101
|
Chang D, Yang X, Fan S, Fan T, Zhang M, Ono M. Engineering of MSCs sheet for the prevention of myocardial ischemia and for left ventricle remodeling. Stem Cell Res Ther 2023; 14:102. [PMID: 37098611 PMCID: PMC10127056 DOI: 10.1186/s13287-023-03322-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 03/29/2023] [Indexed: 04/27/2023] Open
Abstract
Tissue engineering combines cell biology and material science to construct tissues or organs for disease modeling, drug testing, and regenerative medicine. The cell sheet is a newly developed tissue engineering technology that has brought about scaffold-free tissue and shows great application potential. In this review, we summarized recent progress and future possibilities in preclinical research into and clinical applications of cell sheets fabricated by differing cell types from various sources for cardiac tissue repair, and the manufacturing strategies and promising application potential of 3D cell-dense tissue constructed from cell sheets. Special attention was paid to the mechanisms of mesenchymal stem cell (MSC) sheets in the prevention of myocardial ischemia and left ventricle remodeling. Comparing MSCs sheets with other types of cell sheets and 3D cardiac tissues, engineering tissues' potential safety and effectiveness concerns were also discussed.
Collapse
Affiliation(s)
- Dehua Chang
- Department of Cell Therapy in Regenerative Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Xiaotong Yang
- BOE Regenerative Medicine Technology Co., Ltd., No. 9 JiuXianQiao North Road, Beijing, 100015, China
| | - Siyang Fan
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Taibing Fan
- Children Heart Center, Fuwai Central China Cardiovascular Hospital, No. 1 Fuwai Road, Zhengzhou, 450018, China
| | - Mingkui Zhang
- Heart Center, First Hospital of Tsinghua University, No. 6 JiuXianQiao 1st Road, Beijing, 10016, China
| | - Minoru Ono
- Department of Cardiac Surgery, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
102
|
Tonkin D, Yee-Goh A, Katare R. Healing the Ischaemic Heart: A Critical Review of Stem Cell Therapies. Rev Cardiovasc Med 2023; 24:122. [PMID: 39076280 PMCID: PMC11273058 DOI: 10.31083/j.rcm2404122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 07/31/2024] Open
Abstract
Ischaemic heart disease (IHD) remains the leading cause of mortality worldwide. Current pharmaceutical treatments focus on delaying, rather than preventing disease progression. The only curative treatment available is orthotopic heart transplantation, which is greatly limited by a lack of available donors and the possibility for immune rejection. As a result, novel therapies are consistently being sought to improve the quality and duration of life of those suffering from IHD. Stem cell therapies have garnered attention globally owing to their potential to replace lost cardiac cells, regenerate the ischaemic myocardium and to release protective paracrine factors. Despite recent advances in regenerative cardiology, one of the biggest challenges in the clinical translation of cell-based therapies is determining the most efficacious cell type for repair. Multiple cell types have been investigated in clinical trials; with inconsistent methodologies and isolation protocols making it difficult to draw strong conclusions. This review provides an overview of IHD focusing on pathogenesis and complications, followed by a summary of different stem cells which have been trialled for use in the treatment of IHD, and ends by exploring the known mechanisms by which stem cells mediate their beneficial effects on ischaemic myocardium.
Collapse
Affiliation(s)
- Devin Tonkin
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, 9010 Dunedin, New Zealand
| | - Anthony Yee-Goh
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, 9010 Dunedin, New Zealand
| | - Rajesh Katare
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, 9010 Dunedin, New Zealand
| |
Collapse
|
103
|
Maleki B, Noureddini M, Saadat S, Verdi J, Farrokhian A, Ghanbarian H, Cheraghi E, Alani B. Effect of miR-18a-5p, miR-19a-3p, and miR-20a-5p on In Vitro Cardiomyocyte Differentiation of Human Endometrium Tissue-Derived Stem Cells Through Regulation of Smad4 Expression. Rep Biochem Mol Biol 2023; 12:136-146. [PMID: 37724150 PMCID: PMC10505458 DOI: 10.52547/rbmb.12.1.136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/22/2023] [Indexed: 09/20/2023]
Abstract
Background Smad4 regulates the expression of the genes required for heart homeostasis. Regarding the central role of microRNAs in cardiac biology, we investigated the expression of the three Smad4-targeting miRNAs, namely miR-18a-5p, miR-19a-3p, and miR-20a-5p, as well as Smad4 during differentiation of human endometrium-derived mesenchymal stem cells (hEMSCs) into cardiomyocytes (CMs). Methods To evaluate mesenchymal phenotype and multi-lineage differentiation ability of hEMSCs, immunophenotyping by flow cytometry and differentiation into osteoblasts and adipocytes were performed, respectively. For transdifferentiation into CMs, hEMSCs were exposed to a cardiomyogenic medium composed of 5-aza and bFGF for 30 days. The comparison between transcriptional expression levels of Nkx2-5, GATA4, Smad4, TNNT2, TBX5, miR-18a-5p, miR-19a-3p, and miR-20a-5p by qRT-PCR, as well as protein levels of Nkx2-5, Smad4, and cTnT by immunofluorescence staining, was conducted in every 6 days. Results In vitro, the mesenchymal stem cell phenotype of hEMSCs and their potency for differentiation into other MSCs were confirmed. Differentiated hEMSCs had morphological characteristics of CMs. The percentage of positive cells for Nkx2-5, Smad4, and cTnT proteins was increased following induction and culminated on the 24th day. Also, mRNA levels of Nkx2-5, GATA4, Smad4, TNNT2, and TBX5 exhibited the same trend. The expression of investigated miRNAs was significantly decreased sequentially. A significant negative correlation between expressions of Smad4 and investigated miRNAs was observed. Conclusion Our results indicate that miR-18a-5p, miR-19a-3p, and miR-20a-5p are involved in the cardiac differentiation propensity of hEMSCs potentially by regulation of Smad levels. Although, more mechanistic experiments are required to confirm this idea.
Collapse
Affiliation(s)
- Behnaz Maleki
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran.
| | - Mahdi Noureddini
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran.
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| | - Somayeh Saadat
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran.
| | - Javad Verdi
- Department of Applied Cellular Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Alireza Farrokhian
- Department of Cardiology, School of Medicine, Shahid Beheshti Hospital, Kashan University of Medical Sciences, Kashan, Iran.
| | - Hossein Ghanbarian
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ebrahim Cheraghi
- Department of Biology, Faculty of Science, University of Qom, Qom, Iran.
| | - Behrang Alani
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
104
|
Administration of stem cells against cardiovascular diseases with a focus on molecular mechanisms: Current knowledge and prospects. Tissue Cell 2023; 81:102030. [PMID: 36709696 DOI: 10.1016/j.tice.2023.102030] [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: 10/23/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023]
Abstract
Cardiovascular diseases (CVDs) are a serious global concern for public and human health. Despite the emergence of significant therapeutic advances, it is still the leading cause of death and disability worldwide. As a result, extensive efforts are underway to develop practical therapeutic approaches. Stem cell-based therapies could be considered a promising strategy for the treatment of CVDs. The efficacy of stem cell-based therapeutic approaches is demonstrated through recent laboratory and clinical studies due to their inherent regenerative properties, proliferative nature, and their capacity to differentiate into different cells such as cardiomyocytes. These properties could improve cardiovascular functioning leading to heart regeneration. The two most common types of stem cells with the potential to cure heart diseases are induced pluripotent stem cells (iPSCs) and mesenchymal stem cells (MSCs). Several studies have demonstrated the use, efficacy, and safety of MSC and iPSCs-based therapies for the treatment of CVDs. In this study, we explain the application of stem cells, especially iPSCs and MSCs, in the treatment of CVDs with a focus on cellular and molecular mechanisms and then discuss the advantages, disadvantages, and perspectives of using this technology in the treatment of these diseases.
Collapse
|
105
|
Kishino Y, Fukuda K. Unlocking the Pragmatic Potential of Regenerative Therapies in Heart Failure with Next-Generation Treatments. Biomedicines 2023; 11:biomedicines11030915. [PMID: 36979894 PMCID: PMC10046277 DOI: 10.3390/biomedicines11030915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/28/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
Patients with chronic heart failure (HF) have a poor prognosis due to irreversible impairment of left ventricular function, with 5-year survival rates <60%. Despite advances in conventional medicines for HF, prognosis remains poor, and there is a need to improve treatment further. Cell-based therapies to restore the myocardium offer a pragmatic approach that provides hope for the treatment of HF. Although first-generation cell-based therapies using multipotent cells (bone marrow-derived mononuclear cells, mesenchymal stem cells, adipose-derived regenerative cells, and c-kit-positive cardiac cells) demonstrated safety in preclinical models of HF, poor engraftment rates, and a limited ability to form mature cardiomyocytes (CMs) and to couple electrically with existing CMs, meant that improvements in cardiac function in double-blind clinical trials were limited and largely attributable to paracrine effects. The next generation of stem cell therapies uses CMs derived from human embryonic stem cells or, increasingly, from human-induced pluripotent stem cells (hiPSCs). These cell therapies have shown the ability to engraft more successfully and improve electromechanical function of the heart in preclinical studies, including in non-human primates. Advances in cell culture and delivery techniques promise to further improve the engraftment and integration of hiPSC-derived CMs (hiPSC-CMs), while the use of metabolic selection to eliminate undifferentiated cells will help minimize the risk of teratomas. Clinical trials of allogeneic hiPSC-CMs in HF are now ongoing, providing hope for vast numbers of patients with few other options available.
Collapse
Affiliation(s)
| | - Keiichi Fukuda
- Correspondence: ; Tel.: +81-3-5363-3874; Fax: +81-3-5363-3875
| |
Collapse
|
106
|
Moskowitzova K, Fauza DO. Transamniotic stem cell therapy (TRASCET): An emerging minimally invasive strategy for intrauterine stem cell delivery. Semin Perinatol 2023; 47:151728. [PMID: 36990923 DOI: 10.1016/j.semperi.2023.151728] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Transamniotic stem cell therapy (TRASCET) is an emerging strategy for prenatal stem cell therapy involving the least invasive method described to date of delivering select stem cells to virtually any anatomical site in the fetus, including the blood and bone marrow, as well as to fetal annexes, including the placenta. Such broad therapeutic potential derives, to a large extent, from unique routing patterns following stem cell delivery into the amniotic fluid, which have commonalities with naturally occurring fetal cell kinetics. First reported experimentally only less than a decade ago, TRASCET has yet to be attempted clinically, though a first clinical trial appears imminent. Despite significant experimental advances, much promise and perhaps excessive publicity, most cell-based therapies have yet to deliver meaningful large-scale impact to patient care. The few exceptions typically consist of therapies based on the amplification of the normal biological role played by the given cells in their natural environment. Therein lays much of the appeal of TRASCET, in that it, too, is in essence a magnification of naturally occurring processes in the distinctive environment of the maternal-fetal unit. As much as fetal stem cells possess unique characteristics compared with other stem cells, so does the fetus when compared with any other age group, converging into a scenario that enables therapeutic paradigms exclusive to prenatal life. This review summarizes the diversity of applications and biological responses associated with the TRASCET principle.
Collapse
Affiliation(s)
- Kamila Moskowitzova
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, 300 Longwood Avenue - Fegan 3, Boston, MA 02115, USA
| | - Dario O Fauza
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, 300 Longwood Avenue - Fegan 3, Boston, MA 02115, USA.
| |
Collapse
|
107
|
Menasché P. Mesenchymal Stromal Cell Therapy for Heart Failure: Never Stop DREAMing. J Am Coll Cardiol 2023; 81:864-866. [PMID: 36858706 DOI: 10.1016/j.jacc.2022.12.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 03/03/2023]
Affiliation(s)
- Philippe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, AP-HP, Université Paris Cité, Inserm, PARCC, Paris, France.
| |
Collapse
|
108
|
Sanchez-Petitto G, Rezvani K, Daher M, Rafei H, Kebriaei P, Shpall EJ, Olson A. Umbilical Cord Blood Transplantation: Connecting Its Origin to Its Future. Stem Cells Transl Med 2023; 12:55-71. [PMID: 36779789 PMCID: PMC9985112 DOI: 10.1093/stcltm/szac086] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 10/16/2022] [Indexed: 02/14/2023] Open
Abstract
Transplantation of umbilical cord blood (UCB) is an attractive alternative source of hematopoietic stem cells (HSCs). The unique properties of cord blood and its distinct immune tolerance and engraftment kinetics compared to bone marrow (BM) and peripheral blood progenitor cells, permit a wider disparity in human leukocyte antigen levels between a cord blood donor and recipient after an unrelated umbilical cord blood transplant (UCBT). In addition, it is readily available and has a lowered risk of graft-versus-host disease (GvHD), with similar long-term clinical outcomes, compared to BM transplants. However, the relatively low number of cells administered by UCB units, as well as the associated delayed engraftment and immune reconstitution, pose limitations to the wide application of UCBT. Research into several aspects of UCBT has been evaluated, including the ex vivo expansion of cord blood HSCs and the process of fucosylation to enhance engraftment. Additionally, UCB has also been used in the treatment of several neurodegenerative and cardiovascular disorders with varying degrees of success. In this article, we will discuss the biology, clinical indications, and benefits of UCBT in pediatric and adult populations. We will also discuss future directions for the use of cord blood.
Collapse
Affiliation(s)
- Gabriela Sanchez-Petitto
- Department of Stem Cell Transplant and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplant and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - May Daher
- Department of Stem Cell Transplant and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Hind Rafei
- Department of Stem Cell Transplant and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplant and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplant and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Amanda Olson
- Department of Stem Cell Transplant and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
109
|
Szydlak R. Mesenchymal stem cells in ischemic tissue regeneration. World J Stem Cells 2023; 15:16-30. [PMID: 36909782 PMCID: PMC9993139 DOI: 10.4252/wjsc.v15.i2.16] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/10/2022] [Accepted: 01/19/2023] [Indexed: 02/21/2023] Open
Abstract
Diseases caused by ischemia are one of the leading causes of death in the world. Current therapies for treating acute myocardial infarction, ischemic stroke, and critical limb ischemia do not complete recovery. Regenerative therapies opens new therapeutic strategy in the treatment of ischemic disorders. Mesenchymal stem cells (MSCs) are the most promising option in the field of cell-based therapies, due to their secretory and immunomodulatory abilities, that contribute to ease inflammation and promote the regeneration of damaged tissues. This review presents the current knowledge of the mechanisms of action of MSCs and their therapeutic effects in the treatment of ischemic diseases, described on the basis of data from in vitro experiments and preclinical animal studies, and also summarize the effects of using these cells in clinical trial settings. Since the obtained therapeutic benefits are not always satisfactory, approaches aimed at enhancing the effect of MSCs in regenerative therapies are presented at the end.
Collapse
Affiliation(s)
- Renata Szydlak
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kraków 31-034, Poland
| |
Collapse
|
110
|
Li Y, Huang J, Wang J, Xia S, Ran H, Gao L, Feng C, Gui L, Zhou Z, Yuan J. Human umbilical cord-derived mesenchymal stem cell transplantation supplemented with curcumin improves the outcomes of ischemic stroke via AKT/GSK-3β/β-TrCP/Nrf2 axis. J Neuroinflammation 2023; 20:49. [PMID: 36829224 PMCID: PMC9951499 DOI: 10.1186/s12974-023-02738-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 02/16/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Human umbilical cord-derived mesenchymal stem cell (hUC-MSC) engraftment is a promising therapy for acute ischemic stroke (AIS). However, the harsh ischemic microenvironment limits the therapeutic efficacy of hUC-MSC therapy. Curcumin is an anti-inflammatory agent that could improve inflammatory microenvironment. However, whether it enhances the neuroprotective efficacy of hUC-MSC transplantation is still unknown. In the present study, we investigated the therapeutic efficacy and the possible mechanism of combined curcumin and hUC-MSC treatment in AIS. METHODS Middle cerebral artery occlusion (MCAO) mice and oxygen glucose deprivation (OGD) microglia were administrated hUC-MSCs with or without curcumin. Neurological deficits assessment, brain water content and TTC were used to assess the therapeutic effects of combined treatment. To elucidate the mechanism, MCAO mice and OGD microglia were treated with AKT inhibitor MK2206, GSK3β activator sodium nitroprusside (SNP), GSK3β inhibitor TDZD-8 and Nrf2 gene knockout were used. Immunofluorescence, flow cytometric analysis, WB and RT-PCR were used to evaluate the microglia polarization and the expression of typical oxidative mediators, inflammatory cytokines and the AKT/GSK-3β/β-TrCP/Nrf2 pathway protein. RESULTS Compared with the solo hUC-MSC-grafted or curcumin groups, combined curcumin-hUC-MSC therapy significantly improved the functional performance outcomes, diminished the infarct volumes and the cerebral edema. The combined treatment promoted anti-inflammatory microglia polarization via Nrf2 pathway and decreased the expression of ROS, oxidative mediators and pro-inflammatory cytokines, while elevating the expression of the anti-inflammatory cytokines. Nrf2 knockout abolished the antioxidant stress and anti-inflammation effects mediated with combined treatment. Moreover, the combined treatment enhanced the phosphorylation of AKT and GSK3β, inhibited the β-TrCP nucleus translocation, accompanied with Nrf2 activation in the nucleus. AKT inhibitor MK2206 activated GSK3β and β-TrCP and suppressed Nrf2 phosphorylation in nucleus, whereas MK2206 with the GSK3β inhibitor TDZD-8 reversed these phenomena. Furthermore, combined treatment followed by GSK3β inhibition with TDZD-8 restricted β-TrCP nucleus accumulation, which facilitated Nrf2 expression. CONCLUSIONS We have demonstrated that combined curcumin-hUC-MSC therapy exerts anti-inflammation and antioxidant stress efficacy mediated by anti-inflammatory microglia polarization via AKT/GSK-3β/β-TrCP/Nrf2 axis and an improved neurological function after AIS.
Collapse
Affiliation(s)
- Yuan Li
- grid.410570.70000 0004 1760 6682Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Chongqing, 400038 China
| | - Jialu Huang
- grid.410570.70000 0004 1760 6682Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Chongqing, 400038 China
| | - Jie Wang
- grid.410570.70000 0004 1760 6682Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Chongqing, 400038 China
| | - Simin Xia
- grid.410570.70000 0004 1760 6682Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Chongqing, 400038 China
| | - Hong Ran
- grid.410570.70000 0004 1760 6682Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Chongqing, 400038 China
| | - Lenyu Gao
- grid.410570.70000 0004 1760 6682Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Chongqing, 400038 China ,grid.410570.70000 0004 1760 6682Department of Traditional Chinese Medicine and Rheumatology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Chengjian Feng
- Department of Medical Engineering, 958th Hospital of the People’s Liberation Army, Chongqing, 400038 China
| | - Li Gui
- grid.410570.70000 0004 1760 6682Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Chongqing, 400038 China
| | - Zhenhua Zhou
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Chongqing, 400038, China.
| | - Jichao Yuan
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Chongqing, 400038, China.
| |
Collapse
|
111
|
Yan TT, Xiao R, Wang Y, Lin GA, Zheng Y, Zhao H, Li WJ, Shang XZ, Meng JS, Hu DS, Li S, Wang C, Lin ZC, Chen HC, Zhao DY, Tang D. [A prospective study on application of human umbilical cord mesenchymal stem cells combined with autologous Meek microskin transplantation in patients with extensive burns]. ZHONGHUA SHAO SHANG YU CHUANG MIAN XIU FU ZA ZHI 2023; 39:114-121. [PMID: 36878520 DOI: 10.3760/cma.j.cn501225-20220728-00319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Objective: To investigate the effects of human umbilical cord mesenchymal stem cells (hUCMSCs) combined with autologous Meek microskin transplantation on patients with extensive burns. Methods: The prospective self-controlled study was conducted. From May 2019 to June 2022, 16 patients with extensive burns admitted to the 990th Hospital of PLA Joint Logistics Support Force met the inclusion criteria, while 3 patients were excluded according to the exclusion criteria, and 13 patients were finally selected, including 10 males and 3 females, aged 24-61 (42±13) years. A total of 20 trial areas (40 wounds, with area of 10 cm×10 cm in each wound) were selected. Two adjacent wounds in each trial area were divided into hUCMSC+gel group applied with hyaluronic acid gel containing hUCMSCs and gel only group applied with hyaluronic acid gel only according to the random number table, with 20 wounds in each group. Afterwards the wounds in two groups were transplanted with autologous Meek microskin grafts with an extension ratio of 1∶6. In 2, 3, and 4 weeks post operation, the wound healing was observed, the wound healing rate was calculated, and the wound healing time was recorded. The specimen of wound secretion was collected for microorganism culture if there was purulent secretion on the wound post operation. In 3, 6, and 12 months post operation, the scar hyperplasia in wound was assessed using the Vancouver scar scale (VSS). In 3 months post operation, the wound tissue was collected for hematoxylin-eosin (HE) staining to observe the morphological changes and for immunohistochemical staining to observe the positive expressions of Ki67 and vimentin and to count the number of positive cells. Data were statistically analyzed with paired samples t test and Bonferronni correction. Results: In 2, 3, and 4 weeks post operation, the wound healing rates in hUCMSC+gel group were (80±11)%, (84±12)%, and (92±9)%, respectively, which were significantly higher than (67±18)%, (74±21)%, and (84±16)% in gel only group (with t values of 4.01, 3.52, and 3.66, respectively, P<0.05). The wound healing time in hUCMSC+gel group was (31±11) d, which was significantly shorter than (36±13) d in gel only group (t=-3.68, P<0.05). The microbiological culture of the postoperative wound secretion specimens from the adjacent wounds in 2 groups was identical, with negative results in 4 trial areas and positive results in 16 trial areas. In 3, 6, and 12 months post operation, the VSS scores of wounds in gel only group were 7.8±1.9, 6.7±2.1, and 5.4±1.6, which were significantly higher than 6.8±1.8, 5.6±1.6, and 4.0±1.4 in hUCMSC+gel group, respectively (with t values of -4.79, -4.37, and -5.47, respectively, P<0.05). In 3 months post operation, HE staining showed an increase in epidermal layer thickness and epidermal crest in wound in hUCMSC+gel group compared with those in gel only group, and immunohistochemical staining showed a significant increase in the number of Ki67 positive cells in wound in hUCMSC+gel group compared with those in gel only group (t=4.39, P<0.05), with no statistically significant difference in the number of vimentin positive cells in wound between the 2 groups (P>0.05). Conclusions: The application of hyaluronic acid gel containing hUCMSCs to the wound is simple to perform and is therefore a preferable route. Topical application of hUCMSCs can promote healing of the autologous Meek microskin grafted area in patients with extensive burns, shorten wound healing time, and alleviate scar hyperplasia. The above effects may be related to the increased epidermal thickness and epidermal crest, and active cell proliferation.
Collapse
Affiliation(s)
- T T Yan
- Military Burn Center, the 990th Hospital of PLA Joint Logistics Support Force, Zhumadian 463002, China
| | - R Xiao
- Military Burn Center, the 990th Hospital of PLA Joint Logistics Support Force, Zhumadian 463002, China
| | - Y Wang
- Beijing Zhongjing Hi-Tech Biotechnology Co., Beijing 100089, China
| | - G A Lin
- Military Burn Center, the 990th Hospital of PLA Joint Logistics Support Force, Zhumadian 463002, China
| | - Y Zheng
- Military Burn Center, the 990th Hospital of PLA Joint Logistics Support Force, Zhumadian 463002, China
| | - H Zhao
- Henan Cellular Industry Technology Research Institute Co., Zhengzhou 450121, China
| | - W J Li
- Military Burn Center, the 990th Hospital of PLA Joint Logistics Support Force, Zhumadian 463002, China
| | - X Z Shang
- Military Burn Center, the 990th Hospital of PLA Joint Logistics Support Force, Zhumadian 463002, China
| | - J S Meng
- Military Burn Center, the 990th Hospital of PLA Joint Logistics Support Force, Zhumadian 463002, China
| | - D S Hu
- Military Burn Center, the 990th Hospital of PLA Joint Logistics Support Force, Zhumadian 463002, China
| | - S Li
- Military Burn Center, the 990th Hospital of PLA Joint Logistics Support Force, Zhumadian 463002, China
| | - C Wang
- Military Burn Center, the 990th Hospital of PLA Joint Logistics Support Force, Zhumadian 463002, China
| | - Z C Lin
- Military Burn Center, the 990th Hospital of PLA Joint Logistics Support Force, Zhumadian 463002, China
| | - H C Chen
- Military Burn Center, the 990th Hospital of PLA Joint Logistics Support Force, Zhumadian 463002, China
| | - D Y Zhao
- Military Burn Center, the 990th Hospital of PLA Joint Logistics Support Force, Zhumadian 463002, China
| | - D Tang
- Department of Burns & Plastic Surgery, PLA General Hospital of Central Theater Command, Wuhan 430012, China
| |
Collapse
|
112
|
Empagliflozin-Pretreated Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Attenuated Heart Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:7747727. [PMID: 36852325 PMCID: PMC9966826 DOI: 10.1155/2023/7747727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/20/2023]
Abstract
Objective Small extracellular vesicles derived from mesenchymal stem cells (MSCs) play important roles in cardiac protection. Studies have shown that the cardiovascular protection of sodium-glucose cotransporter 2 inhibitor (SGLT2i) is independent of its hypoglycemic effect. This study is aimed at investigating whether small extracellular vesicles derived from MSCs pretreated with empagliflozin (EMPA) has a stronger cardioprotective function after myocardial infarction (MI) and to explore the underlying mechanisms. Methods and Results We evaluated the effects of EMPA on MSCs and the effects of EMPA-pretreated MSCs-derived small extracellular vesicles (EMPA-sEV) on myocardial apoptosis, angiogenesis, and cardiac function after MI in vitro and in vivo. The small extracellular vesicles of control MSCs (MSC-sEV) and EMPA-pretreated MSCs were extracted, respectively. Small extracellular vesicles were cocultured with apoptotic H9c2 cells induced by H2O2 or injected into the infarcted area of the Sprague-Dawley (SD) rat myocardial infarction model. EMPA increased the cell viability, migration ability, and inhibited apoptosis and senescence of MSCs. In vitro, EMPA-sEV inhibited apoptosis of H9c2 cells compared with the control group (MSC-sEV). In the SD rat model of MI, EMPA-sEV inhibited myocardial apoptosis and promoted angiogenesis in the infarct marginal areas compared with the MSC-sEV. Meanwhile, EMPA-sEV reduced infarct size and improved cardiac function. Through small extracellular vesicles (miRNA) sequencing, we found several differentially expressed miRNAs, among which miR-214-3p was significantly elevated in EMPA-sEV. Coculture of miR-214-3p high expression MSC-derived small extracellular vesicles with H9c2 cells produced similar protective effects. In addition, miR-214-3p was found to promote AKT phosphorylation in H9c2 cells. Conclusions Our data suggest that EMPA-sEV significantly improve cardiac repair after MI by inhibiting myocardial apoptosis. miR-214-3p at least partially mediated the myocardial protection of EMPA-sEV through the AKT signaling pathway.
Collapse
|
113
|
Shaik R, Xu J, Wang Y, Hong Y, Zhang G. Fibrin-Enriched Cardiac Extracellular Matrix Hydrogel Promotes In Vitro Angiogenesis. ACS Biomater Sci Eng 2023; 9:877-888. [PMID: 36630688 PMCID: PMC10064974 DOI: 10.1021/acsbiomaterials.2c01148] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Angiogenesis is essential for cardiac repair after myocardial infarction. Promoting angiogenesis has been demonstrated as an effective approach for myocardial infarction treatment. Several different strategies for inducing myocardial angiogenesis have been explored, including exogenous delivery of angiogenic genes, proteins, microRNAs, cells, and extracellular vesicles. Various types of injectable hydrogels have been investigated for cardiac tissue repair. One of the most promising injectable hydrogels in cardiac regeneration is a cardiac extracellular matrix hydrogel that is derived from decellularized porcine myocardium. It can be delivered minimally invasively via transendocardial delivery. The safety and efficacy of cardiac extracellular matrix hydrogels have been shown in small and large animal myocardial infarction models as well as clinical trials. The main mechanisms underlying the therapeutic benefits of cardiac extracellular matrix hydrogels have been elucidated and involved in the modulation of the immune response, downregulation of pathways related to heart failure progression and fibrosis, upregulation of genes important for cardiac muscle contraction, and enhancing cardiomyocyte differentiation and maturation from stem cells. However, no potent capillary network formation induced by cardiac extracellular matrix hydrogels has been reported. In this study, we tested the feasibility of incorporating a fibrin matrix into cardiac extracellular matrix hydrogels to improve the angiogenic properties of the hydrogel. Our in vitro results demonstrate that fibrin-enriched cardiac extracellular matrix hydrogels can induce robust endothelial cell tube formation from human umbilical vein endothelial cells and promote the sprouting of human mesenchymal stem cell spheroids. The obtained information from this study is very critical toward the future in vivo evaluation of fibrin-enriched cardiac extracellular matrix hydrogels in promoting myocardial angiogenesis.
Collapse
Affiliation(s)
- Rubia Shaik
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Jiazhu Xu
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Yong Wang
- Department of Biomedical Engineering, Pennsylvania State University, State College, University Park, Pennsylvania 16801, United States
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Ge Zhang
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
114
|
Mesenchymal Stem Cells and Their Exocytotic Vesicles. Int J Mol Sci 2023; 24:ijms24032085. [PMID: 36768406 PMCID: PMC9916886 DOI: 10.3390/ijms24032085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
Mesenchymal stem cells (MSCs), as a kind of pluripotent stem cells, have attracted much attention in orthopedic diseases, geriatric diseases, metabolic diseases, and sports functions due to their osteogenic potential, chondrogenic differentiation ability, and adipocyte differentiation. Anti-inflammation, anti-fibrosis, angiogenesis promotion, neurogenesis, immune regulation, and secreted growth factors, proteases, hormones, cytokines, and chemokines of MSCs have been widely studied in liver and kidney diseases, cardiovascular and cerebrovascular diseases. In recent years, many studies have shown that the extracellular vesicles of MSCs have similar functions to MSCs transplantation in all the above aspects. Here we review the research progress of MSCs and their exocrine vesicles in recent years.
Collapse
|
115
|
Pathogenic Role of Adipose Tissue-Derived Mesenchymal Stem Cells in Obesity and Obesity-Related Inflammatory Diseases. Cells 2023; 12:cells12030348. [PMID: 36766689 PMCID: PMC9913687 DOI: 10.3390/cells12030348] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/12/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023] Open
Abstract
Adipose tissue-derived mesenchymal stem cells (ASCs) are adult stem cells, endowed with self-renewal, multipotent capacities, and immunomodulatory properties, as mesenchymal stem cells (MSCs) from other origins. However, in a pathological context, ASCs like MSCs can exhibit pro-inflammatory properties and attract inflammatory immune cells at their neighborhood. Subsequently, this creates an inflammatory microenvironment leading to ASCs' or MSCs' dysfunctions. One such example is given by obesity where adipogenesis is impaired and insulin resistance is initiated. These opposite properties have led to the classification of MSCs into two categories defined as pro-inflammatory ASC1 or anti-inflammatory ASC2, in which plasticity depends on the micro-environmental stimuli. The aim of this review is to (i) highlight the pathogenic role of ASCs during obesity and obesity-related inflammatory diseases, such as rheumatoid arthritis, multiple sclerosis, psoriasis, inflammatory bowel disease, and cancer; and (ii) describe some of the mechanisms leading to ASCs dysfunctions. Thus, the role of soluble factors, adhesion molecules; TLRs, Th17, and Th22 cells; γδ T cells; and immune checkpoint overexpression will be addressed.
Collapse
|
116
|
Wei Q, Liu X, Su JL, Wang YX, Chu ZQ, Ma K, Huang QL, Li HH, Fu XB, Zhang CP. Small extracellular vesicles from mesenchymal stem cells: A potential Weapon for chronic non-healing wound treatment. Front Bioeng Biotechnol 2023; 10:1083459. [PMID: 36704302 PMCID: PMC9872203 DOI: 10.3389/fbioe.2022.1083459] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023] Open
Abstract
Chronic non-healing wounds have posed a severe threat to patients mentally and physically. Behavior dysregulation of remaining cells at wound sites is recognized as the chief culprit to destroy healing process and hinders wound healing. Therefore, regulating and restoring normal cellular behavior is the core of chronic non-healing wound treatment. In recent years, the therapy with mesenchymal stem cells (MSCs) has become a promising option for chronic wound healing and the efficacy has increasingly been attributed to their exocrine functions. Small extracellular vesicles derived from MSCs (MSC-sEVs) are reported to benefit almost all stages of wound healing by regulating the cellular behavior to participate in the process of inflammatory response, angiogenesis, re-epithelization, and scarless healing. Here, we describe the characteristics of MSC-sEVs and discuss their therapeutic potential in chronic wound treatment. Additionally, we also provide an overview of the application avenues of MSC-sEVs in wound treatment. Finally, we summarize strategies for large-scale production and engineering of MSC-sEVs. This review may possibly provide meaningful guidance for chronic wound treatment with MSC-sEVs.
Collapse
Affiliation(s)
- Qian Wei
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Xi Liu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Jian-Long Su
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Ya-Xi Wang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Zi-Qiang Chu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Kui Ma
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese, PLA General Hospital, Beijing, China
- Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Research Unit of Trauma Care, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China
| | - Qi-Lin Huang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Hai-Hong Li
- Department of Wound Repair, Institute of Wound Repair and Regeneration Medicine, Southern University of Science and Technology Hospital, Southern University of Science and Technology School of Medicine, Shenzhen, China
| | - Xiao-Bing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese, PLA General Hospital, Beijing, China
- Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Research Unit of Trauma Care, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China
| | - Cui-Ping Zhang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese, PLA General Hospital, Beijing, China
- Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Research Unit of Trauma Care, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China
| |
Collapse
|
117
|
Labuz DF, Whitlock AE, Kycia I, Zurakowski D, Fauza DO. Early functional analysis on the pulmonary hemodynamic effects of Transamniotic Stem Cell Therapy (TRASCET) in the nitrofen model of congenital diaphragmatic hernia. J Pediatr Surg 2023; 58:8-13. [PMID: 36280468 DOI: 10.1016/j.jpedsurg.2022.09.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/16/2022] [Indexed: 01/09/2023]
Abstract
PURPOSE Transamniotic stem cell therapy (TRASCET) with mesenchymal stem cells (MSCs) has been shown to impact pulmonary vascular development and remodeling in experimental congenital diaphragmatic hernia (CDH), with secondary structural cardiac effects. We sought to determine whether TRASCET has any functional impact on term fetal pulmonary hemodynamics in the nitrofen model. METHODS Time-dated pregnant rat dams (n = 13) received nitrofen on gestational day 9 (E9) to induce fetal CDH. Fetuses (n = 155) were divided into three groups: untreated (n = 45), and two groups receiving volume-matched intra-amniotic injections on E17 of either saline (sham; n = 46), or a suspension of amniotic fluid-derived MSCs (afMSCs) (TRASCET; n = 64). Donor afMSCs were syngeneic, phenotyped by flow cytometry, and "primed" by exposure to interferon-gamma and interleukin-1beta prior to administration in vivo. At term (E21), fetuses underwent Doppler flow assessment at the mid-pulmonary artery and 4-chamber echocardiogram. Pulmonary vascular resistance was estimated by pulmonary artery acceleration time (PAAT), max velocity (MaxV) and velocity time integral (VTI). Cardiac function was assessed by global longitudinal strain (GLS) and ejection fraction (EF) using speckle analyses. Healthy fetuses (n = 11) served as additional controls. Statistical analysis was by the Mann-Whitney U test RESULTS: High resolution ultrasound data could be obtained from 8 to 13 fetuses per group. The PAAT and the PAAT normalized to cardiac cycle time were significantly improved by TRASCET compared to both untreated and sham-treated CDH (p = 0.004 to <0.001 in all pairwise comparisons). The flow profile sharpness (MaxV:VTI) was increased in untreated (p = 0.06) and sham (p = 0.01) groups but normalized by TRASCET (p<0.01). There was no difference in GLS between TRASCET and either the untreated or sham groups (p = 0.25 to p = 0.93). CONCLUSION Transamniotic stem cell therapy improves pulmonary vascular resistance in early term fetuses in the Nitrofen model of congenital diaphragmatic hernia. Further focus on the functional pulmonary hemodynamic impact of this therapy is justified. LEVEL OF EVIDENCE N/A (animal and laboratory study).
Collapse
Affiliation(s)
- Daniel F Labuz
- Department of Surgery, Boston Children's Hospital/ Harvard Medical School, Boston, MA, United States
| | - Ashlyn E Whitlock
- Department of Surgery, Boston Children's Hospital/ Harvard Medical School, Boston, MA, United States
| | - Ina Kycia
- Department of Surgery, Boston Children's Hospital/ Harvard Medical School, Boston, MA, United States
| | - David Zurakowski
- Department of Surgery, Boston Children's Hospital/ Harvard Medical School, Boston, MA, United States
| | - Dario O Fauza
- Department of Surgery, Boston Children's Hospital/ Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
118
|
Qi T, Xu X, Guo Y, Xia Y, Peng L, Li C, Ding F, Gao C, Fan M, Yu M, Zhao H, He Y, Li W, Hai C, Gao E, Zhang X, Gao F, Fan Y, Yan W, Tao L. CSF2RB overexpression promotes the protective effects of mesenchymal stromal cells against ischemic heart injury. Theranostics 2023; 13:1759-1773. [PMID: 37064880 PMCID: PMC10091875 DOI: 10.7150/thno.81336] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/26/2023] [Indexed: 04/18/2023] Open
Abstract
Aims: The invasive intramyocardial injection of mesenchymal stromal cells (MSCs) allows for limited repeat injections and shows poor therapeutic efficacy against ischemic heart failure. Intravenous injection is an alternative method because this route allows for repeated, noninvasive, and easy delivery. However, the lack of targeting of MSCs hinders the ability of these cells to accumulate in the ischemic area after intravenous injections. We investigated whether and how the overexpression of colony-stimulating factor 2 receptor beta subunit (CSF2RB) may regulate the cardiac homing of MSCs and their cardioprotective effects against ischemic heart failure. Methods and Results: Adult mice were subjected to myocardial ischemia/reperfusion (MI/R) or sham operations. We observed significantly higher CSF2 protein expression and secretion by the ischemic heart from 1 day to 2 weeks after MI/R. Mouse adipose tissue-derived MSCs (ADSCs) were infected with adenovirus harboring CSF2RB or control adenovirus. Enhanced green fluorescent protein (EGFP)-labeled ADSCs were intravenously injected into MI/R mice every three days for a total of 7 times. Compared with ADSCs infected with control adenovirus, intravenously delivered ADSCs overexpressing CSF2RB exhibited markedly increased cardiac homing. Histological analysis revealed that CSF2RB overexpression significantly enhanced the ADSC-mediated proangiogenic, antiapoptotic, and antifibrotic effects. More importantly, ADSCs overexpressing CSF2RB significantly increased the left ventricular ejection fraction and cardiac contractility/relaxation in MI/R mice. In vitro experiments demonstrated that CSF2RB overexpression increases the migratory capacity and reduces the hypoxia/reoxygenation-induced apoptosis of ADSCs. We identified STAT5 phosphorylation as the key mechanism underlying the effects of CSF2RB on promoting ADSC migration and inhibiting ADSC apoptosis. RNA sequencing followed by cause-effect analysis revealed that CSF2RB overexpression increases the expression of the ubiquitin ligase RNF4. Coimmunoprecipitation and coimmunostaining experiments showed that RNF4 binds to phosphorylated STAT5. RNF4 knockdown reduced STAT5 phosphorylation as well as the antiapoptotic and promigratory actions of ADSCs overexpressing CSF2RB. Conclusions: We demonstrate for the first time that CSF2RB overexpression optimizes the efficacy of intravenously delivered MSCs in the treatment of ischemic heart injury by increasing the response of the MSCs to a CSF2 gradient and CSF2RB-dependent STAT5/RNF4 activation.
Collapse
Affiliation(s)
- Tingting Qi
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xiaoming Xu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yongzhen Guo
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
- School of Public Management, Northwest University, Xi'an 710127, China
| | - Yunlong Xia
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Lu Peng
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Fengyue Ding
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Chao Gao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Miaomiao Fan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Min Yu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Huishou Zhao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yuan He
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Wenli Li
- Department of Toxicology, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Shanxi Key Lab of Free Radical Biology and Medicine, School of Public Health, The Fourth Military Medical University, Xi'an 710032, China
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Chunxu Hai
- Department of Toxicology, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Shanxi Key Lab of Free Radical Biology and Medicine, School of Public Health, The Fourth Military Medical University, Xi'an 710032, China
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Erhe Gao
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Xing Zhang
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Feng Gao
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yanhong Fan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
- ✉ Corresponding authors: Ling Tao, MD, PhD, Professor and Chief of Department of Cardiology, Xijing Hospital, The Fourth Military Medical University. 127 West Changle Rd, Xi'an, China, 710032. Tel.: +86-29-84771692; +86-29-84775183; Fax: +86-29-84771692; E-mail: . Wenjun Yan, MD, PhD, Associate Professor, Department of Cardiology, Xijing Hospital, The Fourth Military Medical University. 127 West Changle Rd, Xi'an, China, 710032. Tel.: +86-29-84775183; Fax: +86-29-84771692; E-mail: . Yanhong Fan, MD, PhD, Associate Professor, Department of Cardiology, Xijing Hospital, The Fourth Military Medical University. 127 West Changle Rd, Xi'an, China, 710032. Tel.: +86-29-84775183; Fax: +86-29-84771692;
| | - Wenjun Yan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
- ✉ Corresponding authors: Ling Tao, MD, PhD, Professor and Chief of Department of Cardiology, Xijing Hospital, The Fourth Military Medical University. 127 West Changle Rd, Xi'an, China, 710032. Tel.: +86-29-84771692; +86-29-84775183; Fax: +86-29-84771692; E-mail: . Wenjun Yan, MD, PhD, Associate Professor, Department of Cardiology, Xijing Hospital, The Fourth Military Medical University. 127 West Changle Rd, Xi'an, China, 710032. Tel.: +86-29-84775183; Fax: +86-29-84771692; E-mail: . Yanhong Fan, MD, PhD, Associate Professor, Department of Cardiology, Xijing Hospital, The Fourth Military Medical University. 127 West Changle Rd, Xi'an, China, 710032. Tel.: +86-29-84775183; Fax: +86-29-84771692;
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
- ✉ Corresponding authors: Ling Tao, MD, PhD, Professor and Chief of Department of Cardiology, Xijing Hospital, The Fourth Military Medical University. 127 West Changle Rd, Xi'an, China, 710032. Tel.: +86-29-84771692; +86-29-84775183; Fax: +86-29-84771692; E-mail: . Wenjun Yan, MD, PhD, Associate Professor, Department of Cardiology, Xijing Hospital, The Fourth Military Medical University. 127 West Changle Rd, Xi'an, China, 710032. Tel.: +86-29-84775183; Fax: +86-29-84771692; E-mail: . Yanhong Fan, MD, PhD, Associate Professor, Department of Cardiology, Xijing Hospital, The Fourth Military Medical University. 127 West Changle Rd, Xi'an, China, 710032. Tel.: +86-29-84775183; Fax: +86-29-84771692;
| |
Collapse
|
119
|
Extracellular magnetic labeling of biomimetic hydrogel-induced human mesenchymal stem cell spheroids with ferumoxytol for MRI tracking. Bioact Mater 2023; 19:418-428. [PMID: 35574059 PMCID: PMC9079175 DOI: 10.1016/j.bioactmat.2022.04.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/20/2022] [Accepted: 04/24/2022] [Indexed: 02/07/2023] Open
Abstract
Labeling of mesenchymal stem cells (MSCs) with superparamagnetic iron oxide nanoparticles (SPIONs) has emerged as a potential method for magnetic resonance imaging (MRI) tracking of transplanted cells in tissue repair studies and clinical trials. Labeling of MSCs using clinically approved SPIONs (ferumoxytol) requires the use of transfection reagents or magnetic field, which largely limits their clinical application. To overcome this obstacle, we established a novel and highly effective method for magnetic labeling of MSC spheroids using ferumoxytol. Unlike conventional methods, ferumoxytol labeling was done in the formation of a mechanically tunable biomimetic hydrogel-induced MSC spheroids. Moreover, the labeled MSC spheroids exhibited strong MRI T2 signals and good biosafety. Strikingly, the encapsulated ferumoxytol was localized in the extracellular matrix (ECM) of the spheroids instead of the cytoplasm, minimizing the cytotoxicity of ferumoxytol and maintaining the viability and stemness properties of biomimetic hydrogel-induced MSC spheroids. This demonstrates the potential of this method for post-transplantation MRI tracking in the clinic. An extracellular magnetic labeling method was developed for MSC spheroids using ferumoxytol. Ferumoxytol encapsulated into abundant ECM proteins network of MSC spheroids ensured this method is stable and durable. Uniformly sized magnetic spheroids induced by mechanically tunable biomimetic hydrogels promoted MSCs stemness properties. Magnetically labeled MSC spheroids exhibited superior MRI imaging both in vitro and in vivo.
Collapse
|
120
|
Gao L, Qiu F, Cao H, Li H, Dai G, Ma T, Gong Y, Luo W, Zhu D, Qiu Z, Zhu P, Chu S, Yang H, Liu Z. Therapeutic delivery of microRNA-125a-5p oligonucleotides improves recovery from myocardial ischemia/reperfusion injury in mice and swine. Theranostics 2023; 13:685-703. [PMID: 36632217 PMCID: PMC9830430 DOI: 10.7150/thno.73568] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 12/12/2022] [Indexed: 01/04/2023] Open
Abstract
Rationale: Clinical application of mesenchymal stem cells (MSCs) and MSC-derived exosomes (MSC-Exos) to alleviate myocardial ischemia/reperfusion (I/R) injury is compromised by the low cell engraftment rate and uncontrolled exosomal content. As one of their active ingredients, single-component microRNA therapy may have more inherent advantages. We sought to find an ideal microRNA candidate and determine whether it could reproduce the cardioprotective effects of MSCs and MSC-Exos. Methods: Cardiac function and myocardial remodeling in MSC, MSC-Exo, or microRNA oligonucleotide-treated mouse hearts were investigated after I/R injury. The effects of microRNA oligonucleotides on cardiac cells (macrophages, cardiomyocytes, fibroblasts, and endothelial cells) and their downstream mechanisms were confirmed. Large animals were also employed to investigate the safety of microRNA therapy. Results: The results showed that microRNA-125a-5p (miR-125a-5p) is enriched in MSC-Exos, and intramyocardial delivery of their modified oligonucleotides (agomir) in mouse I/R myocardium, as well as MSCs or MSC-Exos, exerted obvious cardioprotection by increasing cardiac function and limiting adverse remodeling. In addition, miR-125a-5p agomir treatment increased M2 macrophage polarization, promoted angiogenesis, and attenuated fibroblast proliferation and activation, which subsequently contributed to the improvements in cardiomyocyte apoptosis and inflammation. Mechanistically, Klf13, Tgfbr1, and Daam1 are considered the targets of miR-125a-5p for regulating the function of macrophages, fibroblasts, and endothelial cells, respectively. Similar results were observed following miR-125a-5p agomir treatment in a porcine model, with no increase in the risk of arrhythmia or hepatic, renal, or cardiac toxicity. Conclusions: This targeted microRNA delivery presents an effective and safe strategy as a stem cell and exosomal therapy in I/R cardiac repair.
Collapse
Affiliation(s)
- Ling Gao
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China.,✉ Corresponding authors: Ling Gao, PhD, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Rd., Shanghai, 200123, China. E-mail: ; Zhongmin Liu, MD, PhD, Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd., Shanghai, 200120, China. E-mail: ; Huangtian Yang, PhD, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, 320 Yueyang Rd., Shanghai, 200031, China. E-mail: ; Shuguang Chu, PhD, Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China. E-mail:
| | - Fan Qiu
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Hao Cao
- Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Hao Li
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Gonghua Dai
- Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Teng Ma
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Yanshan Gong
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Wei Luo
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Dongling Zhu
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Zhixuan Qiu
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Shuguang Chu
- Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,✉ Corresponding authors: Ling Gao, PhD, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Rd., Shanghai, 200123, China. E-mail: ; Zhongmin Liu, MD, PhD, Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd., Shanghai, 200120, China. E-mail: ; Huangtian Yang, PhD, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, 320 Yueyang Rd., Shanghai, 200031, China. E-mail: ; Shuguang Chu, PhD, Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China. E-mail:
| | - Huangtian Yang
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China.,Research Institute of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai 200031, China.,✉ Corresponding authors: Ling Gao, PhD, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Rd., Shanghai, 200123, China. E-mail: ; Zhongmin Liu, MD, PhD, Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd., Shanghai, 200120, China. E-mail: ; Huangtian Yang, PhD, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, 320 Yueyang Rd., Shanghai, 200031, China. E-mail: ; Shuguang Chu, PhD, Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China. E-mail:
| | - Zhongmin Liu
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China.,Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Research Institute of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Shanghai Institute of Stem Cell Research and Clinical translation, Shanghai East Hospital, Tongji University, Shanghai 200120, China.,✉ Corresponding authors: Ling Gao, PhD, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Rd., Shanghai, 200123, China. E-mail: ; Zhongmin Liu, MD, PhD, Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd., Shanghai, 200120, China. E-mail: ; Huangtian Yang, PhD, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, 320 Yueyang Rd., Shanghai, 200031, China. E-mail: ; Shuguang Chu, PhD, Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China. E-mail:
| |
Collapse
|
121
|
Evaluation of the Optimal Manufacturing Protocols and Therapeutic Properties of Mesenchymal Stem/Stromal Cells Derived from Wharton's Jelly. Int J Mol Sci 2022; 24:ijms24010652. [PMID: 36614096 PMCID: PMC9820979 DOI: 10.3390/ijms24010652] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/03/2023] Open
Abstract
Wharton's jelly (WJ) from the umbilical cord (UC) is a good source of mesenchymal stem/stromal cells (MSCs), which can be isolated and used in therapy. Current knowledge shows that even small changes in the cell environment may result in obtaining a subpopulation of cells with different therapeutic properties. For this reason, the conditions of UC transportation, cell isolation, and cultivation and the banking of cells destined for clinical use should be unified and optimized. In this project, we tried various protocols for cell vs. bioptat isolation, banking, and transport in order to determine the most optimal. The most efficient isolation method of WJ-MSCs was chopping the whole umbilical matrix with a scalpel after vessel and lining membrane removal. The optimal solution for short term cell transportation was a multi-electrolyte fluid without glucose. Considering the use of WJ-MSCs in cell therapies, it was important to investigate the soluble secretome of both WJ bioptats and WJ-MSCs. WJ-MSCs secreted higher levels of cytokines and chemokines than WJ bioptats. WJ-MSCs secreted HGF, CCL2, ICAM-1, BDNF, and VEGF. Since these cells might be used in treating neurodegenerative disorders, we investigated the impact of cerebrospinal fluid (CSF) on WJ-MSCs' features. In the presence of CSF, the cells expressed consecutive neural markers both at the protein and gene level: nestin, β-III-tubulin, S-100-β, GFAP, and doublecortin. Based on the obtained results, a protocol for manufacturing an advanced-therapy medicinal product was composed.
Collapse
|
122
|
Rodríguez-Eguren A, Gómez-Álvarez M, Francés-Herrero E, Romeu M, Ferrero H, Seli E, Cervelló I. Human Umbilical Cord-Based Therapeutics: Stem Cells and Blood Derivatives for Female Reproductive Medicine. Int J Mol Sci 2022; 23:ijms232415942. [PMID: 36555583 PMCID: PMC9785531 DOI: 10.3390/ijms232415942] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/04/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
There are several conditions that lead to female infertility, where traditional or conventional treatments have limited efficacy. In these challenging scenarios, stem cell (SC) therapies have been investigated as alternative treatment strategies. Human umbilical cord (hUC) mesenchymal stem cells (hUC-MSC), along with their secreted paracrine factors, extracts, and biomolecules, have emerged as promising therapeutic alternatives in regenerative medicine, due to their remarkable potential to promote anti-inflammatory and regenerative processes more efficiently than other autologous treatments. Similarly, hUC blood derivatives, such as platelet-rich plasma (PRP), or isolated plasma elements, such as growth factors, have also demonstrated potential. This literature review aims to summarize the recent therapeutic advances based on hUC-MSCs, hUC blood, and/or other plasma derivatives (e.g., extracellular vesicles, hUC-PRP, and growth factors) in the context of female reproductive medicine. We present an in-depth analysis of the principal molecules mediating tissue regeneration, compiling the application of these therapies in preclinical and clinical studies, within the context of the human reproductive tract. Despite the recent advances in bioengineering strategies that sustain delivery and amplify the scope of the therapeutic benefits, further clinical trials are required prior to the wide implementation of these alternative therapies in reproductive medicine.
Collapse
Affiliation(s)
- Adolfo Rodríguez-Eguren
- IVI Foundation, Health Research Institute La Fe, 46026 Valencia, Spain
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 05610, USA
| | | | - Emilio Francés-Herrero
- IVI Foundation, Health Research Institute La Fe, 46026 Valencia, Spain
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Mónica Romeu
- Gynecological Service, Consortium General University Hospital of Valencia, 46014 Valencia, Spain
| | - Hortensia Ferrero
- IVI Foundation, Health Research Institute La Fe, 46026 Valencia, Spain
| | - Emre Seli
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 05610, USA
- IVIRMA New Jersey, Basking Ridge, NJ 07920, USA
| | - Irene Cervelló
- IVI Foundation, Health Research Institute La Fe, 46026 Valencia, Spain
- Correspondence: or
| |
Collapse
|
123
|
Kędziora A, Konstanty-Kalandyk J, Litwinowicz R, Mazur P, Kapelak B, Piątek J. Hybrid techniques for myocardial regeneration: state of the art and future perspectives. ADVANCES IN INTERVENTIONAL CARDIOLOGY 2022; 18:360-365. [PMID: 36967853 PMCID: PMC10031663 DOI: 10.5114/aic.2022.121124] [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: 11/01/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
Myocardium has a limited proliferative capacity, and adult hearts are considered incapable of regenerating after injury. A significant loss in the viable myocardium eventually diminishes the heart's ability to contract synchronously, leading to heart failure. Despite the development in interventional and pharmacological treatment for ischemic heart disease and heart failure, there is a significant number of highly symptomatic patients. For these individuals, treatments that stimulate myocardial regeneration can offer alleviation of dyspnea and angina and improvement in quality of life. Stem cells are known to promote neovascularization and endothelial repair. Various stem cell lines have been investigated over the years to establish those with the highest potential to differentiate into cardiomyocytes, including bone marrow-derived mononuclear cells, mesenchymal stromal cells, CD34+, CD133+, endothelial progenitor cells, and adipose-derived mesenchymal stromal cells. Stem cell studies were based on several delivery pathways: infusion into coronary vessels, direct injection into the injured region of the myocardium, and delivery within the novel bioengineered scaffolds. Acellular materials have also been investigated over the years. They demonstrate the therapeutic potential to promote angiogenesis and release of growth factors to improve the restoration of critical components of the extracellular matrix. This review summarizes hybrid cardiac regeneration treatments that combine novel bioengineering techniques with delivery approaches that cardiac surgeons can provide.
Collapse
Affiliation(s)
- Anna Kędziora
- Department of Cardiovascular Surgery and Transplantology, John Paul II Hospital, Krakow, Poland
- Jagiellonian University Medical College, Krakow, Poland
| | - Janusz Konstanty-Kalandyk
- Department of Cardiovascular Surgery and Transplantology, John Paul II Hospital, Krakow, Poland
- Jagiellonian University Medical College, Krakow, Poland
| | - Radosław Litwinowicz
- Jagiellonian University Medical College, Krakow, Poland
- Department of Cardiac Surgery, Regional Specialist Hospital, Grudziadz, Poland
| | - Piotr Mazur
- Department of Cardiovascular Surgery and Transplantology, John Paul II Hospital, Krakow, Poland
- Jagiellonian University Medical College, Krakow, Poland
- Department of Cardiac Surgery, Leipzig Heart Center, Leipzig, Germany
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester MIN, USA
| | - Bogusław Kapelak
- Department of Cardiovascular Surgery and Transplantology, John Paul II Hospital, Krakow, Poland
- Jagiellonian University Medical College, Krakow, Poland
| | - Jacek Piątek
- Department of Cardiovascular Surgery and Transplantology, John Paul II Hospital, Krakow, Poland
- Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
124
|
Stem cell sheet fabrication from human umbilical cord mesenchymal stem cell and Col-T scaffold. Stem Cell Res 2022; 65:102960. [PMID: 36399925 DOI: 10.1016/j.scr.2022.102960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Today, stem cell therapy has been shown to be a remarkable progress and an important application in the regeneration of defective tissues and organs. To deliver stem cells to the injured area, several methods have been proposed such as an intravenous infusion, direct damaged tissue injection, or stem cell sheet transplantation. In this study, we aimed to fabricate a stem cell sheet by culturing human umbilical cord mesenchymal stem cells (hUC-MSCs) on a Col-T scaffold to recover the structure and function of damaged tissues. The results showed that cells reach confluent on the scaffold surface 18 h after seeding. These stem cells were able to survive and proliferate on Col-T scaffold. The average tensile strength of the stem cell sheet was 2.65 MPa. The sheet reached the sterile standards when tested for total bacteria, Candida albicans, Pseudomonas aeruginosa, and Staphylococcus aureus according to Circular number 06/2011/TT-BYT of Vietnam Ministry of Health. In addition, the stem cell sheet was non-toxic when evaluated for exposure toxicity and fluid toxicity according to iSO-10993. Importantly, 5 days after culturing on the Col-T scaffold, the seeded hUC-MSCs were still possessed all properties of MSC such as spindle-shaped, adhesive, could differentiate into mesoderm-derived cells, showed to be CD90, CD105, CD73 positive and CD45, CD34, CD11b, CD19, HLA-DR negative. In summary, our study was successful in creating a stem cell sheet from hUC-MSCs and Col-T scaffold for subsequent in vivo transplantation in the future.
Collapse
|
125
|
Lau H, Han DW, Park J, Lehner E, Kals C, Arzt C, Bayer E, Auer D, Schally T, Grasmann E, Fang H, Lee J, Lee HS, Han J, Gimona M, Rohde E, Bae S, Oh SW. GMP-compliant manufacturing of biologically active cell-derived vesicles produced by extrusion technology. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e70. [PMID: 38938599 PMCID: PMC11080851 DOI: 10.1002/jex2.70] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/08/2022] [Accepted: 11/01/2022] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) released by a variety of cell types have been shown to act as a natural delivery system for bioactive molecules such as RNAs and proteins. EV therapy holds great promise as a safe and cell-free therapy for many immunological and degenerative diseases. However, translation to clinical application is limited by several factors, including insufficient large-scale manufacturing technologies and low yield. We have developed a novel drug delivery platform technology, BioDrone™, based on cell-derived vesicles (CDVs) produced from diverse cell sources by using a proprietary extrusion process. This extrusion technology generates nanosized vesicles in far greater numbers than naturally obtained EVs. We demonstrate that the CDVs are surrounded by a lipid bilayer membrane with a correct membrane topology. Physical, biochemical and functional characterisation results demonstrate the potential of CDVs to act as effective therapeutics. Umbilical cord mesenchymal stem cell (UCMSC)-derived CDVs exhibit a biological activity that is similar to UCMSCs or UCMSC-derived EVs. Lastly, we present the establishment of a GMP-compliant process to allow the production of a large number of UCMSC-CDVs in a reproducible manner. GMP-compliant manufacturing of CDVs will facilitate the preclinical and clinical evaluation of these emerging therapeutics in anti-inflammatory or regenerative medicine. This study also represents a crucial step in the development of this novel drug delivery platform based on CDVs.
Collapse
Affiliation(s)
| | - Dong Woo Han
- BioDrone Research InstituteMDimune Inc.SeoulKorea
| | - Jinhee Park
- BioDrone Research InstituteMDimune Inc.SeoulKorea
| | - Edwine Lehner
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
| | - Carina Kals
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
| | - Claudia Arzt
- Transfer Centre for Extracellular Vesicle Theralytic Technologies (EV‐TT)SalzburgAustria
| | - Elisabeth Bayer
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
| | - Daniela Auer
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
| | - Tanja Schally
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
| | - Eva Grasmann
- Transfer Centre for Extracellular Vesicle Theralytic Technologies (EV‐TT)SalzburgAustria
| | - Han Fang
- Transfer Centre for Extracellular Vesicle Theralytic Technologies (EV‐TT)SalzburgAustria
| | - Jae‐Young Lee
- Department of Ophthalmology, Eunpyeong St. Mary's Hospital, College of MedicineThe Catholic University of KoreaSeoulKorea
| | - Hyun Soo Lee
- Department of Ophthalmology, Eunpyeong St. Mary's Hospital, College of MedicineThe Catholic University of KoreaSeoulKorea
| | - Jinah Han
- BioDrone Therapeutics Inc.SeattleUSA
| | - Mario Gimona
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
- Transfer Centre for Extracellular Vesicle Theralytic Technologies (EV‐TT)SalzburgAustria
- Research Program “Nanovesicular Therapies”Paracelsus Medical UniversitySalzburgAustria
| | - Eva Rohde
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
- Department of Transfusion Medicine, University HospitalSalzburger Landeskliniken GesmbH (SALK) and Paracelsus Medical UniversitySalzburgAustria
| | - Shingyu Bae
- BioDrone Research InstituteMDimune Inc.SeoulKorea
| | - Seung Wook Oh
- BioDrone Research InstituteMDimune Inc.SeoulKorea
- BioDrone Therapeutics Inc.SeattleUSA
| |
Collapse
|
126
|
Baranovskii DS, Klabukov ID, Arguchinskaya NV, Yakimova AO, Kisel AA, Yatsenko EM, Ivanov SA, Shegay PV, Kaprin AD. Adverse events, side effects and complications in mesenchymal stromal cell-based therapies. Stem Cell Investig 2022; 9:7. [PMID: 36393919 PMCID: PMC9659480 DOI: 10.21037/sci-2022-025] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/28/2022] [Indexed: 07/22/2023]
Abstract
Numerous clinical studies have shown a wide clinical potential of mesenchymal stromal cells (MSCs) application. However, recent experience has accumulated numerous reports of adverse events and side effects associated with MSCs therapy. Furthermore, the strategies and methods of MSCs therapy did not change significantly in recent decades despite the clinical impact and awareness of potential complications. An extended understanding of limitations could lead to a wider clinical implementation of safe cell therapies and avoid harmful approaches. Therefore, our objective was to summarize the possible negative effects observed during MSCs-based therapies. We were also aimed to discuss the risks caused by weaknesses in cell processing, including isolation, culturing, and storage. Cell processing and cell culture could dramatically influence cell population profile, change protein expression and cell differentiation paving the way for future negative effects. Long-term cell culture led to accumulation of chromosomal abnormalities. Overdosed antibiotics in culture media enhanced the risk of mycoplasma contamination. Clinical trials reported thromboembolism and fibrosis as the most common adverse events of MSCs therapy. Their delayed manifestation generally depends on the patient's individual phenotype and requires specific awareness during the clinical trials with obligatory inclusion in the patient' informed consents. Finally we prepared the safety checklist, recommended for clinical specialists before administration or planning of MSCs therapy.
Collapse
Affiliation(s)
- Denis S. Baranovskii
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
- Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
| | - Ilya D. Klabukov
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
- Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
- Obninsk Institute for Nuclear Power Engineering of the National Research Nuclear University MEPhI, Obninsk, Russia
| | - Nadezhda V. Arguchinskaya
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Anna O. Yakimova
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Anastas A. Kisel
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Elena M. Yatsenko
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Sergei A. Ivanov
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Peter V. Shegay
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Andrey D. Kaprin
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
- Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
| |
Collapse
|
127
|
Chetty S, Yarani R, Swaminathan G, Primavera R, Regmi S, Rai S, Zhong J, Ganguly A, Thakor AS. Umbilical cord mesenchymal stromal cells-from bench to bedside. Front Cell Dev Biol 2022; 10:1006295. [PMID: 36313578 PMCID: PMC9597686 DOI: 10.3389/fcell.2022.1006295] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/27/2022] [Indexed: 11/27/2022] Open
Abstract
In recent years, mesenchymal stromal cells (MSCs) have generated a lot of attention due to their paracrine and immuno-modulatory properties. mesenchymal stromal cells derived from the umbilical cord (UC) are becoming increasingly recognized as having increased therapeutic potential when compared to mesenchymal stromal cells from other sources. The purpose of this review is to provide an overview of the various compartments of umbilical cord tissue from which mesenchymal stromal cells can be isolated, the differences and similarities with respect to their regenerative and immuno-modulatory properties, as well as the single cell transcriptomic profiles of in vitro expanded and freshly isolated umbilical cord-mesenchymal stromal cells. In addition, we discuss the therapeutic potential and biodistribution of umbilical cord-mesenchymal stromal cells following systemic administration while providing an overview of pre-clinical and clinical trials involving umbilical cord-mesenchymal stromal cells and their associated secretome and extracellular vesicles (EVs). The clinical applications of umbilical cord-mesenchymal stromal cells are also discussed, especially in relation to obstacles and potential solutions for their effective translation from bench to bedside.
Collapse
Affiliation(s)
- Shashank Chetty
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
| | - Reza Yarani
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
- Translational Type 1 Diabetes Research, Department of Clinical, Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Ganesh Swaminathan
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
| | - Rosita Primavera
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
| | - Shobha Regmi
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
| | - Sravanthi Rai
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
| | - Jim Zhong
- Department of Diagnostic and Interventional Radiology, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Abantika Ganguly
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
| | - Avnesh S Thakor
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
| |
Collapse
|
128
|
Maeda S, Kawamura T, Sasaki M, Shimamura K, Shibuya T, Harada A, Honmou O, Sawa Y, Miyagawa S. Intravenous infusion of bone marrow-derived mesenchymal stem cells improves tissue perfusion in a rat hindlimb ischemia model. Sci Rep 2022; 12:16986. [PMID: 36216855 PMCID: PMC9551049 DOI: 10.1038/s41598-022-18485-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 08/12/2022] [Indexed: 12/29/2022] Open
Abstract
Intravenous infusion of stem cells is a minimally invasive cellular delivery method, though a few have been reported in a critical limb-threatening ischemia (CLTI) animal model or patients. In the present study, we hypothesized that intravenous infusion of bone-marrow derived mesenchymal stem cells (MSCs) improves tissue perfusion in a rat hindlimb ischemia model. Hindlimb ischemia was generated in Sprague-Dawley rats by femoral artery removal, then seven days after ischemic induction intravenous infusion of 1 × 106 MSCs (cell group) or vehicle (control group) was performed. As compared with the control, tissue perfusion was significantly increased in the cell group. Histological findings showed that capillary density was significantly increased in the cell group, with infused green fluorescent protein (GFP)-MSCs distributed in the ischemic limb. Furthermore, gene expression of vascular endothelial growth factor (VEGF) was significantly increased in ischemic hindlimb muscle tissues of rats treated with MSC infusion. In conclusion, intravenous infusion of bone-marrow derived MSCs improved tissue perfusion in ischemic hindlimbs through angiogenesis, suggesting that intravenous infusion of MSCs was a promising cell delivery method for treatment of CLTI.
Collapse
Affiliation(s)
- Shusaku Maeda
- grid.136593.b0000 0004 0373 3971Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Takuji Kawamura
- grid.136593.b0000 0004 0373 3971Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Masanori Sasaki
- grid.263171.00000 0001 0691 0855Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido Japan
| | - Kazuo Shimamura
- grid.136593.b0000 0004 0373 3971Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Takashi Shibuya
- grid.136593.b0000 0004 0373 3971Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Akima Harada
- grid.136593.b0000 0004 0373 3971Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Osamu Honmou
- grid.263171.00000 0001 0691 0855Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido Japan
| | - Yoshiki Sawa
- grid.136593.b0000 0004 0373 3971Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Shigeru Miyagawa
- grid.136593.b0000 0004 0373 3971Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita, Osaka 565-0871 Japan
| |
Collapse
|
129
|
Csöbönyeiová M, Beerová N, Klein M, Debreová-Čeháková M, Danišovič Ľ. Cell-Based and Selected Cell-Free Therapies for Myocardial Infarction: How Do They Compare to the Current Treatment Options? Int J Mol Sci 2022; 23:10314. [PMID: 36142245 PMCID: PMC9499607 DOI: 10.3390/ijms231810314] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/03/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Because of cardiomyocyte death or dysfunction frequently caused by myocardial infarction (MI), heart failure is a leading cause of morbidity and mortality in modern society. Paradoxically, only limited and non-curative therapies for heart failure or MI are currently available. As a result, over the past two decades research has focused on developing cell-based approaches promoting the regeneration of infarcted tissue. Cell-based therapies for myocardial regeneration include powerful candidates, such as multipotent stem cells (mesenchymal stem cells (MSCs), bone-marrow-derived stem cells, endothelial progenitor cells, and hematopoietic stem cells) and induced pluripotent stem cells (iPSCs). These possess unique properties, such as potency to differentiate into desired cell types, proliferation capacity, and patient specificity. Preclinical and clinical studies have demonstrated modest improvement in the myocardial regeneration and reduced infarcted areas upon transplantation of pluripotent or multipotent stem cells. Another cell population that need to be considered as a potential source for cardiac regeneration are telocytes found in different organs, including the heart. Their therapeutic effect has been studied in various heart pathologies, such as MI, arrhythmias, or atrial amyloidosis. The most recent cell-free therapeutic tool relies on the cardioprotective effect of complex cargo carried by small membrane-bound vesicles-exosomes-released from stem cells via exocytosis. The MSC/iPSC-derived exosomes could be considered a novel exosome-based therapy for cardiovascular diseases thanks to their unique content. There are also other cell-free approaches, e.g., gene therapy, or acellular cardiac patches. Therefore, our review provides the most recent insights into the novel strategies for myocardial repair based on the regenerative potential of different cell types and cell-free approaches.
Collapse
Affiliation(s)
- Mária Csöbönyeiová
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Nikoleta Beerová
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Martin Klein
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Michaela Debreová-Čeháková
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Ľuboš Danišovič
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| |
Collapse
|
130
|
Feng H, Liu Q, Deng Z, Li H, Zhang H, Song J, Liu X, Liu J, Wen B, Wang T. Human umbilical cord mesenchymal stem cells ameliorate erectile dysfunction in rats with diabetes mellitus through the attenuation of ferroptosis. Stem Cell Res Ther 2022; 13:450. [PMID: 36064453 PMCID: PMC9444126 DOI: 10.1186/s13287-022-03147-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/18/2022] [Indexed: 11/15/2022] Open
Abstract
Background Erectile dysfunction (ED), as one of the most prevalent consequences in male diabetic patients, has a serious impact on men's physical and mental health, and the treatment effect of diabetic mellitus erectile dysfunction (DMED) is often worse. Therefore, the development of a novel therapeutic approach is urgent. As stem cells with high differentiation potential, human umbilical cord mesenchymal stem cells (HUCMSCs) have been widely used in the treatment of diseases in other systems, and are expected to be a promising strategy for the treatment of DMED. In this study, we investigated the role of HUCMSCs in managing erectile function in rat models of type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM) and compared the effects of two different injection methods. Methods T1DM and T2DM ED rats were given labelled HUCMSCs by corpus cavernosum injection and tail vein injection, respectively. ICP and MAP were monitored simultaneously by electrical stimulation four weeks after injection to indicate the erectile function of rats. To track the development and colonisation capabilities of stem cells, we performed EdU assay with penile tissue. The histological changes of the penis were observed by hematoxylin–eosin staining, and Masson’s trichrome staining was conducted to evaluate the smooth muscle content and the degree of fibrosis in the rat penis. Then, we employed specific kits to measure the level of NO, cGMP, MDA, SOD and Fe in penis. Electron transmission microscopy was implemented to observe morphology of mitochondria. Besides, western blot and immunofluorescence staining were performed to demonstrate the expression of ferroptosis-related genes. Results We found that HUCMSCs improved erectile function in T1DM and T2DM ED rats, with no difference in efficacy between corpus cavernosum injection and tail vein injection. The EdU assay revealed that only a tiny percentage of HUCMSCs colonised the corpus cavernosum, while smooth muscle in the penis expanded and collagen decreased following HUCMSC injection. Moreover, the levels of oxidative stress in the penis of the rats given HUCMSCs were dramatically reduced, as was the tissue iron content. HUCMSCs normalised mitochondrial morphology within corpus cavernosum smooth muscle cells (CCSMCs), which were characteristically altered by high glucose. Furthermore, the expression of ferroptosis inhibitory genes SLC7A11 and GPX4 was obviously elevated in CCSMCs after stem cell management, but the abundances of ACSL4, LPCAT3 and ALOX15 showed the polar opposite tendency. Conclusions HUCMSCs can effectively and safely alleviate erectile dysfunction in T1DM and T2DM ED rats, while restoring erectile function by attenuating diabetes-induced ferroptosis in CCSMCs. Additionally, this study provides significant evidence for the development of HUCMSCs as a viable therapeutic strategy for DMED. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03147-w.
Collapse
Affiliation(s)
- Huan Feng
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qi Liu
- Department of Urology, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, China
| | - Zhiyao Deng
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, Guangdong, China
| | - Hao Li
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huajie Zhang
- Department of Urology, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, China
| | - Jingyu Song
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaming Liu
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jihong Liu
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bo Wen
- Department of Urology, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, China.
| | - Tao Wang
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China. .,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, Guangdong, China.
| |
Collapse
|
131
|
Wang P, Deng Z, Li A, Li R, Huang W, Cui J, Chen S, Li B, Zhang S. β-Catenin promotes long-term survival and angiogenesis of peripheral blood mesenchymal stem cells via the Oct4 signaling pathway. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1434-1449. [PMID: 36050404 PMCID: PMC9535028 DOI: 10.1038/s12276-022-00839-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 04/06/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022]
Abstract
Stem cell therapy has been extensively studied to improve heart function following myocardial infarction; however, its therapeutic potency is limited by low rates of engraftment, survival, and differentiation. Here, we aimed to determine the roles of the β-catenin/Oct4 signaling axis in the regulation of long-term survival and angiogenesis of peripheral blood mesenchymal stem cells (PBMSCs). These cells were obtained from rat abdominal aortic blood. We showed that β-catenin promotes the self-renewal, antiapoptotic effects, and long-term survival of PBMSCs by activating the Oct4 pathway through upregulation of the expression of the antiapoptotic factors Bcl2 and survivin and the proangiogenic cytokine bFGF and suppression of the levels of the proapoptotic factors Bax and cleaved caspase-3. β-Catenin overexpression increased Oct4 expression. β-Catenin knockdown suppressed Oct4 expression in PBMSCs. However, β-catenin levels were not affected by Oct4 overexpression or knockdown. Chromatin immunoprecipitation assays proved that β-catenin directly regulates Oct4 transcription in PBMSCs. In vivo, PBMSCs overexpressing β-catenin showed high survival in infarcted hearts and resulted in better myocardial repair. Further functional analysis identified Oct4 as the direct upstream regulator of Ang1, bFGF, HGF, VEGF, Bcl2, and survivin, which cooperatively drive antiapoptosis and angiogenesis of engrafted PBMSCs. These findings revealed the regulation of β-catenin in PBMSCs by the Oct4-mediated antiapoptotic/proangiogenic signaling axis and provide a breakthrough point for improving the long-term survival and therapeutic effects of PBMSCs. Boosting expression of a specific gene has allowed researchers to generate stem cells with increased capacity for tissue repair after a heart attack. Several studies have shown that treatment with a population of circulating cells known as ‘peripheral blood mesenchymal stem cells’ (PBMSCs) can regenerate cardiac tissue. These cells generally have a short lifespan when used therapeutically, but researchers led by Shaoheng Zhang at Jinan University in Guangzhou China have increased long-term survival and performance by boosting expression of the gene encoding β-catenin, a protein that promotes cell survival and proliferation. PBMSCs expressing increased levels of β-catenin preserved heart function in a rat model of heart attack, stimulating blood vessel growth and improving animal survival. This study also reveals proteins regulated by β-catenin, which could potentially be exploited for finer control of PBMSC function.
Collapse
Affiliation(s)
- Pengzhen Wang
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China.,Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Zhanyu Deng
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Aiguo Li
- Department of Orthopaedics, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Rongsen Li
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Weiguang Huang
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Jin Cui
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Songsheng Chen
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Biao Li
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Shaoheng Zhang
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China.
| |
Collapse
|
132
|
Jedrzejewska A, Braczko A, Kawecka A, Hellmann M, Siondalski P, Slominska E, Kutryb-Zajac B, Yacoub MH, Smolenski RT. Novel Targets for a Combination of Mechanical Unloading with Pharmacotherapy in Advanced Heart Failure. Int J Mol Sci 2022; 23:9886. [PMID: 36077285 PMCID: PMC9456495 DOI: 10.3390/ijms23179886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 12/19/2022] Open
Abstract
LVAD therapy is an effective rescue in acute and especially chronic cardiac failure. In several scenarios, it provides a platform for regeneration and sustained myocardial recovery. While unloading seems to be a key element, pharmacotherapy may provide powerful tools to enhance effective cardiac regeneration. The synergy between LVAD support and medical agents may ensure satisfying outcomes on cardiomyocyte recovery followed by improved quality and quantity of patient life. This review summarizes the previous and contemporary strategies for combining LVAD with pharmacotherapy and proposes new therapeutic targets. Regulation of metabolic pathways, enhancing mitochondrial biogenesis and function, immunomodulating treatment, and stem-cell therapies represent therapeutic areas that require further experimental and clinical studies on their effectiveness in combination with mechanical unloading.
Collapse
Affiliation(s)
- Agata Jedrzejewska
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 Street, 80-211 Gdansk, Poland
| | - Alicja Braczko
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 Street, 80-211 Gdansk, Poland
| | - Ada Kawecka
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 Street, 80-211 Gdansk, Poland
| | - Marcin Hellmann
- Department of Cardiac Diagnostics, Medical University of Gdansk, Smoluchowskiego 17, 80-214 Gdansk, Poland
| | - Piotr Siondalski
- Department of Cardiac Surgery, Medical University of Gdansk, Debinki 7 Street, 80-211 Gdansk, Poland
| | - Ewa Slominska
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 Street, 80-211 Gdansk, Poland
| | - Barbara Kutryb-Zajac
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 Street, 80-211 Gdansk, Poland
| | - Magdi H. Yacoub
- Heart Science Centre, Imperial College of London at Harefield Hospital, Harefield UB9 6JH, UK
| | - Ryszard T. Smolenski
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 Street, 80-211 Gdansk, Poland
| |
Collapse
|
133
|
Zhang YU, Ye G, Zhao J, Chen Y, Kong L, Sheng C, Yuan L. Exosomes carried miR-181c-5p alleviates neuropathic pain in CCI rat models. AN ACAD BRAS CIENC 2022; 94:e20210564. [PMID: 35976364 DOI: 10.1590/0001-3765202220210564] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 10/20/2021] [Indexed: 11/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) derived exosomes (Exos) are one of the most promising candidate for the treatment of this condition. However, the underlying molecular mechanism remains uncertain. Here we investigated the therapeutic effect of exosomal miR-181c-5p (ExomiR-181c-5p) on a rat model of neuropathic pain induced by sciatic nerve chronic constriction injury (CCI). In this study NP model was established using the CCI method. NP levels were assessed using PWT and PWL. Microarray analysis and RT-PCR were used to determine the relative expression of miR-181c-5p. MSC-derived exosomes were extracted using the total exosome isolation reagent characterized by WB and NTA. MiR-181c-5p was loading into Exos using electroporation. The inflammation response in microglia cells and CCI rats were assessed by ELISA assay respectively. Our study demonstrates that miR-181c-5p expression was obviously decreased in a time-dependent manner in CCI rats. MiR-181c-5p was effectively electroporated and highly detected in MSC-derived Exos. ExomiR-181c-5p internalized by microglia cells and inhibit the secretion of inflammation factors. ExomiR-181c-5p intrathecal administration alleviated neuropathic pain and neuroinflammation response in CCI rats. Taken together, ExomiR-181c-5p alleviated CCI-induced NP by inhibiting neuropathic inflammation. ExomiR-181c-5p may be a valid alternative for the treatment of neuropathic pain and has vast potential for future development.
Collapse
Affiliation(s)
- Y U Zhang
- Department of Anesthesiology, Ningbo No.6 Hospital, Ningbo 315040, Zhejiang, PR China
| | - Guangyao Ye
- Department of Anesthesiology, Ningbo No.6 Hospital, Ningbo 315040, Zhejiang, PR China
| | - Jingsong Zhao
- Department of Anesthesiology, Ningbo No.6 Hospital, Ningbo 315040, Zhejiang, PR China
| | - Yuebo Chen
- Department of Anesthesiology, Ningbo No.6 Hospital, Ningbo 315040, Zhejiang, PR China
| | - Lingsi Kong
- Department of Anesthesiology, Ningbo No.6 Hospital, Ningbo 315040, Zhejiang, PR China
| | - Chaoxu Sheng
- Department of Anesthesiology, Ningbo No.6 Hospital, Ningbo 315040, Zhejiang, PR China
| | - Liyong Yuan
- Department of Anesthesiology, Ningbo No.6 Hospital, Ningbo 315040, Zhejiang, PR China
| |
Collapse
|
134
|
Wang Y, Zhang Y, Chen K, Liu J, Wu D, Cheng Y, Wang H, Li Y. Insufficient S-adenosylhomocysteine hydrolase compromises the beneficial effect of diabetic BMSCs on diabetic cardiomyopathy. Stem Cell Res Ther 2022; 13:418. [PMID: 35964109 PMCID: PMC9375418 DOI: 10.1186/s13287-022-03099-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/27/2022] [Indexed: 11/23/2022] Open
Abstract
Background Autologous stem cell therapy is a promising strategy for cardiovascular diseases including diabetic cardiomyopathy (DCM), but conclusions from clinical trials were compromised. We assumed that diabetes might induce the dysfunction of stem cells and thus limit its therapeutic effect. This study aimed to compare the effect of diabetes and nondiabetes-derived bone marrow mesenchymal stem cells (BMSCs) transplantation on DCM and explored the potential mechanism. Methods Rats with diabetes were induced using high-fat diets and streptozotocin (STZ) injection. BMSCs harvested from diabetic and nondiabetic rats were infused into DCM rats, and the effects on the heart were identified by echocardiography and histopathology. The inhibition or overexpression of SAHH in nondiabetic and diabetic BMSCs was used to confirm its key role in stem cell activity and cardiac therapy. Results Compared with normal BMSCs, the therapeutic effects of diabetic rat-derived stem cells on improving cardiac function and adverse remodeling were significantly attenuated. In vitro, diabetic BMSCs had lower cell viability and paracrine function than nondiabetic BMSCs. It was further found that diabetic BMSCs had obvious mitochondrial oxidative stress damage and S-adenosylhomocysteine (SAH) accumulation due to S-adenosylhomocysteine hydrolase (SAHH) deficiency. SAHH inhibition by adenosine dialdehyde (ADA) or shSAHH plasmid in normal BMSCs significantly reduced the favorable effects on endothelial cell proliferation and tube-forming capacity. In contrast, SAHH overexpression in diabetic BMSCs significantly improved cellular activity and paracrine function. Transplantation of BMSCs with SAHH overexpression improved cardiac adverse remodeling and angiogenesis. Activation of the Nrf2 signaling pathway may be one of the key mechanisms of SAHH-mediated improvement of stem cell viability and cardiac repair. Conclusions Diabetes leads to compromised bioactivity and repair capacity of BMSCs. Our study suggests that SAHH activation may improve the cardioprotective effect of autologous transplantation of diabetes-derived BMSCs on patients with DCM. Graphical abstract Diabetes induced the inhibition of S-adenosylhomocysteine (SAH) expression and aging phenotype in BMSCs and thus decreased the cell viability and paracrine function. Compared with normal BMSCs, the therapeutic effects of diabetic rat-derived BMSCs on improving cardiac function and adverse remodeling were significantly attenuated. SAHH overexpression in diabetic BMSCs significantly rescued cellular function partly via activating Nrf2/HO-1 signal. Transplantation of diabetic BMSCs with SAHH overexpression improved angiogenesis and cardiac adverse remodeling in rats.![]() Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03099-1.
Collapse
Affiliation(s)
- Ying Wang
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China.,Department of Endocrinology, First Hospital of Harbin, Harbin, People's Republic of China
| | - Yuying Zhang
- Department of Pathology, First Hospital of Harbin, Harbin, People's Republic of China
| | - Kegong Chen
- Department of Thoracic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Future Medical Laboratory, Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Jie Liu
- Department of Endocrinology, First Hospital of Harbin, Harbin, People's Republic of China
| | - Donghong Wu
- Department of Endocrinology, First Hospital of Harbin, Harbin, People's Republic of China
| | - Yao Cheng
- Department of Endocrinology, First Hospital of Harbin, Harbin, People's Republic of China
| | - Hongjie Wang
- Department of Endocrinology, Forth Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Yanbo Li
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China. .,Department of Endocrinology, South China Hospital of Shenzhen University, No. 1 Fuxin Road, Longgang District, Shenzhen, 518116, People's Republic of China.
| |
Collapse
|
135
|
Gong C, Chang L, Sun X, Qi Y, Huang R, Chen K, Wang B, Kang L, Wang L, Xu B. Infusion of two-dose mesenchymal stem cells is more effective than a single dose in a dilated cardiomyopathy rat model by upregulating indoleamine 2,3-dioxygenase expression. Stem Cell Res Ther 2022; 13:409. [PMID: 35962420 PMCID: PMC9373305 DOI: 10.1186/s13287-022-03101-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/28/2022] [Indexed: 11/10/2022] Open
Abstract
Background and aims The therapeutic efficacy of single-dose mesenchymal stromal cell (MSC) therapy for heart failure (HF) remains inconsistent. This study aimed to investigate whether infusion with two-dose human umbilical cord MSC (hUCMSCs) could be therapeutically superior to single-dose therapy in a rat model of dilated cardiomyopathy (DCM) and explored the underlying mechanisms. Methods Male Sprague–Dawley rats were intraperitoneally injected with doxorubicin (DOX) to establish a DCM model and randomized to intravenously receive single-dose or two-dose hUCMSCs at an interval of 14 days. Their left ventricular (LV) systolic and diastolic functions were analyzed by echocardiography. The percentages of Th1, Th2, Th17, and Treg cells in the heart, spleen, lymph nodes, and peripheral blood and the levels of serum cytokines in individual rats were analyzed by flow cytometry and cytometric bead assay, respectively. The degrees of cardiac fibrosis and cardiomyocyte apoptosis were examined by histology. The importance of indoleamine 2,3-dioxygenase (IDO), an activator of Treg differentiation, in the therapeutic effect of hUCMSCs on inflammation and heart function of rats was determined after induction of IDO over-expression (IDO-OE) using IFN-γ (1 ng/ml) and TNF-α (10 ng/ml) stimulation or silencing (IDO-KD) using small interfering RNA (siRNA) technology. Results Compared with the single dose, two-dose hUCMSCs were more effective in improving LV performance, attenuating cardiac dilation, reducing cardiomyocyte apoptosis and cardiac fibrosis. Two-dose hUCMSC therapy significantly increased Treg number in the heart and peripheral blood, accompanied by increased cardiac IDO expression. Compared with the control hUCMSCs, IDO-OE hUCMSCs significantly enhanced Treg and Th2 cell responses and decreased systemic Th17 cell responses and Th1 cell numbers in the mediastinal lymph nodes. Treatment with IDO-OE hUCMSCs significantly improved LV remodeling and dysfunction. However, treatment with IDO-KD hUCMSCs had opposite effects in rats. Conclusions Administration of two-dose hUCMSCs has better therapeutic effects than single-dose therapy for inhibiting myocardial inflammation to improve LV function in DCM rats. These effects are associated with upregulating IDO expression and its systemic anti-inflammatory activities. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03101-w.
Collapse
Affiliation(s)
- Chenyi Gong
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Lei Chang
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China
| | - Xuan Sun
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Yu Qi
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Rong Huang
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Ke Chen
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Bin Wang
- Clinical Stem Cell Centor, Nanjing Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Lina Kang
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Lian Wang
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China. .,Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China.
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China. .,Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
136
|
Hsiao LC, Lin YN, Shyu WC, Ho M, Lu CR, Chang SS, Wang YC, Chen JY, Lu SY, Wu MY, Li KY, Lin YK, Tseng WYI, Su MY, Hsu CT, Tsai CK, Chiu LT, Chen CL, Lin CL, Hu KC, Cho DY, Tsai CH, Chang KC, Jeng LB. First-in-human pilot trial of combined intracoronary and intravenous mesenchymal stem cell therapy in acute myocardial infarction. Front Cardiovasc Med 2022; 9:961920. [PMID: 36017096 PMCID: PMC9395611 DOI: 10.3389/fcvm.2022.961920] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 07/13/2022] [Indexed: 12/17/2022] Open
Abstract
BackgroundAcute ST-elevation myocardial infarction (STEMI) elicits a robust cardiomyocyte death and inflammatory responses despite timely revascularization.ObjectivesThis phase 1, open-label, single-arm, first-in-human study aimed to assess the safety and efficacy of combined intracoronary (IC) and intravenous (IV) transplantation of umbilical cord-derived mesenchymal stem cells (UMSC01) for heart repair in STEMI patients with impaired left ventricular ejection fraction (LVEF 30-49%) following successful reperfusion by percutaneous coronary intervention.MethodsConsenting patients received the first dose of UMSC01 through IC injection 4-5 days after STEMI followed by the second dose of UMSC01 via IV infusion 2 days later. The primary endpoint was occurrence of any treatment-related adverse events and the secondary endpoint was changes of serum biomarkers and heart function by cardiac magnetic resonance imaging during a 12-month follow-up period.ResultsEight patients gave informed consents, of whom six completed the study. None of the subjects experienced treatment-related serious adverse events or major adverse cardiovascular events during IC or IV infusion of UMSC01 and during the follow-up period. The NT-proBNP level decreased (1362 ± 1801 vs. 109 ± 115 pg/mL, p = 0.0313), the LVEF increased (52.67 ± 12.75% vs. 62.47 ± 17.35%, p = 0.0246), and the wall motion score decreased (26.33 ± 5.57 vs. 22.33 ± 5.85, p = 0.0180) at the 12-month follow-up compared to the baseline values. The serial changes of LVEF were 0.67 ± 3.98, 8.09 ± 6.18, 9.04 ± 10.91, and 9.80 ± 7.56 at 1, 3, 6, and 12 months, respectively as compared to the baseline.ConclusionThis pilot study shows that combined IC and IV transplantation of UMSC01 in STEMI patients with impaired LVEF appears to be safe, feasible, and potentially beneficial in improving heart function. Further phase 2 studies are required to explore the effectiveness of dual-route transplantation of UMSC01 in STEMI patients.
Collapse
Affiliation(s)
- Lien-Cheng Hsiao
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Yen-Nien Lin
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Woei-Cherng Shyu
- Department of Neurology and Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
- Ever Supreme Bio Technology Co., Ltd, Taichung, Taiwan
| | - Ming Ho
- Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Chiung-Ray Lu
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Shih-Sheng Chang
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Yu-Chen Wang
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Jan-Yow Chen
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Shang-Yeh Lu
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Mei-Yao Wu
- School of Post-Baccalaureate Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Keng-Yuan Li
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Yu-Kai Lin
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Yih I. Tseng
- Molecular Imaging Center, National Taiwan University, Taipei, Taiwan
| | - Mao-Yuan Su
- Molecular Imaging Center, National Taiwan University, Taipei, Taiwan
| | - Chin-Ting Hsu
- Ever Supreme Bio Technology Co., Ltd, Taichung, Taiwan
| | | | - Lu-Ting Chiu
- Ever Supreme Bio Technology Co., Ltd, Taichung, Taiwan
| | | | - Cheng-Li Lin
- School of Medicine, China Medical University, Taichung, Taiwan
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Kai-Chieh Hu
- School of Medicine, China Medical University, Taichung, Taiwan
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Der-Yang Cho
- School of Medicine, China Medical University, Taichung, Taiwan
- Department of Neurosurgery, China Medical University Hospital, Taichung, Taiwan
| | - Chang-Hai Tsai
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Kuan-Cheng Chang
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
- *Correspondence: Kuan-Cheng Chang,
| | - Long-Bin Jeng
- School of Medicine, China Medical University, Taichung, Taiwan
- Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan
- Long-Bin Jeng,
| |
Collapse
|
137
|
Hoang DM, Pham PT, Bach TQ, Ngo ATL, Nguyen QT, Phan TTK, Nguyen GH, Le PTT, Hoang VT, Forsyth NR, Heke M, Nguyen LT. Stem cell-based therapy for human diseases. Signal Transduct Target Ther 2022; 7:272. [PMID: 35933430 PMCID: PMC9357075 DOI: 10.1038/s41392-022-01134-4] [Citation(s) in RCA: 451] [Impact Index Per Article: 150.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 02/07/2023] Open
Abstract
Recent advancements in stem cell technology open a new door for patients suffering from diseases and disorders that have yet to be treated. Stem cell-based therapy, including human pluripotent stem cells (hPSCs) and multipotent mesenchymal stem cells (MSCs), has recently emerged as a key player in regenerative medicine. hPSCs are defined as self-renewable cell types conferring the ability to differentiate into various cellular phenotypes of the human body, including three germ layers. MSCs are multipotent progenitor cells possessing self-renewal ability (limited in vitro) and differentiation potential into mesenchymal lineages, according to the International Society for Cell and Gene Therapy (ISCT). This review provides an update on recent clinical applications using either hPSCs or MSCs derived from bone marrow (BM), adipose tissue (AT), or the umbilical cord (UC) for the treatment of human diseases, including neurological disorders, pulmonary dysfunctions, metabolic/endocrine-related diseases, reproductive disorders, skin burns, and cardiovascular conditions. Moreover, we discuss our own clinical trial experiences on targeted therapies using MSCs in a clinical setting, and we propose and discuss the MSC tissue origin concept and how MSC origin may contribute to the role of MSCs in downstream applications, with the ultimate objective of facilitating translational research in regenerative medicine into clinical applications. The mechanisms discussed here support the proposed hypothesis that BM-MSCs are potentially good candidates for brain and spinal cord injury treatment, AT-MSCs are potentially good candidates for reproductive disorder treatment and skin regeneration, and UC-MSCs are potentially good candidates for pulmonary disease and acute respiratory distress syndrome treatment.
Collapse
Affiliation(s)
- Duc M Hoang
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam.
| | - Phuong T Pham
- Department of Cellular Therapy, Vinmec High-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
| | - Trung Q Bach
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Anh T L Ngo
- Department of Cellular Therapy, Vinmec High-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
| | - Quyen T Nguyen
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Trang T K Phan
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Giang H Nguyen
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Phuong T T Le
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Van T Hoang
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Nicholas R Forsyth
- Institute for Science & Technology in Medicine, Keele University, Keele, UK
| | - Michael Heke
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Liem Thanh Nguyen
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| |
Collapse
|
138
|
Human Mesenchymal Stem Cell Sheets Improve Uterine Incision Repair in a Rodent Hysterotomy Model. Am J Perinatol 2022; 39:1212-1222. [PMID: 33368093 DOI: 10.1055/s-0040-1721718] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE The study aimed to assess the feasibility of creating and transplanting human umbilical cord mesenchymal stem cell sheets applied to a rat model of hysterotomy, and additionally to determine benefits of human umbilical cord mesenchymal stem cell sheet transplantation in reducing uterine fibrosis and scarring. STUDY DESIGN Human umbilical cord mesenchymal stem cell sheets are generated by culturing human umbilical cord mesenchymal stem cells on thermo-responsive cell culture plates. The temperature-sensitive property of these culture dishes facilitates normal cell culture in a thin contiguous layer and allows for reliable recovery of intact stem cell sheets without use of destructive proteolytic enzymes.We developed a rat hysterotomy model using nude rats. The rat uterus has two distinct horns: one horn provided a control/untreated scarring site, while the second horn was the cell sheet transplantation site.On day 14 following surgery, complete uteri were harvested and subjected to histologic evaluations of all hysterotomy sites. RESULTS The stem cell sheet culture process yielded human umbilical cord mesenchymal stem cell sheets with surface area of approximately 1 cm2.Mean myometrial thickness in the cell sheet-transplanted group was 274 μm compared with 191 μm in the control group (p = 0.02). Mean fibrotic surface area in the human umbilical cord mesenchymal stem cell sheet-transplanted group was 95,861 μm2 compared with 129,185 μm2 in the control group. Compared with control horn sites, cell sheet-transplanted horns exhibited significantly smaller fibrotic-to-normal myometrium ratios (0.18 vs. 0.27, respectively, p = 0.029). Mean number of fibroblasts in cell sheet-transplanted horns was significantly smaller than the control horns (483 vs. 716/mm2, respectively, p = 0.001). CONCLUSION Human umbilical cord mesenchymal stem cell sheet transplantation is feasible in a rat model of hysterotomy. Furthermore, use of stem cell sheets reduces fibroblast infiltration and uterine scar fibrotic tissue formation during hysterotomy healing, potentially mitigating risks of uterine scar formation. KEY POINTS · Stem cell sheet transplanted to hysterotomy promotes myometrial regeneration and reduced fibrotic tissue formation.. · This study demonstrates the feasibility of using human umbilical cord mesenchymal stem cell sheets..
Collapse
|
139
|
Margiana R, Markov A, Zekiy AO, Hamza MU, Al-Dabbagh KA, Al-Zubaidi SH, Hameed NM, Ahmad I, Sivaraman R, Kzar HH, Al-Gazally ME, Mustafa YF, Siahmansouri H. Clinical application of mesenchymal stem cell in regenerative medicine: a narrative review. Stem Cell Res Ther 2022; 13:366. [PMID: 35902958 PMCID: PMC9330677 DOI: 10.1186/s13287-022-03054-0] [Citation(s) in RCA: 180] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/18/2022] [Indexed: 12/16/2022] Open
Abstract
The multipotency property of mesenchymal stem cells (MSCs) has attained worldwide consideration because of their immense potential for immunomodulation and their therapeutic function in tissue regeneration. MSCs can migrate to tissue injury areas to contribute to immune modulation, secrete anti-inflammatory cytokines and hide themselves from the immune system. Certainly, various investigations have revealed anti-inflammatory, anti-aging, reconstruction, and wound healing potentials of MSCs in many in vitro and in vivo models. Moreover, current progresses in the field of MSCs biology have facilitated the progress of particular guidelines and quality control approaches, which eventually lead to clinical application of MSCs. In this literature, we provided a brief overview of immunoregulatory characteristics and immunosuppressive activities of MSCs. In addition, we discussed the enhancement, utilization, and therapeutic responses of MSCs in neural, liver, kidney, bone, heart diseases, and wound healing.
Collapse
Affiliation(s)
- Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Alexander Markov
- Tyumen State Medical University, Tyumen, Russian Federation.,Tyumen Industrial University, Tyumen, Russian Federation
| | - Angelina O Zekiy
- Department of Prosthetic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | | | | | - Noora M Hameed
- Anesthesia Techniques, Al-Nisour University College, Baghdad, Iraq
| | - Irshad Ahmad
- Department of Medical Rehabilitation Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - R Sivaraman
- Department of Mathematics, Dwaraka Doss Goverdhan Doss Vaishnav College, Arumbakkam, University of Madras, Chennai, India
| | - Hamzah H Kzar
- Veterinary Medicine College, Al-Qasim Green University, Al-Qasim, Iraq
| | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Homayoon Siahmansouri
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
140
|
Li T, Zhou L, Fan M, Chen Z, Yan L, Lu H, Jia M, Wu H, Shan L. Human Umbilical Cord-Derived Mesenchymal Stem Cells Ameliorate Skin Aging of Nude Mice Through Autophagy-Mediated Anti-Senescent Mechanism. Stem Cell Rev Rep 2022; 18:2088-2103. [PMID: 35864432 DOI: 10.1007/s12015-022-10418-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2022] [Indexed: 10/17/2022]
Abstract
Skin aging is a currently irreversible process, affected by increased oxidative stress, activated cellular senescence, and lacked regeneration of the dermal layer. Mesenchymal stem cells (MSCs), such as human umbilical cord-derived MSCs (hucMSCs), have pro-regeneration and anti-aging potencies. To explore whether hucMSCs can be used to treat skin aging, this study employed skin-aging model of nude mice to conduct in vivo assays, including biochemical analysis of superoxide dismutase (SOD) and malondialdehyde (MDA), gross observation, histopathological observation, and immunohistochemical analysis. To clarify how hucMSCs work on skin aging, this study employed skin-aging model of human dermal fibroblasts (HDFs) to conduct in vitro assays by applying conditional medium of hucMSCs (CMM), including wound healing assay, senescence staining, flow cytometric oxidative detection, real time PCR, and western blot analysis. The in vivo data demonstrated that hucMSCs dose-dependently removed wrinkles, smoothed skin texture, and increased dermal thickness and collagen production of aged skin by reversing SOD and MDA levels and up-regulating Col-1 and VEGF expressions, indicating anti-oxidative and pro-regenerative effects against skin aging. The in vitro data revealed that hucMSCs significantly reversed the senescence of HDFs by promoting cell migration, inhibiting ROS production, and restoring the overexpressions of oxidative and senescent markers through paracrine mode of action, and the paracrine mechanism was mediated by the inhibition of autophagy. This study provided novel knowledge regarding the anti-aging efficacy and paracrine mechanism of hucMSCs on skin, making hucMSCs-based therapy a promising regime for skin aging treatment.
Collapse
Affiliation(s)
- Ting Li
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Department of Plastic and Aesthetic Center, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Li Zhou
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengqiang Fan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zuxiang Chen
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yan
- Cell Resource Bank and Integrated Cell Preparation Center of Xiaoshan District, Hangzhou Regional Cell Preparation Center (Shangyu Biotechnology Co., Ltd), Hangzhou, China
| | - Haishan Lu
- Department of Dermatology, PLA 903 Hospital, Hangzhou, China
| | - Ming Jia
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huiling Wu
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China. .,Department of Plastic and Aesthetic Center, The First Affiliated Hospital of Zhejiang University, Hangzhou, China.
| | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China. .,Cell Resource Bank and Integrated Cell Preparation Center of Xiaoshan District, Hangzhou Regional Cell Preparation Center (Shangyu Biotechnology Co., Ltd), Hangzhou, China.
| |
Collapse
|
141
|
Qu W, Wang Z, Engelberg-Cook E, Yan D, Siddik AB, Bu G, Allickson JG, Kubrova E, Caplan AI, Hare JM, Ricordi C, Pepine CJ, Kurtzberg J, Pascual JM, Mallea JM, Rodriguez RL, Nayfeh T, Saadi S, Durvasula RV, Richards EM, March K, Sanfilippo FP. Efficacy and Safety of MSC Cell Therapies for Hospitalized Patients with COVID-19: A Systematic Review and Meta-Analysis. Stem Cells Transl Med 2022; 11:688-703. [PMID: 35640138 PMCID: PMC9299515 DOI: 10.1093/stcltm/szac032] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 03/09/2022] [Indexed: 08/10/2023] Open
Abstract
MSC (a.k.a. mesenchymal stem cell or medicinal signaling cell) cell therapies show promise in decreasing mortality in acute respiratory distress syndrome (ARDS) and suggest benefits in treatment of COVID-19-related ARDS. We performed a meta-analysis of published trials assessing the efficacy and adverse events (AE) rates of MSC cell therapy in individuals hospitalized for COVID-19. Systematic searches were performed in multiple databases through November 3, 2021. Reports in all languages, including randomized clinical trials (RCTs), non-randomized interventional trials, and uncontrolled trials, were included. Random effects model was used to pool outcomes from RCTs and non-randomized interventional trials. Outcome measures included all-cause mortality, serious adverse events (SAEs), AEs, pulmonary function, laboratory, and imaging findings. A total of 736 patients were identified from 34 studies, which included 5 RCTs (n = 235), 7 non-randomized interventional trials (n = 370), and 22 uncontrolled comparative trials (n = 131). Patients aged on average 59.4 years and 32.2% were women. When compared with the control group, MSC cell therapy was associated with a reduction in all-cause mortality (RR = 0.54, 95% CI: 0.35-0.85, I 2 = 0.0%), reduction in SAEs (IRR = 0.36, 95% CI: 0.14-0.90, I 2 = 0.0%) and no significant difference in AE rate. A sub-group with pulmonary function studies suggested improvement in patients receiving MSC. These findings support the potential for MSC cell therapy to decrease all-cause mortality, reduce SAEs, and improve pulmonary function compared with conventional care. Large-scale double-blinded, well-powered RCTs should be conducted to further explore these results.
Collapse
Affiliation(s)
- Wenchun Qu
- Corresponding co-authors: Wenchun Qu, MD, PhD, Department of Pain Medicine, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224.
| | - Zhen Wang
- Evidence-Based Practice Center, Mayo Clinic, Rochester, MN, USA
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
| | | | - Dan Yan
- Department of Pain Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | - Guojun Bu
- Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Eva Kubrova
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Arnold I Caplan
- Skeletal Research Center, Biology Department, Case Western Reserve University, Cleveland, OH, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute and Cardiology Division, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Camillo Ricordi
- Department of Surgery, Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Carl J Pepine
- Division of Cardiovascular Medicine, and Center for Regenerative Medicine, University of Florida, Gainesville, FL, USA
| | - Joanne Kurtzberg
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC, USA
| | - Jorge M Pascual
- Division of Pulmonary, Allergy and Sleep Medicine, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Jorge M Mallea
- Division of Pulmonary, Allergy and Sleep Medicine, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | - Tarek Nayfeh
- Evidence-Based Practice Center, Mayo Clinic, Rochester, MN, USA
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
| | - Samer Saadi
- Evidence-Based Practice Center, Mayo Clinic, Rochester, MN, USA
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
| | | | - Elaine M Richards
- Department of Physiology and Functional Genomics, Center of Regenerative Medicine, University of Florida, Gainesville, FL, USA
| | - Keith March
- Division of Cardiovascular Medicine, and Center for Regenerative Medicine, University of Florida, Gainesville, FL, USA
| | - Fred P Sanfilippo
- Fred P. Sanfilippo, MD, PhD, Pathology and Laboratory Medicine, School of Medicine, Emory University, 1518 Clifton Road, 730GCR, Atlanta, GA 30322, USA.
| |
Collapse
|
142
|
Hernandez JC, Yeh DW, Marh J, Choi HY, Kim J, Chopra S, Ding L, Thornton M, Grubbs B, Makowka L, Sher L, Machida K. Activated and nonactivated MSCs increase survival in humanized mice after acute liver injury through alcohol binging. Hepatol Commun 2022; 6:1549-1560. [PMID: 35246968 PMCID: PMC9234635 DOI: 10.1002/hep4.1924] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 01/15/2022] [Accepted: 01/31/2022] [Indexed: 11/08/2022] Open
Abstract
The ability of the liver to regenerate after injury makes it an ideal organ to study for potential therapeutic interventions. Mesenchymal stem cells (MSCs) possess self-renewal and differentiation properties, as well as anti-inflammatory properties that make them an ideal candidate for therapy of acute liver injury. The primary aim of this study is to evaluate the potential for reversal of hepatic injury using human umbilical cord-derived MSCs. Secondary aims include comparison of various methods of administration as well as comparison of activated versus nonactivated human umbilical cord stem cells. To induce liver injury, humanized mice were fed high-cholesterol high-fat liquid diet with alcohol binge drinking. Mice were then treated with either umbilical cord MSCs, activated umbilical cord MSCs, or a placebo and followed for survival. Blood samples were obtained at the end of the binge drinking and at the time of death to measure alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels. Histology of all mouse livers was reported at time of death. Activated MSCs that were injected intravenously, intraperitoneally, or both routes had superior survival compared with nonactivated MSCs and with placebo-treated mice. AST and ALT levels were elevated in all mice before treatment and improved in the mice treated with stem cells. Conclusion: Activated stem cells resulted in marked improvement in survival and in recovery of hepatic chemistries. Activated umbilical cord MSCs should be considered an important area of investigation in acute liver injury.
Collapse
Affiliation(s)
- Juan Carlos Hernandez
- Departments of Molecular Microbiology and ImmunologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Da-Wei Yeh
- Departments of Molecular Microbiology and ImmunologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Joel Marh
- PrimeGenUS Inc.Santa AnaCaliforniaUSA
| | - Hye Yeon Choi
- Departments of Molecular Microbiology and ImmunologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Julia Kim
- PrimeGenUS Inc.Santa AnaCaliforniaUSA
| | - Shefali Chopra
- Department of PathologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Li Ding
- Department of Population and PublicHealth Sciences University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Matthew Thornton
- Department of SurgeryUniversity of Southern CaliforniaLos AngelesCaliforniaUSA.,Childrens Hospital Los AngelesLos AngelesCaliforniaUSA
| | - Brendan Grubbs
- Department of SurgeryUniversity of Southern CaliforniaLos AngelesCaliforniaUSA.,Childrens Hospital Los AngelesLos AngelesCaliforniaUSA
| | | | - Linda Sher
- PrimeGenUS Inc.Santa AnaCaliforniaUSA.,Department of SurgeryUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Keigo Machida
- Departments of Molecular Microbiology and ImmunologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA.,Southern California Research Center for ALPD and CirrhosisLos AngelesCaliforniaUSA
| |
Collapse
|
143
|
Arango-Rodríguez ML, Solarte-David VA, Becerra-Bayona SM, Callegari E, Paez MD, Sossa CL, Vera MEO, Mateus LC, Eduardo Serrano S, Ardila-Roa AK, Viviescas LTG. Role of mesenchymal stromal cells derivatives in diabetic foot ulcers: a controlled randomized phase 1/2 clinical trial. Cytotherapy 2022; 24:1035-1048. [PMID: 36084965 DOI: 10.1016/j.jcyt.2022.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/18/2022] [Accepted: 04/27/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Diabetes-related foot complications have been identified as the most common isolated cause of morbidity among patients with diabetes and the leading cause of amputation. Therefore, new strategies to stimulate skin regeneration may provide a novel therapeutic approach to reduce non-healing ulcer disease. Recently, we demonstrated in proof-of-concept in humans that administration of allogeneic bone marrow mesenchymal stromal cellss derivatives (allo-hBM-MSCDs) is effective in a similar way to the use of allogeneic bone marrow mesenchymal stromal cellss (allo-hBM-MSCs) in grade 2 diabetic foot ulcers (DFUs). AIM To assess the safety and efficacy profile of the allo-hBM-MSCDs relative to the conventional approach (PolyMen® dressing) in 1/2 clinical trial phases in patients with grade 1 and 2 DFUs. METHODS In the present study, we used 2 doses of allo-hBM-MSCDs (1 mL) or 1 dose of allo-hBM-MSCs (1 × 106 cells) intradermally injected around wounds and assessed their safety and effectiveness, relative to the conventional approach (PolyMem dressing). Allo-hBM-MSCDs and allo-hBM-MSCs were produced in a certified Good Manufacturing Practice-type Laboratory. Patients with grade 1 and 2 DFUs were randomized to receive allo-hBM-MSCDs (n=12), allo-hBM-MSCs (n=6) or conventional treatment (PolyMem dressing) (n=10). The wound-healing process was macroscopically evaluated until the complete closure of the ulcers. RESULTS No adverse events were reported. Patients with grade 1 and 2 DFUs treated with either allo-hBM-MSCDs or allo-hBM-MSCs, achieved greater percentages of wound closure, enhanced skin regeneration in shorter times and a greater ulcer-free survival relative to the patients who received conventional treatment. Finally, through proteomic analysis, we elucidated the proteins and growth factors that are secreted by allo-hBM-MSCs and relevant to the wound-healing process. In addition, by combining proteomics with Gene Ontology analysis, we comprehensively classified secreted proteins on both biological process and molecular function. CONCLUSIONS In this phase 1/2 trial, our cumulative results suggest that 2 doses of allo-hBM-MSCDs combined with a wound dressing are a safe and effective treatment for grade 1 and 2 DFUs.
Collapse
Affiliation(s)
- Martha L Arango-Rodríguez
- Banco Multitejidos y Centro de Terapias Avanzadas, Clínica FOSCAL Internacional, Floridablanca, Colombia.
| | - Víctor Alfonso Solarte-David
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia; Facultad de Ingeniería, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia 680003
| | - Silvia M Becerra-Bayona
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia
| | - Eduardo Callegari
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota, USA
| | - Maria D Paez
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota, USA
| | - Claudia L Sossa
- Fundación Oftalmológica de Santander Carlos Ardila Lulle Floridablanca, Colombia; Programa para el Tratamiento y Estudio de Enfermedades Hematológicas y Oncológicas de Santander (PROTEHOS), 681004153 Floridablanca, Colombia
| | | | - Ligia C Mateus
- Fundación Oftalmológica de Santander Carlos Ardila Lulle Floridablanca, Colombia
| | - Sergio Eduardo Serrano
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia
| | - Andrea K Ardila-Roa
- Banco Multitejidos y Centro de Terapias Avanzadas, Clínica FOSCAL Internacional, Floridablanca, Colombia
| | - Lady T Giratá Viviescas
- Banco Multitejidos y Centro de Terapias Avanzadas, Clínica FOSCAL Internacional, Floridablanca, Colombia
| |
Collapse
|
144
|
Association between Mesenchymal Stem Cells and COVID-19 Therapy: Systematic Review and Current Trends. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9346939. [PMID: 35782071 PMCID: PMC9242780 DOI: 10.1155/2022/9346939] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/20/2022] [Accepted: 06/02/2022] [Indexed: 11/17/2022]
Abstract
Background The novel coronavirus first emerged in Wuhan, China, and quickly spread across the globe, spanning various countries and resulting in a worldwide pandemic by the end of December 2019. Given the current advances in treatments available for COVID-19, mesenchymal stem cell (MSC) therapy seems to be a prospective option for management of ARDS observed in COVID-19 patients. This present study is aimed at exploring the therapeutic potential and safety of using MSC obtained by isolation from health cord tissues in the treatment of patients with COVID-19. Methods A systematic search was done based on the guidelines of the PRISMA 2020 statement. A literature search was executed using controlled vocabulary and indexing of trials to evaluate all the relevant studies involving the use of medical subject headings (MeSH) in electronic databases like PubMed, Embase, Scopus, Cochrane Central Register of Controlled Trials (CENTRAL), and clinicaltrials.gov up to 31 December 2021. The protocol was registered in the PROSPERO register with ID CRD42022301666. Findings. After screening finally, 22 remaining articles were included in this systematic review. The studies revealed that MSC exosomes are found to be superior to MSC alone in terms of safety owing to being smaller with a lesser immunological response which leads to free movement in blood capillaries without clumping and also cannot further divide, thus reducing the oncogenic potential of MSC-derived exosomes as compared to MSC only. The studies demonstrated that the lungs healed with the use of exosomes compared to how they presented initially at the hospital. MSCs are found to increase the angiogenesis process and alveolar reepithelization, reducing markers like TNF alpha, TGF beta, and COL I and III, reducing the growth of myofibroblasts and increasing survivability of endothelium leading to attenuated pulmonary fibrosis and even reversing them. Interpretation. We can conclude that the use of mesenchymal stem cells or their derived exosomes is safe and well-tolerated in patients with COVID-19. It improves different parameters of oxygenation and helps in the healing of the lungs. The viral load along with different inflammatory cells and biomarkers of inflammation tend to decrease. Chest X-ray, CT scan, and different radiological tools are used to show improvement and reduced ongoing destructive processes.
Collapse
|
145
|
Huang A, Liu Y, Qi X, Chen S, Huang H, Zhang J, Han Z, Han ZC, Li Z. Intravenously transplanted mesenchymal stromal cells: a new endocrine reservoir for cardioprotection. Stem Cell Res Ther 2022; 13:253. [PMID: 35715868 PMCID: PMC9204704 DOI: 10.1186/s13287-022-02922-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/17/2022] [Indexed: 11/30/2022] Open
Abstract
Background Intravenous administration of mesenchymal stromal cells (MSCs) has an acknowledged competence of cardiac repair, despite a lack of systematic description of the underlying biological mechanisms. The lung, but not the heart, is the main trapped site for intravenously transplanted MSCs, which leaves a spatial gap between intravenously transplanted MSCs and the injured myocardium. How lung-trapped MSCs after intravenous transplantation rejuvenate the injured myocardium remains unknown. Methods MSCs were isolated from human placenta tissue, and DF-MSCs or Gluc-MSCs were generated by transduced with firefly luciferase (Fluc)/enhanced green fluorescence protein (eGFP) or Gaussia luciferase (Gluc) lactadherin fusion protein. The therapeutic efficiency of intravenously transplanted MSCs was investigated in a murine model of doxorubicin (Dox)-induced cardiotoxicity. Trans-organ communication from the lung to the heart with the delivery of blood was investigated by testing the release of MSC-derived extracellular vesicles (MSC-EVs), and the potential miRNA inner MSC-EVs were screened out and verified. The potential therapeutic miRNA inner MSC-EVs were then upregulated or downregulated to assess the further therapeutic efficiency Results Dox-induced cardiotoxicity, characterized by cardiac atrophy, left ventricular dysfunction, and injured myocardium, was alleviated by consecutive doses of MSCs. These cardioprotective effects might be attributed to suppressing GRP78 triggering endoplasmic reticulum (ER) stress-induced apoptosis in cardiomyocytes. Our results confirmed that miR-181a-5p from MSCs-derived EVs (MSC-EVs) inhibited GRP78. Intravenous DF-MSCs were trapped in lung vasculature, secreted a certain number of EVs into serum, which could be confirmed by the detection of eGFP+ EVs. GLuc activity was increased in serum EVs from mice administrated with GLuc-MSCs. MiR-181a-5p, inhibiting GRP78 with high efficacy, was highly expressed in serum EVs and myocardium after injecting consecutive doses of MSCs into mice treated with Dox. Finally, upregulation or downregulation of miR-181a-5p levels in MSC-EVs enhanced or weakened therapeutic effects on Dox-induced cardiotoxicity through modulating ER stress-induced apoptosis. Conclusions This study identifies intravenously transplanted MSCs, as an endocrine reservoir, to secrete cardioprotective EVs into blood continuously and gradually to confer the trans-organ communication that relieves Dox-induced cardiotoxicity. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02922-z.
Collapse
Affiliation(s)
- Anan Huang
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China.,Department of Cardiology, Tianjin Union Medical Center, 190 Jieyuan Road, Tianjin, 300121, China.,The Key Laboratory of Bioactive Materials, Ministry of Education, The College of Life Sciences, Nankai University, Tianjin, China
| | - Yue Liu
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China.,The Key Laboratory of Bioactive Materials, Ministry of Education, The College of Life Sciences, Nankai University, Tianjin, China
| | - Xin Qi
- Department of Cardiology, Tianjin Union Medical Center, 190 Jieyuan Road, Tianjin, 300121, China.
| | - Shang Chen
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China
| | - Haoyan Huang
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China
| | - Jun Zhang
- Department of Pain Medicine, Tianjin Union Medical Center, Nankai University, Tianjin, China
| | - Zhibo Han
- Jiangxi Engineering Research Center for Stem Cell, Shangrao, Jiangxi, China.,Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd, Tianjin, China
| | - Zhong-Chao Han
- Jiangxi Engineering Research Center for Stem Cell, Shangrao, Jiangxi, China.,Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd, Tianjin, China
| | - Zongjin Li
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China. .,The Key Laboratory of Bioactive Materials, Ministry of Education, The College of Life Sciences, Nankai University, Tianjin, China. .,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
146
|
Gao S, Jin Y, Ma J, Wang J, Wang J, Shao Z, Fan T, Zhang M, Chang D. Preclinical study of human umbilical cord mesenchymal stem cell sheets for the recovery of ischemic heart tissue. Stem Cell Res Ther 2022; 13:252. [PMID: 35690871 PMCID: PMC9188245 DOI: 10.1186/s13287-022-02919-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/03/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Human umbilical cord mesenchymal stem cells (hUC-MSCs) have been widely used due to their multipotency, a broad range of sources, painless collection, and compliance with standard amplification. Cell sheet technology is a tissue engineering methodology requiring scaffolds free, and it provides an effective method for cell transplantation. To improve the therapeutic efficacy, we combined hUC-MSCs with cell sheet technology to evaluate the safety and efficacy of hUC-MSC sheets in preclinical studies using appropriate animal models. METHODS hUC-MSC sheets were fabricated by hUC-MSCs from a cell bank established by a standard operation process and quality control. Cytokine secretion, immunoregulation, and angiopoiesis were evaluated in vitro. Oncogenicity and cell diffusion assays of hUC-MSC sheets were conducted to verify the safety of hUC-MSCs sheet transplantation in mice. To provide more meaningful animal experimental data for clinical trials, porcine myocardial infarction (MI) models were established by constriction of the left circumflex, and hUC-MSC sheets were transplanted onto the ischemic area of the heart tissue. Cardiac function was evaluated and compared between the experimental and MI groups. RESULTS The in vitro results showed that hUC-MSC sheets could secrete multiple cellular factors, including VEGF, HGF, IL-6, and IL-8. Peripheral blood mononuclear cells had a lower proliferation rate and lower TNF-α secretion when co-cultured with hUC-MSC sheets. TH1 cells had a smaller proportion after activation. In vivo safety results showed that the hUC-MSCs sheet had no oncogenicity and was mainly distributed on the surface of the ischemic myocardial tissue. Echocardiography showed that hUC-MSC sheets effectively improved the left ventricular ejection fraction (LVEF), and the LVEF significantly changed (42.25 ± 1.23% vs. 66.9 ± 1.10%) in the hUC-MSC transplantation group compared with the MI group (42.52 ± 0.65% vs. 39.55 ± 1.97%) at 9 weeks. The infarct ratio of the hUC-MSCs sheet transplantation groups was also significantly reduced (14.2 ± 4.53% vs. 4.00 ± 2.00%) compared with that of the MI group. CONCLUSION Allogeneic source and cell bank established by the standard operation process and quality control make hUC-MSCs sheet possible to treat MI by off-the-shelf drug with universal quality instead of individualized medical technology.
Collapse
Affiliation(s)
- Shuang Gao
- BOE Regenerative Medicine Technology Co., Ltd., No. 9 JiuXianQiao North Road, Beijing, 100015, China
| | - Yongqiang Jin
- Heart Center, First Hospital of Tsinghua University, No. 6 JiuXianQiao 1st Road, Beijing, 10016, China
| | - Jianlin Ma
- BOE Regenerative Medicine Technology Co., Ltd., No. 9 JiuXianQiao North Road, Beijing, 100015, China
| | - Juan Wang
- BOE Regenerative Medicine Technology Co., Ltd., No. 9 JiuXianQiao North Road, Beijing, 100015, China
| | - Jing Wang
- BOE Regenerative Medicine Technology Co., Ltd., No. 9 JiuXianQiao North Road, Beijing, 100015, China
| | - Zehua Shao
- Heart Center of Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, China
| | - Taibing Fan
- Children Heart Center, Fuwai Central China Cardiovascular Hospital, No. 1 Fuwai Road, Zhengzhou, 450018, China
| | - Mingkui Zhang
- Heart Center, First Hospital of Tsinghua University, No. 6 JiuXianQiao 1st Road, Beijing, 10016, China
| | - Dehua Chang
- Department of Cell Therapy in Regenerative Medicine, The University of Tokyo Hospital, 7-3-1 Honggo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
147
|
Li Y, Yang L, Hu F, Xu J, Ye J, Liu S, Wang L, Zhuo M, Ran B, Zhang H, Ye J, Xiao J. Novel Thermosensitive Hydrogel Promotes Spinal Cord Repair by Regulating Mitochondrial Function. ACS APPLIED MATERIALS & INTERFACES 2022; 14:25155-25172. [PMID: 35618676 DOI: 10.1021/acsami.2c04341] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The repair of spinal cord injury (SCI) is still a tough clinical challenge and needs innovative therapies. Mitochondrial function is significantly compromised after SCI and has emerged as an important factor causing neuronal apoptosis and hindering functional recovery. In this study, umbilical cord mesenchymal stem cells (UCMSC), which are promising seed cells for nerve regeneration, and basic fibroblast growth factor (bFGF) that have been demonstrated to have a variety of effects on neural regeneration were jointly immobilized in extracellular matrix (ECM) and heparin-poloxamer (HP) to create a polymer bioactive system that brings more hope and possibility for the treatment of SCI. Our results in vitro and in vivo showed that the UCMSC-bFGF-ECM-HP thermosensitive hydrogel has good therapeutic effects, mainly in reducing apoptosis and improving the mitochondrial function. It showed promising utility for the functional recovery of impaired mitochondrial function by promoting mitochondrial fusion, reducing pathological mitochondrial fragmentation, increasing mitochondrial energy supply, and improving the metabolism of MDA, LDH, and ROS. In addition, we uncovered a distinct molecular mechanism underlying the protective effects associated with activating p21-activated kinase 1 (PAK1) and mitochondrial sirtuin 4 (SIRT4) by the UCMSC-bFGF-ECM-HP hydrogel. The expansion of new insights into the molecular relationships between PAK1 and SIRT4, which links the mitochondrial function in SCI, can lay the foundation for future applications and help to provide promising interventions of stem-cell-based biological scaffold therapies and potential therapeutic targets for the clinical formulation of SCI treatment strategies.
Collapse
Affiliation(s)
- Yi Li
- Medical College of Soochow University, Suzhou, Jiangsu 215123, China
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi 341000, China
| | - Liangliang Yang
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Fei Hu
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ji Xu
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Junsong Ye
- Subcenter for Stem Cell Clinical Translation, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, China
- Ganzhou Key Laboratory of Stem Cell and Regenerative Medicine, Gannan Medical University, Ganzhou, Jiangxi 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi 341000, China
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, Jiangxi 341000, China
| | - Shuhua Liu
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi 341000, China
| | - Lifeng Wang
- Medical College of Soochow University, Suzhou, Jiangsu 215123, China
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi 341000, China
| | - Ming Zhuo
- Medical College of Soochow University, Suzhou, Jiangsu 215123, China
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi 341000, China
| | - Bing Ran
- Medical College of Soochow University, Suzhou, Jiangsu 215123, China
- Department of Pain, The First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi 341000, China
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Junming Ye
- Medical College of Soochow University, Suzhou, Jiangsu 215123, China
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi 341000, China
| | - Jian Xiao
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| |
Collapse
|
148
|
Afjeh-Dana E, Naserzadeh P, Moradi E, Hosseini N, Seifalian AM, Ashtari B. Stem Cell Differentiation into Cardiomyocytes: Current Methods and Emerging Approaches. Stem Cell Rev Rep 2022; 18:2566-2592. [PMID: 35508757 DOI: 10.1007/s12015-021-10280-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 12/26/2022]
Abstract
Cardiovascular diseases (CVDs) are globally known to be important causes of mortality and disabilities. Common treatment strategies for CVDs, such as pharmacological therapeutics impose serious challenges due to the failure of treatments for myocardial necrosis. By contrast, stem cells (SCs) based therapies are seen to be promising approaches to CVDs treatment. In such approaches, cardiomyocytes are differentiated from SCs. To fulfill SCs complete potential, the method should be appointed to generate cardiomyocytes with more mature structure and well-functioning operations. For heart repairing applications, a greatly scalable and medical-grade cardiomyocyte generation must be used. Nonetheless, there are some challenges such as immune rejection, arrhythmogenesis, tumorigenesis, and graft cell death potential. Herein, we discuss the types of potential SCs, and commonly used methods including embryoid bodies related techniques, co-culture, mechanical stimulation, and electrical stimulation and their applications, advantages and limitations in this field. An estimated 17.9 million people died from CVDs in 2019, representing 32 % of all global deaths. Of these deaths, 85 % were due to heart attack and stroke.
Collapse
Affiliation(s)
- Elham Afjeh-Dana
- Radiation Biology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Parvaneh Naserzadeh
- Radiation Biology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Moradi
- Radiation Biology Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Nasrin Hosseini
- Neuroscience Research Centre, Iran University of Medical Sciences, Tehran, Iran.
| | - Alexander Marcus Seifalian
- Nanotechnology & Regenerative Medicine Commercialisation Centre (NanoRegMed Ltd), London BioScience Innovation Centre, London, UK
| | - Behnaz Ashtari
- Radiation Biology Research Centre, Iran University of Medical Sciences, Tehran, Iran. .,Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran. .,Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
149
|
Wanjiang W, Xin C, Yaxing C, Jie W, Hongyan Z, Fei N, Chengmin L, Chengjian F, Jichao Y, Jiangkai L. Curcumin Improves Human Umbilical Cord-Derived Mesenchymal Stem Cell Survival via ERK1/2 Signaling and Promotes Motor Outcomes After Spinal Cord Injury. Cell Mol Neurobiol 2022; 42:1241-1252. [PMID: 33247374 PMCID: PMC11441298 DOI: 10.1007/s10571-020-01018-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/18/2020] [Indexed: 11/28/2022]
Abstract
Human umbilical cord-derived mesenchymal stem cell (hUC-MSC) transplantation is thought to be a promising strategy for treating spinal cord injury (SCI). However, the low survival rate of transplanted hUC-MSCs limits their clinical application in cell replacement therapy. Curcumin can suppress inflammation after SCI; however, it remains unknown whether curcumin can modulate the survival of transplanted hUC-MSCs. In this study, to investigate whether curcumin could strengthen the therapeutic effects of hUC-MSC transplantation on SCI, we induced hUC-MSC apoptosis with TNF-α, transplanted hUC-MSC into SCI rats, and assessed the antiapoptotic effect and mechanism of curcumin. LDH release analysis and flow cytometry demonstrated that TNF-α led to hUC-MSC apoptosis and that curcumin increased the hUC-MSC survival rate in a dose-dependent manner. In addition, we showed that the phosphorylation levels of ERK1/2, JNK, and P38 were upregulated in apoptotic hUC-MSCs, while curcumin increased the phosphorylation of ERK1/2 but did not activate JNK or P38, and these effects were reversed by the p42/44 antagonist U0126. Furthermore, we found that the motor function scores and number of surviving HNA-positive cells were significantly increased after curcumin and hUC-MSC transplantation therapy 8 weeks post-SCI, while U0126 markedly attenuated these effects. These data confirmed that curcumin suppressed hUC-MSC apoptosis through the ERK1/2 signaling pathway and that combined curcumin and hUC-MSC treatment improved motor function in rats after SCI. The current research provides a strong basis for hUC-MSC replacement therapy in conjunction with curcumin in the treatment and management of SCI in humans.
Collapse
Affiliation(s)
- Wu Wanjiang
- Department of Neurosurgery, Institute of Neurosurgery, Key Laboratory of Neurotrauma Prevention and Treatment, Army Medical University), Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Chongqing, 400038, China
| | - Chen Xin
- Department of Neurosurgery, Institute of Neurosurgery, Key Laboratory of Neurotrauma Prevention and Treatment, Army Medical University), Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Chongqing, 400038, China
| | - Chen Yaxing
- Department of Neurosurgery, Institute of Neurosurgery, Key Laboratory of Neurotrauma Prevention and Treatment, Army Medical University), Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Chongqing, 400038, China
| | - Wang Jie
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Chongqing, 400038, China
| | - Zhang Hongyan
- Department of Neurosurgery, Institute of Neurosurgery, Key Laboratory of Neurotrauma Prevention and Treatment, Army Medical University), Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Chongqing, 400038, China
| | - Ni Fei
- Department of Field Nursing, School of Nursing, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ling Chengmin
- Department of Neurosurgery, Institute of Neurosurgery, Key Laboratory of Neurotrauma Prevention and Treatment, Army Medical University), Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Chongqing, 400038, China
| | - Feng Chengjian
- Department of Medical Engineering, 958th Hospital of the People's Liberation Army, Chongqing, 400038, China
| | - Yuan Jichao
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Chongqing, 400038, China.
| | - Lin Jiangkai
- Department of Neurosurgery, Institute of Neurosurgery, Key Laboratory of Neurotrauma Prevention and Treatment, Army Medical University), Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Chongqing, 400038, China.
| |
Collapse
|
150
|
Yu Z, Wen Y, Jiang N, Li Z, Guan J, Zhang Y, Deng C, Zhao L, Zheng SG, Zhu Y, Su W, Zhuo Y. TNF-α stimulation enhances the neuroprotective effects of gingival MSCs derived exosomes in retinal ischemia-reperfusion injury via the MEG3/miR-21a-5p axis. Biomaterials 2022; 284:121484. [DOI: 10.1016/j.biomaterials.2022.121484] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/20/2022] [Accepted: 03/22/2022] [Indexed: 12/14/2022]
|