1
|
In vitro controlled release of extracellular vesicles for cardiac repair from poly(glycerol sebacate) acrylate-based polymers. Acta Biomater 2020; 115:92-103. [PMID: 32814141 DOI: 10.1016/j.actbio.2020.08.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022]
Abstract
Cell therapy to restore cardiac function in chronic heart failure has been extensively studied. However, its therapeutic value is limited due to poor cell engraftment and survival and the therapeutic outcomes have been attributed to paracrine secretions such as extracellular vesicles (EV). The direct use of EV is an attractive therapeutic strategy and it has been shown that the kinetics of delivery of the EV to the targeted tissue may impact the outcomes. However, there are currently no technologies to deliver EV to the heart in a controlled and tunable manner. The objective of this study was to design a controlled release system, based on a photocurable adhesive polymer, to locally deliver EV to the cardiac tissue. We have first demonstrated that the adhesive polymer, PGSA-g-EG, did not impact the EV bioactivity in vitro and was biocompatible in vivo when tested in a rat model. Importantly, the polymer remained attached to the heart surface for at least 1 month. We have then evaluated and optimized the in vitro release kinetics of the EV from the PGSA-g-EG polymer. Freeze-dried EV formulations were developed to tune the release kinetics and maximize the loading in the polymeric material. Moreover, despite the instability of the EV in aqueous medium at 37°C, the PGSA-g-EG polymer was able to release bioactive EV for at least 14 days. Overall, these results suggest that the PGSA-g-EG is a suitable material to promote the controlled delivery of bioactive EV over an extended period of time. STATEMENT OF SIGNIFICANCE: Extracellular vesicles (EV) are an investigational class of therapeutics that has shown promise to restore cardiac function following an ischemic event. Furthermore, its translation to the clinics is expected to pose less regulatory challenges than cell-based therapies. However, EV therapeutic outcomes are likely to be impacted by the route of administration and the kinetics of delivery to the target tissue. Therefore, there is a need for biomaterial-based technologies to deliver, in a controlled and tunable manner, EV to the heart. The present study describes the use of PGSA-g-EG polymer as an adhesive cardiac patch with potential to enable the controlled delivery of bioactive EV over an extended period of time to the cardiac tissue.
Collapse
|
2
|
Engineering a naturally-derived adhesive and conductive cardiopatch. Biomaterials 2019; 207:89-101. [PMID: 30965152 DOI: 10.1016/j.biomaterials.2019.03.015] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 03/08/2019] [Accepted: 03/13/2019] [Indexed: 12/11/2022]
Abstract
Myocardial infarction (MI) leads to a multi-phase reparative process at the site of damaged heart that ultimately results in the formation of non-conductive fibrous scar tissue. Despite the widespread use of electroconductive biomaterials to increase the physiological relevance of bioengineered cardiac tissues in vitro, there are still several limitations associated with engineering biocompatible scaffolds with appropriate mechanical properties and electroconductivity for cardiac tissue regeneration. Here, we introduce highly adhesive fibrous scaffolds engineered by electrospinning of gelatin methacryloyl (GelMA) followed by the conjugation of a choline-based bio-ionic liquid (Bio-IL) to develop conductive and adhesive cardiopatches. These GelMA/Bio-IL adhesive patches were optimized to exhibit mechanical and conductive properties similar to the native myocardium. Furthermore, the engineered patches strongly adhered to murine myocardium due to the formation of ionic bonding between the Bio-IL and native tissue, eliminating the need for suturing. Co-cultures of primary cardiomyocytes and cardiac fibroblasts grown on GelMA/Bio-IL patches exhibited comparatively better contractile profiles compared to pristine GelMA controls, as demonstrated by over-expression of the gap junction protein connexin 43. These cardiopatches could be used to provide mechanical support and restore electromechanical coupling at the site of MI to minimize cardiac remodeling and preserve normal cardiac function.
Collapse
|
3
|
Samal R, Sappa PK, Gesell Salazar M, Wenzel K, Reinke Y, Völker U, Felix SB, Hammer E, Könemann S. Global secretome analysis of resident cardiac progenitor cells from wild-type and transgenic heart failure mice: Why ambience matters. J Cell Physiol 2018; 234:10111-10122. [PMID: 30575044 DOI: 10.1002/jcp.27677] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 10/04/2018] [Indexed: 01/08/2023]
Abstract
Resident cardiac progenitor cells (CPCs) have gained attention in cardiac regenerative medicine primarily due to their paracrine activity. In our current study we determined the role of pathological conditions such as heart failure on the autocrine-paracrine action of stem cell antigen-1 (Sca-1) expressing CPC. This comparative secretome profiling of Sca-1+ cells derived from transgenic heart failure (αMHC-cyclin-T1/Gαq overexpression [Cyc] cells) versus healthy (wild-type [Wt] cells) mice, achieved via mass-spectrometric quantification, enabled the identification of over 700 proteins. Our results demonstrate that the heart failure milieu caused a 2-fold enrichment of extracellular matrix proteins (ECM) like biglycan, versican, collagen XII, and angiogenic factors like heparan sulfate proteoglycan 2, plasminogen activator inhibitor 1 in the secretome. We further elucidated the direct influence of the secretome on the functional behavior of Sca-1 + cells via in vitro tube forming assay. Secreted factors present in the diseased milieu induced tube formation in Cyc cells (1.7-fold; p < 0.01) when compared with Wt cells after 24 hr of exposure. The presence of conditioned media moderately increased the proliferation of Cyc cells but had a more pronounced effect on Wt cells. Overall, these findings revealed global modifications in the secretory activity of adult Sca-1 + cells in the heart failure milieu. The secretion of ECM proteins and angiogenic factors, which are crucial for cardiac remodeling and recovery, was notably enriched in the supernatant of Cyc cells. Thus, during heart failure the microenvironment of Sca-1 + cells might favor angiogenesis and proliferation suggesting their potential to recover the damaged heart.
Collapse
Affiliation(s)
- Rasmita Samal
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research, partner site Greifswald), Greifswald, Germany
| | - Praveen Kumar Sappa
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,Department of Hematology and Oncology, Internal Medicine C, University Greifswald, Greifswald, Germany
| | - Manuela Gesell Salazar
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Kristin Wenzel
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research, partner site Greifswald), Greifswald, Germany
| | - Yvonne Reinke
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research, partner site Greifswald), Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research, partner site Greifswald), Greifswald, Germany
| | - Stephan Burkhard Felix
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research, partner site Greifswald), Greifswald, Germany
| | - Elke Hammer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research, partner site Greifswald), Greifswald, Germany
| | - Stephanie Könemann
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research, partner site Greifswald), Greifswald, Germany
| |
Collapse
|
4
|
Bei Y, Zhou Q, Sun Q, Xiao J. Telocytes in cardiac regeneration and repair. Semin Cell Dev Biol 2016; 55:14-21. [PMID: 26826525 DOI: 10.1016/j.semcdb.2016.01.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 01/24/2016] [Indexed: 02/08/2023]
Abstract
Telocytes (TCs) are a novel type of stromal cells reported by Popescu's group in 2010. The unique feature that distinguishes TCs from other "classical" stromal cells is their extremely long and thin telopodes (Tps). As evidenced by electron microscopy, TCs are widely distributed in almost all tissues and organs. TCs contribute to form a three-dimensional interstitial network and play as active regulators in intercellular communication via homocellular/heterocellular junctions or shed vesicles. Interestingly, increasing evidence suggests the potential role of TCs in regenerative medicine. Although the heart retains some limited endogenous regenerative capacity, cardiac regenerative and repair response is however insufficient to make up the loss of cardiomyocytes upon injury. Developing novel strategies to increase cardiomyocyte renewal and repair is of great importance for the treatment of cardiac diseases. In this review, we focus on the role of TCs in cardiac regeneration and repair. We particularly describe the intercellular communication between TCs and cardiomyocytes, stem/progenitor cells, endothelial cells, and fibroblasts. Also, we discuss the current knowledge about TCs in cardiac repair after myocardial injury, as well as their potential roles in cardiac development and aging. TC-based therapy or TC-derived exosome delivery might be used as novel therapeutic strategies to promote cardiac regeneration and repair.
Collapse
Affiliation(s)
- Yihua Bei
- Regeneration and Aging Lab, Experimental Center of Life Sciences, School of Life Science, Shanghai University, Shanghai 200444, China; Shanghai Key Laboratory of Bio-Energy Crops, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Qiulian Zhou
- Regeneration and Aging Lab, Experimental Center of Life Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Qi Sun
- Regeneration and Aging Lab, Experimental Center of Life Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Junjie Xiao
- Regeneration and Aging Lab, Experimental Center of Life Sciences, School of Life Science, Shanghai University, Shanghai 200444, China; Shanghai Key Laboratory of Bio-Energy Crops, School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
5
|
Albulescu R, Tanase C, Codrici E, Popescu DI, Cretoiu SM, Popescu LM. The secretome of myocardial telocytes modulates the activity of cardiac stem cells. J Cell Mol Med 2015; 19:1783-94. [PMID: 26176909 PMCID: PMC4549029 DOI: 10.1111/jcmm.12624] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 05/06/2015] [Indexed: 02/05/2023] Open
Abstract
Telocytes (TCs) are interstitial cells that are present in numerous organs, including the heart interstitial space and cardiac stem cell niche. TCs are completely different from fibroblasts. TCs release extracellular vesicles that may interact with cardiac stem cells (CSCs) via paracrine effects. Data on the secretory profile of TCs and the bidirectional shuttle vesicular signalling mechanism between TCs and CSCs are scarce. We aimed to characterize and understand the in vitro effect of the TC secretome on CSC fate. Therefore, we studied the protein secretory profile using supernatants from mouse cultured cardiac TCs. We also performed a comparative secretome analysis using supernatants from rat cultured cardiac TCs, a pure CSC line and TCs-CSCs in co-culture using (i) high-sensitivity on-chip electrophoresis, (ii) surface-enhanced laser desorption/ionization time-of-flight mass spectrometry and (iii) multiplex analysis by Luminex-xMAP. We identified several highly expressed molecules in the mouse cardiac TC secretory profile: interleukin (IL)-6, VEGF, macrophage inflammatory protein 1α (MIP-1α), MIP-2 and MCP-1, which are also present in the proteome of rat cardiac TCs. In addition, rat cardiac TCs secrete a slightly greater number of cytokines, IL-2, IL-10, IL-13 and some chemokines like, GRO-KC. We found that VEGF, IL-6 and some chemokines (all stimulated by IL-6 signalling) are secreted by cardiac TCs and overexpressed in co-cultures with CSCs. The expression levels of MIP-2 and MIP-1α increased twofold and fourfold, respectively, when TCs were co-cultured with CSCs, while the expression of IL-2 did not significantly differ between TCs and CSCs in mono culture and significantly decreased (twofold) in the co-culture system. These data suggest that the TC secretome plays a modulatory role in stem cell proliferation and differentiation.
Collapse
Affiliation(s)
- Radu Albulescu
- Biochemistry-Proteomics Department, Victor Babeş National Institute of PathologyBucharest, Romania
- National Institute for Chemical Pharmaceutical Research & DevelopmentBucharest, Romania
| | - Cristiana Tanase
- Biochemistry-Proteomics Department, Victor Babeş National Institute of PathologyBucharest, Romania
| | - Elena Codrici
- Biochemistry-Proteomics Department, Victor Babeş National Institute of PathologyBucharest, Romania
| | - Daniela I Popescu
- Biochemistry-Proteomics Department, Victor Babeş National Institute of PathologyBucharest, Romania
| | - Sanda M Cretoiu
- Division of Cell Biology and Histology, Carol Davila University of Medicine and PharmacyBucharest, Romania
- Department of Ultrastructural Pathology, Victor Babeş National Institute of PathologyBucharest, Romania
| | - Laurentiu M Popescu
- Division of Cell Biology and Histology, Carol Davila University of Medicine and PharmacyBucharest, Romania
- Department of Advanced Studies, Victor Babeş National Institute of PathologyBucharest, Romania
- * Correspondence to: Laurentiu M. POPESCU, MD, PhD, E-mail:
| |
Collapse
|
6
|
Choi HW, Zhou T, Singh M, Jabbour GE. Recent developments and directions in printed nanomaterials. NANOSCALE 2015; 7:3338-55. [PMID: 25366473 DOI: 10.1039/c4nr03915g] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
In this review, we survey several recent developments in printing of nanomaterials for contacts, transistors, sensors of various kinds, light-emitting diodes, solar cells, memory devices, and bone and organ implants. The commonly used nanomaterials are classified according to whether they are conductive, semiconducting/insulating or biological in nature. While many printing processes are covered, special attention is paid to inkjet printing and roll-to-roll printing in light of their complexity and popularity. In conclusion, we present our view of the future development of this field.
Collapse
Affiliation(s)
- Hyung Woo Choi
- Department of Chemical and Materials Engineering, University of Nevada, Reno, NV 89557, USA
| | | | | | | |
Collapse
|
7
|
Hua P, Liu J, Tao J, Liu J, Yang S. Influence of caspase-3 silencing on the proliferation and apoptosis of rat bone marrow mesenchymal stem cells under hypoxia. Int J Clin Exp Med 2015; 8:1624-1633. [PMID: 25932091 PMCID: PMC4402738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 01/05/2015] [Indexed: 06/04/2023]
Abstract
AIMS To investigate the effects of caspase-3 silencing on the proliferation and apoptosis of rat bone marrow mesenchymal stem cells (MSCs) under hypoxia. METHODS Rat bone marrow MSCs were transfected with a recombinant shRNA lentivirus targeting caspase-3 expression. Protein expression of caspase-3 was measured by western blotting. Cell proliferation was measured with MTS, and the cell cycle was analyzed by flow cytometry. The apoptosis rate was measured at various time points under hypoxia. Apoptotic morphology was assessed by Hoechst 33258 staining. mRNA levels of caspase-3, Bcl-2, and Bax were measured by real-time PCR. RESULTS Western blotting showed that the rat MSCs were stably transfected with the shRNA targeting caspase-3 by a significant reduction of caspase-3 expression. Silencing of caspase-3 expression resulted in a significant increase of MSC proliferation (P < 0.05), an increase of cells in S-phase (52.66 ± 0.30%), and a significant decrease of apoptotic MSCs (P < 0.05). These effects exhibited a slow increase during hypoxic culture. Furthermore, caspase-3 silencing significantly down-regulated mRNA expression of caspase-3 (P < 0.01) and Bax (P < 0.01), and up-regulated Bcl-2 mRNA expression (p < 0.01), thereby increasing the ratio of Bcl-2/Bax (P < 0.05). CONCLUSION Caspase-3 silencing modulates the cell cycle of MSCs, promotes cell proliferation, and enhances the anti-apoptotic capacity of MSCs under hypoxia in vitro.
Collapse
Affiliation(s)
- Ping Hua
- Department of Cardiovascular Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen UniversityGuangzhou 510120, China
| | - Jialiang Liu
- Department of Cardiac-Thoracic Surgery, Chengdu Fifth People’s HospitalChengdu 611130, China
| | - Jun Tao
- Department of Cardiovascular Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen UniversityGuangzhou 510120, China
| | - Jianyang Liu
- Department of Cardiovascular Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen UniversityGuangzhou 510120, China
| | - Songran Yang
- Department of Experimental Psychology, University of OxfordOxford OX1 3UD, United Kingdom
| |
Collapse
|
8
|
Lu ZX, Mao LL, Lian F, He J, Zhang WT, Dai CY, Xue S, Lu WG, Zhu HS. Cardioprotective activity of placental growth factor in a rat model of acute myocardial infarction: nanoparticle-based delivery versus direct myocardial injection. BMC Cardiovasc Disord 2014; 14:53. [PMID: 24742302 PMCID: PMC4014437 DOI: 10.1186/1471-2261-14-53] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 04/04/2014] [Indexed: 12/13/2022] Open
Abstract
Background To comparatively evaluate the cardioprotective activity of placental growth factor (PGF) delivered through direct injection and a nanoparticle-based system respectively and to study the underlying mechanisms in a rat model of acute myocardial infarction (AMI). Methods Poly lactic-co-glycolic acid (PLGA)-based PGF-carrying nanoparticles (PGF-PLGANPs) were created. The mean size and morphology of particles were analyzed with particle size analyzer and transmission electronic microscopy (TEM). Encapsulation efficiency and sustained-release dose curve were analyzed by ELISA. Sprague-Dawley rats were randomized into four groups (n = 10). While animals in the first group were left untreated as controls, those in the other 3 groups underwent surgical induction of AMI, followed by treatment with physiological saline, PGF, and PGF-PLGANPs, respectively. Cardiac function was evaluated by transthoracic echocardiography at 4 weeks after treatment. At 6 weeks, rats were sacrificed, infarction size was analyzed with Masson trichrome staining, and protein contents of TIMP-2, MT1-MMP and MMP-2 at the infarction border were determined by immunohistochemistry and western blotting analysis. Results PGF was released for at least 15 days, showing successful preparation of PGF-PLGANPs. Coronary artery ligation successfully induced AMI. Compared to physiological saline control, PGF, injected to the myocardium either as a nude molecule or in a form of nanoparticles, significantly reduced infarction size, improved cardiac function, and elevated myocardial expression of TIMP-2, MT1-MMP, and MMP-2 (P < 0.05). The effect of PGF-PLGANPs was more pronounced than that of non-encapsulated PGF (P < 0.05). Conclusion Target PGF delivery to myocardium may improve cardiac function after AMI in rats. PLGA-based nanoparticles appear to be a better approach to delivery PGF. PGF exerts its cardioprotective effect at least partially through regulating metalloproteinase-mediated myocardial tissue remodeling.
Collapse
Affiliation(s)
| | | | - Feng Lian
- Department of Cardiovascular Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pu-Jian Rd, Shanghai 200127, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
9
|
Lee JW, Lee SH, Youn YJ, Ahn MS, Kim JY, Yoo BS, Yoon J, Kwon W, Hong IS, Lee K, Kwan J, Park KS, Choi D, Jang YS, Hong MK. A randomized, open-label, multicenter trial for the safety and efficacy of adult mesenchymal stem cells after acute myocardial infarction. J Korean Med Sci 2014; 29:23-31. [PMID: 24431901 PMCID: PMC3890472 DOI: 10.3346/jkms.2014.29.1.23] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 11/06/2013] [Indexed: 12/20/2022] Open
Abstract
Recent studies suggest that the intracoronary administration of bone marrow (BM)-derived mesenchymal stem cells (MSCs) may improve left ventricular function in patients with acute myocardial infarction (AMI). However, there is still argumentative for the safety and efficacy of MSCs in the AMI setting. We thus performed a randomized pilot study to investigate the safety and efficacy of MSCs in patients with AMI. Eighty patients with AMI after successful reperfusion therapy were randomly assigned and received an intracoronary administration of autologous BM-derived MSCs into the infarct related artery at 1 month. During follow-up period, 58 patients completed the trial. The primary endpoint was changes in left ventricular ejection fraction (LVEF) by single-photon emission computed tomography (SPECT) at 6 month. We also evaluated treatment-related adverse events. The absolute improvement in the LVEF by SPECT at 6 month was greater in the BM-derived MSCs group than in the control group (5.9% ± 8.5% vs 1.6% ± 7.0%; P=0.037). There was no treatment-related toxicity during intracoronary administration of MSCs. No significant adverse cardiovascular events occurred during follow-up. In conclusion, the intracoronary infusion of human BM-derived MSCs at 1 month is tolerable and safe with modest improvement in LVEF at 6-month follow-up by SPECT. (ClinicalTrials.gov registration number: NCT01392105).
Collapse
Affiliation(s)
- Jun-Won Lee
- Division of Cardiology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Seung-Hwan Lee
- Division of Cardiology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Young-Jin Youn
- Division of Cardiology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Min-Soo Ahn
- Division of Cardiology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Jang-Young Kim
- Division of Cardiology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Byung-Su Yoo
- Division of Cardiology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Junghan Yoon
- Division of Cardiology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Woocheol Kwon
- Department of Radiology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - In-Soo Hong
- Department of Radiology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Kyounghoon Lee
- Department of Cardiology, Gachon Medical School, Gil Medical Center, Incheon, Korea
| | - Jun Kwan
- Division of Cardiology, Department of Internal Medicine, Inha University Hospital, Incheon, Korea
| | - Keum Soo Park
- Division of Cardiology, Department of Internal Medicine, Inha University Hospital, Incheon, Korea
| | - Donghoon Choi
- Yonsei Cardiovascular Center and Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Yang Soo Jang
- Yonsei Cardiovascular Center and Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Mun K. Hong
- Division of Cardiology, Department of Medicine, St. Luke's Roosevelt Hospital, Columbia University College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
10
|
Cardiac stem cell niche, MMP9, and culture and differentiation of embryonic stem cells. Methods Mol Biol 2013; 1035:153-63. [PMID: 23959989 DOI: 10.1007/978-1-62703-508-8_13] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Embryonic stem cells (ESC) are totipotent, self-renewing, and clonogenic, having potential to differentiate into a wide variety of cell types. Due to regenerative capability, it has tremendous potential for treating myocardial infarction (death of myocardial tissue) and type 1 diabetes (death of pancreatic beta cells). Understanding the components regulating ESC differentiation is the key to unlock the regenerative potential of ESC-based therapies. Both the stiffness of extracellular matrix (ECM) and surrounding niche/microenvironment play pivotal roles in ESC differentiation. Matrix metalloproteinase-9 (MMP9) induces fibrosis that causes stiffness of the ECM and impairs differentiation of cardiac stem cells into cardiomyocytes. Here, we describe the method of ESC culture and differentiation, and the expression of MMP9 and its inhibitor, tissue inhibitor of metalloproteinase-4 (TIMP4) in differentiating ESC.
Collapse
|