related diseases. However, their therapeutic effects are limited by their inability to cross cellular barriers. Cell-penetrating peptides (CPPs), which can transport cargoes into the cell, could resolve this issue, as would be discussed in this review. KEY FINDINGS CPPs have been successfully exploited in vitro and in vivo for peptide/protein delivery to treat a wide range of diseases involving oxidative stress, inflammatory processes and apoptosis. Their in vivo applications are still limited due to some fundamental issues of CPPs, including nonspecificity, proteolytic instability, potential toxicity and immunogenicity. SUMMARY Totally, CPPs could potentially help to manage the diseases involving oxidative stress, inflammatory response and apoptosis by delivering peptides/proteins that could selectively reach proper intracellular targets. More studies to overcome related CPP limitations and confirm the efficacy and safety of this strategy are needed before their clinical usage.
Collapse
Affiliation(s)
- Issa Sadeghian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Biotechnology Incubator, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Javad Raee
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Manica Negahdaripour
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
5
|
Xu Z, Lin L, Fan Y, Huselstein C, De Isla N, He X, Chen Y, Li Y. Secretome of Mesenchymal Stem Cells from Consecutive Hypoxic Cultures Promotes Resolution of Lung Inflammation by Reprogramming Anti-Inflammatory Macrophages. Int J Mol Sci 2022; 23:ijms23084333. [PMID: 35457151 PMCID: PMC9032661 DOI: 10.3390/ijms23084333] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 02/06/2023] Open
Abstract
The secretome from hypoxia-preconditioned mesenchymal stem cells (MSCs) has been shown to promote resolution of inflammation and alleviate acute lung injury (ALI) through its immunomodulatory function. However, the effects of consecutive hypoxic culture on immunomodulatory function of the MSCs secretome are largely unclarified. Here, we intend to investigate the effects of consecutive hypoxia on therapeutic efficacy of conditioned medium derived from MSCs (MSCs-CM) in alleviating ALI. Human umbilical cord-derived MSCs (UC-MSCs) were consecutively cultured in 21% O2 (Nor-MSCs) or in 1% O2 (Hypo-MSCs) from passage 0. Their conditioned medium (Nor-CM and Hypo-CM respectively) was collected and administered into ALI models. Our findings confirmed that Hypo-MSCs exhibited increased proliferation ability and decreased cell senescence compared with Nor-MSCs. Consecutive hypoxia promoted UC-MSCs to secrete immunomodulatory cytokines, such as insulin-like growth factor 1(IGF1), IL10, TNFα-stimulated gene 6(TSG6), TGFβ, and prostaglandin E2 (PGE2). Both Nor-CM and Hypo-CM could effectively limit lung inflammation, promote efferocytosis and modulate anti-inflammatory polarization of lung macrophages in ALI models. Moreover, the effects of Hypo-CM were more potent than Nor-CM. Taken together, our findings indicate that consecutive hypoxic cultures could not only promote both proliferation and quality of UC-MSCs, but also enhance the therapeutic efficacy of their secretome in mitigating lung inflammation by promoting efferocytosis and anti-inflammatory polarization of macrophages.
Collapse
Affiliation(s)
- Zhihong Xu
- Department of Pathophysiology, Hubei Province Key Laboratory of Allergy and Immunology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China; (Z.X.); (L.L.); (Y.F.); (X.H.); (Y.C.)
| | - Lulu Lin
- Department of Pathophysiology, Hubei Province Key Laboratory of Allergy and Immunology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China; (Z.X.); (L.L.); (Y.F.); (X.H.); (Y.C.)
| | - Yuxuan Fan
- Department of Pathophysiology, Hubei Province Key Laboratory of Allergy and Immunology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China; (Z.X.); (L.L.); (Y.F.); (X.H.); (Y.C.)
| | - Céline Huselstein
- UMR 7365 CNRS, Medical School, University of Lorraine, 54505 Nancy, France; (C.H.); (N.D.I.)
| | - Natalia De Isla
- UMR 7365 CNRS, Medical School, University of Lorraine, 54505 Nancy, France; (C.H.); (N.D.I.)
| | - Xiaohua He
- Department of Pathophysiology, Hubei Province Key Laboratory of Allergy and Immunology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China; (Z.X.); (L.L.); (Y.F.); (X.H.); (Y.C.)
| | - Yun Chen
- Department of Pathophysiology, Hubei Province Key Laboratory of Allergy and Immunology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China; (Z.X.); (L.L.); (Y.F.); (X.H.); (Y.C.)
| | - Yinping Li
- Department of Pathophysiology, Hubei Province Key Laboratory of Allergy and Immunology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China; (Z.X.); (L.L.); (Y.F.); (X.H.); (Y.C.)
- Correspondence: ; Tel.: +86-27-6875-8727; Fax: +86-27-6875-9222
| |
Collapse
|
6
|
Suppressing Pyroptosis Augments Post-Transplant Survival of Stem Cells and Cardiac Function Following Ischemic Injury. Int J Mol Sci 2021; 22:ijms22157946. [PMID: 34360711 PMCID: PMC8348609 DOI: 10.3390/ijms22157946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/20/2021] [Accepted: 07/20/2021] [Indexed: 12/11/2022] Open
Abstract
The acute demise of stem cells following transplantation significantly compromises the efficacy of stem cell-based cell therapeutics for infarcted hearts. As the stem cells transplanted into the damaged heart are readily exposed to the hostile environment, it can be assumed that the acute death of the transplanted stem cells is also inflicted by the same environmental cues that caused massive death of the host cardiac cells. Pyroptosis, a highly inflammatory form of programmed cell death, has been added to the list of important cell death mechanisms in the damaged heart. However, unlike the well-established cell death mechanisms such as necrosis or apoptosis, the exact role and significance of pyroptosis in the acute death of transplanted stem cells have not been explored in depth. In the present study, we found that M1 macrophages mediate the pyroptosis in the ischemia/reperfusion (I/R) injured hearts and identified miRNA-762 as an important regulator of interleukin 1β production and subsequent pyroptosis. Delivery of exogenous miRNA-762 prior to transplantation significantly increased the post-transplant survival of stem cells and also significantly ameliorated cardiac fibrosis and heart functions following I/R injury. Our data strongly suggest that suppressing pyroptosis can be an effective adjuvant strategy to enhance the efficacy of stem cell-based therapeutics for diseased hearts.
Collapse
|
7
|
Ju S, Lim L, Wi K, Park C, Ki YJ, Choi DH, Song H. LRP5 Regulates HIF-1α Stability via Interaction with PHD2 in Ischemic Myocardium. Int J Mol Sci 2021; 22:ijms22126581. [PMID: 34205318 PMCID: PMC8235097 DOI: 10.3390/ijms22126581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/12/2021] [Accepted: 06/16/2021] [Indexed: 12/22/2022] Open
Abstract
Low-density lipoprotein receptor-related protein 5 (LRP5) has been studied as a co-receptor for Wnt/β-catenin signaling. However, its role in the ischemic myocardium is largely unknown. Here, we show that LRP5 may act as a negative regulator of ischemic heart injury via its interaction with prolyl hydroxylase 2 (PHD2), resulting in hypoxia-inducible factor-1α (HIF-1α) degradation. Overexpression of LRP5 in cardiomyocytes promoted hypoxia-induced apoptotic cell death, whereas LRP5-silenced cardiomyocytes were protected from hypoxic insult. Gene expression analysis (mRNA-seq) demonstrated that overexpression of LRP5 limited the expression of HIF-1α target genes. LRP5 promoted HIF-1α degradation, as evidenced by the increased hydroxylation and shorter stability of HIF-1α under hypoxic conditions through the interaction between LRP5 and PHD2. Moreover, the specific phosphorylation of LRP5 at T1492 and S1503 is responsible for enhancing the hydroxylation activity of PHD2, resulting in HIF-1α degradation, which is independent of Wnt/β-catenin signaling. Importantly, direct myocardial delivery of adenoviral constructs, silencing LRP5 in vivo, significantly improved cardiac function in infarcted rat hearts, suggesting the potential value of LRP5 as a new target for ischemic injury treatment.
Collapse
Affiliation(s)
- Sujin Ju
- Department of Biochemistry and Molecular Biology, Chosun University School of Medicine, Gwangju 61452, Korea; (S.J.); (K.W.)
| | - Leejin Lim
- Cancer Mutation Research Center, Chosun University, Gwangju 61452, Korea;
| | - Kwanhwan Wi
- Department of Biochemistry and Molecular Biology, Chosun University School of Medicine, Gwangju 61452, Korea; (S.J.); (K.W.)
| | - Changwon Park
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA;
| | - Young-Jae Ki
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju 61452, Korea; (Y.-J.K.); (D.-H.C.)
| | - Dong-Hyun Choi
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju 61452, Korea; (Y.-J.K.); (D.-H.C.)
| | - Heesang Song
- Department of Biochemistry and Molecular Biology, Chosun University School of Medicine, Gwangju 61452, Korea; (S.J.); (K.W.)
- Correspondence: ; Tel.: +82-62-230-6290
| |
Collapse
|
8
|
Wilhelm C, Scherzad A, Bregenzer M, Meyer T, Gehrke T, Kleinsasser N, Hagen R, Hackenberg S. Interaction of head and neck squamous cell carcinoma cells and mesenchymal stem cells under hypoxia and normoxia. Oncol Lett 2020; 20:229. [PMID: 32968451 DOI: 10.3892/ol.2020.12092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) exhibit strong tropism towards tumor tissue. While MSCs generally surround tumors, they can also infiltrate tumors and thereby influence their proliferation. Interactions between MSCs and tumor cells are usually tested under normoxia, but the majority of solid tumors, including head and neck squamous cell carcinoma (HNSCC), are also characterized by hypoxic areas. Hence, the present study aimed to assess the interaction between MSCs and tumor cells under hypoxic conditions. MSCs were cultivated under normoxia and hypoxia, and conditioned media were used to cultivate the HNSCC cell line FaDu. The cell cycle distribution and viability of MSCs and the proliferation of FaDu cells were analyzed under normoxia and hypoxia, and changes in cytokine levels in the conditioned media were evaluated. No cell cycle changes were observed for MSCs after 24 h of cultivation under hypoxia, but the cell viability had declined. Hypoxia also led to a decrease in the proliferation of FaDu cells; however, FaDu cells proliferated faster after 48 h under hypoxia compared with normoxic conditions. This effect was reversed after incubation under normoxia for 72 h and hypoxia for 72 h. While these changes constituted a trend, these differences were not statistically significant. A cytokine assay showed an increase in interleukin (IL)-6 in the hypoxic medium. Overall, the results indicated that there was an interaction between MSCs and tumor cells. The presence or absence of oxygen seemed to influence the functionality of MSCs and their protumorigenic properties, in which IL-6 was identified as a potential mediator. Since MSCs are a component of the tumor stroma, further in vitro and in vivo studies are needed to investigate this interaction in order to develop novel approaches for tumor therapy.
Collapse
Affiliation(s)
- Christian Wilhelm
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Julius Maximilian University of Wuerzburg, D-97080 Wuerzburg, Germany
| | - Agmal Scherzad
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Julius Maximilian University of Wuerzburg, D-97080 Wuerzburg, Germany
| | - Maximilian Bregenzer
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Julius Maximilian University of Wuerzburg, D-97080 Wuerzburg, Germany
| | - Till Meyer
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Julius Maximilian University of Wuerzburg, D-97080 Wuerzburg, Germany
| | - Thomas Gehrke
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Julius Maximilian University of Wuerzburg, D-97080 Wuerzburg, Germany
| | - Norbert Kleinsasser
- Department of Otorhinolaryngology, Head and Neck Surgery, Kepler University, A-4020 Linz, Austria
| | - Rudolf Hagen
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Julius Maximilian University of Wuerzburg, D-97080 Wuerzburg, Germany
| | - Stephan Hackenberg
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Julius Maximilian University of Wuerzburg, D-97080 Wuerzburg, Germany
| |
Collapse
|
9
|
Abstract
Stem cells (SCs) are discovered long back but the idea that SCs possess therapeutic potential came up just a few decades back. In a past decade stem cell therapy is highly emerged and displayed tremendous potential for the treatment of a wide range of diseases and disorders such as blindness and vision impairment, type I diabetes, infertility, HIV, etc. SCs are very susceptible to destruction after transplantation into the host because of the inability to sustain elevated stress conditions inside the damaged tissue/organ. Heat shock proteins (HSPs) are molecular chaperones/stress proteins expressed in response to stress (elevated temperature, harmful chemicals, ischemia, viruses, etc) inside a living cell. HSPs protect the cell from damage by assisting in the proper folding of cellular proteins. This review briefly summarises different types of HSPs, their classification, cellular functions as well as the role of HSPs in regulating SC self-renewal and survival in the transplanted host. Applications of HSP modulated SCs in regenerative medicine and for the treatment of ischemic heart disease, myocardial infarction (MI), osteoarthritis, ischemic stroke, spinocerebellar ataxia type 3 (SCA3), leukemia, hepatic ischemia-reperfusion injury, Graft-versus-host disease (GVHD) and Parkinson's disease (PD) are discussed. In order to provide potential insights in understanding molecular mechanisms related to SCs in vertebrates, correlations between HSPs and SCs in cnidarians and planarians are also reviewed. There is a need to advance research in order to validate the use of HSPs for SC therapy and establish effective treatment strategies.
Collapse
|
10
|
Kim N, Ullah I, Chung K, Lee D, Cha MJ, Ban H, Choi CS, Kim S, Hwang KC, Kumar P, Lee SK. Targeted Delivery of Recombinant Heat Shock Protein 27 to Cardiomyocytes Promotes Recovery from Myocardial Infarction. Mol Pharm 2020; 17:2034-2043. [PMID: 32364395 DOI: 10.1021/acs.molpharmaceut.0c00192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ischemic heart disease, especially myocardial infarction (MI), is the leading cause of death worldwide. Apoptotic mechanisms are thought to play a significant role in cardiomyocyte death after MI. Increased production of heat shock proteins (Hsps) in cardiomyocytes is a normal response to promote tolerance and to reduce cell damage. Hsp27 is considered to be a therapeutic option for the treatment of ischemic heart disease due to its protective effects on hypoxia-induced apoptosis. Despite its antiapoptotic effects, the lack of strategies to deliver Hsp27 to the heart tissue in vivo limits its clinical applicability. In this study, we utilized an antibody against the angiotensin II type 1 (AT1) receptor, which is expressed immediately after ischemia/reperfusion in the heart of MI rats. To achieve cardiomyocyte-targeted Hsp27 delivery after ischemia/reperfusion, we employed the immunoglobulin-binding dimer ZZ, a modified domain of protein A, in conjunction with the AT1 receptor antibody. Using the AT1 receptor antibody, we achieved systemic delivery of ZZ-TAT-GFP fusion protein into the heart of MI rats. This approach enabled selective delivery of Hsp27 to cardiomyocytes, rescued cells from apoptosis, reduced the area of fibrosis, and improved cardiac function in the rat MI model, thus suggesting its applicability as a cardiomyocyte-targeted protein delivery system to inhibit apoptosis induced by ischemic injury.
Collapse
Affiliation(s)
- Nahyeon Kim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 133-791, Korea.,Samsung Bioepis, Incheon 21987, Korea
| | - Irfan Ullah
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 133-791, Korea.,Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, Connecticut 06510, United States
| | - Kunho Chung
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 133-791, Korea.,Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, Connecticut 06510, United States
| | - Dahye Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 133-791, Korea.,Green Cross Cell Co., Yongin 16924, Korea
| | - Min-Ji Cha
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Incheon 22711, Korea
| | - Hongseok Ban
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 133-791, Korea.,Ildong Pharmaceutical Co., Ltd., Hwaseong 445-710, Korea
| | - Chang Seon Choi
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 133-791, Korea.,Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, Connecticut 06510, United States
| | - Sunghwa Kim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 133-791, Korea.,LG Household & Health Care, Seoul 150-721, Korea
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Incheon 22711, Korea
| | - Priti Kumar
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, Connecticut 06510, United States
| | - Sang-Kyung Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 133-791, Korea
| |
Collapse
|
11
|
Shende P, Gandhewar N. Current Trend and Pro-survival Approaches for Augmenting Stem Cell Viability. Curr Pharm Biotechnol 2020; 21:1154-1164. [PMID: 32297579 DOI: 10.2174/1389201021666200416130253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Stem cells are of two types: embryonic and adult stem cells and they act as a repair system by replenishing body tissue. Stem cells differentiate into different types of cells, such as neural, hematopoietic, adipose, etc. and are used for the treatment of various conditions like myocardial infarction, spinal cord injury, Parkinson's disease and diabetes. METHODS This article focuses on recent research development that addresses the viability issues of stem cells. The efficiency of transplanted stem cells reduces due to conditions like hypoxia, inflammation, nutrient deprivation, immunogenicity, extracellular matrix loss on delivery and mechanical stress. RESULTS To increase the viability of stem cells, techniques like scaffolds of stem cells with hydrogel or alginate, pre-conditioning, different routes of administration and encapsulation, are implemented. CONCLUSION For the protection of stem cells against apoptosis, different pathways, namely Phosphoinositide 3-Kinase (PI3K/AKT), Hypoxia-Inducible Factor (HIF1), Mitogen-Activated Protein Kinases (MAPK) and Hippo, are discussed. DISCUSSION Activation of the PI3K/AKT pathway decreases the concentration of apoptotic factors, while the HIF pathway protects stem cells against the micro-environment of tissue (hypoxia).
Collapse
Affiliation(s)
- Pravin Shende
- Shobhaben Pratapbhai Patel School Pharmacy and Technology Management SVKM'S NMIMS, V.L Mehta Road, Vile Parle(W), Mumbai, India
| | - Nivedita Gandhewar
- Shobhaben Pratapbhai Patel School Pharmacy and Technology Management SVKM'S NMIMS, V.L Mehta Road, Vile Parle(W), Mumbai, India
| |
Collapse
|
12
|
Ticagrelor Enhances Release of Anti-Hypoxic Cardiac Progenitor Cell-Derived Exosomes Through Increasing Cell Proliferation In Vitro. Sci Rep 2020; 10:2494. [PMID: 32051439 PMCID: PMC7016113 DOI: 10.1038/s41598-020-59225-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 01/27/2020] [Indexed: 12/13/2022] Open
Abstract
Despite the widespread clinical use of cardioprotection by long-term direct antagonism of P2Y12 receptor, underlying mechanisms are unclear. Here, we identify how release of pro-survival exosomes from human cardiac-derived mesenchymal progenitor cells (hCPCs) is regulated by clinically relevant dose of ticagrelor (1 μM), an oral selective and reversible non-thienopyridine P2Y12 inhibitor. Ticagrelor-induced enhancement of exosome levels is related to increased mitotic activity of hCPCs. We show a drug-response threshold above which the effects on hCPCs are lost due to higher dose of ticagrelor and larger adenosine levels. While it is known that pan-Aurora kinase inhibitor halts cell proliferation through dephosphorylation of histone H3 residue Ser10, we demonstrate that it also prevents ticagrelor-induced effects on release of cardiac progenitor cell-derived exosomes delivering anti-apoptotic HSP70. Indeed, sustained pre-treatment of cardiomyocytes with exosomes released from explant-derived hCPCs exposed to low-dose ticagrelor attenuated hypoxia-induced apoptosis through acute phosphorylation of ERK42/44. Our data indicate that ticagrelor can be leveraged to modulate release of anti-hypoxic exosomes from resident hCPCs.
Collapse
|
13
|
Kim OH, Yoon OJ, Lee HJ. Silk fibroin scaffolds potentiate immunomodulatory function of human mesenchymal stromal cells. Biochem Biophys Res Commun 2019; 519:323-329. [PMID: 31506179 DOI: 10.1016/j.bbrc.2019.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 09/04/2019] [Indexed: 12/19/2022]
Abstract
Although mesenchymal stromal cells (MSCs) show great potential for use in regenerative medicine, their therapeutic efficacy remains limited because of their low adaptation efficiency and viability observed in clinical trials. To potentiate the adaptation and survival efficiency of MSCs after administration in vivo, silk fibroin nanofibers (SFNs) were applied as a scaffold. SFNs are biocompatible, biodegradable polymers with tunable architectures and mechanical properties. Treatment with interferon (IFN)-γ for 18 h increased the expression of immunomodulatory functional cytokines, IDO and COX2 in MSCs. Further, the MSCs grown on SFN sheets showed enhanced IDO1 and COX2 expression following IFN-γ treatment. MSCs showed significantly greater migratory ability on SFN sheets than on glass surfaces or PLGA control sheets. Though IFN-γ treatment slightly reduced the migration ability of MSCs cultured on glass or poly(lactic-co-glycolic acid) (PLGA) nanofiber sheets, it did not alter MSC motility on SFN sheets. Furthermore, MSCs cultured on SFN sheets dramatically suppressed TNF-α secretion from lipopolysaccharide-activated murine splenocytes, suggesting that the immunomodulatory function of MSCs was enhanced by the SFN sheets. Taken together, these data demonstrate that SFN sheets potentiate the reparative and regenerative properties of MSCs.
Collapse
Affiliation(s)
- Ok-Hyeon Kim
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul, 06974, South Korea
| | - Ok Ja Yoon
- Da Vinci College of General Education, Chung-Ang University, Seoul, 06974, South Korea
| | - Hyun Jung Lee
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul, 06974, South Korea.
| |
Collapse
|
14
|
Zeng Y, Li B, Li T, Liu W, Ran C, Penson RT, Poznansky MC, Du Y, Chen H. CD90 low MSCs modulate intratumoral immunity to confer antitumor activity in a mouse model of ovarian cancer. Oncotarget 2019; 10:4479-4491. [PMID: 31320999 PMCID: PMC6633895 DOI: 10.18632/oncotarget.27065] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 06/19/2019] [Indexed: 12/15/2022] Open
Abstract
Both anti-tumoral and pro-tumoral effects of mesenchymal stem cells (MSCs) in preclinical treatment of ovarian cancer have been controversially demonstrated. In this study, we profiled the phenotypes of mouse compact bone-derived MSCs (CB-MSCs) and bone marrow-derived MSCs (BM-MSCs) and found that CB-MSCs expressed lower CD90 compared to BM-MSCs. We examined gene expression of immune regulating cytokines of CB-MSCs in 2D and 3D culture and under stimulation with TLR4 agonist LPS or immune activator VIC-008. Our data showed that when CB-MSCs were cultured in simulated in vivo 3D condition, CD90 expression was further decreased. Moreover, gene expressions of immune activating cytokines IL-12, IL-21, IFNγ and a pro-inflammatory cytokine CXCL10 in CB-MSCs were increased in 3D culture whereas gene expression of anti-inflammatory cytokines IL-10 and CCL5 were downregulated. Stimulation of CB-MSCs by LPS or VIC-008 presented similar profile of the cytokine gene expressions to that in 3D culture which might benefit the anti-tumor efficacy of CD90low MSCs. The anti-tumor effects of CD90low CB-MSCs alone or in combination with VIC-008 were evaluated in a syngeneic orthotopic mouse model of ovarian cancer. Treatment that combines CB-MSCs and VIC-008 significantly decreased tumor growth and prolonged mouse survival. This was associated with the increase of activated anti-tumoral CD4+ and CD8+ T cells and the decrease of Treg cells in the tumor microenvironment. Taken together, our study demonstrates the synergistic anti-tumoral efficacy by application of CB-MSCs combined with immune activator VIC-008 and provides new insight into CD90low MSCs as a new anti-tumor arsenal.
Collapse
Affiliation(s)
- Yang Zeng
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston 02114, USA
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston 02215, USA
| | - Binghao Li
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston 02114, USA
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Tao Li
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston 02114, USA
- Jiangsu Key Laboratory of Clinical Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Wei Liu
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China
| | - Chongzhao Ran
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown 02129, USA
| | - Richard T. Penson
- Medical Gynecologic Oncology, Gillette Center for Women's Cancers, Massachusetts General Hospital and Harvard Medical School, Boston 02114, USA
| | - Mark C. Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston 02114, USA
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China
| | - Huabiao Chen
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston 02114, USA
| |
Collapse
|
15
|
Kim R, Kim P, Lee CY, Lee S, Yun H, Lee MY, Kim J, Baek K, Chang W. Multiple Combination of Angelica gigas Extract and Mesenchymal Stem Cells Enhances Therapeutic Effect. Biol Pharm Bull 2019; 41:1748-1756. [PMID: 30504677 DOI: 10.1248/bpb.b18-00193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Alternative medicines attract attention because stroke is rarely expected to make a full recovery with the most advanced medical technology. Angelica gigas (AG) is a well-known herbal medicine as a neuroprotective agent. The present study introduced mesenchymal stem cells (MSCs) to identify for the advanced treatment of the cerebrovascular disease. The objective of this research is validation of the enhanced effects of multiple combined treatment of AG extract with MSCs on stroke through angiogenesis. Our results confirmed that AG extract with MSCs improved the neovascularization increasing expression of angiogenesis-regulated molecules. The changes of brain and the behavioral ability showed the increased effects of AG extract with MSCs. As a result, AG extract and MSCs may synergistically increase the therapeutic potential by enhancing neovascularization. This mixed approach provides a new experimental protocol of herbal medicine therapy for the treatment of a variety of diseases including stroke, trauma, and spinal cord injury.
Collapse
Affiliation(s)
- Ran Kim
- Department of Biology Education, College of Education, Pusan National University
| | - Pilseog Kim
- Department of Biology Education, College of Education, Pusan National University
| | - Chang Youn Lee
- Department of Integrated Omics for Biomedical Sciences, Yonsei University
| | - Seokyeon Lee
- Department of Biology Education, College of Education, Pusan National University
| | | | - Min Young Lee
- Department of Molecular Physiology, College of Pharmacy, Kyungpook National University
| | - Jongmin Kim
- Department of Life Systems, Sookmyung Women's University
| | - Kyungmin Baek
- Department of Cardiovascular and Neurologic Disease, College of Oriental Medicine, Daegu Hanny University
| | - Woochul Chang
- Department of Biology Education, College of Education, Pusan National University
| |
Collapse
|
16
|
Kuljanin M, Elgamal RM, Bell GI, Xenocostas A, Lajoie GA, Hess DA. Human Multipotent Stromal Cell Secreted Effectors Accelerate Islet Regeneration. Stem Cells 2019; 37:516-528. [DOI: 10.1002/stem.2976] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/12/2018] [Accepted: 12/28/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Miljan Kuljanin
- Don Rix Protein Identification Facility, Department of Biochemistry, Schulich School of Medicine & Dentistry; Western University; London Ontario Canada
- Krembil Centre for Stem Cell Biology, Molecular Medicine Research Laboratories; Robarts Research Institute; London Ontario Canada
| | - Ruth M. Elgamal
- Krembil Centre for Stem Cell Biology, Molecular Medicine Research Laboratories; Robarts Research Institute; London Ontario Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry; Western University; London Ontario Canada
| | - Gillian I. Bell
- Krembil Centre for Stem Cell Biology, Molecular Medicine Research Laboratories; Robarts Research Institute; London Ontario Canada
| | - Anargyros Xenocostas
- Department of Medicine, Division of Haematology, Schulich School of Medicine and Dentistry; Western University; London Ontario Canada
| | - Gilles A. Lajoie
- Don Rix Protein Identification Facility, Department of Biochemistry, Schulich School of Medicine & Dentistry; Western University; London Ontario Canada
| | - David A. Hess
- Krembil Centre for Stem Cell Biology, Molecular Medicine Research Laboratories; Robarts Research Institute; London Ontario Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry; Western University; London Ontario Canada
| |
Collapse
|
17
|
Chae DS, Lee CY, Lee J, Seo HH, Choi CH, Lee S, Hwang KC. Priming stem cells with protein kinase C activator enhances early stem cell-chondrocyte interaction by increasing adhesion molecules. Biol Res 2018; 51:41. [PMID: 30384862 PMCID: PMC6211543 DOI: 10.1186/s40659-018-0191-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 10/16/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) can be defined as degradation of articular cartilage of the joint, and is the most common degenerative disease. To regenerate the damaged cartilage, different experimental approaches including stem cell therapy have been tried. One of the major limitations of stem cell therapy is the poor post-transplantation survival of the stem cells. Anoikis, where insufficient matrix support and adhesion to extracellular matrix causes apoptotic cell death, is one of the main causes of the low post-transplantation survival rate of stem cells. Therefore, enhancing the initial interaction of the transplanted stem cells with chondrocytes could improve the therapeutic efficacy of stem cell therapy for OA. Previously, protein kinase C activator phorbol 12-myristate 13-acetate (PMA)-induced increase of mesenchymal stem cell adhesion via activation of focal adhesion kinase (FAK) has been reported. In the present study, we examine the effect PMA on the adipose-derived stem cells (ADSCs) adhesion and spreading to culture substrates, and further on the initial interaction between ADSC and chondrocytes. RESULTS PMA treatment increased the initial adhesion of ADSC to culture substrate and cellular spreading with increased expression of adhesion molecules, such as FAK, vinculin, talin, and paxillin, at both RNA and protein level. Priming of ADSC with PMA increased the number of ADSCs attached to confluent layer of cultured chondrocytes compared to that of untreated ADSCs at early time point (4 h after seeding). CONCLUSION Taken together, the results of this study suggest that priming ADSCs with PMA can increase the initial interaction with chondrocytes, and this proof of concept can be used to develop a non-invasive therapeutic approach for treating OA. It may also accelerate the regeneration process so that it can relieve the accompanied pain faster in OA patients. Further in vivo studies examining the therapeutic effect of PMA pretreatment of ADSCs for articular cartilage damage are required.
Collapse
Affiliation(s)
- Dong-Sik Chae
- Department of Medicine, The Graduate School, Yonsei University, Seoul, South Korea.,Department of Orthopedic Surgery, International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon, South Korea
| | - Chang Youn Lee
- Department of Integrated Omics for Biomedical Sciences, Yonsei University, Seoul, South Korea
| | - Jiyun Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, South Korea
| | - Hyang-Hee Seo
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, South Korea
| | - Chong-Hyuk Choi
- Department of Orthopedic Surgery, Yonsei University College of Medicine, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, South Korea.
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, South Korea.
| |
Collapse
|
18
|
Song Y, Zhong C, Wang X. Heat shock protein 70: A promising therapeutic target for myocardial ischemia–reperfusion injury. J Cell Physiol 2018; 234:1190-1207. [DOI: 10.1002/jcp.27110] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 06/29/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Yan‐Jun Song
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure, Laboratory of Heart Center, Department of Cardiology, Heart Center, Zhujiang Hospital Southern Medical University Guangzhou China
- School of Laboratory Medicine and Biotechnology Southern Medical University Guangzhou China
| | - Chong‐Bin Zhong
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure, Laboratory of Heart Center, Department of Cardiology, Heart Center, Zhujiang Hospital Southern Medical University Guangzhou China
| | - Xian‐Bao Wang
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure, Laboratory of Heart Center, Department of Cardiology, Heart Center, Zhujiang Hospital Southern Medical University Guangzhou China
| |
Collapse
|
19
|
Andreeva ER, Buravkova LB. The Role of Interplay of Mesenchymal Stromal Cells and Macrophages in Physiological and Reparative Tissue Remodeling. ACTA ACUST UNITED AC 2018. [DOI: 10.1134/s0362119718010036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
20
|
Hu M, Guo G, Huang Q, Cheng C, Xu R, Li A, Liu N, Liu S. The harsh microenvironment in infarcted heart accelerates transplanted bone marrow mesenchymal stem cells injury: the role of injured cardiomyocytes-derived exosomes. Cell Death Dis 2018; 9:357. [PMID: 29500342 PMCID: PMC5834521 DOI: 10.1038/s41419-018-0392-5] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 12/11/2022]
Abstract
Stem cell therapy can be used to repair and regenerate damaged hearts tissue; nevertheless, the low survival rate of transplanted cells limits their therapeutic efficacy. Recently, it has been proposed that exosomes regulate multiple cellular processes by mediating cell survival and communication among cells. The following study investigates whether injured cardiomyocytes-derived exosomes (cardiac exosomes) affect the survival of transplanted bone marrow mesenchymal stem cells (BMSCs) in infarcted heart. To mimic the harsh microenvironment in infarcted heart that the cardiomyocytes or transplanted BMSCs encounter in vivo, cardiomyocytes conditioned medium and cardiac exosomes collected from H2O2-treated cardiomyocytes culture medium were cultured with BMSCs under oxidative stress in vitro. Cardiomyocytes conditioned medium and cardiac exosomes significantly accelerated the injury of BMSCs induced by H2O2; increased cleaved caspase-3/caspase-3 and apoptotic percentage, and decreased the ratio of Bcl-2/Bax and cell viability in those cells. Next, we explored the role of cardiac exosomes in the survival of transplanted BMSCs in vivo by constructing a Rab27a knockout (KO) mice model by a transcription activator-like effector nuclease (TALEN) genome-editing technique; Rab27a is a family of GTPases, which has critical role in secretion of exosomes. Male mouse GFP-modified BMSCs were implanted into the viable myocardium bordering the infarction in Rab27a KO and wild-type female mice. The obtained results showed that the transplanted BMSCs survival in infarcted heart was increased in Rab27a KO mice by the higher level of Y-chromosome Sry DNA, GFP mRNA, and the GFP fluorescence signal intensity. To sum up, these findings revealed that the injured cardiomyocytes-derived exosomes accelerate transplanted BMSCs injury in infarcted heart, thus highlighting a new mechanism underlying the survival of transplanted cells after myocardial infarction.
Collapse
Affiliation(s)
- Ming Hu
- Guangzhou Institute of Cardiovascular Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
| | - Guixian Guo
- Guangzhou Institute of Cardiovascular Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
| | - Qiang Huang
- Guangzhou Institute of Cardiovascular Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
| | - Chuanfang Cheng
- Guangzhou Institute of Cardiovascular Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
| | - Ruqin Xu
- Guangzhou Institute of Cardiovascular Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
| | - Aiqun Li
- Guangzhou Institute of Cardiovascular Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
| | - Ningning Liu
- Guangzhou Institute of Cardiovascular Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, China.
| | - Shiming Liu
- Guangzhou Institute of Cardiovascular Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, China.
| |
Collapse
|
21
|
Wu R, Hu X, Wang J. Concise Review: Optimized Strategies for Stem Cell-Based Therapy in Myocardial Repair: Clinical Translatability and Potential Limitation. Stem Cells 2018; 36:482-500. [PMID: 29330880 DOI: 10.1002/stem.2778] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 12/28/2017] [Accepted: 12/31/2017] [Indexed: 12/15/2022]
Abstract
Ischemic heart diseases (IHDs) remain major public health problems with high rates of morbidity and mortality worldwide. Despite significant advances, current therapeutic approaches are unable to rescue the extensive and irreversible loss of cardiomyocytes caused by severe ischemia. Over the past 16 years, stem cell-based therapy has been recognized as an innovative strategy for cardiac repair/regeneration and functional recovery after IHDs. Although substantial preclinical animal studies using a variety of stem/progenitor cells have shown promising results, there is a tremendous degree of skepticism in the clinical community as many stem cell trials do not confer any beneficial effects. How to accelerate stem cell-based therapy toward successful clinical application attracts considerate attention. However, many important issues need to be fully addressed. In this Review, we have described and compared the effects of different types of stem cells with their dose, delivery routes, and timing that have been routinely tested in recent preclinical and clinical findings. We have also discussed the potential mechanisms of action of stem cells, and explored the role and underlying regulatory components of stem cell-derived secretomes/exosomes in myocardial repair. Furthermore, we have critically reviewed the different strategies for optimizing both donor stem cells and the target cardiac microenvironments to enhance the engraftment and efficacy of stem cells, highlighting their clinical translatability and potential limitation. Stem Cells 2018;36:482-500.
Collapse
Affiliation(s)
- Rongrong Wu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Xinyang Hu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Jian'an Wang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| |
Collapse
|
22
|
Ma JL, Guo WL, Chen XM. Overexpressing microRNA-150 attenuates hypoxia-induced human cardiomyocyte cell apoptosis by targeting glucose-regulated protein-94. Mol Med Rep 2018; 17:4181-4186. [PMID: 29328381 PMCID: PMC5802188 DOI: 10.3892/mmr.2018.8375] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 06/13/2017] [Indexed: 01/10/2023] Open
Abstract
MicroRNA (miR)-150 has been demonstrated to protect the heart from ischemic injury. However, the protective effect of miR-150 in hypoxia-injured cardiomyocytes remains unclear. The present study aimed to investigate the target gene of miR-150 and the underlying molecular mechanisms of miR-150 in hypoxia-induced cardiomyocyte apoptosis. Using the hypoxia model of human cardiomyocytes (HCMs) in vitro, it was demonstrated that miR-150 was markedly inhibited in HCMs after hypoxia treatment. Overexpressing miR-150 significantly decreased hypoxia-induced HCM death and apoptosis. In addition, GRP94 was revealed to be a direct target of miR-150. Additionally, GRP94 was demonstrated to be involved in hypoxia-induced HCM apoptosis, and the protein expression levels of GRP94 were increased in HCMs in the presence of hypoxia. These findings demonstrated that miR-150 is involved in hypoxia-mediated gene regulation and apoptosis in HCMs. Furthermore, GRP94 knockout increased the cell viability of hypoxia-impaired HCMs with miR-150 mimic or miR-150 inhibitor transfection. In conclusion, miR-150 may serve a protective role in cardiomyocyte hypoxia injury, and the underlying mechanism was mediated, at least partially, by inhibiting GRP94 expression. These findings may provide a novel insight for the therapy of hypoxia-induced myocardial I/R injury.
Collapse
Affiliation(s)
- Jian-Lin Ma
- Department of Emergency, Binzhou City Central Hospital, Binzhou, Shandong 251700, P.R. China
| | - Wen-Ling Guo
- Department of Obstetrics, Binzhou City Central Hospital, Binzhou, Shandong 251700, P.R. China
| | - Xue-Mei Chen
- Department of Obstetrics, Binzhou City Central Hospital, Binzhou, Shandong 251700, P.R. China
| |
Collapse
|
23
|
Heat-Shock Protein 70 Overexpression in Adipose-Derived Stem Cells Enhances Fat Graft Survival. Ann Plast Surg 2017; 78:460-466. [PMID: 28106629 DOI: 10.1097/sap.0000000000000968] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Autologous fat grafting is a prevalent technique used for soft-tissue augmentation; however, the poor survival rate of the grafted tissue remains a drawback of this method. Although adipose-derived stem cells (ASCs) are an attractive candidate for enhancing graft retention, the poor posttransplantation viability of these cells limits their application. Here we investigated whether overexpression of the antiapoptotic protein heat-shock protein 70 (Hsp70) could enhance ASCs' therapeutic potential for fat transplant survival. METHODS Recombinant adenoviral vectors were used to overexpress Hsp70 in ASCs isolated from a healthy woman. The Hsp70 expression was assessed by quantitative real-time polymerase chain reaction and Western blot analyses. The adipose tissue granules aspirated from another woman were mixed with ASCs expressing green fluorescent protein (GFP)-tagged Hsp70 (group A) or GFP alone (group B), untreated ASCs (group C), and phosphate-buffered saline (group D). Fat mixtures were then injected subcutaneously into the backs of nude mice, and graft survival was compared after 3 months. RESULTS Adipose-derived stem cells transduced with recombinant adenoviral vectors exhibited significantly increased Hsp70 expression in vitro. Meanwhile, weight retention analyses demonstrated that fat grafts using the group A cell population exhibited significantly higher survival rates than the other treatment groups in vivo. Moreover, histological analyses revealed that fat grafts containing GFP-Hsp70-expressing ASCs yielded significantly lower levels of tissue fibrosis and fat cysts/vacuoles, higher capillary densities, and increased numbers of viable adipocytes than the control groups. CONCLUSIONS Our data indicate that Hsp70 overexpression enhances the efficacy of ASC therapy by improving the survival and quality of the transplanted fat tissues.
Collapse
|
24
|
Selaginella bryopteris Aqueous Extract Improves Stability and Function of Cryopreserved Human Mesenchymal Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:8530656. [PMID: 28811868 PMCID: PMC5546052 DOI: 10.1155/2017/8530656] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/02/2017] [Accepted: 06/15/2017] [Indexed: 02/07/2023]
Abstract
The effective long-term cryopreservation of human mesenchymal stem cells (MSCs) is an essential prerequisite step and represents a critical approach for their sustained supply in basic research, regenerative medicine, and tissue engineering applications. Therefore, attempts have been made in the present investigation to formulate a freezing solution consisting of a combination of Selaginella bryopteris water-soluble extract with and without dimethyl sulfoxide (Me2SO) for the efficient long-term storage of human umbilical cord blood- (hUCB-) derived MSCs. The cryopreservation experiment using the formulated freezing solution was further performed with hUCB MSCs in a controlled rate freezer. A significant increase in postthaw cell viability and cell attachment of MSCs was achieved with freezing medium containing Selaginella bryopteris water extract along with 10% Me2SO as compared to the freezing medium containing Me2SO (10% v/v) alone. Furthermore, the decreasing apoptotic events and reactive oxygen species production along with increasing expression of heat shock proteins also confirmed the beneficial effect of Selaginella bryopteris water extract. The beneficial effect of Selaginella bryopteris water extract was validated by its ability to render postpreservation high cell viability. In conclusion, the formulated freezing solution has been demonstrated to be effective for the standardization of cryopreservation protocol for hMSCs.
Collapse
|
25
|
Mullick M, Venkatesh K, Sen D. d-Alanine 2, Leucine 5 Enkephaline (DADLE)-mediated DOR activation augments human hUCB-BFs viability subjected to oxidative stress via attenuation of the UPR. Stem Cell Res 2017; 22:20-28. [DOI: 10.1016/j.scr.2017.05.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 05/15/2017] [Accepted: 05/21/2017] [Indexed: 01/16/2023] Open
|
26
|
Husak Z, Dworzak MN. Chronic stress induces CD99, suppresses autophagy, and affects spontaneous adipogenesis in human bone marrow stromal cells. Stem Cell Res Ther 2017; 8:83. [PMID: 28420430 PMCID: PMC5395812 DOI: 10.1186/s13287-017-0532-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 02/20/2017] [Accepted: 03/09/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bone marrow-derived mesenchymal stromal cells (MSCs) are multipotent cells with a high constitutive level of autophagy and low expression of CD99. Under certain conditions, MSCs may develop tumorigenic properties. However, these transformation-induced conditions are largely unknown. Recently, we have identified an association between Hsp70, a main participant in cellular stress response and tumorigenesis, and CD99. Preliminary observations had revealed upregulation of both proteins in stressed long-term cultured MSCs. And so we hypothesized that CD99 is implicated in stress-induced mechanisms of cellular transformation in MSCs. Hence, we investigated the effects of prolonged stress on MSCs and the role of CD99 and autophagy in their survival. METHODS Human telomerase reverse transcriptase (hTERT) overexpressing immortalized MSCs and primary bone marrow stromal cells were used to investigate the influence of long-term serum deprivation and hypoxia on growth and differentiation of MSCs. Cell proliferation and apoptosis were evaluated using flow cytometry, differentiation capabilities of MSCs were assessed by immunohistochemical staining followed by microscopic examination. CD99, Hsp70 expression were analyzed using flow cytometry, western blotting, and reverse transcriptase polymerase chain reaction. Autophagy was explored with specific inhibitors using cell morphology examination and western blotting. RESULTS Chronic stress factors are able to change the morphology of MSCs and to inhibit spontaneous differentiation into adipocyte lineage. Furthermore, CD99 elevation and downregulation of p53 and p21 accompanied defective autophagy, which is usually associated with tumor formation. We found that inhibition of autophagy by chloroquine promoted cell detachment and modulated CD99 expression level whereas incorporation of CD99 recombinant protein into the cells suppressed autophagy. CONCLUSIONS Obtained results provide a model for chronic stress-induced transformation of MSCs via CD99 and may therefore be highly relevant to mesenchymal tumorigenesis.
Collapse
Affiliation(s)
- Zvenyslava Husak
- St. Anna Kinderkrebsforschung, Children’s Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
| | - Michael N. Dworzak
- St. Anna Kinderkrebsforschung, Children’s Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
- St. Anna Kinderspital, Kinderspitalgasse 6, 1090 Vienna, Austria
| |
Collapse
|
27
|
SRT1720 promotes survival of aged human mesenchymal stem cells via FAIM: a pharmacological strategy to improve stem cell-based therapy for rat myocardial infarction. Cell Death Dis 2017; 8:e2731. [PMID: 28383554 PMCID: PMC5477573 DOI: 10.1038/cddis.2017.107] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 01/24/2017] [Accepted: 02/03/2017] [Indexed: 02/07/2023]
Abstract
SIRT1 has been proved to rejuvenate and improve the therapeutic efficacy of aged rat mesenchymal stem cells (MSCs). Herein, we investigate the protective effect of pretreatment with SIRT1 activator SRT1720 on aged human MSCs (hMSCs). The optimized pretreatment condition for aged hMSCs was determined to be 0.5 μM SRT1720 for 24 h by monitoring the survival of aged hMSCs subjected to serum deprivation±hypoxia and±500 μM hydrogen peroxide (H2O2). Pretreatment with these conditions increased the survival of aged hMSCs 1 day (2.7-fold) and 3 days (1.9-fold) after being transplanted into a rat myocardial infarction (MI) model created by ligation of the left anterior descending (LAD) coronary artery. Transplantation with SRT1720 pretreated aged hMSCs achieved increased left ventricular ejection fraction (58.9±3.6 versus 52.8±5%) and angiogenesis with reduced fibrosis of rat hearts as compared to DMSO pretreated group 28 days following MI. Unbiased transcriptome analysis conducted on aged hMSCs under oxidative stress indicated the Fas apoptosis inhibitory molecule (FAIM) was significantly upregulated following SRT1720 pretreatment (14.9±0.2-folds). Moreover, the anti-apoptotic effect of SRT1720 was mitigated by FAIM knockdown with a small interfering RNA-targeted FAIM. These results indicated that pretreatment with SRT1720 improves survival of aged hMSCs, and enhances their therapeutic efficacy for rat myocardial infarction (MI). Upregulation of FAIM possibly involves in the mechanisms of the protective effects.
Collapse
|
28
|
Acute Hypoxic Stress Affects Migration Machinery of Tissue O 2-Adapted Adipose Stromal Cells. Stem Cells Int 2016; 2016:7260562. [PMID: 28115943 PMCID: PMC5225392 DOI: 10.1155/2016/7260562] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 11/01/2016] [Accepted: 11/16/2016] [Indexed: 12/17/2022] Open
Abstract
The ability of mesenchymal stromal (stem) cells (MSCs) to be mobilised from their local depot towards sites of injury and to participate in tissue repair makes these cells promising candidates for cell therapy. Physiological O2 tension in an MSC niche in vivo is about 4-7%. However, most in vitro studies of MSC functional activity are performed at 20% O2. Therefore, this study focused on the effects of short-term hypoxic stress (0.1% O2, 24 h) on adipose tissue-derived MSC motility at tissue-related O2 level. No significant changes in integrin expression were detected after short-term hypoxic stress. However, O2 deprivation provoked vimentin disassembly and actin polymerisation and increased cell stiffness. In addition, hypoxic stress induced the downregulation of ACTR3, DSTN, MACF1, MID1, MYPT1, NCK1, ROCK1, TIAM1, and WASF1 expression, the products of which are known to be involved in leading edge formation and cell translocation. These changes were accompanied by the attenuation of targeted and nontargeted migration of MSCs after short-term hypoxic exposure, as demonstrated in scratch and transwell migration assays. These results indicate that acute hypoxic stress can modulate MSC function in their native milieu, preventing their mobilisation from sites of injury.
Collapse
|
29
|
Feyen DA, Gaetani R, Doevendans PA, Sluijter JP. Stem cell-based therapy: Improving myocardial cell delivery. Adv Drug Deliv Rev 2016; 106:104-115. [PMID: 27133386 DOI: 10.1016/j.addr.2016.04.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/19/2016] [Accepted: 04/20/2016] [Indexed: 12/15/2022]
Abstract
Stem cell-based therapies form an exciting new class of medicine that attempt to provide the body with the building blocks required for the reconstruction of damaged organs. However, delivering cells to the correct location, while preserving their integrity and functional properties, is a complex undertaking. These challenges have led to the development of a highly dynamic interdisciplinary research field, wherein medical, biological, and chemical sciences have collaborated to develop strategies to overcome the physiological barriers imposed on the cellular therapeutics. In this respect, improving the acute retention and subsequent survival of stem cells is key to effectively increase the effect of the therapy, while proper tissue integration is imperative for stem cells to functionally replace lost cells in damaged organs. In this review, we will use the heart as an example to highlight the current knowledge of therapeutic stem cell utilization, the existing pitfalls and limitations, and the approaches that have been developed to overcome them.
Collapse
|
30
|
Andreeva ER, Lobanova MV, Udartseva OO, Buravkova LB. Response of Adipose Tissue-Derived Stromal Cells in Tissue-Related O2 Microenvironment to Short-Term Hypoxic Stress. Cells Tissues Organs 2015; 200:307-15. [PMID: 26407140 DOI: 10.1159/000438921] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2015] [Indexed: 12/19/2022] Open
Abstract
A microenvironment low in O2 ('physiological' hypoxia) governs the functions of perivascular multipotent mesenchymal stromal cells, defining their involvement in tissue physiological homeostasis and regenerative remodelling. Acute hypoxic stress is considered as one of the important factors inducing tissue damage. Here, we evaluate the influence of short-term hypoxia (1% O2 for 24 h) on perivascular adipose tissue-derived cells (ASCs) permanently expanded in tissue-related O2 (5%) microenvironment. After hypoxic exposure, ASCs retained high viability, stromal cell morphology and mesenchymal phenotype (CD73+, CD90+, CD105+ and CD45-). Mild oxidative damage was unveiled as elevation of reactive oxygen species and thiobarbituric acid-active products, while no reduction in the activity of the antioxidant enzymes catalase and glutathione peroxidase and a 20% statistically significant increase in superoxide dismutase activity was detected. Expression of hypoxia-inducible factor (HIF)-1α and HIF-3α isoforms was differently regulated. HIF-1α displayed transient up-regulation, with maximum levels 30 min after acute hypoxic exposure, while HIF-3α was significantly up-regulated after 24 h. Up-regulation of ERK7, MEK1 and c-fos, and down-regulation of MKK6, p53, CCNA2, CCNB1 and CCNB2 were observed after 24 h of oxygen deprivation. Acute hypoxic exposure did not affect the gene expression of other mitogen-activated protein kinases (MAPKs) and MAPK kinases, MAPK/ERK kinase-interacting proteins, MAPK-activated transcription factors and scaffolding proteins. Significant stimulation of vascular endothelial growth factor α and interleukin-6 production was detected in ASC-conditioned medium. Thus, tissue O2-adapted ASCs are resistant to hypoxic stress, which can ensure their effective involvement in the regeneration of tissue damage under significant oxygen deprivation.
Collapse
Affiliation(s)
- Elena R Andreeva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | | | | | | |
Collapse
|
31
|
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have demonstrated significant potentials for the treatment of inflammatory bowel disease. Clinical feasible methods to individually document the MSC recruitment to intestinal mucosa is lacking. Here, we proposed that endomicroscopy could noninvasively track MSCs in vivo at cellular resolution. METHOD Isolated Sprague Dawley rat MSC was characterized, fluorescently labeled, and imaged ex vivo using an endomicroscope. Then enhanced green fluorescent protein (eGFP)-labeled MSC was tracked in vivo, and acquired images were compared with immunofluorescence, immunohistology, and fluorescent in situ hybridization results. RESULTS Endomicroscopy visualized clearly the eGFP-labeled or carboxyfluorescein succinimidyl ester-stained MSC ex vivo. Endomicroscopy using the FIVE1 system could track eGFP-labeled MSC with distinct in vivo features. Immunofluorescence, immunohistochemistry, and fluorescent in situ hybridization confirmed the presence of eGFP-positive cells. In vivo endomicroscopy could quantify the transplanted MSCs that homed to colonic mucosa of the recipient rat in multiple models, including the rat-to-rat allograft, human-to-rat xenograft, hypoxia-induced MSC, and busulfan immunosuppressed recipient rat models. After hypoxia induction, there was a trend of enhanced rat MSC homing to the inflamed mucosa as visualized by endomicroscopy (114.1 in hypoxia group versus 34.3 in other 3 groups combined, t = 2.14, P = 0.0644). CONCLUSIONS Endomicroscopy is a novel and promising tool to track transplanted MSCs to the colonic mucosa. This clinical available noninvasive cellular tracking method may provide new insight to individualize each recipient's regimen in the future.
Collapse
|
32
|
Hua P, Liu JY, Tao J, Yang SR. Application and Progress of Combined Mesenchymal Stem Cell Transplantation in the Treatment of Ischemic Cardiomyopathy. BIOMED RESEARCH INTERNATIONAL 2015; 2015:568502. [PMID: 26295041 PMCID: PMC4532814 DOI: 10.1155/2015/568502] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/07/2015] [Indexed: 11/29/2022]
Abstract
Treatment of ischemic cardiomyopathy caused by myocardial infarction (MI) using mesenchymal stem cell (MSC) transplantation is a widely researched field, with promising clinical application. However, the low survival rate of transplanted cells has a severe impact on treatment outcome. Currently, research is focused on investigating the strategy of combining genetic engineering, tissue engineering materials, and drug/hypoxia preconditioning to improve ischemic cardiomyopathy treatment outcome using MSC transplantation treatment (MSCTT). This review discusses the application and progress of these techniques.
Collapse
Affiliation(s)
- Ping Hua
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jian-Yang Liu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jun Tao
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Song-Ran Yang
- Department of Experimental Psychology, University of Oxford, Oxford OX1 3UD, UK
| |
Collapse
|
33
|
Qin J, Yuan F, Peng Z, Ye K, Yang X, Huang L, Jiang M, Lu X. Periostin enhances adipose-derived stem cell adhesion, migration, and therapeutic efficiency in Apo E deficient mice with hind limb ischemia. Stem Cell Res Ther 2015. [PMID: 26204963 PMCID: PMC4533765 DOI: 10.1186/s13287-015-0126-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Introduction Therapeutic angiogenesis by transplantation of autologous/allogeneic adipose-derived stem cells (ADSCs) is a potential approach for severe ischemic diseases. However, poor viability, adhesion, migration and differentiation limit the therapeutic efficiency after the cells were transplanted into the targeted area. Periostin, an extracellular matrix protein, exhibits a critical role in wound repair as well as promotes cell adhesion, survival, and angiogenesis. Method ADSCs were obtained and genetically engineered with periostin gene (P-ADSCs). The viability, proliferation, migration, and apoptosis of P-ADSCs under hypoxia were analyzed. Moreover, P-ADSCs were implanted into Apo E deficient mice with hind limb ischemia. The Laser Doppler perfusion index, immunofluorescence, and histological pathology assay were tested to evaluate the therapeutic effects. The associated molecular mechanism of periostin on the proliferation, adhesion, migration, and differentiation of ADSCs was also analyzed. Results The in vitro studies have shown that periostin-transfected ADSCs (P-ADSCs) promoted viability, proliferation, and migration of ADSCs. Apoptosis of ADSCs was inhibited under hypoxic conditions. The Laser Doppler perfusion index was significantly higher in the P-ADSCs group compared with that in the ADSC and control groups after 4 weeks. Immunofluorescence and histological pathology assay showed that the P-ADSCs were in and around the ischemic sites, and some cells differentiated into capillaries and endothelium. Microvessel densities were significantly improved in P-ADSCs group compared with those in the control group. The molecular mechanisms that provide the beneficial effects of periostin were connected with the upregulated expression of integrinβ1/FAK/PI3K/Akt/eNOS signal pathway and the increased secretion of growth factors. Conclusion Overexpression of periostin by gene transfection on ADSCs promotes survival, migration, and therapeutic efficiency, which will bring new insights into the treatment of critical limb ischemia. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0126-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jinbao Qin
- Department of Vascular Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai JiaoTong University, School of Medicine, Shanghai, 200011, People's Republic of China.
| | - Fukang Yuan
- Department of Vascular Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai JiaoTong University, School of Medicine, Shanghai, 200011, People's Republic of China.
| | - Zhiyou Peng
- Department of Vascular Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai JiaoTong University, School of Medicine, Shanghai, 200011, People's Republic of China.
| | - Kaichuang Ye
- Department of Vascular Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai JiaoTong University, School of Medicine, Shanghai, 200011, People's Republic of China.
| | - Xinrui Yang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai JiaoTong University, School of Medicine, Shanghai, 200011, People's Republic of China.
| | - Lijia Huang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai JiaoTong University, School of Medicine, Shanghai, 200011, People's Republic of China.
| | - Mier Jiang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai JiaoTong University, School of Medicine, Shanghai, 200011, People's Republic of China.
| | - Xinwu Lu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai JiaoTong University, School of Medicine, Shanghai, 200011, People's Republic of China. .,Vascular Center of Shanghai JiaoTong University, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
34
|
Chang W, Kim R, Park SI, Jung YJ, Ham O, Lee J, Kim JH, Oh S, Lee MY, Kim J, Park MS, Chung YA, Hwang KC, Maeng LS. Enhanced Healing of Rat Calvarial Bone Defects with Hypoxic Conditioned Medium from Mesenchymal Stem Cells through Increased Endogenous Stem Cell Migration via Regulation of ICAM-1 Targeted-microRNA-221. Mol Cells 2015; 38:643-50. [PMID: 26062554 PMCID: PMC4507031 DOI: 10.14348/molcells.2015.0050] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/22/2015] [Accepted: 04/24/2015] [Indexed: 01/08/2023] Open
Abstract
The use of conditioned medium from mesenchymal stem cells may be a feasible approach for regeneration of bone defects through secretion of various components of mesenchymal stem cells such as cytokines, chemokines, and growth factors. Mesenchymal stem cells secrete and accumulate multiple factors in conditioned medium under specific physiological conditions. In this study, we investigated whether the conditioned medium collected under hypoxic condition could effectively influence bone regeneration through enhanced migration and adhesion of endogenous mesenchymal stem cells. Cell migration and adhesion abilities were increased through overexpression of intercellular adhesion molecule-1 in hypoxic conditioned medium treated group. Intercellular adhesion molecule-1 was upregulated by microRNA-221 in mesenchymal stem cells because microRNAs are key regulators of various biological functions via gene expression. To investigate the effects in vivo, evaluation of bone regeneration by computed tomography and histological assays revealed that osteogenesis was enhanced in the hypoxic conditioned medium group relative to the other groups. These results suggest that behavioral changes of endogenous mesenchymal stem cells through microRNA-221 targeted-intercellular adhesion molecule-1 expression under hypoxic conditions may be a potential treatment for patients with bone defects.
Collapse
Affiliation(s)
- Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, Busan 609-735,
Korea
| | - Ran Kim
- Department of Biology Education, College of Education, Pusan National University, Busan 609-735,
Korea
| | - Sang In Park
- Institute of Catholic Integrative Medicine, Incheon St. Mary’s Hospital, The Catholic University of Korea, College of Medicine, Incheon 403-720,
Korea
| | - Yu Jin Jung
- EIT/LOFUS Research Center, International St. Mary’s Hospital, Catholic Kwandong University, Incheon 404-834,
Korea
| | - Onju Ham
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752,
Korea
| | - Jihyun Lee
- Department of Biology Education, College of Education, Pusan National University, Busan 609-735,
Korea
| | - Ji Hyeong Kim
- Department of Biology Education, College of Education, Pusan National University, Busan 609-735,
Korea
| | - Sekyung Oh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305,
USA
| | - Min Young Lee
- Department of Molecular Physiology, College of Pharmacy, Kyungpook National University, Daegu 702-701,
Korea
| | - Jongmin Kim
- Department of Life Systems, Sookmyung Women’s University, Seoul 140-742,
Korea
| | - Moon-Seo Park
- Department of Biology Education, College of Education, Pusan National University, Busan 609-735,
Korea
| | - Yong-An Chung
- Institute of Catholic Integrative Medicine, Incheon St. Mary’s Hospital, The Catholic University of Korea, College of Medicine, Incheon 403-720,
Korea
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangwon 210-701,
Korea
- Catholic Kwandong University International, St. Mary’s Hospital, Incheon 404-834,
Korea
| | - Lee-So Maeng
- Institute of Catholic Integrative Medicine, Incheon St. Mary’s Hospital, The Catholic University of Korea, College of Medicine, Incheon 403-720,
Korea
| |
Collapse
|
35
|
Park J, Kim B, Han J, Oh J, Park S, Ryu S, Jung S, Shin JY, Lee BS, Hong BH, Choi D, Kim BS. Graphene oxide flakes as a cellular adhesive: prevention of reactive oxygen species mediated death of implanted cells for cardiac repair. ACS NANO 2015; 9:4987-99. [PMID: 25919434 DOI: 10.1021/nn507149w] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Mesenchymal stem cell (MSC) implantation has emerged as a potential therapy for myocardial infarction (MI). However, the poor survival of MSCs implanted to treat MI has significantly limited the therapeutic efficacy of this approach. This poor survival is primarily due to reactive oxygen species (ROS) generated in the ischemic myocardium after the restoration of blood flow. ROS primarily causes the death of implanted MSCs by inhibiting the adhesion of the MSCs to extracellular matrices at the lesion site (i.e., anoikis). In this study, we proposed the use of graphene oxide (GO) flakes to protect the implanted MSCs from ROS-mediated death and thereby improve the therapeutic efficacy of the MSCs. GO can adsorb extracellular matrix (ECM) proteins. The survival of MSCs, which had adhered to ECM protein-adsorbed GO flakes and were subsequently exposed to ROS in vitro or implanted into the ischemia-damaged and reperfused myocardium, significantly exceeded that of unmodified MSCs. Furthermore, the MSC engraftment improved by the adhesion of MSCs to GO flakes prior to implantation enhanced the paracrine secretion from the MSCs following MSC implantation, which in turn promoted cardiac tissue repair and cardiac function restoration. This study demonstrates that GO can effectively improve the engraftment and therapeutic efficacy of MSCs used to repair the injury of ROS-abundant ischemia and reperfusion by protecting implanted cells from anoikis.
Collapse
Affiliation(s)
- Jooyeon Park
- †School of Chemical and Biological Engineering, Seoul National University, Seoul 151-744, Republic of Korea
| | | | - Jin Han
- †School of Chemical and Biological Engineering, Seoul National University, Seoul 151-744, Republic of Korea
| | | | - Subeom Park
- ⊥Department of Chemistry, Seoul National University, Seoul 151-747, Republic of Korea
| | - Seungmi Ryu
- §Interdisciplinary Program of Bioengineering, Seoul National University, Seoul 151-744, Republic of Korea
| | | | - Jung-Youn Shin
- †School of Chemical and Biological Engineering, Seoul National University, Seoul 151-744, Republic of Korea
| | | | - Byung Hee Hong
- ⊥Department of Chemistry, Seoul National University, Seoul 151-747, Republic of Korea
| | | | - Byung-Soo Kim
- †School of Chemical and Biological Engineering, Seoul National University, Seoul 151-744, Republic of Korea
- §Interdisciplinary Program of Bioengineering, Seoul National University, Seoul 151-744, Republic of Korea
- ∥Bio-MAX Institute, Institute of Chemical Processes, Seoul National University, Seoul 151-744, Republic of Korea
| |
Collapse
|
36
|
Activation of NRG1-ERBB4 signaling potentiates mesenchymal stem cell-mediated myocardial repairs following myocardial infarction. Cell Death Dis 2015; 6:e1765. [PMID: 25996292 PMCID: PMC4669719 DOI: 10.1038/cddis.2015.91] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 01/14/2015] [Accepted: 02/16/2015] [Indexed: 12/31/2022]
Abstract
Mesenchymal stem cell (MSC) transplantation has achieved only modest success in the treatment of ischemic heart disease owing to poor cell viability in the diseased microenvironment. Activation of the NRG1 (neuregulin1)-ERBB4 (v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 4) signaling pathway has been shown to stimulate mature cardiomyocyte cell cycle re-entry and cell division. In this connection, we aimed to determine whether overexpression of ERBB4 in MSCs can enhance their cardio-protective effects following myocardial infarction. NRG1, MSCs or MSC-ERBB4 (MSC with ERBB4 overexpression), were transplanted into mice following myocardial infarction. Superior to that of MSCs and solely NRG1, MSC-ERBB4 transplantation significantly preserved heart functions accompanied with reduced infarct size, enhanced cardiomyocyte division and less apoptosis during early phase of infarction. The transduction of ERBB4 into MSCs indeed increased cell mobility and apoptotic resistance under hypoxic and glucose-deprived conditions via a PI3K/Akt signaling pathway in the presence of NRG1. Unexpectedly, introduction of ERBB4 into MSC in turn potentiates NRG1 synthesis and secretion, thus forming a novel NRG1-ERBB4-NRG1 autocrine loop. Conditioned medium of MSC-ERBB4 containing elevated NRG1, promoted cardiomyocyte growth and division, whereas neutralization of NRG1 blunted this proliferation. These findings collectively suggest that ERBB4 overexpression potentiates MSC survival in the infarcted heart, enhances NRG1 generation to restore declining NRG1 in the infarcted region and stimulates cardiomyocyte division. ERBB4 has an important role in MSC-mediated myocardial repairs.
Collapse
|
37
|
Pretreatment of Adipose Derived Stem Cells with Curcumin Facilitates Myocardial Recovery via Antiapoptosis and Angiogenesis. Stem Cells Int 2015; 2015:638153. [PMID: 26074974 PMCID: PMC4436501 DOI: 10.1155/2015/638153] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 12/28/2014] [Accepted: 12/29/2014] [Indexed: 02/04/2023] Open
Abstract
The poor survival rate of transplanted stem cells in ischemic myocardium has limited their therapeutic efficacy. Curcumin has potent antioxidant property. This study investigates whether prior curcumin treatment protects stem cells from oxidative stress injury and improves myocardial recovery following cells transplantation. Autologous Sprague-Dawley rat adipose derived mesenchymal stem cells (ADSCs) were pretreated with or without curcumin. The hydrogen peroxide/serum deprivation (H2O2/SD) medium was used to mimic the ischemic condition in vitro. Cytoprotective effects of curcumin on ADSCs were evaluated. Curcumin pretreatment significantly increased cell viability and VEGF secretion, and decreased cell injury and apoptosis via regulation of PTEN/Akt/p53 and HO-1 signal proteins expression. The therapeutic potential of ADSCs implantation was investigated in myocardial ischemia-reperfusion injury (IRI) model. Transplantation of curcumin pretreated ADSCs not only resulted in better heart function, higher cells retention, and smaller infarct size, but also decreased myocardial apoptosis, promoted neovascularization, and increased VEGF level in ischemic myocardium. Together, priming of ADSCs with curcumin improved tolerance to oxidative stress injury and resulted in enhancement of their therapeutic potential of ADSCs for myocardial repair. Curcumin pretreatment is a promising adjuvant strategy for stem cells transplantation in myocardial restoration.
Collapse
|
38
|
Andreeva ER, Pogodina MV, Buravkova LB. Hypoxic stress as an activation trigger of multipotent mesenchymal stromal cells. ACTA ACUST UNITED AC 2015. [DOI: 10.1134/s0362119715020024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
39
|
Lee S, Choi E, Cha MJ, Hwang KC. Cell adhesion and long-term survival of transplanted mesenchymal stem cells: a prerequisite for cell therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:632902. [PMID: 25722795 PMCID: PMC4333334 DOI: 10.1155/2015/632902] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/19/2015] [Indexed: 12/13/2022]
Abstract
The literature provides abundant evidence that mesenchymal stem cells (MSCs) are an attractive resource for therapeutics and have beneficial effects in regenerating injured tissues due to their self-renewal ability and broad differentiation potential. Although the therapeutic potential of MSCs has been proven in both preclinical and clinical studies, several questions have not yet been addressed. A major limitation to the use of MSCs in clinical applications is their poor viability at the site of injury due to the harsh microenvironment and to anoikis driven by the loss of cell adhesion. To improve the survival of the transplanted MSCs, strategies to regulate apoptotic signaling and enhance cell adhesion have been developed, such as pretreatment with cytokines, growth factors, and antiapoptotic molecules, genetic modifications, and hypoxic preconditioning. More appropriate animal models and a greater understanding of the therapeutic mechanisms of MSCs will be required for their successful clinical application. Nevertheless, the development of stem cell therapies using MSCs has the potential to treat degenerative diseases. This review discusses various approaches to improving MSC survival by inhibiting anoikis.
Collapse
Affiliation(s)
- Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 210-701, Republic of Korea
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City 404-834, Republic of Korea
| | - Eunhyun Choi
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 210-701, Republic of Korea
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City 404-834, Republic of Korea
| | - Min-Ji Cha
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 210-701, Republic of Korea
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City 404-834, Republic of Korea
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 210-701, Republic of Korea
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City 404-834, Republic of Korea
| |
Collapse
|
40
|
Liang Y, Lin Q, Zhu J, Li X, Fu Y, Zou X, Liu X, Tan H, Deng C, Yu X, Shan Z, Yuan W. The caspase-8 shRNA-modified mesenchymal stem cells improve the function of infarcted heart. Mol Cell Biochem 2014; 397:7-16. [PMID: 25060909 DOI: 10.1007/s11010-014-2165-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Accepted: 07/14/2014] [Indexed: 12/15/2022]
Abstract
The beneficial effects of mesenchymal stem cells (MSCs) in cardiac cell therapy are greatly limited due to poor survival after transplantation into ischemic hearts. Here, we investigated whether caspase 8 small hairpin RNA (shRNA) modification enhance human MSCs (hMSCs) survival and improve infarcted heart function. Recombinant adenovirus encoding pre-miRNA-155-designed caspase 8 shRNA was prepared to inhibit caspase 8 expression in hMSCs. The effect of caspase 8 shRNA modification on protecting hMSCs from apoptosis under the conditions of serum deprivation and hypoxia was tested by Annexin V/PI staining and caspase 8 activity assay. The caspase 8 shRNA-modified and superparamagnetic iron oxide (SPIO)-labeled hMSCs were injected into the border zone of the infarcted region of rat heart. Echocardiography and Masson trichrome staining were performed to assess heart function and cardiac fibrosis. Our results showed that adenovirus-mediated caspase 8 shRNA could efficiently inhibit caspase 8 expression in hMSCs. Knock-down of caspase 8 expression lead to inhibition of hMSCs apoptosis, reduction of caspase 8 activity and up-regulations of HGF, IGF-1 and Bcl-2. Transplantation of caspase 8 shRNA-modified hMSCs could significantly improve infracted heart function, attenuate cardiac fibrosis. Consistently, the rate of cardiomyocyte apoptosis and caspase 8 activity were significantly decreased, and the survival rate of transplanted hMSCs was markedly elevated in the myocardium receiving caspase 8 shRNA-modified hMSCs transplantation. Together, our findings implicated the therapeutic potential of caspase 8 shRNA-modified hMSCs in improving the infarcted heart function.
Collapse
Affiliation(s)
- Yeyou Liang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Buravkova LB, Andreeva ER, Gogvadze V, Zhivotovsky B. Mesenchymal stem cells and hypoxia: where are we? Mitochondrion 2014; 19 Pt A:105-12. [PMID: 25034305 DOI: 10.1016/j.mito.2014.07.005] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 07/09/2014] [Indexed: 12/23/2022]
Abstract
Multipotent mesenchymal stromal cells (MSCs) are involved in the organization and maintenance of tissue integrity. MSCs have also attracted attention as a promising tool for cell therapy and regenerative medicine. However, their usage is limited due to cell impairment induced by an extremely harsh microenvironment during transplantation ex vivo. The microenvironment of MSCs in tissue depots is characterized by rather low oxygen consumption, demonstrating that MSCs might be quite resistant to oxygen limitation. However, accumulated data revealed that the response of MSCs to hypoxic conditions is rather controversial, demonstrating both damaging and ameliorating effects. Here, we make an attempt to summarize recent knowledge on the survival of MSCs under low oxygen conditions of varying duration and severity and to elucidate the mechanisms of MSC resistance/sensitivity to hypoxic impact.
Collapse
Affiliation(s)
- L B Buravkova
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia; Faculty of Basic Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia
| | - E R Andreeva
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - V Gogvadze
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177 Stockholm, Sweden
| | - B Zhivotovsky
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177 Stockholm, Sweden.
| |
Collapse
|
42
|
Kim H, Han JW, Lee JY, Choi YJ, Sohn YD, Song M, Yoon YS. Diabetic Mesenchymal Stem Cells Are Ineffective for Improving Limb Ischemia Due to Their Impaired Angiogenic Capability. Cell Transplant 2014; 24:1571-84. [PMID: 25008576 DOI: 10.3727/096368914x682792] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The purpose of this study was to investigate the effects of diabetes on mesenchymal stem cells (MSCs) in terms of their angiogenic and therapeutic potential for repairing tissue ischemia. We culture-isolated MSCs from streptozotocin-induced diabetic rats (D-MSCs) and compared their proliferation, differentiation, and angiogenic effects with those from normal rats (N-MSCs). The angiogenic effects of MSCs were evaluated by real-time PCR, in vitro tube formation assay, and transplantation of the MSCs into a hindlimb ischemia model followed by laser Doppler perfusion imaging. The number of MSCs derived from diabetic rats was smaller, and their proliferation rate was slower than N-MSCs. Upon induction of differentiation, the osteogenic and angiogenic differentiation of D-MSCs were aberrant compared to N-MSCs. The expression of angiogenic factors was lower in D-MSCs than N-MSCs. D-MSCs cocultured with endothelial cells resulted in decreased tube formation compared to N-MSCs. D-MSCs were ineffective to improve hindlimb ischemia and showed lower capillary density and angiogenic gene expression in ischemic limbs than N-MSCs. D-MSCs have defective proliferation and angiogenic activities and are ineffective for repairing hindlimb ischemia. Newer measures are needed before MSCs can be employed as a source for autologous cell therapy.
Collapse
Affiliation(s)
- Hyongbum Kim
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Liu X, Chen H, Zhu W, Chen H, Hu X, Jiang Z, Xu Y, Zhou Y, Wang K, Wang L, Chen P, Hu H, Wang C, Zhang N, Ma Q, Huang M, Hu D, Zhang L, Wu R, Wang Y, Xu Q, Yu H, Wang J. Transplantation of SIRT1-engineered aged mesenchymal stem cells improves cardiac function in a rat myocardial infarction model. J Heart Lung Transplant 2014; 33:1083-92. [PMID: 25034794 DOI: 10.1016/j.healun.2014.05.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 04/24/2014] [Accepted: 05/27/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Previous studies have demonstrated that biological aging has a negative influence on the therapeutic effects of mesenchymal stem cells (MSCs)-based therapy. Using a rat myocardial infarction (MI) model, we tested the hypothesis that silent mating type information regulation 2 homolog 1 (SIRT1) may ameliorate the phenotype and improve the function of aged MSCs and thus enhance the efficacy of aged MSCs-based therapy. METHODS Sixty female rats underwent left anterior descending coronary artery ligation and were randomly assigned to receiving: intramyocardial injection of cell culture medium (DMEM group); SIRT1 overexpression vector-treated aged MSCs (SIRT1-aged MSCs group) obtained from aged male SD rats or empty vector-treated aged MSCs (vector-aged MSCs group). Another 20 sham-operated rats that underwent open-chest surgery without coronary ligation or any other intervention served as controls. RESULTS SIRT1-aged MSC group exhibited enhanced blood vessel density in the border zone of MI hearts, which was associated with reduced cardiac remodeling, leading to improved cardiac performance. Consistent with the in vivo data, our in vitro experiments also demonstrated that SIRT1 overexpression ameliorated aged MSCs senescent phenotype and recapitulated the pro-angiogenesis property of MSCs and conferred the anti-stress response capabilities, as indicated by increases in pro-angiogenic factors, angiopoietin 1 (Ang1) and basic fibroblast growth factor (bFGF), expressions and a decrease in anti-angiogenic factor thrombospondin-1 (TBS1) at mRNA levels, and increases in Bcl-2/Bax ratio at protein level. CONCLUSIONS Up-regulating SIRT1 expression could enhance the efficacy of aged MSCs-based therapy for MI as it relates to the amelioration of senescent phenotype and hence improved biological function of aged MSCs.
Collapse
Affiliation(s)
- Xianbao Liu
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huiqiang Chen
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Zhu
- Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Han Chen
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyang Hu
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhi Jiang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yinchuan Xu
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Zhou
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kan Wang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lihan Wang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Panpan Chen
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hengxun Hu
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen Wang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Na Zhang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qunchao Ma
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingyuan Huang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dexing Hu
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ling Zhang
- Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Rongrong Wu
- Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaping Wang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiyuan Xu
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Yu
- Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian'an Wang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
44
|
McGinley LM, McMahon J, Stocca A, Duffy A, Flynn A, O'Toole D, O'Brien T. Mesenchymal stem cell survival in the infarcted heart is enhanced by lentivirus vector-mediated heat shock protein 27 expression. Hum Gene Ther 2014; 24:840-51. [PMID: 23987185 DOI: 10.1089/hum.2011.009] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Mesenchymal stem cell (MSC) therapy offers the potential to promote recovery after myocardial infarction (MI). However, therapeutic efficacy may be limited by poor survival and retention of transplanted cells. A combination of gene and cell therapy has the capacity to prevent donor cell death and augment the reparative and regenerative effects of cell transfer. The present study investigates the effect of exogenous heat shock protein 27 (Hsp27) expression in MSCs in an in vitro model of ischemia and in an in vivo rat MI model and aims to determine if this could enhance the therapeutic benefit associated with cell delivery. Hsp27 overexpression by lentivirus vector modification resulted in increased MSC survival in vitro and in vivo. Furthermore, decreased apoptosis in the infarcted tissue and improved cardiac function was observed in the Hsp27 group, enhancing the therapeutic effect of MSCs. Together, these data demonstrate that ex vivo genetic modification-specifically Hsp27 overexpression-offers the possibility of enhancing the efficacy of MSC therapy in MI.
Collapse
Affiliation(s)
- Lisa M McGinley
- 1 Regenerative Medicine Institute, National University of Ireland , Galway, Galway, Ireland
| | | | | | | | | | | | | |
Collapse
|
45
|
Richardson JD, Nelson AJ, Zannettino ACW, Gronthos S, Worthley SG, Psaltis PJ. Optimization of the cardiovascular therapeutic properties of mesenchymal stromal/stem cells-taking the next step. Stem Cell Rev Rep 2014; 9:281-302. [PMID: 22529015 DOI: 10.1007/s12015-012-9366-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Despite current treatment options, cardiac failure is associated with significant morbidity and mortality highlighting a compelling clinical need for novel therapeutic approaches. Based on promising pre-clinical data, stem cell therapy has been suggested as a possible therapeutic strategy. Of the candidate cell types evaluated, mesenchymal stromal/stem cells (MSCs) have been widely evaluated due to their ease of isolation and ex vivo expansion, potential allogeneic utility and capacity to promote neo-angiogenesis and endogenous cardiac repair. However, the clinical application of MSCs for mainstream cardiovascular use is currently hindered by several important limitations, including suboptimal retention and engraftment and restricted capacity for bona fide cardiomyocyte regeneration. Consequently, this has prompted intense efforts to advance the therapeutic properties of MSCs for cardiovascular disease. In this review, we consider the scope of benefit from traditional plastic adherence-isolated MSCs and the lessons learned from their conventional use in preclinical and clinical studies. Focus is then given to the evolving strategies aimed at optimizing MSC therapy, including discussion of cell-targeted techniques that encompass the preparation, pre-conditioning and manipulation of these cells ex vivo, methods to improve their delivery to the heart and innovative substrate-directed strategies to support their interaction with the host myocardium.
Collapse
Affiliation(s)
- James D Richardson
- Cardiovascular Research Centre, Royal Adelaide Hospital and Department of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | | | | | | | | | | |
Collapse
|
46
|
Buravkova LB, Rylova YV, Andreeva ER, Kulikov AV, Pogodina MV, Zhivotovsky B, Gogvadze V. Low ATP level is sufficient to maintain the uncommitted state of multipotent mesenchymal stem cells. Biochim Biophys Acta Gen Subj 2013; 1830:4418-25. [PMID: 23742825 DOI: 10.1016/j.bbagen.2013.05.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 05/02/2013] [Accepted: 05/20/2013] [Indexed: 12/16/2022]
Abstract
BACKGROUND Multipotent mesenchymal stromal cells (MMSCs) are minimally differentiated precursors with great potential to transdifferentiate. These cells are quite resistant to oxygen limitation, suggesting that a hypoxic milieu can be physiological for MMSCs. METHODS Human MMSCs isolated from adipose tissue were grown at various oxygen concentrations. Alteration in cell immunophenotype was determined by flow cytometry after staining with specific antibodies. Concentrations of glucose and lactate were determined using the Biocon colorimetric test. Cellular respiration was assessed using oxygen electrode. The modes of cell death were analyzed by flow cytometry after staining with Annexin V and propidium iodide. RESULTS We found that permanent oxygen deprivation attenuated cellular ATP levels in these cells, diminishing mitochondrial ATP production but stimulating glycolytic ATP production. At the same time, permanent hypoxia did not affect MMSCs' viability, stimulated their proliferation and reduced their capacity to differentiate. Further, permanent hypoxia decreased spontaneous cell death by MMSCs. CONCLUSIONS Under hypoxic conditions glycolysis provides sufficient energy to maintain MMSCs in an uncommitted state. GENERAL SIGNIFICANCE These findings are of interest not only for scientific reasons, but also in practical terms. Oxygen concentration makes an essential contribution to MMSC physiology and should be taken into account in the setting of protocols for cellular therapy.
Collapse
Affiliation(s)
- L B Buravkova
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, Moscow, Russia
| | | | | | | | | | | | | |
Collapse
|
47
|
Rennert RC, Sorkin M, Garg RK, Gurtner GC. Stem cell recruitment after injury: lessons for regenerative medicine. Regen Med 2013; 7:833-50. [PMID: 23164083 DOI: 10.2217/rme.12.82] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tissue repair and regeneration are thought to involve resident cell proliferation as well as the selective recruitment of circulating stem and progenitor cell populations through complex signaling cascades. Many of these recruited cells originate from the bone marrow, and specific subpopulations of bone marrow cells have been isolated and used to augment adult tissue regeneration in preclinical models. Clinical studies of cell-based therapies have reported mixed results, however, and a variety of approaches to enhance the regenerative capacity of stem cell therapies are being developed based on emerging insights into the mechanisms of progenitor cell biology and recruitment following injury. This article discusses the function and mechanisms of recruitment of important bone marrow-derived stem and progenitor cell populations following injury, as well as the emerging therapeutic applications targeting these cells.
Collapse
Affiliation(s)
- Robert C Rennert
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 257 Campus Drive West, Hagey Building GK-201, Stanford, CA 94305-5148, USA
| | | | | | | |
Collapse
|
48
|
Park JM, Je JH, Wu WH, Jee HJ, Lee SK, Lee MG. Attenuation of contact hypersensitivity by cell-permeable heat shock protein 70 in BALB/c mouse model. Exp Dermatol 2012; 21:969-71. [DOI: 10.1111/exd.12044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2012] [Indexed: 10/27/2022]
Affiliation(s)
- Jin Mo Park
- Department of Dermatology and Cutaneous Biology Research Institute; Severance Hospital, Yonsei University College of Medicine; Seoul; Korea
| | - Jeong Hwan Je
- Department of Dermatology and Cutaneous Biology Research Institute; Severance Hospital, Yonsei University College of Medicine; Seoul; Korea
| | - Wen Hao Wu
- Department of Dermatology and Cutaneous Biology Research Institute; Severance Hospital, Yonsei University College of Medicine; Seoul; Korea
| | - Hyun Joong Jee
- Department of Dermatology and Cutaneous Biology Research Institute; Severance Hospital, Yonsei University College of Medicine; Seoul; Korea
| | - Sang-Kyou Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Translational Research Center for Protein Function Control; Yonsei University; Seoul; Korea
| | - Min-Geol Lee
- Department of Dermatology and Cutaneous Biology Research Institute; Severance Hospital, Yonsei University College of Medicine; Seoul; Korea
| |
Collapse
|
49
|
Wen Z, Zheng S, Zhou C, Yuan W, Wang J, Wang T. Bone marrow mesenchymal stem cells for post-myocardial infarction cardiac repair: microRNAs as novel regulators. J Cell Mol Med 2012; 16:657-71. [PMID: 22004043 PMCID: PMC3822837 DOI: 10.1111/j.1582-4934.2011.01471.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Transplantation of bone marrow-derived mesenchymal stem cells (MSCs) is safe and may improve cardiac function and structural remodelling in patients following myocardial infarction (MI). Cardiovascular cell differentiation and paracrine effects to promote endogenous cardiac regeneration, neovascularization, anti-inflammation, anti-apoptosis, anti-remodelling and cardiac contractility, may contribute to MSC-based cardiac repair following MI. However, current evidence indicates that the efficacy of MSC transplantation was unsatisfactory, due to the poor viability and massive death of the engrafted MSCs in the infarcted myocardium. MicroRNAs are short endogenous, conserved, non-coding RNAs and important regulators involved in numerous facets of cardiac pathophysiologic processes. There is an obvious involvement of microRNAs in almost every facet of putative repair mechanisms of MSC-based therapy in MI, such as stem cell differentiation, neovascularization, apoptosis, cardiac remodelling, cardiac contractility and arrhythmias, and others. It is proposed that therapeutic modulation of individual cardiovascular microRNA of MSCs, either mimicking or antagonizing microRNA actions, will hopefully enhance MSC therapeutic efficacy. In addition, MSCs may be manipulated to enhance functional microRNA expression or to inhibit aberrant microRNA levels in a paracrine manner. We hypothesize that microRNAs may be used as novel regulators in MSC-based therapy in MI and MSC transplantation by microRNA regulation may represent promising therapeutic strategy for MI patients in the future.
Collapse
Affiliation(s)
- Zhuzhi Wen
- The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
50
|
Lee SY, Ham O, Cha MJ, Song BW, Choi E, Kim IK, Chang W, Lim S, Lee CY, Park JH, Lee J, Bae Y, Seo HH, Choi E, Jang Y, Hwang KC. The promotion of cardiogenic differentiation of hMSCs by targeting epidermal growth factor receptor using microRNA-133a. Biomaterials 2012; 34:92-9. [PMID: 23069713 DOI: 10.1016/j.biomaterials.2012.09.069] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2012] [Accepted: 09/26/2012] [Indexed: 01/08/2023]
Abstract
Human bone marrow-derived mesenchymal stem cells (hMSCs) are an attractive candidate for cell therapy in heart disease. Low survival and incomplete electromechanical integration between resident cardiomyocytes and transplanted hMSCs remain unsolved. In order for an infarcted heart to tolerate transplantation, differentiation capacity in stem cells must be reinforced. In this study, we found that compound 56, an epidermal growth factor receptor (EGFR) inhibitor, promotes cardiogenic differentiation of hMSCs and the transplantation of hMSCs treated with compound 56 resulted in enhancement of heart functions. Furthermore, hMSCs transfected with microRNA-133a (miR-133a), which targets EGFR, were observed to express cardiac-specific markers. We also discovered that luciferase activity is exclusively decreased by targeting EGFR in hMSCs transfected with miR-133a mimic. These results suggest that EGFR plays a key role in the regulation of cardiogenic differentiation in hMSCs.
Collapse
Affiliation(s)
- Se-Yeon Lee
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|