1
|
Montague EC, Ozcan B, Sefton E, Wulkan F, Alibhai FJ, Laflamme MA. Human pluripotent stem cell-based cardiac repair: Lessons learned and challenges ahead. Adv Drug Deliv Rev 2025; 222:115594. [PMID: 40334814 DOI: 10.1016/j.addr.2025.115594] [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: 11/04/2024] [Revised: 05/01/2025] [Accepted: 05/03/2025] [Indexed: 05/09/2025]
Abstract
The transplantation of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) and hPSC-derived cardiac progenitors (hPSC-CPs) represents a promising strategy for regenerating hearts damaged by myocardial infarction (MI). After nearly two decades of experience testing these cell populations in various small- and large-animal MI models, multiple clinical trials have recently been initiated. In this review, we consider the principal lessons learned from preclinical experience with hPSC-CMs and -CPs, focusing on three conclusions that have been supported by the majority of reported transplantation studies. First, hPSC-CMs and -CPs stably engraft in injured hearts and partially remuscularize the infarct scar, but more progress is needed to improve graft cell retention and survival. Second, the transplantation of hPSC-CMs and -CPs has been found to improve contractile function in infarcted hearts, but the mechanistic basis for these effects remains incompletely elucidated. Third, the graft tissue formed by these cells can integrate and activate synchronously with host myocardium, but this capacity for electromechanical integration has been associated with an elevated risk of graft-related arrhythmias. Here, we summarize the preclinical evidence supporting these three observations, identify the relevant gaps and barriers to translation, and summarize ongoing efforts to improve the safety and efficacy of hPSC-CM- and -CP-based regenerative therapies.
Collapse
Affiliation(s)
- E Coulter Montague
- Department of Biomedical Engineering, University of Toronto, ON, Canada; McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Bilgehan Ozcan
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Elana Sefton
- Department of Biomedical Engineering, University of Toronto, ON, Canada; McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Fanny Wulkan
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Faisal J Alibhai
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Michael A Laflamme
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada; Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
2
|
Ramezani M, Baheiraei N, Bathaie SZ, Razavi M, Naderi N. Alginate hydrogel-encapsulated bone marrow-derived mesenchymal stem cells and crocin improve cardiac function in a rat model of myocardial infarction. Int J Biol Macromol 2025; 306:141548. [PMID: 40023415 DOI: 10.1016/j.ijbiomac.2025.141548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/02/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
Cardiovascular diseases (CVDs), particularly myocardial infarction (MI), are the leading cause of mortality worldwide and significantly contribute to morbidity. This study incorporated varying concentrations of crocin (CRO) into alginate hydrogel (ALG) to enhance cardiac function. Following synthesizing the hydrogel, it was characterized through a series of experiments, including morphological assessment, rheological analysis, cytocompatibility testing, and cellular viability evaluation. The therapeutic efficacy of the synthesized hydrogel in combination with bone-derived mesenchymal stem cells (BMSCs), was then investigated in a rat model of MI using echocardiography, histology, and immunohistochemistry. The results indicated that the prepared hydrogels exhibited adequate porosity and favorable rheological properties. Notably, CRO at lower concentrations significantly improved the viability of BMSCs. To evaluate the therapeutic potential in vivo, the ALG/CRO hydrogel loaded with BMSCs was implanted into the MI region of the rat model. The findings demonstrate that the ALG/CRO hydrogel can significantly reduce scar thickness and promote angiogenesis, thereby improving the recovery of cardiac function. Consequently, the ALG/CRO hydrogel has the potential to serve as an injectable carrier for the delivery of cells aimed at cardiac regeneration.
Collapse
Affiliation(s)
- Mina Ramezani
- Tissue Engineering and Applied Cell Sciences Division, Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Nafiseh Baheiraei
- Tissue Engineering and Applied Cell Sciences Division, Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - S Zahra Bathaie
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Mehdi Razavi
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, University of Central Florida College of Medicine, Orlando, Florida 32827, USA; Department of Material Sciences and Engineering, University of Central Florida, Orlando, Florida 32816, USA; Biomedical Engineering Program, Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, Florida 32816, USA.
| | - Nasim Naderi
- Cardiovascular research center, Rajaie cardiovascular institiue, Tehran, Iran.
| |
Collapse
|
3
|
da Rocha VP, Mansano BSDM, Dos Santos CFC, Teixeira ILA, de Oliveira HA, Vieira SS, Antonio EL, Izar MCDO, Fonseca FAH, Serra AJ. How long does the biological effect of a red light-emitting diode last on adipose-derived mesenchymal stem cells? Photochem Photobiol 2025; 101:206-214. [PMID: 38888236 DOI: 10.1111/php.13983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/16/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Abstract
This research investigated the duration of the influence of red light-emitting diodes (LED, 630 nm; output power: 2452.5 mW; laser beam: 163.5 cm2; irradiance: 15 mW/cm2; radiant exposure: 4 J/cm2) on different periods after irradiation (6, 12, 24, 48, and 72 h) on adipose-derived mesenchymal stem cells' (AdMSCs) metabolism and paracrine factors. AdMSCs were irradiated three times every 48 h. Twenty-four hours after the last irradiation, there was a higher MTT absorbance, followed by a decrease after 48 h. The cells' secretome showed increased levels of IL-6 and VEGF after 12 and 24 h, but this was reversed after 48 h. Additionally, LED irradiation resulted in higher levels of nitrite and did not affect oxidative stress markers. LED irradiation had significant effects on AdMSCs after 24 h compared to other groups and its control group.
Collapse
Affiliation(s)
- Vitor Pocani da Rocha
- Department of Medicine, Cardiology Division, Federal University of Sao Paulo, São Paulo, SP, Brazil
| | | | | | | | | | - Stella Sousa Vieira
- Department of Medicine, Cardiology Division, Federal University of Sao Paulo, São Paulo, SP, Brazil
| | - Ednei Luiz Antonio
- Department of Medicine, Cardiology Division, Federal University of Sao Paulo, São Paulo, SP, Brazil
| | | | | | - Andrey Jorge Serra
- Department of Medicine, Cardiology Division, Federal University of Sao Paulo, São Paulo, SP, Brazil
| |
Collapse
|
4
|
Wang Y, Dong H, Dong T, Zhao L, Fan W, Zhang Y, Yao W. Treatment of cytokine release syndrome-induced vascular endothelial injury using mesenchymal stem cells. Mol Cell Biochem 2024; 479:1149-1164. [PMID: 37392343 DOI: 10.1007/s11010-023-04785-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/04/2023] [Indexed: 07/03/2023]
Abstract
Cytokine release syndrome (CRS) is an acute systemic inflammatory reaction in which hyperactivated immune cells suddenly release a large amount of cytokines, leading to exaggerated inflammatory responses, multiple organ dysfunction, and even death. Although palliative treatment strategies have significantly reduced the overall mortality, novel targeted treatment regimens with superior therapy efficacy are urgently needed. Vascular endothelial cells (ECs) are important target cells of systemic inflammation, and their destruction is considered to be the initiating event underlying many serious complications of CRS. Mesenchymal stem/stromal cells (MSCs) are multipotent cells with self-renewing differentiation capacity and immunomodulatory properties. MSC transplantation can effectively suppress the activation of immune cells, reduce the bulk release of cytokines, and repair damaged tissues and organs. Here, we review the molecular mechanisms underlying CRS-induced vascular endothelial injury and discuss potential treatments using MSCs. Preclinical studies demonstrate that MSC therapy can effectively repair endothelium damage and thus reduce the incidence and severity of ensuing CRS-induced complications. This review highlights the therapeutic role of MSCs in fighting against CRS-induced EC damage, and summarizes the possible therapeutic formulations of MSCs for improved efficacy in future clinical trials.
Collapse
Affiliation(s)
- Yuyan Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Yangtze University, Jingzhou, China
- Health Science Center, Yangtze University, Jingzhou, China
| | - Haibo Dong
- Wuhan Optics Valley Vcanbiopharma Co., Ltd, Wuhan, China
- Key Industrial Base for Stem Cell Engineering Products, Tianjin, China
| | - Tengyun Dong
- Wuhan Optics Valley Vcanbiopharma Co., Ltd, Wuhan, China
- Key Industrial Base for Stem Cell Engineering Products, Tianjin, China
| | - Lulu Zhao
- Wuhan Optics Valley Vcanbiopharma Co., Ltd, Wuhan, China
- Key Industrial Base for Stem Cell Engineering Products, Tianjin, China
| | - Wen Fan
- Department of Laboratory Medicine, The First Affiliated Hospital of Yangtze University, Jingzhou, China.
| | - Yu Zhang
- Wuhan Optics Valley Vcanbiopharma Co., Ltd, Wuhan, China.
- Key Industrial Base for Stem Cell Engineering Products, Tianjin, China.
- Haihe Laboratory of Cell Ecosystem, Tianjin, China.
| | - Weiqi Yao
- Wuhan Optics Valley Vcanbiopharma Co., Ltd, Wuhan, China.
- Key Industrial Base for Stem Cell Engineering Products, Tianjin, China.
- Department of Biology and Medicine, Hubei University of Technology, Wuhan, China.
| |
Collapse
|
5
|
Zhang HZ, Chae DS, Kim SW. ASC and SVF Cells Synergistically Induce Neovascularization in Ischemic Hindlimb Following Cotransplantation. Int J Mol Sci 2021; 23:185. [PMID: 35008610 PMCID: PMC8745515 DOI: 10.3390/ijms23010185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 01/29/2023] Open
Abstract
Previously, we reported the angio-vasculogenic properties of human stromal vascular fraction (SVF) and adipose tissue-derived mesenchymal stem cells (ASCs). In this study, we investigated whether the combination of ASCs and SVF cells exhibited synergistic angiogenic properties. We conducted quantitative (q)RT-PCR, Matrigel plug, tube formation assays, and in vivo therapeutic assays using an ischemic hind limb mouse model. Immunohistochemical analysis was also conducted. qRT-PCR results revealed that FGF-2 was highly upregulated in ASCs compared with SVF, while PDGF-b and VEGF-A were highly upregulated in SVF. Conditioned medium from mixed cultures of ASCs and SVF (A+S) cells showed higher Matrigel tube formation and endothelial cell proliferation in vitro. A+S cell transplantation into ischemic mouse hind limbs strongly prevented limb loss and augmented blood perfusion compared with SVF cell transplantation. Transplanted A+S cells also showed high capillary density, cell proliferation, angiogenic cytokines, and anti-apoptotic potential in vivo compared with transplanted SVF. Our data indicate that A+S cell transplantation results in synergistic angiogenic therapeutic effects. Accordingly, A+S cell injection could be an alternative therapeutic strategy for treating ischemic diseases.
Collapse
Affiliation(s)
- Hong Zhe Zhang
- Department of Cardiology, College of Medicine, Dong-A University, Busan 49201, Korea;
| | - Dong-Sik Chae
- Department of Orthopedic Surgery, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Korea
| | - Sung-Whan Kim
- Department of Medicine, College of Medicine, Catholic Kwandong University, Gangneung 25601, Korea
| |
Collapse
|
6
|
Silver SE, Barrs RW, Mei Y. Transplantation of Human Pluripotent Stem Cell-Derived Cardiomyocytes for Cardiac Regenerative Therapy. Front Cardiovasc Med 2021; 8:707890. [PMID: 34820426 PMCID: PMC8606657 DOI: 10.3389/fcvm.2021.707890] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/20/2021] [Indexed: 01/14/2023] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide and bears an immense economic burden. Late-stage heart failure often requires total heart transplantation; however, due to donor shortages and lifelong immunosuppression, alternative cardiac regenerative therapies are in high demand. Human pluripotent stem cells (hPSCs), including human embryonic and induced pluripotent stem cells, have emerged as a viable source of human cardiomyocytes for transplantation. Recent developments in several mammalian models of cardiac injury have provided strong evidence of the therapeutic potential of hPSC-derived cardiomyocytes (hPSC-CM), showing their ability to electromechanically integrate with host cardiac tissue and promote functional recovery. In this review, we will discuss recent developments in hPSC-CM differentiation and transplantation strategies for delivery to the heart. We will highlight the mechanisms through which hPSC-CMs contribute to heart repair, review major challenges in successful transplantation of hPSC-CMs, and present solutions that are being explored to address these limitations. We end with a discussion of the clinical use of hPSC-CMs, including hurdles to clinical translation, current clinical trials, and future perspectives on hPSC-CM transplantation.
Collapse
Affiliation(s)
- Sophia E. Silver
- Bioengineering Department, Clemson University, Clemson, SC, United States
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Ryan W. Barrs
- Bioengineering Department, Clemson University, Clemson, SC, United States
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC, United States
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
7
|
Kim YJ, Kim SW, Lee JR, Um SH, Joung YK, Bhang SH. Comparing the cytotoxic effect of light-emitting and organic light-emitting diodes based light therapy on human adipose-derived stem cells. J IND ENG CHEM 2021. [DOI: 10.1016/j.jiec.2021.07.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
8
|
Oxygen Delivery Approaches to Augment Cell Survival After Myocardial Infarction: Progress and Challenges. Cardiovasc Toxicol 2021; 22:207-224. [PMID: 34542796 DOI: 10.1007/s12012-021-09696-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/11/2021] [Indexed: 10/20/2022]
Abstract
Myocardial infarction (MI), triggered by blockage of a coronary artery, remains the most common cause of death worldwide. After MI, the capability of providing sufficient blood and oxygen significantly decreases in the heart. This event leads to depletion of oxygen from cardiac tissue and consequently leads to massive cardiac cell death due to hypoxemia. Over the past few decades, many studies have been carried out to discover acceptable approaches to treat MI. However, very few have addressed the crucial role of efficient oxygen delivery to the injured heart. Thus, various strategies were developed to increase the delivery of oxygen to cardiac tissue and improve its function. Here, we have given an overall discussion of the oxygen delivery mechanisms and how the current technologies are employed to treat patients suffering from MI, including a comprehensive view on three major technical approaches such as oxygen therapy, hemoglobin-based oxygen carriers (HBOCs), and oxygen-releasing biomaterials (ORBs). Although oxygen therapy and HBOCs have shown promising results in several animal and clinical studies, they still have a few drawbacks which limit their effectiveness. More recent studies have investigated the efficacy of ORBs which may play a key role in the future of oxygenation of cardiac tissue. In addition, a summary of conducted studies under each approach and the remaining challenges of these methods are discussed.
Collapse
|
9
|
Increased Myocardial Retention of Mesenchymal Stem Cells Post-MI by Pre-Conditioning Exercise Training. Stem Cell Rev Rep 2021; 16:730-741. [PMID: 32306279 DOI: 10.1007/s12015-020-09970-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Stem cell (SC) therapy is a promising approach to improve post-myocardial infarction (MI) cardiac remodeling, but the proinflammatory microenvironment may lead to SC loss and, therefore, may have a negative impact on therapy. It appears that exercise training (ET) improves myocardial microenvironment for SC transplantation. Therefore, we tested the effect of ET on post-infarction retention of adipose-derived SCs (ADSCs) and its combined effects on the inflammatory microenvironment. Fischer-344 female rats were randomized to one of the following groups: Sham; sedentary coronary occlusion who did not receive ADSCs (sMI); sedentary coronary occlusion who received ADSCs; exercise coronary occlusion who received ADSCs. Rats were trained nine weeks prior to MI, followed by ADSCs transplantation. The MI led to left ventricle (LV) dilation and dysfunction, myocardial hypertrophy and fibrosis, and increased proinflammatory profile compared to Sham rats. Conversely, ADSCs transplanted rats exhibited, better morphological and functional LV parameters; inhibition of myocardial hypertrophy and fibrosis; and attenuation of proinflammatory cytokines (interleukins 1β and 10, tumor necrosis factor α, and transforming growth factor β) in the myocardium compared to sMI rats. Interestingly, ET enhanced the effect of ADSCs on interleukin 10 expression. There was a correlation between cytokine expression and myocardial ADSCs retention. The. ET enhanced the beneficial effects of ADSCs in infarcted myocardium, which was associated with higher ADSCs retention. These findings highlight the importance of ET in myocardial retention of ADSCs and attenuation of cardiac remodeling post-infarction. Cytokine analysis suggests improvement in ET-linked myocardial microenvironment based on its anti-inflammatory action.
Collapse
|
10
|
Sim WS, Park BW, Ban K, Park HJ. In Situ Preconditioning of Human Mesenchymal Stem Cells Elicits Comprehensive Cardiac Repair Following Myocardial Infarction. Int J Mol Sci 2021; 22:1449. [PMID: 33535594 PMCID: PMC7867207 DOI: 10.3390/ijms22031449] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 01/04/2023] Open
Abstract
Human bone marrow-derived mesenchymal stem cells (BM-MSCs), represented as a population of adult stem cells, have long been considered as one of the most promising sources for cell-based cardiac regenerative therapy. However, their clinical use has been significantly hampered by low survival and poor retention following administration into failing hearts. Here, to improve the therapeutic effectiveness of BM-MSCs, we examined a novel therapeutic platform named in situ preconditioning in a rat myocardial infarction (MI) model. In situ preconditioning was induced by a combinatory treatment of BM-MSCs with genetically engineered hepatocyte growth factor-expressing MSCs (HGF-eMSCs) and heart-derived extracellular matrix (hdECM) hydrogel. Subsequently, our results demonstrated that in situ preconditioning with cell mixture substantially improved the survival/retention of BM-MSCs in the MI-induced rat hearts. Enhanced retention of BM-MSCs ultimately led to a significant cardiac function improvement, which was derived from the protection of myocardium and enhancement of vessel formation in the MI hearts. The results provide compelling evidence that in situ preconditioning devised to improve the therapeutic potential of BM-MSCs can be an effective strategy to achieve cardiac repair of MI hearts.
Collapse
Affiliation(s)
- Woo-Sup Sim
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea; (W.-S.S.); (B.-W.P.)
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea
| | - Bong-Woo Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea; (W.-S.S.); (B.-W.P.)
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Hun-Jun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea; (W.-S.S.); (B.-W.P.)
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea
- Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
11
|
Madsen SD, Giler MK, Bunnell BA, O'Connor KC. Illuminating the Regenerative Properties of Stem Cells In Vivo with Bioluminescence Imaging. Biotechnol J 2020; 16:e2000248. [PMID: 33089922 DOI: 10.1002/biot.202000248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/17/2020] [Indexed: 11/10/2022]
Abstract
Preclinical animal studies are essential to the development of safe and effective stem cell therapies. Bioluminescence imaging (BLI) is a powerful tool in animal studies that enables the real-time longitudinal monitoring of stem cells in vivo to elucidate their regenerative properties. This review describes the application of BLI in preclinical stem cell research to address critical challenges in producing successful stem cell therapeutics. These challenges include stem cell survival, proliferation, homing, stress response, and differentiation. The applications presented here utilize bioluminescence to investigate a variety of stem and progenitor cells in several different in vivo models of disease and implantation. An overview of luciferase reporters is provided, along with the advantages and disadvantages of BLI. Additionally, BLI is compared to other preclinical imaging modalities and potential future applications of this technology are discussed in emerging areas of stem cell research.
Collapse
Affiliation(s)
- Sean D Madsen
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, 70118, USA.,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Margaret K Giler
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, 70118, USA.,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA.,Department of Pharmacology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Kim C O'Connor
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, 70118, USA.,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| |
Collapse
|
12
|
Selvakumar D, Clayton ZE, Chong JJH. Robust Cardiac Regeneration: Fulfilling the Promise of Cardiac Cell Therapy. Clin Ther 2020; 42:1857-1879. [PMID: 32943195 DOI: 10.1016/j.clinthera.2020.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE We review the history of cardiac cell therapy, highlighting lessons learned from initial adult stem cell (ASC) clinical trials. We present pluripotent stem cell-derived cardiomyocytes (PSC-CMs) as a leading candidate for robust regeneration of infarcted myocardium but identify several issues that must be addressed before successful clinical translation. METHODS We conducted an unstructured literature review of PubMed-listed articles, selecting the most comprehensive and relevant research articles, review articles, clinical trials, and basic or translation articles in the field of cardiac cell therapy. Articles were identified using the search terms adult stem cells, pluripotent stem cells, cardiac stem cell, and cardiac regeneration or from references of relevant articles, Articles were prioritized and selected based on their impact, originality, or potential clinical applicability. FINDINGS Since its inception, the ASC therapy field has been troubled by conflicting preclinical data, academic controversies, and inconsistent trial designs. These issues have damaged perceptions of cardiac cell therapy among investors, the academic community, health care professionals, and, importantly, patients. In hindsight, the key issue underpinning these problems was the inability of these cell types to differentiate directly into genuine cardiomyocytes, rendering them unable to replace damaged myocardium. Despite this, beneficial effects through indirect paracrine or immunomodulatory effects remain possible and continue to be investigated. However, in preclinical models, PSC-CMs have robustly remuscularized infarcted myocardium with functional, force-generating cardiomyocytes. Hence, PSC-CMs have now emerged as a leading candidate for cardiac regeneration, and unpublished reports of first-in-human delivery of these cells have recently surfaced. However, the cardiac cell therapy field's history should serve as a cautionary tale, and we identify several translational hurdles that still remain. Preclinical solutions to issues such as arrhythmogenicity, immunogenicity, and poor engraftment rates are needed, and next-generation clinical trials must draw on robust knowledge of mechanistic principles of the therapy. IMPLICATIONS The clinical transplantation of functional stem cell-derived heart tissue with seamless integration into native myocardium is a lofty goal. However, considerable advances have been made during the past 2 decades. Currently, PSC-CMs appear to be the best prospect to reach this goal, but several hurdles remain. The history of adult stem cell trials has taught us that shortcuts cannot be taken without dire consequences, and it is essential that progress not be hurried and that a worldwide, cross-disciplinary approach be used to ensure safe and effective clinical translation.
Collapse
Affiliation(s)
- Dinesh Selvakumar
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Zoe E Clayton
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - James J H Chong
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia.
| |
Collapse
|
13
|
Abstract
Stem cells (SCs) are discovered long back but the idea that SCs possess therapeutic potential came up just a few decades back. In a past decade stem cell therapy is highly emerged and displayed tremendous potential for the treatment of a wide range of diseases and disorders such as blindness and vision impairment, type I diabetes, infertility, HIV, etc. SCs are very susceptible to destruction after transplantation into the host because of the inability to sustain elevated stress conditions inside the damaged tissue/organ. Heat shock proteins (HSPs) are molecular chaperones/stress proteins expressed in response to stress (elevated temperature, harmful chemicals, ischemia, viruses, etc) inside a living cell. HSPs protect the cell from damage by assisting in the proper folding of cellular proteins. This review briefly summarises different types of HSPs, their classification, cellular functions as well as the role of HSPs in regulating SC self-renewal and survival in the transplanted host. Applications of HSP modulated SCs in regenerative medicine and for the treatment of ischemic heart disease, myocardial infarction (MI), osteoarthritis, ischemic stroke, spinocerebellar ataxia type 3 (SCA3), leukemia, hepatic ischemia-reperfusion injury, Graft-versus-host disease (GVHD) and Parkinson's disease (PD) are discussed. In order to provide potential insights in understanding molecular mechanisms related to SCs in vertebrates, correlations between HSPs and SCs in cnidarians and planarians are also reviewed. There is a need to advance research in order to validate the use of HSPs for SC therapy and establish effective treatment strategies.
Collapse
|
14
|
Abstract
Stem cell therapy offers a breakthrough opportunity for the improvement of ischemic heart diseases. Numerous clinical trials and meta-analyses appear to confirm its positive but variable effects on heart function. Whereas these trials widely differed in design, cell type, source, and doses reinjected, cell injection route and timing, and type of cardiac disease, crucial key factors that may favour the success of cell therapy emerge from the review of their data. Various types of cell have been delivered. Injection of myoblasts does not improve heart function and is often responsible for severe ventricular arrythmia occurrence. Using bone marrow mononuclear cells is a misconception, as they are not stem cells but mainly a mix of various cells of hematopoietic lineages and stromal cells, only containing very low numbers of cells that have stem cell-like features; this likely explain the neutral results or at best the modest improvement in heart function reported after their injection. The true existence of cardiac stem cells now appears to be highly discredited, at least in adults. Mesenchymal stem cells do not repair the damaged myocardial tissue but attenuate post-infarction remodelling and contribute to revascularization of the hibernated zone surrounding the scar. CD34+ stem cells - likely issued from pluripotent very small embryonic-like (VSEL) stem cells - emerge as the most convincing cell type, inducing structural and functional repair of the ischemic myocardial area, providing they can be delivered in large amounts via intra-myocardial rather than intra-coronary injection, and preferentially after myocardial infarct rather than chronic heart failure.
Collapse
Affiliation(s)
- Philippe Hénon
- CellProthera SAS and Institut de Recherche en Hématologie et Transplantation, CellProthera SAS 12 rue du Parc, 68100, Mulhouse, France.
| |
Collapse
|
15
|
Multi-Spheroid-Loaded Human Acellular Dermal Matrix Carrier Preserves Its Spheroid Shape and Improves In Vivo Adipose-Derived Stem Cell Delivery and Engraftment. Tissue Eng Regen Med 2020; 17:271-283. [PMID: 32314311 DOI: 10.1007/s13770-020-00252-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/09/2020] [Accepted: 03/15/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Current in vivo adult stem cell delivery presents limited clinical effects due to poor engraftment and survival. To overcome current challenges in cell delivery and promote surgical cell delivery for soft tissue repair, a multi-spheroid-loaded thin sectioned acellular dermal matrix (tsADM) carrier which preserves loaded spheroids' three-dimensional (3D) structure, was developed. METHODS Adipose-derived stem cells (ASCs) were used for spheroid delivery. After generating spheroids in 3D cell culture dishes, spheroid plasticity and survival in-between coverslips were evaluated. Spheroids were loaded onto tsADM, their shape changes were followed up for 14 days, and then imaged. Spheroid adhesion stability to tsADM against shear stress was also evaluated. Finally, cell delivery efficacy was compared with cell-seeded tsADM by in vivo implantation and histological evaluation. RESULTS Spheroids withstood cyclic compression stress and maintained their 3D shape without fusion after 48 h of culture in-between coverslips. Cell survival improved when spheroids were cultured on tsADM in-between the coverslips. Spheroid-loaded tsADM with coverslips maintained their spheroid outline for 14 days of culture whereas without coverslips, the group lost their outline due to spreading after 4 days in culture. Spheroids loaded onto tsADMs were more stable after six rather than 3 days in culture. Spheroid-loaded tsADMs showed about a 2.96-fold higher ASCs transplantation efficacy than cell-seeded tsADMs after 2 weeks of in vivo transplantation. CONCLUSION These results indicate that transplantation of spheroid-loaded tsADMs significantly improved cell delivery. These findings suggest that a combined approach with other cells, drugs, and nanoparticles may improve cell delivery and therapeutic efficacy.
Collapse
|
16
|
Panahi M, Rahimi B, Rahimi G, Yew Low T, Saraygord-Afshari N, Alizadeh E. Cytoprotective effects of antioxidant supplementation on mesenchymal stem cell therapy. J Cell Physiol 2020; 235:6462-6495. [PMID: 32239727 DOI: 10.1002/jcp.29660] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 02/15/2020] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) are earmarked as perfect candidates for cell therapy and tissue engineering due to their capacity to differentiate into different cell types. However, their potential for application in regenerative medicine declines when the levels of the reactive oxygen and nitrogen species (RONS) increase from the physiological levels, a phenomenon which is at least inevitable in ex vivo cultures and air-exposed damaged tissues. Increased levels of RONS can alter the patterns of osteogenic and adipogenic differentiation and inhibit proliferation, as well. Besides, oxidative stress enhances senescence and cell death, thus lowering the success rates of the MSC engraftment. Hence, in this review, we have selected some representatives of antioxidants and newly emerged nano antioxidants in three main categories, including chemical compounds, biometabolites, and protein precursors/proteins, which are proved to be effective in the treatment of MSCs. We will focus on how antioxidants can be applied to optimize the clinical usage of the MSCs and their associated signaling pathways. We have also reviewed several paralleled properties of some antioxidants and nano antioxidants which can be simultaneously used in real-time imaging, scaffolding techniques, and other applications in addition to their primary antioxidative function.
Collapse
Affiliation(s)
- Mohammad Panahi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahareh Rahimi
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Golbarg Rahimi
- Department of Cellular and Molecular Biology, University of Esfahan, Esfahan, Iran
| | - Teck Yew Low
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Neda Saraygord-Afshari
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Effat Alizadeh
- Drug Applied Research Center and Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Klama-Baryła A, Rojczyk E, Kitala D, Łabuś W, Smętek W, Wilemska-Kucharzewska K, Kucharzewski M. Preparation of placental tissue transplants and their application in skin wound healing and chosen skin bullous diseases - Stevens-Johnson syndrome and toxic epidermal necrolysis treatment. Int Wound J 2020; 17:491-507. [PMID: 31943788 DOI: 10.1111/iwj.13305] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/17/2019] [Indexed: 12/20/2022] Open
Abstract
Unique properties of amniotic membrane make it a promising source for tissue engineering and a clinically useful alternative for patients suffering from chronic wounds including, for example, ulcers, burns, ocular surface damages and wounds occurring in the course of bullous diseases like stevens-johnson syndrome and toxic epidermal necrolysis. Its use has many advantages over standard wound care, as it contains pluripotent cells, nutrients, anti-fibrotic and anti-inflammatory cytokines, growth factors and extracellular matrix (ECM) proteins. Placental tissues can be prepared as a medical component, an advanced therapy medicinal product or a tissue graft. In addition to basic preparation procedures such as washing, rinsing, cutting, drying and sterilisation, there are many optional steps such as perforation, crosslinking and decellularisation. Finally, transplants should be properly stored-in cryopreserved or dehydrated form. In recent years, many studies including basic science and clinical trials have proven the potential to expand the use of amniotic membrane and amnion-derived cells to the fields of orthopaedics, dentistry, surgery, urology, vascular tissue engineering and even oncology. In this review, we discuss the role of placental tissues in skin wound healing and in the treatment of various diseases, with particular emphasis on bullous diseases. We also describe some patented procedures for placental tissue grafts preparation.
Collapse
Affiliation(s)
- Agnieszka Klama-Baryła
- The Burn Centre of Stanisław Sakiel in Siemianowice Śląskie, Siemianowice Śląskie, Poland
| | - Ewa Rojczyk
- Department of Descriptive and Topographic Anatomy, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, Zabrze, Poland
| | - Diana Kitala
- The Burn Centre of Stanisław Sakiel in Siemianowice Śląskie, Siemianowice Śląskie, Poland
| | - Wojciech Łabuś
- The Burn Centre of Stanisław Sakiel in Siemianowice Śląskie, Siemianowice Śląskie, Poland
| | - Wojciech Smętek
- The Burn Centre of Stanisław Sakiel in Siemianowice Śląskie, Siemianowice Śląskie, Poland
| | | | - Marek Kucharzewski
- Department of Descriptive and Topographic Anatomy, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, Zabrze, Poland
| |
Collapse
|
18
|
Jaquenod De Giusti C, Santalla M, Das S. Exosomal non-coding RNAs (Exo-ncRNAs) in cardiovascular health. J Mol Cell Cardiol 2019; 137:143-151. [PMID: 31669445 DOI: 10.1016/j.yjmcc.2019.09.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 09/05/2019] [Accepted: 09/27/2019] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EVs) play a role in the pathophysiological processes and in different diseases, including cardiovascular disease. Out of several categories of EVs, exosomes (smallest - 30 to 150 nm) are gaining most of the focus as the next generation of biomarkers and in therapeutic strategies. This is because exosomes can be differentiated from other types of EVs based on the expression of tetraspanin molecules on the surface. More importantly, exosomes can be traced back to the cell of origin by identifying the unique cellular marker(s) on the exosomal surface. Recently, several researchs have demonstrated an important and underappreciated mechanism of paracrine cell-cell communication involving exosomal transfer, and its subsequent functional impact on recipient cells. Exosomes are enriched in proteins, mRNAs, miRNAs, and other non-coding RNAs, which can potentially alter myocardial function. Additionally, different stages of tissue damage can also be identified by measuring these bioactive molecules in the circulation. There are several aspects of this new concept still unknown. Therefore, in this review, we have summarized the knowledge we have so far and highlighted the potential of this novel concept of next generation biomarkers and therapeutic intervention.
Collapse
Affiliation(s)
- Carolina Jaquenod De Giusti
- Centro de Investigaciones Cardiovasculares UNLP-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina.
| | - Manuela Santalla
- Centro de Investigaciones Cardiovasculares UNLP-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina; Departamento de Ciencias Básicas y Experimentales, Universidad Nacional del Noroeste de Buenos Aires, Pergamino, Argentina
| | - Samarjit Das
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
19
|
Heo JS, Pyo S, Lim JY, Yoon DW, Kim BY, Kim JH, Kim GJ, Lee SG, Kim J. Biological effects of melatonin on human adipose‑derived mesenchymal stem cells. Int J Mol Med 2019; 44:2234-2244. [PMID: 31573052 PMCID: PMC6844604 DOI: 10.3892/ijmm.2019.4356] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 09/11/2019] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are capable of differentiating into other cell types and exhibit immunomodulatory effects. MSCs are affected by several intrinsic and extrinsic signaling modulators, including growth factors, cytokines, extracellular matrix and hormones. Melatonin, produced by the pineal gland, is a hormone that regulates sleep cycles. Recent studies have shown that melatonin improves the therapeutic effects of stem cells. The present study aimed to investigate whether melatonin enhances the biological activities of human adipose-derived MSCs. The results demonstrated that treatment with melatonin promoted cell proliferation by inducing SRY-box transcription factor 2 gene expression and preventing replicative senescence. In addition, melatonin exerted anti-adipogenic effects on MSCs. PCR analysis revealed that the expression of the CCAAT enhancer binding protein a gene, a key transcription factor in adipogenesis, was decreased following melatonin treatment, resulting in reduced adipogenic differentiation in an in vitro assay. The present study also examined the effect of melatonin on the immunomodulatory response using a co-culture system of human peripheral blood mononuclear cells and MSCs. Activated T cells were strongly inhibited following melatonin exposure compared with those in the control group. Finally, the favorable effects of melatonin on MSCs were confirmed using luzindole, a selective melatonin receptor antagonist. The proliferation-promoting, anti-inflammatory effects of melatonin suggested that melatonin-treated MSCs may be used for effective cell therapy.
Collapse
Affiliation(s)
- June Seok Heo
- Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| | - Sangshin Pyo
- Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| | - Ja-Yun Lim
- Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| | - Dae Wui Yoon
- Department of Biomedical Laboratory Science, College of Health Science, Jungwon University, Geosan, Chungbuk 28024, Republic of Korea
| | - Bo Yong Kim
- Department of Health and Environmental Science, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| | - Jin-Hee Kim
- Department of Biomedical Laboratory Science, College of Health Science, Cheongju University, Cheongju, North Chungcheong 28497, Republic of Korea
| | - Gi Jin Kim
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Seung Gwan Lee
- Department of Health and Environmental Science, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| | - Jinkwan Kim
- Department of Biomedical Laboratory Science, College of Health Science, Jungwon University, Geosan, Chungbuk 28024, Republic of Korea
| |
Collapse
|
20
|
Streeter BW, Xue J, Xia Y, Davis ME. Electrospun Nanofiber-Based Patches for the Delivery of Cardiac Progenitor Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:18242-18253. [PMID: 31021079 DOI: 10.1021/acsami.9b04473] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Congenital heart disease is the number one cause of birth defect-related death because it often leads to right ventricular heart failure (RVHF). One promising avenue to combat this RVHF is the use of cardiac patches composed of stem cells and scaffolds. Herein, we demonstrate a reparative cardiac patch by combining neonatal or child c-kit+ progenitor cells (CPCs) with a scaffold composed of electrospun polycaprolactone nanofibers. We examined different parameters of the patch, including the alignment, composition, and surface properties of the nanofibers, as well as the age of the CPCs. The patch based on uniaxially aligned nanofibers successfully aligned the CPCs. With the inclusion of gelatin in the nanofiber matrix and/or coating of fibronectin on the surface of the nanofibers, the metabolism of both neonatal and child CPCs was generally enhanced. The conditioned media collected from both patches based on aligned and random nanofibers could reduce the fibrotic gene expression in rat cardiac fibroblasts, following stimulation with transforming growth factor β. Furthermore, the conditioned media collected from the nanofiber-based patches could lead to the formation of tubes of human umbilical vein endothelial cells, indicating the pro-angiogenic capability of the patch. Taken together, the electrospun nanofiber-based patches are a suitable delivery vehicle for CPCs and can confer reparative benefit through anti-fibrotic and pro-angiogenic paracrine signaling.
Collapse
Affiliation(s)
- Benjamin W Streeter
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Jiajia Xue
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Younan Xia
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
- School of Chemistry and Biochemistry, School of Chemical and Biological Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
- Division of Cardiology , Emory University School of Medicine , Atlanta , Georgia 30322 , United States
- Children's Heart Research and Outcomes (HeRO) Center , Children's Healthcare of Atlanta and Emory University , Atlanta , Georgia 30322 , United States
| |
Collapse
|
21
|
Madonna R, Angelucci S, Di Giuseppe F, Doria V, Giricz Z, Görbe A, Ferdinandy P, De Caterina R. Proteomic analysis of the secretome of adipose tissue-derived murine mesenchymal cells overexpressing telomerase and myocardin. J Mol Cell Cardiol 2019; 131:171-186. [PMID: 31055035 DOI: 10.1016/j.yjmcc.2019.04.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 02/06/2019] [Accepted: 04/18/2019] [Indexed: 12/25/2022]
Abstract
RATIONALE Understanding mechanisms of the therapeutic effects of stem/progenitor cells, among which adipose tissue-derived mesenchymal stromal cells (AT-MSCs), has important implications for clinical use. Since the majority of such cells die within days or weeks after transplantation and do not persist in the transplanted organ or tissue, their effects appear to be largely mediated by paracrine signaling pathways, and are enhanced by overexpression of the antisenescent protein telomerase reverse transcriptase (TERT), and the anti-apoptotic transcription factor myocardin (MYOCD). AIM By a proteomic approach combining two-dimensional gel electrophoresis (2DE) with matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF/TOF) mass spectrometry, we aimed at analyzing how soluble and vesicular secretomes of aged murine AT-MSCs and their angiogenic function are modulated by the overexpression of TERT and MYOCD. METHODS We cultured murine mock-transduced AT-MSCs and "rejuvenated" AT-MSCs overexpressing TERT and MYOCD (rTMAT-MSCs) harvested from 1-year-old male C57BL/6 mice. We established proteomes from 3 mock-transduced AT-MSCs and rTMAT-MSCs cultures in serum-free conditions, as well as their corresponding conditioned medium (CM) and exosome-enriched fractions (Exo+). RESULTS AND CONCLUSIONS Proteomic analysis revealed a 2-fold increase of matrix metalloproteinase-2 (MMP-2) and its inhibitor metalloproteinase inhibitor 2 (TIMP2) in the CM - but not in the Exo + - of rTMAT-MSCs as compared to mock-transduced AT-MSCs. At the functional level, rTMAT-MSCs-CM, and - to a lesser extent - its Exo + fraction, increased tube formation of human vein endothelial cells (HUVECs), which could be blocked by anti-MMP2 and enhanced by anti-TIMP2 antibodies, respectively. Altogether, our results identify MMP2 and its inhibitor TIMP2 as novel candidates by which rTMAT-MSCs can support angiogenesis. Our strategy also illustrates the usefulness of comparative targeted proteomic approach to decipher molecular pathways underlying rTMAT-MSCs properties.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Center of Aging Sciences and Translational Medicine - CESI-Met and Institute of Cardiology, "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy; Department of Internal Medicine, Cardiology, The University of Texas Health Science Center at Houston, Houston, Texas, United States; Department of Neurosciences, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti-Pescara, Chieti, Italy.
| | - Stefania Angelucci
- Department of Medical, Oral & Biotechnological Sciences, Dentistry and Biotechnology, and Aging Research Center and Translational Medicine, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Stem TeCh Group, Via L Polacchi 13, Chieti, Italy
| | - Fabrizio Di Giuseppe
- Department of Medical, Oral & Biotechnological Sciences, Dentistry and Biotechnology, and Aging Research Center and Translational Medicine, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Stem TeCh Group, Via L Polacchi 13, Chieti, Italy
| | - Vanessa Doria
- Center of Aging Sciences and Translational Medicine - CESI-Met and Institute of Cardiology, "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy; Department of Neurosciences, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti-Pescara, Chieti, Italy
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Raffaele De Caterina
- Center of Aging Sciences and Translational Medicine - CESI-Met and Institute of Cardiology, "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy; Institute of Cardiology, University of Pisa, Pisa, Italy; Department of Neurosciences, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti-Pescara, Chieti, Italy.
| |
Collapse
|
22
|
Su N, Jiang LY, Wang X, Gao PL, Zhou J, Wang CY, Luo Y. Membrane-Binding Adhesive Particulates Enhance the Viability and Paracrine Function of Mesenchymal Cells for Cell-Based Therapy. Biomacromolecules 2019; 20:1007-1017. [PMID: 30616345 DOI: 10.1021/acs.biomac.8b01624] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Understanding the fundamental cell-material interactions is essential to designing functional materials for biomedical applications. Although mesenchymal stromal cells (MSCs) are known to secrete cytokines and exosomes that are effective to treat degenerative diseases, the inherent property of biomaterials to modulate the therapeutic function of MSCs remains to be investigated. Here, a multivalent cell-membrane adhesive conjugate was generated through polyamindoamine (PAMAM) and an oligopeptide, IKVAV, and the conjugate was further complexed with hyaluronic acid (HA). The adhesive particulates were used to coat the surface of adipose-derived mesenchymal stromal cells (Ad-MSCs) and studied in the MSC spheroid culture. The analysis showed that the adhesive complexes formed via PAMAM conjugates and HA significantly promoted the proliferation and the gene expression of pro-angiogenesis cytokines in MSCs; the production of anti-inflammatory miRNAs in exosomes could also be elevated. The transplantation of the Ad-MSCs primed with PAMAM-IKVAV/HA composite particulates in a rat myocardial infarction model further demonstrated the beneficial effects of membrane-binding materials on improving the cell retention and tissue angiogenesis. The new function of membrane-binding adhesive materials potentially provides useful ways to improve cell-based therapy.
Collapse
Affiliation(s)
- Ni Su
- Department of Biomedical Engineering, College of Engineering , Peking University Room 206, Fangzheng Building, 298 Chengfu Road , Haidian District, Beijing 100871 , China
| | - Li-Yang Jiang
- Department of Biomedical Engineering, College of Engineering , Peking University Room 206, Fangzheng Building, 298 Chengfu Road , Haidian District, Beijing 100871 , China
| | - Xi Wang
- Department of Biomedical Engineering, College of Engineering , Peking University Room 206, Fangzheng Building, 298 Chengfu Road , Haidian District, Beijing 100871 , China
| | - Peng-Lai Gao
- Department of Biomedical Engineering, College of Engineering , Peking University Room 206, Fangzheng Building, 298 Chengfu Road , Haidian District, Beijing 100871 , China
| | - Jin Zhou
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center , Academy of Military Medical Sciences , 27 Taiping Road , Haidian District, Beijing 100039 , China
| | - Chang-Yong Wang
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center , Academy of Military Medical Sciences , 27 Taiping Road , Haidian District, Beijing 100039 , China
| | - Ying Luo
- Department of Biomedical Engineering, College of Engineering , Peking University Room 206, Fangzheng Building, 298 Chengfu Road , Haidian District, Beijing 100871 , China
| |
Collapse
|
23
|
Aloisi A, Pisignano D, Rinaldi R. Nanotechnologies for Neurosciences. POLITO SPRINGER SERIES 2019:81-98. [DOI: 10.1007/978-3-319-71048-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
24
|
Streeter BW, Davis ME. Therapeutic Cardiac Patches for Repairing the Myocardium. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1144:1-24. [DOI: 10.1007/5584_2018_309] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
25
|
Bordoni M, Rey F, Fantini V, Pansarasa O, Di Giulio AM, Carelli S, Cereda C. From Neuronal Differentiation of iPSCs to 3D Neuro-Organoids: Modelling and Therapy of Neurodegenerative Diseases. Int J Mol Sci 2018; 19:E3972. [PMID: 30544711 PMCID: PMC6321164 DOI: 10.3390/ijms19123972] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/27/2018] [Accepted: 12/07/2018] [Indexed: 12/15/2022] Open
Abstract
In the last decade, the advances made into the reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) led to great improvements towards their use as models of diseases. In particular, in the field of neurodegenerative diseases, iPSCs technology allowed to culture in vitro all types of patient-specific neural cells, facilitating not only the investigation of diseases' etiopathology, but also the testing of new drugs and cell therapies, leading to the innovative concept of personalized medicine. Moreover, iPSCs can be differentiated and organized into 3D organoids, providing a tool which mimics the complexity of the brain's architecture. Furthermore, recent developments in 3D bioprinting allowed the study of physiological cell-to-cell interactions, given by a combination of several biomaterials, scaffolds, and cells. This technology combines bio-plotter and biomaterials in which several types of cells, such as iPSCs or differentiated neurons, can be encapsulated in order to develop an innovative cellular model. IPSCs and 3D cell cultures technologies represent the first step towards the obtainment of a more reliable model, such as organoids, to facilitate neurodegenerative diseases' investigation. The combination of iPSCs, 3D organoids and bioprinting will also allow the development of new therapeutic approaches. Indeed, on the one hand they will lead to the development of safer and patient-specific drugs testing but, also, they could be developed as cell-therapy for curing neurodegenerative diseases with a regenerative medicine approach.
Collapse
Affiliation(s)
- Matteo Bordoni
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, 27100 Pavia, Italy.
| | - Federica Rey
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, via A. di Rudinì 8, 20142 Milan, Italy.
| | - Valentina Fantini
- Department of Brain and Behavioural Sciences, University of Pavia, 27100 Pavia, Italy.
- Laboratory of Neurobiology and Neurogenetic, Golgi-Cenci Foundation, 20081 Abbiategrasso, Italy.
| | - Orietta Pansarasa
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, 27100 Pavia, Italy.
| | - Anna Maria Di Giulio
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, via A. di Rudinì 8, 20142 Milan, Italy.
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of MilanVia Giovanni Battista Grassi, 74, 20157 Milan, Italy.
| | - Stephana Carelli
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, via A. di Rudinì 8, 20142 Milan, Italy.
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of MilanVia Giovanni Battista Grassi, 74, 20157 Milan, Italy.
| | - Cristina Cereda
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, 27100 Pavia, Italy.
- Laboratory of Neurobiology and Neurogenetic, Golgi-Cenci Foundation, 20081 Abbiategrasso, Italy.
| |
Collapse
|
26
|
Allogeneic Mesenchymal Stem Cells and Biomaterials: The Perfect Match for Cardiac Repair? Int J Mol Sci 2018; 19:ijms19103236. [PMID: 30347686 PMCID: PMC6213975 DOI: 10.3390/ijms19103236] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022] Open
Abstract
Coronary heart disease is the leading cause of death worldwide with huge socio-economic consequences. Cell therapy, and particularly mesenchymal stem cells (MSC), are considered a promising option to treat this disorder, due to their robust trophic and immunomodulatory properties. However, limitations such as their low rate of engraftment and poor survival after administration into the heart have precluded their large-scale clinical use. Nevertheless, the combination of MSC with polymer-made scaffolds or hydrogels has proven to enhance their retention and, therefore, their efficacy. Additionally, their allogeneic use could permit the creation of ready-to-use cell patches able to improve their feasibility and promote their application in clinical settings. In this review, the experimental and clinical results derived from the use of MSC in cardiac pathology, as well as advances in the bioengineering field to improve the potential of therapeutic cells, are extensively discussed. Additionally, the current understanding of the heart response to the allogeneic MSC transplants is addressed.
Collapse
|
27
|
Wang O, Ismail A, Fabian FM, Lin H, Li Q, Elowsky C, Carlson MA, Burgess W, Velander WH, Kidambi S, Lei Y. A totally recombinant fibrin matrix for mesenchymal stem cell culture and delivery. J Biomed Mater Res A 2018; 106:3135-3142. [PMID: 30152030 DOI: 10.1002/jbm.a.36508] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 06/15/2018] [Accepted: 07/12/2018] [Indexed: 01/14/2023]
Abstract
Mesenchymal stem cells (MSCs) have been widely studied for tissue engineering and treating diseases in laboratories, clinical trials, and clinics. Fibrin matrices are often used to culture MSCs or increase the retention of MSCs at the injection site. However, fibrins made with the human plasma derived fibrinogen have high cost and risk of human pathogen transmission. In this article, we studied if fibrin matrices made with recombinant human fibrinogen, recombinant human thrombin, and recombinant human factor XIII could be used to culture and deliver MSCs. We systematically investigated the relationships between the fibrin matrix formulation, its nanostructure, and the behaviors of the cells in the matrix including the cell morphology, viability, and growth. We found that the fibrinogen concentration significantly affected the matrix structure and cell behaviors. We then used an optimized fibrin matrix to deliver human MSCs into mice subcutaneously. We found that the matrix could significantly enhance the retention of MSCs at the injection site. To our best knowledge, this is the first study on using fibrin matrices made with entirely recombinant proteins for culturing and delivering MSCs. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 3135-3142, 2018.
Collapse
Affiliation(s)
- Ou Wang
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska.,Biomedical Engineering Program, University of Nebraska, Lincoln, Nebraska
| | - Ayman Ismail
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska
| | - Frank Marco Fabian
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska
| | - Haishuang Lin
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska
| | - Qiang Li
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska
| | - Christian Elowsky
- Department of Agronomy and Horticulture, University of Nebraska, Lincoln, Nebraska
| | - Mark A Carlson
- Department of Surgery, University of Nebraska Medical Center and the Omaha VA Medical Center, Omaha, Nebraska
| | - Wilson Burgess
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska
| | - William H Velander
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska
| | - Srivatsan Kidambi
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska.,Biomedical Engineering Program, University of Nebraska, Lincoln, Nebraska.,Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska.,Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Yuguo Lei
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska.,Biomedical Engineering Program, University of Nebraska, Lincoln, Nebraska.,Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska.,Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
28
|
Wu KH, Wang SY, Xiao QR, Yang Y, Huang NP, Mo XM, Sun J. Efficient generation of functional cardiomyocytes from human umbilical cord-derived virus-free induced pluripotent stem cells. Cell Tissue Res 2018; 374:275-283. [PMID: 29961217 DOI: 10.1007/s00441-018-2875-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/19/2018] [Indexed: 11/25/2022]
Abstract
We have previously demonstrated that human umbilical cord-derived mesenchymal stem cells (UC-MSCs) can differentiate into cardiomyocyte-like cells. However, no contracting cells were observed during differentiation. In this study, we generated induced pluripotent stem cells (iPSCs) from UC-MSCs using mRNA reprogramming and focused on the differentiation of reprogrammed iPSCs into functional cardiomyocytes. For cardiac differentiation, the spontaneously contracting cell clusters were present on day 8 of differentiation. Immunostaining studies and cardiac-specific gene expression confirmed the cardiomyocyte phenotype of the differentiated cells. Electrophysiology studies indicated that iPSCs derived from UC-MSCs had a capacity for differentiation into nodal-, atrial-, and ventricular-like phenotypes based on action potential characteristics, and the derived cardiomyocytes exhibited responsiveness to β-adrenergic and muscarinic stimulations. Moreover, the derived cardiomyocytes displayed spontaneous intracellular Ca2+ transients. These results demonstrate that functional cardiomyocytes can be generated from reprogrammed UC-MSCs, and the methodology described here will serve as a useful protocol to obtain functional cardiomyocytes from human mesenchymal stem cells.
Collapse
Affiliation(s)
- Kai Hong Wu
- Cardiovascular Center, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, People's Republic of China.
| | - Su Yun Wang
- Cardiovascular Center, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, People's Republic of China
| | - Qian Ru Xiao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China
| | - Yu Yang
- Cardiovascular Center, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, People's Republic of China
| | - Ning Ping Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China
| | - Xu Ming Mo
- Cardiovascular Center, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, People's Republic of China
| | - Jian Sun
- Cardiovascular Center, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, People's Republic of China
| |
Collapse
|
29
|
Piaggesi A, Låuchli S, Bassetto F, Biedermann T, Marques A, Najafi B, Palla I, Scarpa C, Seimetz D, Triulzi I, Turchetti G, Vaggelas A. Advanced therapies in wound management: cell and tissue based therapies, physical and bio-physical therapies smart and IT based technologies. J Wound Care 2018; 27:S1-S137. [DOI: 10.12968/jowc.2018.27.sup6a.s1] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Alberto Piaggesi
- Prof, Director, EWMA Scientific Recorder (Editor), Diabetic Foot Section of the Pisa University Hospital, Department of Endocrinology and Metabolism, University of Pisa, Lungarno Pacinotti 43, 56126 Pisa, Italy
| | - Severin Låuchli
- Chief of Dermatosurgery and Woundcare, EWMA Immediate Past President (Co-editor), Department of Dermatology, University Hospital, Zurich, Råmistrasse 100, 8091 Zärich, Schwitzerland
| | - Franco Bassetto
- Prof, Head of Department, Clinic of Plastic and Reconstructive Surgery, University of Padova, Via Giustiniani, 35100 Padova
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, August Forel-Strasse 7, 8008 Zürich, Switzerland
| | - Alexandra Marques
- University of Minho, 3B's Research Group in Biomaterials, Biodegradables and Biomimetics, Avepark - Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal
| | - Bijan Najafi
- Professor of Surgery, Director of Clinical Research, Division of Vascular Surgery and Endovascular Therapy, Director of Interdisciplinary Consortium on Advanced Motion Performance (iCAMP), Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, MS: BCM390, Houston, TX 77030-3411, US
| | - Ilaria Palla
- Institute of Management, Sant'Anna School of Advanced Studies, Piazza Martiri della Libertà, 33, 56127 Pisa, Italy
| | - Carlotta Scarpa
- Clinic of Plastic and Reconstructive Surgery, University of Padova, Via Giustiniani, 35100 Padova
| | - Diane Seimetz
- Founding Partner, Biopharma Excellence, c/o Munich Technology Center, Agnes-Pockels-Bogen 1, 80992 Munich, Germany
| | - Isotta Triulzi
- Institute of Management, Sant'Anna School of Advanced Studies, Piazza Martiri della Libertà, 33, 56127 Pisa, Italy
| | - Giuseppe Turchetti
- Fulbright Scholar, Institute of Management, Sant'Anna School of Advanced Studies, Piazza Martiri della Libertà, 33, 56127 Pisa, Italy
| | - Annegret Vaggelas
- Consultant, Biopharma Excellence, c/o Munich Technology Center, Agnes-Pockels-Bogen 1, 80992 Munich, Germany
| |
Collapse
|
30
|
Tauroursodeoxycholic Acid Protects against the Effects of P-Cresol-Induced Reactive Oxygen Species via the Expression of Cellular Prion Protein. Int J Mol Sci 2018; 19:ijms19020352. [PMID: 29370069 PMCID: PMC5855574 DOI: 10.3390/ijms19020352] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/16/2018] [Accepted: 01/22/2018] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) could be a promising solution in the treatment of various diseases including chronic kidney disease (CKD). However, endoplasmic reticulum (ER) stress induced by ischemia in the area of application limits the integration and survival of MSCs in patients. In our study, we generated ER stress-induced conditions in MSCs using P-cresol. As P-cresol is a toxic compound accumulated in the body of CKD patients and induces apoptosis and inflammation through reactive oxygen species (ROS), we observed ER stress-induced MSC apoptosis activated by oxidative stress, which in turn resulted from ROS generation. To overcome stress-induced apoptosis, we investigated the protective effects of tauroursodeoxycholic acid (TUDCA), a bile acid, on ER stress in MSCs. In ER stress, TUDCA treatment of MSCs reduced ER stress-associated protein activation, including GRP78, PERK, eIF2α, ATF4, IRE1α, and CHOP. Next, to explore the protective mechanism adopted by TUDCA, TUDCA-mediated cellular prion protein (PrPC) activation was assessed. We confirmed that PrPC expression significantly increased ROS, which was eliminated by superoxide dismutase and catalase in MSCs. These findings suggest that TUDCA protects from inflammation and apoptosis in ER stress via PrPC expression. Our study demonstrates that TUDCA protects MSCs against inflammation and apoptosis in ER stress by PrPC expression in response to P-cresol exposure.
Collapse
|
31
|
Tseng TC, Wong CW, Hsieh FY, Hsu SH. Biomaterial Substrate-Mediated Multicellular Spheroid Formation and Their Applications in Tissue Engineering. Biotechnol J 2017; 12. [DOI: 10.1002/biot.201700064] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/01/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Ting-Chen Tseng
- Institute of Polymer Science and Engineering; National Taiwan University; Taipei Taiwan
| | - Chui-Wei Wong
- Institute of Polymer Science and Engineering; National Taiwan University; Taipei Taiwan
| | - Fu-Yu Hsieh
- Institute of Polymer Science and Engineering; National Taiwan University; Taipei Taiwan
| | - Shan-hui Hsu
- Institute of Polymer Science and Engineering; National Taiwan University; Taipei Taiwan
- Institute of Cellular and System Medicine; National Health Research Institutes; Miaoli Taiwan
| |
Collapse
|
32
|
Engineered M13 Nanofiber Accelerates Ischemic Neovascularization by Enhancing Endothelial Progenitor Cells. Tissue Eng Regen Med 2017; 14:787-802. [PMID: 30603528 DOI: 10.1007/s13770-017-0074-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/08/2017] [Accepted: 07/16/2017] [Indexed: 12/21/2022] Open
Abstract
Dysfunction or loss of blood vessel causes several ischemic diseases. Although endothelial progenitor cells (EPCs) are a promising source for cell-based therapy, ischemia-induced pathophysiological condition limits the recovery rate by causing drastic cell death. To overcome this issue, we attempted to develop a cell-targeted peptide delivery and priming system to enhance EPC-based neovascularization using an engineered M13 bacteriophage harboring nanofibrous tubes displaying ~2700 multiple functional motifs. The M13 nanofiber was modified by displaying RGD, which is an integrin-docking peptide, on the minor coat protein, and by mutilayering SDKP motifs, which are the key active sites for thymosin β4, on the major coat protein. The engineered M13 nanofiber dramatically enhanced ischemic neovascularization by activating intracellular and extracellular processes such as proliferation, migration, and tube formation in the EPCs. Furthermore, transplantation of the primed EPCs with the M13 nanofiber harboring RGD and SDKP facilitated functional recovery and neovascularization in a murine hindlimb ischemia model. Overall, this study demonstrates the effectiveness of the M13 nanofiber-based novel peptide delivery and priming strategy in promoting EPC bioactivity and neovessel regeneration. To our knowledge, this is first report on M13 nanofibers harboring dual functional motifs, the use of which might be a novel strategy for stem and progenitor cell therapy against cardiovascular ischemic diseases.
Collapse
|
33
|
Xu JY, Chen GH, Yang YJ. Exosomes: A Rising Star in Falling Hearts. Front Physiol 2017; 8:494. [PMID: 28751864 PMCID: PMC5508217 DOI: 10.3389/fphys.2017.00494] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 06/28/2017] [Indexed: 12/20/2022] Open
Abstract
Although exosomes were previously recognized as a mechanism for discharging useless cellular components, growing evidence has elucidated their roles in conveying information between cells. They contribute to cell-cell communication by carrying nucleic acids, proteins and lipids that can, in turn, regulate behavior of the target cells. Recent research suggested that exosomes extensively participate in progression of diverse cardiovascular diseases (CVDs), such as myocardial infarction, cardiomyopathy, pulmonary arterial hypertension and others. Here, we summarize effects of exosome-derived molecules (mainly microRNAs and proteins) on cardiac function, to examine their potential applications as biomarkers or therapeutics in CVDs.
Collapse
Affiliation(s)
- Jun-Yan Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing, China
| | - Gui-Hao Chen
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing, China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing, China
| |
Collapse
|
34
|
Xu HL, Yu WZ, Lu CT, Li XK, Zhao YZ. Delivery of growth factor-based therapeutics in vascular diseases: Challenges and strategies. Biotechnol J 2017; 12. [PMID: 28296342 DOI: 10.1002/biot.201600243] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 01/27/2017] [Accepted: 02/09/2017] [Indexed: 12/18/2022]
Abstract
Either cardiovascular or peripheral vascular diseases have become the major cause of morbidity and mortality worldwide. Recently, growth factors therapeutics, whatever administrated in form of exogenous growth factors or their relevant genes have been discovered to be an effective strategy for the prevention and therapy of vascular diseases, because of their promoting angiogenesis. Besides, as an alternative, stem cell-based therapy has been also developed in view of their paracrine-mediated effect or ability of differentiation toward angiogenesis-related cells under assistance of growth factors. Despite of being specific and potent, no matter growth factors or stem cells-based therapy, their full clinical transformation is limited from bench to bedside. In this review, the potential choices of therapeutic modes based on types of different growth factors or stem cells were firstly summarized for vascular diseases. The confronted various challenges such as lack of non-invasive delivery method, the physiochemical challenge, the short half-life time, and poor cell survival, were carefully analyzed for these therapeutic modes. Various strategies to overcome these limitations are put forward from the perspective of drug delivery. The expertised design of a suitable delivery form will undoubtedly provide valuable insight into their clinical application in the regenerative medicine.
Collapse
Affiliation(s)
- He-Lin Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Wen-Ze Yu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Cui-Tao Lu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Xiao-Kun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
- Collaborative Innovation Center of Biomedical Science by Wenzhou University & Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Ying-Zheng Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| |
Collapse
|
35
|
Lee JH, Han YS, Lee SH. Potentiation of biological effects of mesenchymal stem cells in ischemic conditions by melatonin via upregulation of cellular prion protein expression. J Pineal Res 2017; 62. [PMID: 28095625 DOI: 10.1111/jpi.12385] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/11/2017] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) are promising candidates for stem cell-based therapy in ischemic diseases. However, ischemic injury induces pathophysiological conditions, such as oxidative stress and inflammation, which diminish therapeutic efficacy of MSC-based therapy by reducing survival and functionality of transplanted MSCs. To overcome this problem, we explored the effects of melatonin on the proliferation, resistance to oxidative stress, and immunomodulatory properties of MSCs. Treatment with melatonin enhanced MSC proliferation and self-renewal via upregulation of cellular prion protein (PrPC ) expression. Melatonin diminished the extent of MSC apoptosis in oxidative stress conditions by regulating the levels of apoptosis-associated proteins, such as BCL-2, BAX, PARP-1, and caspase-3, in a PrPC -dependent manner. In addition, melatonin regulated the immunomodulatory effects of MSCs via the PrPC -IDO axis. In a murine hind-limb ischemia model, melatonin-stimulated MSCs improved the blood flow perfusion, limb salvage, and vessel regeneration by lowering the extent of apoptosis of affected local cells and transplanted MSCs as well as by reducing infiltration of macrophages. These melatonin-mediated therapeutic effects were inhibited by silencing of PrPC expression. Our findings for the first time indicate that melatonin promotes MSC functionality and enhances MSC-mediated neovascularization in ischemic tissues through the upregulation of PrPC expression. In conclusion, melatonin-treated MSCs could provide a therapeutic strategy for vessel regeneration in ischemic disease, and the targeting of PrPC levels may prove instrumental for MSC-based therapies.
Collapse
Affiliation(s)
- Jun Hee Lee
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Yong-Seok Han
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
- Departments of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, Korea
| |
Collapse
|
36
|
Yoon YM, Lee JH, Yun SP, Han YS, Yun CW, Lee HJ, Noh H, Lee SJ, Han HJ, Lee SH. Tauroursodeoxycholic acid reduces ER stress by regulating of Akt-dependent cellular prion protein. Sci Rep 2016; 6:39838. [PMID: 28004805 PMCID: PMC5177936 DOI: 10.1038/srep39838] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 11/28/2016] [Indexed: 11/09/2022] Open
Abstract
Although mesenchymal stem cells (MSCs) are a promising cell source for regenerative medicine, ischemia-induced endoplasmic reticulum (ER) stress induces low MSC engraftment and limits their therapeutic efficacy. To overcome this, we investigated the protective effect of tauroursodeoxycholic acid (TUDCA), a bile acid, on ER stress in MSCs in vitro and in vivo. In ER stress conditions, TUDCA treatment of MSCs reduced the activation of ER stress-associated proteins, including GRP78, PERK, eIF2α, ATF4, IRE1α, JNK, p38, and CHOP. In particular, TUDCA inhibited the dissociation between GRP78 and PERK, resulting in reduced ER stress-mediated cell death. Next, to explore the ER stress protective mechanism induced by TUDCA treatment, TUDCA-mediated cellular prion protein (PrPC) activation was assessed. TUDCA treatment increased PrPC expression, which was regulated by Akt phosphorylation. Manganese-dependent superoxide dismutase (MnSOD) expression also increased significantly in response to signaling through the TUDCA-Akt axis. In a murine hindlimb ischemia model, TUDCA-treated MSC transplantation augmented the blood perfusion ratio, vessel formation, and transplanted cell survival more than untreated MSC transplantation did. Augmented functional recovery following MSC transplantation was blocked by PrPC downregulation. This study is the first to demonstrate that TUDCA protects MSCs against ER stress via Akt-dependent PrPC and Akt-MnSOD pathway.
Collapse
Affiliation(s)
- Yeo Min Yoon
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Jun Hee Lee
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Seung Pil Yun
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Yong-Seok Han
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Chul Won Yun
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul 151-741, Republic of Korea
| | - Hyunjin Noh
- Department of Internal Medicine, Hyonam Kidney Laboratory, Soonchunhyang University, Seoul, Republic of Korea
| | - Sei-Jung Lee
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul 151-741, Republic of Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul 151-741, Republic of Korea
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea.,Departments of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, 330-930, Republic of Korea
| |
Collapse
|
37
|
Han YS, Lee JH, Yoon YM, Yun CW, Noh H, Lee SH. Hypoxia-induced expression of cellular prion protein improves the therapeutic potential of mesenchymal stem cells. Cell Death Dis 2016; 7:e2395. [PMID: 27711081 PMCID: PMC5133977 DOI: 10.1038/cddis.2016.310] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/30/2016] [Accepted: 09/05/2016] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are ‘adult' multipotent cells that promote regeneration of injured tissues in vivo. However, differences in oxygenation levels between normoxic culture conditions (21% oxygen) and both the MSC niche (2–8% oxygen) and ischemic injury-induced oxidative stress conditions in vivo have resulted in low efficacy of MSC therapies in both pre-clinical and clinical studies. To address this issue, we examined the effectiveness of hypoxia preconditioning (2% oxygen) for enhancing the bioactivity and tissue-regenerative potential of adipose-derived MSCs. Hypoxia preconditioning enhanced the proliferative potential of MSCs by promoting the expression of normal cellular prion protein (PrPC). In particular, hypoxia preconditioning-mediated MSC proliferation was regulated by PrPC-dependent JAK2 and STAT3 activation. In addition, hypoxia preconditioning-induced PrPC regulated superoxide dismutase and catalase activity, and inhibited oxidative stress-induced apoptosis via inactivation of cleaved caspase-3. In a murine hindlimb ischemia model, hypoxia preconditioning enhanced the survival and proliferation of transplanted MSCs, ultimately resulting in improved functional recovery of the ischemic tissue, including the ratio of blood flow perfusion, limb salvage, and neovascularization. These results suggest that Hypo-MSC offer a therapeutic strategy for accelerated neovasculogenesis in ischemic diseases, and that PrPC comprises a potential target for MSC-based therapies.
Collapse
Affiliation(s)
- Yong-Seok Han
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Jun Hee Lee
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham School of Medicine, Baltimore, AL 35294, USA
| | - Yeo Min Yoon
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Chul Won Yun
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Hyunjin Noh
- Department of Internal Medicine, Soonchunhyang University, Seoul, Republic of Korea.,Hyonam Kidney Laboratory, Soonchunhyang University, Seoul, Republic of Korea
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea.,Departments of Biochemistry, Soonchunhyang University College of Medicine, Cheonan 330-930, Republic of Korea
| |
Collapse
|
38
|
Moya A, Larochette N, Paquet J, Deschepper M, Bensidhoum M, Izzo V, Kroemer G, Petite H, Logeart-Avramoglou D. Quiescence Preconditioned Human Multipotent Stromal Cells Adopt a Metabolic Profile Favorable for Enhanced Survival under Ischemia. Stem Cells 2016; 35:181-196. [PMID: 27578059 DOI: 10.1002/stem.2493] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 08/11/2016] [Accepted: 08/21/2016] [Indexed: 12/20/2022]
Abstract
A major impediment to the development of therapies with mesenchymal stem cells/multipotent stromal cells (MSC) is the poor survival and engraftment of MSCs at the site of injury. We hypothesized that lowering the energetic demand of MSCs by driving them into a quiescent state would enhance their survival under ischemic conditions. Human MSCs (hMSCs) were induced into quiescence by serum deprivation (SD) for 48 hours. Such preconditioned cells (SD-hMSCs) exhibited reduced nucleotide and protein syntheses compared to unpreconditioned hMSCs. SD-hMSCs sustained their viability and their ATP levels upon exposure to severe, continuous, near-anoxia (0.1% O2 ) and total glucose depletion for up to 14 consecutive days in vitro, as they maintained their hMSC multipotential capabilities upon reperfusion. Most importantly, SD-hMSCs showed enhanced viability in vivo for the first week postimplantation in mice. Quiescence preconditioning modified the energy-metabolic profile of hMSCs: it suppressed energy-sensing mTOR signaling, stimulated autophagy, promoted a shift in bioenergetic metabolism from oxidative phosphorylation to glycolysis and upregulated the expression of gluconeogenic enzymes, such as PEPCK. Since the presence of pyruvate in cell culture media was critical for SD-hMSC survival under ischemic conditions, we speculate that these cells may utilize some steps of gluconeogenesis to overcome metabolic stress. These findings support that SD preconditioning causes a protective metabolic adaptation that might be taken advantage of to improve hMSC survival in ischemic environments. Stem Cells 2017;35:181-196.
Collapse
Affiliation(s)
- Adrien Moya
- Laboratory of Bioengineering and Bioimaging for Osteo-Articular tissues, UMR 7052, CNRS, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Nathanaël Larochette
- Laboratory of Bioengineering and Bioimaging for Osteo-Articular tissues, UMR 7052, CNRS, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Joseph Paquet
- Laboratory of Bioengineering and Bioimaging for Osteo-Articular tissues, UMR 7052, CNRS, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Mickael Deschepper
- Laboratory of Bioengineering and Bioimaging for Osteo-Articular tissues, UMR 7052, CNRS, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Morad Bensidhoum
- Laboratory of Bioengineering and Bioimaging for Osteo-Articular tissues, UMR 7052, CNRS, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Valentina Izzo
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Cell Biology and Metabolomics platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,INSERM, U1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Cell Biology and Metabolomics platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,INSERM, U1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital Q2:07, Stockholm, Sweden
| | - Hervé Petite
- Laboratory of Bioengineering and Bioimaging for Osteo-Articular tissues, UMR 7052, CNRS, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Delphine Logeart-Avramoglou
- Laboratory of Bioengineering and Bioimaging for Osteo-Articular tissues, UMR 7052, CNRS, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
39
|
Maass M, Krausgrill B, Eschrig S, Kaluschke T, Urban K, Peinkofer G, Plenge TG, Oeckenpöhler S, Raths M, Ladage D, Halbach M, Hescheler J, Müller-Ehmsen J. Intramyocardially Transplanted Neonatal Cardiomyocytes (NCMs) Show Structural and Electrophysiological Maturation and Integration and Dose-Dependently Stabilize Function of Infarcted Rat Hearts. Cell Transplant 2016; 26:157-170. [PMID: 27539827 DOI: 10.3727/096368916x692870] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Cardiac cell replacement therapy is a promising therapy to improve cardiac function in heart failure. Persistence, structural and functional maturation, and integration of transplanted cardiomyocytes into recipients' hearts are crucial for a safe and efficient replacement of lost cells. We studied histology, electrophysiology, and quantity of intramyocardially transplanted rat neonatal cardiomyocytes (NCMs) and performed a detailed functional study with repeated invasive (pressure-volume catheter) and noninvasive (echocardiography) analyses of infarcted female rat hearts including pharmacological stress before and 3 weeks after intramyocardial injection of 5 × 106 (low NCM) or 25 × 106 (high NCM) syngeneic male NCMs or medium as placebo (Ctrl). Quantitative real-time polymerase chain reaction (PCR) for Y-chromosome confirmed a fivefold higher persisting male cell number in high NCM versus low NCM after 3 weeks. Sharp electrode measurements within viable slices of recipient hearts demonstrated that transplanted NCMs integrate into host myocardium and mature to an almost adult phenotype, which might be facilitated through gap junctions between host myocardium and transplanted NCMs as indicated by connexin43 in histology. Ejection fraction of recipient hearts was severely impaired after ligation of left anterior descending (LAD; pressure-volume catheter: 39.2 ± 3.6%, echocardiography: 39.9 ± 1.4%). Repeated analyses revealed a significant further decline within 3 weeks in Ctrl and a dose-dependent stabilization in cell-treated groups. Consistently, stabilized cardiac function/morphology in cell-treated groups was seen in stroke volume, cardiac output, ventricle length, and wall thickness. Our findings confirm that cardiac cell replacement is a promising therapy for ischemic heart disease since immature cardiomyocytes persist, integrate, and mature after intramyocardial transplantation, and they dose-dependently stabilize cardiac function after myocardial infarction.
Collapse
|
40
|
Alemdar N, Leijten J, Camci-Unal G, Hjortnaes J, Ribas J, Paul A, Mostafalu P, Gaharwar AK, Qiu Y, Sonkusale S, Liao R, Khademhosseini A. Oxygen-Generating Photo-Cross-Linkable Hydrogels Support Cardiac Progenitor Cell Survival by Reducing Hypoxia-Induced Necrosis. ACS Biomater Sci Eng 2016; 3:1964-1971. [DOI: 10.1021/acsbiomaterials.6b00109] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Neslihan Alemdar
- Biomaterials Innovation Research Center, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- Harvard-MIT Division of Health Sciences
and Technology, Massachusetts Institute of Technology, 77 Massachusetts
Avenue, Cambridge, Massachusetts 02139, United States
| | - Jeroen Leijten
- Biomaterials Innovation Research Center, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- Harvard-MIT Division of Health Sciences
and Technology, Massachusetts Institute of Technology, 77 Massachusetts
Avenue, Cambridge, Massachusetts 02139, United States
- Department of Developmental BioEngineering, MIRA Institute for Biomedical
Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Gulden Camci-Unal
- Biomaterials Innovation Research Center, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- Harvard-MIT Division of Health Sciences
and Technology, Massachusetts Institute of Technology, 77 Massachusetts
Avenue, Cambridge, Massachusetts 02139, United States
| | - Jesper Hjortnaes
- Biomaterials Innovation Research Center, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- Harvard-MIT Division of Health Sciences
and Technology, Massachusetts Institute of Technology, 77 Massachusetts
Avenue, Cambridge, Massachusetts 02139, United States
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Joao Ribas
- Biomaterials Innovation Research Center, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- Harvard-MIT Division of Health Sciences
and Technology, Massachusetts Institute of Technology, 77 Massachusetts
Avenue, Cambridge, Massachusetts 02139, United States
- Doctoral Program in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Arghya Paul
- Biomaterials Innovation Research Center, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- Harvard-MIT Division of Health Sciences
and Technology, Massachusetts Institute of Technology, 77 Massachusetts
Avenue, Cambridge, Massachusetts 02139, United States
| | - Pooria Mostafalu
- Department
of Electrical and Computer and Engineering, Tufts University, Medford Massachusetts 02155, United States
| | - Akhilesh K. Gaharwar
- Biomaterials Innovation Research Center, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- Harvard-MIT Division of Health Sciences
and Technology, Massachusetts Institute of Technology, 77 Massachusetts
Avenue, Cambridge, Massachusetts 02139, United States
| | - Yiling Qiu
- Cardiac Muscle Research Laboratory, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Sameer Sonkusale
- Department
of Electrical and Computer and Engineering, Tufts University, Medford Massachusetts 02155, United States
| | - Ronglih Liao
- Cardiac Muscle Research Laboratory, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- Harvard-MIT Division of Health Sciences
and Technology, Massachusetts Institute of Technology, 77 Massachusetts
Avenue, Cambridge, Massachusetts 02139, United States
- Wyss Institute
for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
- Department of Physics, King Abdulaziz University, Jeddah, 21569, Saudi Arabia
| |
Collapse
|
41
|
Microtissues in Cardiovascular Medicine: Regenerative Potential Based on a 3D Microenvironment. Stem Cells Int 2016; 2016:9098523. [PMID: 27073399 PMCID: PMC4814701 DOI: 10.1155/2016/9098523] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/01/2016] [Accepted: 02/21/2016] [Indexed: 02/06/2023] Open
Abstract
More people die annually from cardiovascular diseases than from any other cause. In particular, patients who suffer from myocardial infarction may be affected by ongoing adverse remodeling processes of the heart that may ultimately lead to heart failure. The introduction of stem and progenitor cell-based applications has raised substantial hope for reversing these processes and inducing cardiac regeneration. However, current stem cell therapies using single-cell suspensions have failed to demonstrate long-lasting efficacy due to the overall low retention rate after cell delivery to the myocardium. To overcome this obstacle, the concept of 3D cell culture techniques has been proposed to enhance therapeutic efficacy and cell engraftment based on the simulation of an in vivo-like microenvironment. Of great interest is the use of so-called microtissues or spheroids, which have evolved from their traditional role as in vitro models to their novel role as therapeutic agents. This review will provide an overview of the therapeutic potential of microtissues by addressing primarily cardiovascular regeneration. It will accentuate their advantages compared to other regenerative approaches and summarize the methods for generating clinically applicable microtissues. In addition, this review will illustrate the unique properties of the microenvironment within microtissues that makes them a promising next-generation therapeutic approach.
Collapse
|
42
|
Han D, Huang W, Li X, Gao L, Su T, Li X, Ma S, Liu T, Li C, Chen J, Gao E, Cao F. Melatonin facilitates adipose-derived mesenchymal stem cells to repair the murine infarcted heart via the SIRT1 signaling pathway. J Pineal Res 2016; 60:178-92. [PMID: 26607398 DOI: 10.1111/jpi.12299] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 11/19/2015] [Indexed: 12/23/2022]
Abstract
Mesenchymal stem cells (MSCs)-based therapy provides a promising therapy for the ischemic heart disease (IHD). However, engrafted MSCs are subjected to acute cell death in the ischemic microenvironment, characterized by excessive inflammation and oxidative stress in the host's infarcted myocardium. Melatonin, an indole, which is produced by many organs including pineal gland, has been shown to protect bone marrow MSCs against apoptosis although the mechanism of action remains elusive. Using a murine model of myocardial infarction (MI), this study was designed to evaluate the impact of melatonin on adipose-derived mesenchymal stem cells (AD-MSCs)-based therapy for MI and the underlying mechanism involved with a focus on silent information regulator 1(SIRT1) signaling. Our results demonstrated that melatonin promoted functional survival of AD-MSCs in infarcted heart and provoked a synergetic effect with AD-MSCs to restore heart function. This in vivo effect of melatonin was associated with alleviated inflammation, apoptosis, and oxidative stress in infarcted heart. In vitro studies revealed that melatonin exert cytoprotective effects on AD-MSCs against hypoxia/serum deprivation (H/SD) injury via attenuating inflammation, apoptosis, and oxidative stress. Mechanistically, melatonin enhanced SIRT1 signaling, which was accompanied with the increased expression of anti-apoptotic protein Bcl2, and decreased the expression of Ac-FoxO1, Ac-p53, Ac-NF-ΚB, and Bax. Taken together, our findings indicated that melatonin facilitated AD-MSCs-based therapy in MI, possibly through promoting survival of AD-MSCs via SIRT1 signaling. Our data support the promise of melatonin as a novel strategy to improve MSC-based therapy for IHD, possibly through SIRT1 signaling evocation.
Collapse
Affiliation(s)
- Dong Han
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Wei Huang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiang Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lei Gao
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Tao Su
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiujuan Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Sai Ma
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tong Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jiangwei Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Erhe Gao
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Feng Cao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
43
|
Hsieh FY, Tseng TC, Hsu SH. Self-healing hydrogel for tissue repair in the central nervous system. Neural Regen Res 2016; 10:1922-3. [PMID: 26889169 PMCID: PMC4730805 DOI: 10.4103/1673-5374.169624] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
- Fu-Yu Hsieh
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, China
| | - Ting-Chen Tseng
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, China
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, China; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan, China; Research and Development Center for Medical Devices, National Taiwan University, Taipei, Taiwan, China
| |
Collapse
|
44
|
Gaetani R, Yin C, Srikumar N, Braden R, Doevendans PA, Sluijter JPG, Christman KL. Cardiac-Derived Extracellular Matrix Enhances Cardiogenic Properties of Human Cardiac Progenitor Cells. Cell Transplant 2015; 25:1653-1663. [PMID: 26572770 DOI: 10.3727/096368915x689794] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The use of biomaterials has been demonstrated as a viable strategy to promote cell survival and cardiac repair. However, limitations on combinational cell-biomaterial therapies exist, as cellular behavior is influenced by the microenvironment and physical characteristics of the material. Among the different scaffolds employed for cardiac tissue engineering, a myocardial matrix hydrogel has been shown to promote cardiogenesis in murine cardiac progenitor cells (mCPCs) in vitro. In this study, we investigated the influence of the hydrogel on Sca-1-like human fetal and adult CPCs (fCPCs and aCPCs) when encapsulated in three-dimensional (3D) material in vitro. fCPCs encapsulated in the myocardial matrix showed an increase in the gene expression level of cardiac markers GATA-4 and MLC2v and the vascular marker vascular endothelial growth factor receptor 2 (VEGFR2) after 4 days in culture, and a significant increase in GATA-4 up to 1 week. Increased gene expression levels of Nkx2.5, MEF2c, VEGFR2, and CD31 were also observed when aCPCs were cultured in the matrix compared to collagen. Cell survival was sustained in both hydrogels up to 1 week in culture with the myocardial matrix capable of enhancing the expression of the proliferation marker Ki-67 after 4 days in culture. When encapsulated CPCs were treated with H2O2, an improved survival of the cells cultured in the myocardial matrix was observed. Finally, we evaluated the use of the myocardial matrix as hydrogel for in vivo cell transplantation and demonstrated that the gelation properties of the hydrogel are not influenced by the cells. In summary, we showed that the myocardial matrix hydrogel promotes human CPC cardiogenic potential, proliferation, and survival and is a favorable hydrogel for 3D in vitro culture. Furthermore, we demonstrated the in vivo applicability of the matrix as a potential vehicle for cell transplantation.
Collapse
Affiliation(s)
- Roberto Gaetani
- Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Madonna R, Petrov L, Teberino MA, Manzoli L, Karam JP, Renna FV, Ferdinandy P, Montero-Menei CN, Ylä-Herttuala S, De Caterina R. Transplantation of adipose tissue mesenchymal cells conjugated with VEGF-releasing microcarriers promotes repair in murine myocardial infarction. Cardiovasc Res 2015; 108:39-49. [PMID: 26187727 DOI: 10.1093/cvr/cvv197] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 06/30/2015] [Indexed: 11/14/2022] Open
Abstract
RATIONALE Engraftment and survival of transplanted stem or stromal cells in the microenvironment of host tissues may be improved by combining such cells with scaffolds to delay apoptosis and enhance regenerative properties. AIMS We examined whether poly(lactic-co-glycolic acid) pharmacologically active microcarriers (PAMs) releasing vascular endothelial growth factor (VEGF) enhance survival, differentiation, and angiogenesis of adipose tissue-mesenchymal stromal cells (AT-MSCs). We analysed the efficacy of transplanted AT-MSCs conjugated with PAMs in a murine model of acute myocardial infarction (AMI). METHODS AND RESULTS We used fibronectin-coated (empty) PAMs or VEGF-releasing PAMs covered with murine AT-MSCs. Twelve-month-old C57 mice underwent coronary artery ligation to induce AMI, and were randomized into five treatment groups: AMI control (saline 20 µL, n = 7), AMI followed by intramyocardial injection with AT-MSCs (2.5 × 10(5) cells/20 µL, n = 5), or concentrated medium (CM) from AT-MSCs (20 µL, n = 8), or AT-MSCs (2.5 × 10(5) cells/20 µL) conjugated with empty PAMs (n = 7), or VEGF-releasing PAMs (n = 8). Sham-operated mice (n = 7) were used as controls. VEGF-releasing PAMs increased proliferation and angiogenic potential of AT-MSCs, but did not impact their osteogenic or adipogenic differentiation. AT-MSCs conjugated with VEGF-releasing PAMs inhibited apoptosis, decreased fibrosis, increased arteriogenesis and the number of cardiac-resident Ki-67 positive cells, and improved myocardial fractional shortening compared with AT-MSCs alone when transplanted into the infarcted hearts of C57 mice. With the exception of fractional shortening, all such effects of AT-MSCs conjugated with VEGF-PAMs were paralleled by the injection of CM. CONCLUSIONS AT-MSCs conjugated with VEGF-releasing PAMs exert paracrine effects that may have therapeutic applications.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Center of Excellence on Aging, 'G. d'Annunzio' University, Chieti, Italy
| | - Lyubomir Petrov
- Biocenter Kuopio, A. I. Virtanen Institute for Molecular Sciences, Kuopio, Finland
| | - Maria Anna Teberino
- Institute of Cardiology, Department of Neurosciences, Imaging, and Clinical Sciences, 'G. d'Annunzio' University, Chieti, Italy
| | - Lamberto Manzoli
- Department of Medicine and Aging Sciences, 'G. d'Annunzio' University, Chieti, Italy
| | - Jean-Pierre Karam
- INSERM U 1066, Micro et nanomédecine biomimétiques, LUNAM, Université d'Angers, Angers, France
| | - Francesca Vera Renna
- Institute of Cardiology, Department of Neurosciences, Imaging, and Clinical Sciences, 'G. d'Annunzio' University, Chieti, Italy
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Claudia N Montero-Menei
- INSERM U 1066, Micro et nanomédecine biomimétiques, LUNAM, Université d'Angers, Angers, France
| | - Seppo Ylä-Herttuala
- Biocenter Kuopio, A. I. Virtanen Institute for Molecular Sciences, Kuopio, Finland
| | - Raffaele De Caterina
- Center of Excellence on Aging, 'G. d'Annunzio' University, Chieti, Italy Institute of Cardiology, Department of Neurosciences, Imaging, and Clinical Sciences, 'G. d'Annunzio' University, Chieti, Italy
| |
Collapse
|
46
|
Kim J, Shapiro L, Flynn A. The clinical application of mesenchymal stem cells and cardiac stem cells as a therapy for cardiovascular disease. Pharmacol Ther 2015; 151:8-15. [DOI: 10.1016/j.pharmthera.2015.02.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 02/11/2015] [Indexed: 12/18/2022]
|
47
|
Tseng TC, Tao L, Hsieh FY, Wei Y, Chiu IM, Hsu SH. An Injectable, Self-Healing Hydrogel to Repair the Central Nervous System. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:3518-24. [PMID: 25953204 DOI: 10.1002/adma.201500762] [Citation(s) in RCA: 390] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/04/2015] [Indexed: 05/24/2023]
Abstract
An injectable, self-healing hydrogel (≈1.5 kPa) is developed for healing nerve-system deficits. Neurosphere-like progenitors proliferate in the hydrogel and differentiate into neuron-like cells. In the zebrafish injury model, the central nervous system function is partially rescued by injection of the hydrogel and significantly rescued by injection of the neurosphere-laden hydrogel. The self-healing hydrogel may thus potentially repair the central nervous system.
Collapse
Affiliation(s)
- Ting-Chen Tseng
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, 10617, Taiwan, R.O.C
| | - Lei Tao
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, P. R. China
| | - Fu-Yu Hsieh
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, 10617, Taiwan, R.O.C
| | - Yen Wei
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, P. R. China
| | - Ing-Ming Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan, R.O.C
| | - Shan-hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, 10617, Taiwan, R.O.C
| |
Collapse
|
48
|
Lin ZB, Qian B, Yang YZ, Zhou K, Sun J, Mo XM, Wu KH. Isolation, Characterization and Cardiac Differentiation of Human Thymus Tissue Derived Mesenchymal Stromal Cells. J Cell Biochem 2015; 116:1205-12. [PMID: 25535722 DOI: 10.1002/jcb.25072] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 12/18/2014] [Indexed: 01/08/2023]
Affiliation(s)
- Ze Bang Lin
- Department of Cardiothoracic Surgery; Nanjing Children‘s Hospital; Nanjing Medical University; Nanjing China
| | - Bo Qian
- Department of Cardiothoracic Surgery; Nanjing Children‘s Hospital; Nanjing Medical University; Nanjing China
| | - Yu Zhong Yang
- Department of Cardiothoracic Surgery; Nanjing Children‘s Hospital; Nanjing Medical University; Nanjing China
| | - Kai Zhou
- Department of Cardiothoracic Surgery; Nanjing Children‘s Hospital; Nanjing Medical University; Nanjing China
| | - Jian Sun
- Department of Cardiothoracic Surgery; Nanjing Children‘s Hospital; Nanjing Medical University; Nanjing China
| | - Xu Ming Mo
- Department of Cardiothoracic Surgery; Nanjing Children‘s Hospital; Nanjing Medical University; Nanjing China
| | - Kai Hong Wu
- Department of Cardiothoracic Surgery; Nanjing Children‘s Hospital; Nanjing Medical University; Nanjing China
| |
Collapse
|
49
|
Savi M, Bocchi L, Fiumana E, Karam JP, Frati C, Bonafé F, Cavalli S, Morselli PG, Guarnieri C, Caldarera CM, Muscari C, Montero-Menei CN, Stilli D, Quaini F, Musso E. Enhanced engraftment and repairing ability of human adipose-derived stem cells, conveyed by pharmacologically active microcarriers continuously releasing HGF and IGF-1, in healing myocardial infarction in rats. J Biomed Mater Res A 2015; 103:3012-25. [DOI: 10.1002/jbm.a.35442] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 02/09/2015] [Accepted: 02/19/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Monia Savi
- Department of Life Sciences; University of Parma; Parco Area delle Scienze 11/A 43124 Parma Italy
| | - Leonardo Bocchi
- Department of Life Sciences; University of Parma; Parco Area delle Scienze 11/A 43124 Parma Italy
| | - Emanuela Fiumana
- National Institute for Cardiovascular Research; Bologna Italy
- Department of Biomedical and Neuromotor Sciences; University of Bologna; Via Irnerio 48, 40126 Bologna Italy
| | - Jean-Pierre Karam
- Department of Biomedical and Neuromotor Sciences; University of Bologna; Via Irnerio 48, 40126 Bologna Italy
- UMR S-1066 F-49933; LUNAM University; Angers France
- INSERM U1066; MINT “Micro Et Nanomédecines Biomimétiques” F-49933; Angers France
| | - Caterina Frati
- Department of Clinical and Experimental Medicine; University of Parma; Via A. Gramsci 14 43126 Parma Italy
| | - Francesca Bonafé
- National Institute for Cardiovascular Research; Bologna Italy
- Department of Biomedical and Neuromotor Sciences; University of Bologna; Via Irnerio 48, 40126 Bologna Italy
| | - Stefano Cavalli
- Department of Clinical and Experimental Medicine; University of Parma; Via A. Gramsci 14 43126 Parma Italy
| | - Paolo G. Morselli
- Department of Specialist; Diagnostic and Experimental Medicine, University of Bologna; Bologna Italy
| | - Carlo Guarnieri
- National Institute for Cardiovascular Research; Bologna Italy
- Department of Biomedical and Neuromotor Sciences; University of Bologna; Via Irnerio 48, 40126 Bologna Italy
| | - Claudio M. Caldarera
- National Institute for Cardiovascular Research; Bologna Italy
- Department of Biomedical and Neuromotor Sciences; University of Bologna; Via Irnerio 48, 40126 Bologna Italy
| | - Claudio Muscari
- National Institute for Cardiovascular Research; Bologna Italy
- Department of Biomedical and Neuromotor Sciences; University of Bologna; Via Irnerio 48, 40126 Bologna Italy
| | - Claudia N. Montero-Menei
- UMR S-1066 F-49933; LUNAM University; Angers France
- INSERM U1066; MINT “Micro Et Nanomédecines Biomimétiques” F-49933; Angers France
| | - Donatella Stilli
- Department of Life Sciences; University of Parma; Parco Area delle Scienze 11/A 43124 Parma Italy
- National Institute for Cardiovascular Research; Bologna Italy
| | - Federico Quaini
- National Institute for Cardiovascular Research; Bologna Italy
- Department of Clinical and Experimental Medicine; University of Parma; Via A. Gramsci 14 43126 Parma Italy
| | - Ezio Musso
- Department of Life Sciences; University of Parma; Parco Area delle Scienze 11/A 43124 Parma Italy
- National Institute for Cardiovascular Research; Bologna Italy
| |
Collapse
|
50
|
Boyle PM, Karathanos TV, Trayanova NA. "Beauty is a light in the heart": the transformative potential of optogenetics for clinical applications in cardiovascular medicine. Trends Cardiovasc Med 2015; 25:73-81. [PMID: 25453984 PMCID: PMC4336805 DOI: 10.1016/j.tcm.2014.10.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 10/04/2014] [Accepted: 10/05/2014] [Indexed: 11/15/2022]
Abstract
Optogenetics is an exciting new technology in which viral gene or cell delivery is used to inscribe light sensitivity in excitable tissue to enable optical control of bioelectric behavior. Initial progress in the fledgling domain of cardiac optogenetics has included in vitro expression of various light-sensitive proteins in cell monolayers and transgenic animals to demonstrate an array of potentially useful applications, including light-based pacing, silencing of spontaneous activity, and spiral wave termination. In parallel to these developments, the cardiac modeling community has developed a versatile computational framework capable of realistically simulating optogenetics in biophysically detailed, patient-specific representations of the human heart, enabling the exploration of potential clinical applications in a predictive virtual platform. Toward the ultimate goal of assessing the feasibility and potential impact of optogenetics-based therapies in cardiovascular medicine, this review provides (1) a detailed synopsis of in vivo, in vitro, and in silico developments in the field and (2) a critical assessment of how existing clinical technology for gene/cell delivery and intra-cardiac illumination could be harnessed to achieve such lofty goals as light-based arrhythmia termination.
Collapse
Affiliation(s)
- Patrick M Boyle
- Institute for Computational Medicine, Johns Hopkins University, 316 Hackerman Hall, 3400 N Charles Street, Baltimore, MD 21218; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD.
| | - Thomas V Karathanos
- Institute for Computational Medicine, Johns Hopkins University, 316 Hackerman Hall, 3400 N Charles Street, Baltimore, MD 21218; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD
| | - Natalia A Trayanova
- Institute for Computational Medicine, Johns Hopkins University, 316 Hackerman Hall, 3400 N Charles Street, Baltimore, MD 21218; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD
| |
Collapse
|