1
|
Kishino Y, Tohyama S, Morita Y, Soma Y, Tani H, Okada M, Kanazawa H, Fukuda K. Cardiac Regenerative Therapy Using Human Pluripotent Stem Cells for Heart Failure: A State-of-the-Art Review. J Card Fail 2023; 29:503-513. [PMID: 37059512 DOI: 10.1016/j.cardfail.2022.10.433] [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: 07/14/2022] [Revised: 10/07/2022] [Accepted: 10/09/2022] [Indexed: 04/16/2023]
Abstract
Heart transplantation (HT) is the only definitive treatment available for patients with end-stage heart failure who are refractory to medical and device therapies. However, HT as a therapeutic option, is limited by a significant shortage of donors. To overcome this shortage, regenerative medicine using human pluripotent stem cells (hPSCs), such as human embryonic stem cells and human-induced pluripotent stem cells (hiPSCs), has been considered an alternative to HT. Several issues, including the methods of large-scale culture and production of hPSCs and cardiomyocytes, the prevention of tumorigenesis secondary to contamination of undifferentiated stem cells and non-cardiomyocytes, and the establishment of an effective transplantation strategy in large-animal models, need to be addressed to fulfill this unmet need. Although post-transplantation arrhythmia and immune rejection remain problems, the ongoing rapid technological advances in hPSC research have been directed toward the clinical application of this technology. Cell therapy using hPSC-derived cardiomyocytes is expected to serve as an integral component of realistic medicine in the near future and is being potentially viewed as a treatment that would revolutionize the management of patients with severe heart failure.
Collapse
Affiliation(s)
- Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan.
| | - Yuika Morita
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Yusuke Soma
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Hidenori Tani
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Marina Okada
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Hideaki Kanazawa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
2
|
Abdolahzadeh H, Rad NK, Shpichka A, Golroo R, Rahi K, Timashev P, Hassan M, Vosough M. Progress and promise of cell sheet assisted cardiac tissue engineering in regenerative medicine. Biomed Mater 2023; 18. [PMID: 36758240 DOI: 10.1088/1748-605x/acbad4] [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: 09/26/2022] [Accepted: 02/09/2023] [Indexed: 02/11/2023]
Abstract
Cardiovascular diseases (CVDs) are the most common leading causes of premature deaths in all countries. To control the harmful side effects of CVDs on public health, it is necessary to understand the current and prospective strategies in prevention, management, and monitoring CVDs.In vitro,recapitulating of cardiac complex structure with its various cell types is a challenging topic in tissue engineering. Cardiac tissue engineering (CTE) is a multi-disciplinary strategy that has been considered as a novel alternative approach for cardiac regenerative medicine and replacement therapies. In this review, we overview various cell types and approaches in cardiac regenerative medicine. Then, the applications of cell-sheet-assisted CTE in cardiac diseases were discussed. Finally, we described how this technology can improve cardiac regeneration and function in preclinical and clinical models.
Collapse
Affiliation(s)
- Hadis Abdolahzadeh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Khoshdel Rad
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Anastasia Shpichka
- World-Class Research Center 'Digital Biodesign and Personalized Healthcare', Sechenov University, Moscow, Russia.,Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.,Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Reihaneh Golroo
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Kosar Rahi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Peter Timashev
- World-Class Research Center 'Digital Biodesign and Personalized Healthcare', Sechenov University, Moscow, Russia.,Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.,Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Chemistry Department, Lomonosov Moscow State University, Moscow, Russia.,Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
3
|
Smart surface-based cell sheet engineering for regenerative medicine. TRENDS IN CHEMISTRY 2022. [DOI: 10.1016/j.trechm.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
4
|
Post AD, Buchan S, John M, Safavi-Naeini P, Cosgriff-Hernández E, Razavi M. Reconstituting electrical conduction in soft tissue: the path to replace the ablationist. Europace 2021; 23:1892-1902. [PMID: 34477862 DOI: 10.1093/europace/euab187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/08/2021] [Indexed: 11/13/2022] Open
Abstract
Cardiac arrhythmias are a leading cause of morbidity and mortality in the developed world. A common mechanism underlying many of these arrhythmias is re-entry, which may occur when native conduction pathways are disrupted, often by myocardial infarction. Presently, re-entrant arrhythmias are most commonly treated with antiarrhythmic drugs and myocardial ablation, although both treatment methods are associated with adverse side effects and limited efficacy. In recent years, significant advancements in the field of biomaterials science have spurred increased interest in the development of novel therapies that enable restoration of native conduction in damaged or diseased myocardium. In this review, we assess the current landscape of materials-based approaches to eliminating re-entrant arrhythmias. These approaches potentially pave the way for the eventual replacement of myocardial ablation as a preferred therapy for such pathologies.
Collapse
Affiliation(s)
- Allison D Post
- Electrophysiology Clinical Research and Innovations, Texas Heart Institute, 6770 Bertner Avenue, Houston, TX 77030, USA
| | - Skylar Buchan
- Electrophysiology Clinical Research and Innovations, Texas Heart Institute, 6770 Bertner Avenue, Houston, TX 77030, USA
| | - Mathews John
- Electrophysiology Clinical Research and Innovations, Texas Heart Institute, 6770 Bertner Avenue, Houston, TX 77030, USA
| | - Payam Safavi-Naeini
- Electrophysiology Clinical Research and Innovations, Texas Heart Institute, 6770 Bertner Avenue, Houston, TX 77030, USA
| | | | - Mehdi Razavi
- Electrophysiology Clinical Research and Innovations, Texas Heart Institute, 6770 Bertner Avenue, Houston, TX 77030, USA.,Department of Cardiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
5
|
Guo R, Wan F, Morimatsu M, Xu Q, Feng T, Yang H, Gong Y, Ma S, Chang Y, Zhang S, Jiang Y, Wang H, Chang D, Zhang H, Ling Y, Lan F. Cell sheet formation enhances the therapeutic effects of human umbilical cord mesenchymal stem cells on myocardial infarction as a bioactive material. Bioact Mater 2021; 6:2999-3012. [PMID: 33732969 PMCID: PMC7941025 DOI: 10.1016/j.bioactmat.2021.01.036] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Stem cell-based therapy has been used to treat ischaemic heart diseases for two decades. However, optimal cell types and transplantation methods remain unclear. This study evaluated the therapeutic effects of human umbilical cord mesenchymal stem cell (hUCMSC) sheet on myocardial infarction (MI). METHODS hUCMSCs expressing luciferase were generated by lentiviral transduction for in vivo bio-luminescent imaging tracking of cells. We applied a temperature-responsive cell culture surface-based method to form the hUCMSC sheet. Cell retention was evaluated using an in vivo bio-luminescent imaging tracking system. Unbiased transcriptional profiling of infarcted hearts and further immunohistochemical assessment of monocyte and macrophage subtypes were used to determine the mechanisms underlying the therapeutic effects of the hUCMSC sheet. Echocardiography and pathological analyses of heart sections were performed to evaluate cardiac function, angiogenesis and left ventricular remodelling. RESULTS When transplanted to the infarcted mouse hearts, hUCMSC sheet significantly improved the retention and survival compared with cell suspension. At the early stage of MI, hUCMSC sheet modulated inflammation by decreasing Mcp1-positive monocytes and CD68-positive macrophages and increasing Cx3cr1-positive non-classical macrophages, preserving the cardiomyocytes from acute injury. Moreover, the extracellular matrix produced by hUCMSC sheet then served as bioactive scaffold for the host cells to graft and generate new epicardial tissue, providing mechanical support and routes for revascularsation. These effects of hUCMSC sheet treatment significantly improved the cardiac function at days 7 and 28 post-MI. CONCLUSIONS hUCMSC sheet formation dramatically improved the biological functions of hUCMSCs, mitigating adverse post-MI remodelling by modulating the inflammatory response and providing bioactive scaffold upon transplantation into the heart. TRANSLATIONAL PERSPECTIVE Due to its excellent availability as well as superior local cellular retention and survival, allogenic transplantation of hUCMSC sheets can more effectively acquire the biological functions of hUCMSCs, such as modulating inflammation and enhancing angiogenesis. Moreover, the hUCMSC sheet method allows the transfer of an intact extracellular matrix without introducing exogenous or synthetic biomaterial, further improving its clinical applicability.
Collapse
Affiliation(s)
- Rui Guo
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Feng Wan
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
- Department of Cardiovascular Surgery, Tongji University East Hospital, Shanghai, 200120, China
| | - Masatoshi Morimatsu
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Qing Xu
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Tian Feng
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Hang Yang
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Yichen Gong
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Shuhong Ma
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Yun Chang
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Siyao Zhang
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Youxu Jiang
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Heqing Wang
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
- Department of Cardiovascular Surgery, Tongji University East Hospital, Shanghai, 200120, China
| | - Dehua Chang
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
- Department of Cardiac Surgery, The University of Tokyo Hospital, Tokyo, 113-8655, Japan
| | - Hongjia Zhang
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Yunpeng Ling
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Feng Lan
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, China
| |
Collapse
|
6
|
Abstract
Heart failure is a life-threatening disease prevalent worldwide. Cardiac transplantation is the last resort for patients with severe heart failure, but donor shortages represent a critical issue. Cardiac regenerative therapy is beneficial, but it is currently unsuitable as a substitute for cardiac transplantation. Human induced pluripotent stem cells (hiPSCs) are excellent sources for the generation of terminally differentiated cells. The preparation of a large number of pure cardiomyocytes (CMs) is the major premise for translational studies. To control the quality of the generated CMs, an efficient differentiation method, purification strategy, and mass-scale culture must be developed. Metabolic purification and large-scale culture systems have been established, and pure hiPSC-derived CMs of clinical grade are now available for translational research. The most critical challenge in cell therapy is the engraftment of transplanted cells. To overcome the low engraftment ratio of single CMs, aggregations of CMs are developed as cardiac spheroids. A cardiac transplantation device with domed tips and lateral holes has been developed for the transplantation of cardiac spheroids. Large animal models are necessary as the next step in the process toward clinical application. The transplant device has successfully been used to inject cardiac spheroids uniformly into myocardial layers in swine, and this approach is progressing toward clinical use. Remaining issues include immunological rejection and arrhythmia, which will require further investigation to establish safe and effective transplantation. This review summarizes the present status and future challenges of cardiac regenerative therapies.
Collapse
|
7
|
Monsanto MM, Wang BJ, Ehrenberg ZR, Echeagaray O, White KS, Alvarez R, Fisher K, Sengphanith S, Muliono A, Gude NA, Sussman MA. Enhancing myocardial repair with CardioClusters. Nat Commun 2020; 11:3955. [PMID: 32769998 PMCID: PMC7414230 DOI: 10.1038/s41467-020-17742-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 07/14/2020] [Indexed: 12/15/2022] Open
Abstract
Cellular therapy to treat heart failure is an ongoing focus of intense research, but progress toward structural and functional recovery remains modest. Engineered augmentation of established cellular effectors overcomes impediments to enhance reparative activity. Such 'next generation' implementation includes delivery of combinatorial cell populations exerting synergistic effects. Concurrent isolation and expansion of three distinct cardiac-derived interstitial cell types from human heart tissue, previously reported by our group, prompted design of a 3D structure that maximizes cellular interaction, allows for defined cell ratios, controls size, enables injectability, and minimizes cell loss. Herein, mesenchymal stem cells (MSCs), endothelial progenitor cells (EPCs) and c-Kit+ cardiac interstitial cells (cCICs) when cultured together spontaneously form scaffold-free 3D microenvironments termed CardioClusters. scRNA-Seq profiling reveals CardioCluster expression of stem cell-relevant factors, adhesion/extracellular-matrix molecules, and cytokines, while maintaining a more native transcriptome similar to endogenous cardiac cells. CardioCluster intramyocardial delivery improves cell retention and capillary density with preservation of cardiomyocyte size and long-term cardiac function in a murine infarction model followed 20 weeks. CardioCluster utilization in this preclinical setting establish fundamental insights, laying the framework for optimization in cell-based therapeutics intended to mitigate cardiomyopathic damage.
Collapse
Affiliation(s)
- Megan M Monsanto
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Bingyan J Wang
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Zach R Ehrenberg
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Oscar Echeagaray
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Kevin S White
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Roberto Alvarez
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Kristina Fisher
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Sharon Sengphanith
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Alvin Muliono
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Natalie A Gude
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Mark A Sussman
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA.
| |
Collapse
|
8
|
Adams JC, Bell PD, Bodine SC, Brooks HL, Bunnett N, Joe B, Keehan KH, Kleyman TR, Marette A, Morty RE, Ramírez JM, Thomsen MB, Yates BJ, Zucker IH. An American Physiological Society cross-journal Call for Papers on "Deconstructing Organs: Single-Cell Analyses, Decellularized Organs, Organoids, and Organ-on-a-Chip Models". Am J Physiol Lung Cell Mol Physiol 2020; 319:L266-L272. [PMID: 32609556 PMCID: PMC7473938 DOI: 10.1152/ajplung.00311.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Josephine C Adams
- School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - P Darwin Bell
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sue C Bodine
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Heddwen L Brooks
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona
| | - Nigel Bunnett
- Department of Molecular Pathobiology, New York University, New York, New York
| | - Bina Joe
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio.,Center for Hypertension and Personalized Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | | | - Thomas R Kleyman
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - André Marette
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Hôpital Laval, Quebec City, Quebec, Canada.,Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, Justus Liebig University Giessen, member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Jan-Marino Ramírez
- Department of Neurological Surgery, University of Washington Medical Center, Seattle, Washington.,Center on Human Development and Disability, University of Washington, Seattle, Washington.,Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington, Seattle, Washington
| | - Morten B Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bill J Yates
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Irving H Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
9
|
Abstract
The spectrum of ischemic heart diseases, encompassing acute myocardial infarction to heart failure, represents the leading cause of death worldwide. Although extensive progress in cardiovascular diagnoses and therapy has been made, the prevalence of the disease continues to increase. Cardiac regeneration has a promising perspective for the therapy of heart failure. Recently, extracellular matrix (ECM) has been shown to play an important role in cardiac regeneration and repair after cardiac injury. There is also evidence that the ECM could be directly used as a drug to promote cardiomyocyte proliferation and cardiac regeneration. Increasing evidence supports that applying ECM biomaterials to maintain heart function recovery is an important approach to apply the concept of cardiac regenerative medicine to clinical practice in the future. Here, we will introduce the essential role of cardiac ECM in cardiac regeneration and summarize the approaches of delivering ECM biomaterials to promote cardiac repair in this review.
Collapse
Affiliation(s)
- Haotong Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Minghui Bao
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
10
|
Kishino Y, Fujita J, Tohyama S, Okada M, Tanosaki S, Someya S, Fukuda K. Toward the realization of cardiac regenerative medicine using pluripotent stem cells. Inflamm Regen 2020; 40:1. [PMID: 31938077 PMCID: PMC6956487 DOI: 10.1186/s41232-019-0110-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 12/19/2019] [Indexed: 01/08/2023] Open
Abstract
Heart transplantation (HT) is the only radical treatment available for patients with end-stage heart failure that is refractory to optimal medical treatment and device therapies. However, HT as a therapeutic option is limited by marked donor shortage. To overcome this difficulty, regenerative medicine using human-induced pluripotent stem cells (hiPSCs) has drawn increasing attention as an alternative to HT. Several issues including the preparation of clinical-grade hiPSCs, methods for large-scale culture and production of hiPSCs and cardiomyocytes, prevention of tumorigenesis secondary to contamination of undifferentiated stem cells and non-cardiomyocytes, and establishment of an effective transplantation strategy need to be addressed to fulfill this unmet medical need. The ongoing rapid technological advances in hiPSC research have been directed toward the clinical application of this technology, and currently, most issues have been satisfactorily addressed. Cell therapy using hiPSC-derived cardiomyocytes is expected to serve as an integral component of realistic medicine in the near future and is being potentially viewed as a treatment that would revolutionize the management of patients with severe heart failure.
Collapse
Affiliation(s)
- Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582 Japan
| | - Jun Fujita
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582 Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582 Japan
| | - Marina Okada
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582 Japan
| | - Sho Tanosaki
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582 Japan
| | - Shota Someya
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582 Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582 Japan
| |
Collapse
|
11
|
Stem cell-derived cell sheet transplantation for heart tissue repair in myocardial infarction. Stem Cell Res Ther 2020; 11:19. [PMID: 31915074 PMCID: PMC6950817 DOI: 10.1186/s13287-019-1536-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/30/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022] Open
Abstract
Stem cell-derived sheet engineering has been developed as the next-generation treatment for myocardial infarction (MI) and offers attractive advantages in comparison with direct stem cell transplantation and scaffold tissue engineering. Furthermore, induced pluripotent stem cell-derived cell sheets have been indicated to possess higher potential for MI therapy than other stem cell-derived sheets because of their capacity to form vascularized networks for fabricating thickened human cardiac tissue and their long-term therapeutic effects after transplantation in MI. To date, stem cell sheet transplantation has exhibited a dramatic role in attenuating cardiac dysfunction and improving clinical manifestations of heart failure in MI. In this review, we retrospectively summarized the current applications and strategy of stem cell-derived cell sheet technology for heart tissue repair in MI.
Collapse
|
12
|
Wei Z, Gerecht S. A self-healing hydrogel as an injectable instructive carrier for cellular morphogenesis. Biomaterials 2018; 185:86-96. [PMID: 30236839 PMCID: PMC6432635 DOI: 10.1016/j.biomaterials.2018.09.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/06/2018] [Accepted: 09/02/2018] [Indexed: 12/24/2022]
Abstract
Transplantation of progenitor cells can accelerate tissue healing and regenerative processes. Nonetheless, direct cell delivery fails to support survival of transplanted cells or long-term treatment of vascular related diseases due to compromised vasculature and tissue conditions. Using injectable hydrogels that cross-link in situ, could protect cells in vivo, but their sol-gel transition is time-dependent and difficult to precisely control. Hydrogels with self-healing properties are proposed to address these limitations, yet current self-healing hydrogels lack bio-functionality, hindering the morphogenesis of delivered cells into a tissue structure. Here we establish a gelatin (Gtn)-based self-healing hydrogel cross-linked by oxidized dextran (Odex) as an injectable carrier for delivery of endothelial progenitors. The dynamic imine cross-links between Gtn and Odex confer the self-healing ability to the Gtn-l-Odex hydrogels following syringe injection. The self-healing Gtn-l-Odex not only protects the progenitors from injected shear force but it also allows controllable spatial/temporal placement of the cells. Moreover, owing to the cell-adhesive and proteolytic sites of Gtn, the Gtn-l-Odex hydrogels support complex vascular network formation from the endothelial progenitors, both in vitro and in vivo. This is the first report of injectable, self-healing hydrogels with biological properties promoting vascular morphogenesis, which holds great promise for accelerating the success of regenerative therapies.
Collapse
Affiliation(s)
- Zhao Wei
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology Physical-Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology Physical-Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
13
|
Yoshimoto T, Serikawa M, Higa K, Kitamura K, Kasahara M, Yamamoto M, Abe S. Effect of Mesenchymal Cells on Myoblast Sheets Embedded in Collagen Gel. THE BULLETIN OF TOKYO DENTAL COLLEGE 2018; 59:87-95. [DOI: 10.2209/tdcpublication.2017-0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | | | - Kazunari Higa
- Department of Ophthalmology/Cornea Center, Tokyo Dental College Ichikawa General Hospital
| | | | | | | | | |
Collapse
|
14
|
Okitsu K, Iritakenishi T, Iura A, Kuri M, Fujino Y. Femoral nerve block with propofol sedation versus general anesthesia in patients with severe cardiac dysfunction undergoing autologous myoblast sheet transplantation. J Anesth 2017; 31:672-677. [PMID: 28608253 DOI: 10.1007/s00540-017-2376-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/27/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE Regional anesthesia is more favorable than general anesthesia in patients with severe comorbidity; however, data on the superiority of peripheral nerve blocks over general anesthesia in patients with severe cardiac dysfunction are lacking. We aimed to demonstrate that peripheral nerve blocks reduce perioperative analgesic requirements and promote faster recovery compared to general anesthesia. METHODS We retrospectively evaluated intraoperative blood pressure, perioperative medications, and postoperative recovery in patients who underwent skeletal muscle harvesting for autologous myoblast sheet transplantation. We compared patients who received general anesthesia (group G, n = 27) to those who received femoral nerve block with propofol sedation (group B, n = 22). RESULTS Left ventricular ejection fraction was 24% on average, with no significant difference between groups. Compared with group G, a lower dose of propofol was used intraoperatively (1.25 versus 2.0 µg/mL, respectively; P < 0.001) and fewer patients required opioids (13.6 versus 100%, P < 0.01) in group B. Additionally, the lowest intraoperative mean blood pressure was higher (54 versus 48 mmHg, respectively; P = 0.02) in group B. More patients received postoperative analgesic drugs (51.9 versus 13.6%, P = 0.01) and they received them more frequently (1 [0-3] versus 0 [0-1], P = 0.02) in group G. The length of heart care unit stay was shorter in group B than group G (0 [0-18.5] versus 17 [0-47] h, respectively; P < 0.0001). CONCLUSIONS Femoral nerve block with sedation was more beneficial than general anesthesia in patients with severe cardiac dysfunction who underwent skeletal muscle harvesting for autologous myoblast sheet transplantation.
Collapse
Affiliation(s)
- Kenta Okitsu
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan.
| | - Takeshi Iritakenishi
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Akira Iura
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Michioki Kuri
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Yuji Fujino
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| |
Collapse
|
15
|
Liu GD, Zhou SF, Ding XC, Fang CL, Mi SY, Gao XC, Han Q. Soluble expression of recomb inant cMyc, Klf4, Oct4, and Sox2 proteins in bacteria and transduction into living cells. Int J Ophthalmol 2017; 10:560-566. [PMID: 28503428 DOI: 10.18240/ijo.2017.04.10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 12/08/2016] [Indexed: 01/07/2023] Open
Abstract
AIM To develop a new method to produce recombinant reprogramming proteins, cMyc, Klf4, Oct4, and Sox2, in soluble format with low cost for the generation of induced pluripotent stem cells (iPSCs). METHODS A short polypeptide sequence derived from the HIV trans-activator of transcription protein (TAT) and the nucleus localization signal (NLS) polypeptide were fused to the N terminus of the reprogramming proteins and they were constructed into pCold-SUMO vector which can extremely improve the solubility of recombinant proteins. Then these vector plasmids were transformed into E. coli BL21 (DE3) Chaperone competent cells for amplification. The solubility of these recombinant proteins was determined by SDS-PAGE and Coomassie brilliant blue staining. The recombinant proteins were purified by Ni-NTA resin and identified by Western blot. The transduction of these proteins into HEK 293T cells were evaluated by immunofluorescence staining. RESULTS These four reprogramming proteins could be produced in soluble format in pCold-SUMO expression vector system with the assistance of chaperone proteins in bacteria. The proteins were purified successfully with a purity of over 70% with a relative high transduction rate into 293 cells. CONCLUSION The results in the present study indicate the four important reprogramming proteins, cMyc, Klf4, Oct4, and Sox2, can be produced in soluble format in bacteria with low cost. Our new method thus might be expected to greatly contribute to the future study of iPSCs.
Collapse
Affiliation(s)
- Guo-Dan Liu
- Department of Ophthalmology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Shi-Feng Zhou
- Department of Emergency, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Xu-Chen Ding
- Department of Ophthalmology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Chun-Lai Fang
- Department of Ophthalmology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Shu-Yong Mi
- Department of Ophthalmology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Xiang-Chun Gao
- Department of Ophthalmology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Qing Han
- Department of Ophthalmology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| |
Collapse
|
16
|
Weigel T, Pfister T, Schmitz T, Jannasch M, Schürlein S, Hijailan RA, Walles H, Hansmann J. Flexible tissue-like electrode as a seamless tissue-electronic interface. ACTA ACUST UNITED AC 2017. [DOI: 10.1515/bnm-2017-0002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractCurrent implantable electrodes facilitate only a low cellular infiltration impairing the long-term integration into the host’s tissue. To accomplish a seamless electronic-tissue interface, conductive three-dimensional (3D) scaffolds were generated by carbonization of electro-spun fiber meshes. When introducing NaCl particles as porogens, tailored tissue-like electrodes were generated. Characterization of the porous 3D fiber electrodes demonstrated improved material and electrical characteristics compared to standard carbon fiber meshes or flat gold surfaces. The feasibility of the porous 3D electrodes was assessed by cell culture experiments, confirming the migration of cells into the electrode and the formation of contracting cardiomyocyte clusters. Finally, a complex cardiac co-culture system proved the integration of the tissue into the 3D electrode in long-term culture of 7 weeks. These results strengthen the development of tissue-like 3D scaffolds as alternative to two-dimensional (2D) electrodes.
Collapse
|
17
|
Rücker C, Kirch H, Pullig O, Walles H. Strategies and First Advances in the Development of Prevascularized Bone Implants. CURRENT MOLECULAR BIOLOGY REPORTS 2016; 2:149-157. [PMID: 27617188 PMCID: PMC4996880 DOI: 10.1007/s40610-016-0046-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Despite the great regenerative potential of human bone, large bone defects are a serious condition. Commonly, large defects are caused by trauma, bone disease, malignant tumor removal, and infection or medication-related osteonecrosis. Large defects necessitate clinical treatment in the form of autologous bone transplantation or implantation of biomaterials as well as the application of other available methods that enhance bone defect repair. The development and application of prevascularized bone implants are closely related to the development animal models and require dedicated methods in order to reliably predict possible clinical outcomes and the efficacy of implants. Cell sheet engineering, 3D-printing, arteriovenous loops, and naturally derived decellularized scaffolds and their respective testings in animal models are presented as alternative to the autologous bone graft in this article.
Collapse
Affiliation(s)
- Christoph Rücker
- Translational Center Würzburg ‘Regenerative therapies in oncology and musculoskeletal diseases’, Würzburg Branch of the Fraunhofer-Institute Interfacial Engineering and Biotechnology, IGB, Würzburg, Germany
| | - Holger Kirch
- Department Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
| | - Oliver Pullig
- Translational Center Würzburg ‘Regenerative therapies in oncology and musculoskeletal diseases’, Würzburg Branch of the Fraunhofer-Institute Interfacial Engineering and Biotechnology, IGB, Würzburg, Germany
| | - Heike Walles
- Translational Center Würzburg ‘Regenerative therapies in oncology and musculoskeletal diseases’, Würzburg Branch of the Fraunhofer-Institute Interfacial Engineering and Biotechnology, IGB, Würzburg, Germany
- Department Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
18
|
Sajgalik P, Grupper A, Edwards BS, Kushwaha SS, Stulak JM, Joyce DL, Joyce LD, Daly RC, Kara T, Schirger JA. Current Status of Left Ventricular Assist Device Therapy. Mayo Clin Proc 2016; 91:927-40. [PMID: 27378038 DOI: 10.1016/j.mayocp.2016.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 04/22/2016] [Accepted: 05/03/2016] [Indexed: 02/05/2023]
Abstract
Congestive heart failure (HF) remains a serious burden in the Western World. Despite advances in pharmacotherapy and resynchronization, many patients have progression to end-stage HF. These patients may be candidates for heart transplant or left ventricular assist device (LVAD) therapy. Heart transplants are limited by organ shortages and in some cases by patient comorbidities; therefore, LVAD therapy is emerging as a strategy of bridge to transplant or as a destination therapy in patients ineligible for transplant. Patients initially ineligible for a transplant may, in certain cases, become eligible for transplant after physiologic improvement with LVAD therapy, and a small number of patients with an LVAD may have sufficient recovery of myocardial function to allow device explantation. This clinically oriented review will describe (1) the most frequently used pump types and aspects of the continuous-flow physiology and (2) the clinical indications for and the shift toward the use of LVADs in less sick patients with HF. Additionally, we review complications of LVAD therapy and project future directions in this field. We referred to the Interagency Registry for Mechanically Assisted Circulatory Support, landmark trials, and results from recently published studies as major sources in obtaining recent outcomes, and we searched for related published literature via PubMed. This review focuses primarily on clinical practice for primary care physicians and non-HF cardiologists in the United States.
Collapse
Affiliation(s)
- Pavol Sajgalik
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN; Department of Internal Medicine, Cardioangiology, International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Avishay Grupper
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Brook S Edwards
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | | | - John M Stulak
- Division of Cardiovascular Surgery, Mayo Clinic, Rochester, MN
| | - David L Joyce
- Division of Cardiovascular Surgery, Mayo Clinic, Rochester, MN
| | - Lyle D Joyce
- Division of Cardiovascular Surgery, Mayo Clinic, Rochester, MN
| | - Richard C Daly
- Division of Cardiovascular Surgery, Mayo Clinic, Rochester, MN
| | - Tomas Kara
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN; Department of Internal Medicine, Cardioangiology, International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - John A Schirger
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN.
| |
Collapse
|
19
|
Cell Therapy in Ischemic Heart Disease: Interventions That Modulate Cardiac Regeneration. Stem Cells Int 2016; 2016:2171035. [PMID: 26880938 PMCID: PMC4736413 DOI: 10.1155/2016/2171035] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/26/2015] [Accepted: 11/10/2015] [Indexed: 12/15/2022] Open
Abstract
The incidence of severe ischemic heart disease caused by coronary obstruction has progressively increased. Alternative forms of treatment have been studied in an attempt to regenerate myocardial tissue, induce angiogenesis, and improve clinical conditions. In this context, cell therapy has emerged as a promising alternative using cells with regenerative potential, focusing on the release of paracrine and autocrine factors that contribute to cell survival, angiogenesis, and tissue remodeling. Evidence of the safety, feasibility, and potential effectiveness of cell therapy has emerged from several clinical trials using different lineages of adult stem cells. The clinical benefit, however, is not yet well established. In this review, we discuss the therapeutic potential of cell therapy in terms of regenerative and angiogenic capacity after myocardial ischemia. In addition, we addressed nonpharmacological interventions that may influence this therapeutic practice, such as diet and physical training. This review brings together current data on pharmacological and nonpharmacological approaches to improve cell homing and cardiac repair.
Collapse
|
20
|
RUSU E, NECULA LG, NEAGU AI, ALECU M, STAN C, ALBULESCU R, TANASE CP. Current status of stem cell therapy: opportunities and limitations. Turk J Biol 2016; 40:955-967. [DOI: 10.3906/biy-1506-95] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
21
|
Guyette JP, Charest JM, Mills RW, Jank BJ, Moser PT, Gilpin SE, Gershlak JR, Okamoto T, Gonzalez G, Milan DJ, Gaudette GR, Ott HC. Bioengineering Human Myocardium on Native Extracellular Matrix. Circ Res 2015; 118:56-72. [PMID: 26503464 DOI: 10.1161/circresaha.115.306874] [Citation(s) in RCA: 243] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 10/26/2015] [Indexed: 12/12/2022]
Abstract
RATIONALE More than 25 million individuals have heart failure worldwide, with ≈4000 patients currently awaiting heart transplantation in the United States. Donor organ shortage and allograft rejection remain major limitations with only ≈2500 hearts transplanted each year. As a theoretical alternative to allotransplantation, patient-derived bioartificial myocardium could provide functional support and ultimately impact the treatment of heart failure. OBJECTIVE The objective of this study is to translate previous work to human scale and clinically relevant cells for the bioengineering of functional myocardial tissue based on the combination of human cardiac matrix and human induced pluripotent stem cell-derived cardiomyocytes. METHODS AND RESULTS To provide a clinically relevant tissue scaffold, we translated perfusion-decellularization to human scale and obtained biocompatible human acellular cardiac scaffolds with preserved extracellular matrix composition, architecture, and perfusable coronary vasculature. We then repopulated this native human cardiac matrix with cardiomyocytes derived from nontransgenic human induced pluripotent stem cells and generated tissues of increasing 3-dimensional complexity. We maintained such cardiac tissue constructs in culture for 120 days to demonstrate definitive sarcomeric structure, cell and matrix deformation, contractile force, and electrical conduction. To show that functional myocardial tissue of human scale can be built on this platform, we then partially recellularized human whole-heart scaffolds with human induced pluripotent stem cell-derived cardiomyocytes. Under biomimetic culture, the seeded constructs developed force-generating human myocardial tissue and showed electrical conductivity, left ventricular pressure development, and metabolic function. CONCLUSIONS Native cardiac extracellular matrix scaffolds maintain matrix components and structure to support the seeding and engraftment of human induced pluripotent stem cell-derived cardiomyocytes and enable the bioengineering of functional human myocardial-like tissue of multiple complexities.
Collapse
Affiliation(s)
- Jacques P Guyette
- From the Center for Regenerative Medicine (J.P.G., J.M.C., B.J.J., P.T.M., S.E.G., T.O., G.G., H.C.O.), Cardiovascular Research Center (R.W.M., D.J.M.), Division of Cardiology (D.J.M.), and Division of Thoracic Surgery, Department of Surgery (H.C.O.), Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA (J.P.G., B.J.J., P.T.M., S.E.G., G.G., H.C.O.); Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA (J.R.G., G.R.G.); and Harvard Stem Cell Institute, Cambridge, MA (H.C.O.)
| | - Jonathan M Charest
- From the Center for Regenerative Medicine (J.P.G., J.M.C., B.J.J., P.T.M., S.E.G., T.O., G.G., H.C.O.), Cardiovascular Research Center (R.W.M., D.J.M.), Division of Cardiology (D.J.M.), and Division of Thoracic Surgery, Department of Surgery (H.C.O.), Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA (J.P.G., B.J.J., P.T.M., S.E.G., G.G., H.C.O.); Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA (J.R.G., G.R.G.); and Harvard Stem Cell Institute, Cambridge, MA (H.C.O.)
| | - Robert W Mills
- From the Center for Regenerative Medicine (J.P.G., J.M.C., B.J.J., P.T.M., S.E.G., T.O., G.G., H.C.O.), Cardiovascular Research Center (R.W.M., D.J.M.), Division of Cardiology (D.J.M.), and Division of Thoracic Surgery, Department of Surgery (H.C.O.), Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA (J.P.G., B.J.J., P.T.M., S.E.G., G.G., H.C.O.); Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA (J.R.G., G.R.G.); and Harvard Stem Cell Institute, Cambridge, MA (H.C.O.)
| | - Bernhard J Jank
- From the Center for Regenerative Medicine (J.P.G., J.M.C., B.J.J., P.T.M., S.E.G., T.O., G.G., H.C.O.), Cardiovascular Research Center (R.W.M., D.J.M.), Division of Cardiology (D.J.M.), and Division of Thoracic Surgery, Department of Surgery (H.C.O.), Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA (J.P.G., B.J.J., P.T.M., S.E.G., G.G., H.C.O.); Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA (J.R.G., G.R.G.); and Harvard Stem Cell Institute, Cambridge, MA (H.C.O.)
| | - Philipp T Moser
- From the Center for Regenerative Medicine (J.P.G., J.M.C., B.J.J., P.T.M., S.E.G., T.O., G.G., H.C.O.), Cardiovascular Research Center (R.W.M., D.J.M.), Division of Cardiology (D.J.M.), and Division of Thoracic Surgery, Department of Surgery (H.C.O.), Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA (J.P.G., B.J.J., P.T.M., S.E.G., G.G., H.C.O.); Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA (J.R.G., G.R.G.); and Harvard Stem Cell Institute, Cambridge, MA (H.C.O.)
| | - Sarah E Gilpin
- From the Center for Regenerative Medicine (J.P.G., J.M.C., B.J.J., P.T.M., S.E.G., T.O., G.G., H.C.O.), Cardiovascular Research Center (R.W.M., D.J.M.), Division of Cardiology (D.J.M.), and Division of Thoracic Surgery, Department of Surgery (H.C.O.), Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA (J.P.G., B.J.J., P.T.M., S.E.G., G.G., H.C.O.); Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA (J.R.G., G.R.G.); and Harvard Stem Cell Institute, Cambridge, MA (H.C.O.)
| | - Joshua R Gershlak
- From the Center for Regenerative Medicine (J.P.G., J.M.C., B.J.J., P.T.M., S.E.G., T.O., G.G., H.C.O.), Cardiovascular Research Center (R.W.M., D.J.M.), Division of Cardiology (D.J.M.), and Division of Thoracic Surgery, Department of Surgery (H.C.O.), Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA (J.P.G., B.J.J., P.T.M., S.E.G., G.G., H.C.O.); Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA (J.R.G., G.R.G.); and Harvard Stem Cell Institute, Cambridge, MA (H.C.O.)
| | - Tatsuya Okamoto
- From the Center for Regenerative Medicine (J.P.G., J.M.C., B.J.J., P.T.M., S.E.G., T.O., G.G., H.C.O.), Cardiovascular Research Center (R.W.M., D.J.M.), Division of Cardiology (D.J.M.), and Division of Thoracic Surgery, Department of Surgery (H.C.O.), Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA (J.P.G., B.J.J., P.T.M., S.E.G., G.G., H.C.O.); Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA (J.R.G., G.R.G.); and Harvard Stem Cell Institute, Cambridge, MA (H.C.O.)
| | - Gabriel Gonzalez
- From the Center for Regenerative Medicine (J.P.G., J.M.C., B.J.J., P.T.M., S.E.G., T.O., G.G., H.C.O.), Cardiovascular Research Center (R.W.M., D.J.M.), Division of Cardiology (D.J.M.), and Division of Thoracic Surgery, Department of Surgery (H.C.O.), Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA (J.P.G., B.J.J., P.T.M., S.E.G., G.G., H.C.O.); Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA (J.R.G., G.R.G.); and Harvard Stem Cell Institute, Cambridge, MA (H.C.O.)
| | - David J Milan
- From the Center for Regenerative Medicine (J.P.G., J.M.C., B.J.J., P.T.M., S.E.G., T.O., G.G., H.C.O.), Cardiovascular Research Center (R.W.M., D.J.M.), Division of Cardiology (D.J.M.), and Division of Thoracic Surgery, Department of Surgery (H.C.O.), Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA (J.P.G., B.J.J., P.T.M., S.E.G., G.G., H.C.O.); Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA (J.R.G., G.R.G.); and Harvard Stem Cell Institute, Cambridge, MA (H.C.O.)
| | - Glenn R Gaudette
- From the Center for Regenerative Medicine (J.P.G., J.M.C., B.J.J., P.T.M., S.E.G., T.O., G.G., H.C.O.), Cardiovascular Research Center (R.W.M., D.J.M.), Division of Cardiology (D.J.M.), and Division of Thoracic Surgery, Department of Surgery (H.C.O.), Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA (J.P.G., B.J.J., P.T.M., S.E.G., G.G., H.C.O.); Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA (J.R.G., G.R.G.); and Harvard Stem Cell Institute, Cambridge, MA (H.C.O.)
| | - Harald C Ott
- From the Center for Regenerative Medicine (J.P.G., J.M.C., B.J.J., P.T.M., S.E.G., T.O., G.G., H.C.O.), Cardiovascular Research Center (R.W.M., D.J.M.), Division of Cardiology (D.J.M.), and Division of Thoracic Surgery, Department of Surgery (H.C.O.), Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA (J.P.G., B.J.J., P.T.M., S.E.G., G.G., H.C.O.); Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA (J.R.G., G.R.G.); and Harvard Stem Cell Institute, Cambridge, MA (H.C.O.).
| |
Collapse
|
22
|
Garcia-Gomez I, Gudehithlu KP, Arruda JAL, Singh AK. Autologous tissue patch rich in stem cells created in the subcutaneous tissue. World J Stem Cells 2015; 7:1127-1136. [PMID: 26435772 PMCID: PMC4584236 DOI: 10.4252/wjsc.v7.i8.1127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/14/2015] [Accepted: 07/17/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether we could create natural autologous tissue patches in the subcutaneous space for organ repair.
METHODS: We implanted the following three types of inert foreign bodies in the subcutaneous tissue of rats to produce autologous tissue patches of different geometries: (1) a large-sized polyvinyl tube (L = 25 mm, internal diameter = 7 mm) sealed at both ends by heat application for obtaining a large flat piece of tissue patch for organ repair; (2) a fine polyvinyl tubing (L = 25 mm, internal diameter = 3 mm) for creating cylindrically shaped grafts for vascular or nerve repair; and (3) a slurry of polydextran particle gel for inducing a bladder-like tissue. Implantation of inert materials was carried out by making a small incision on one or either side of the thoracic-lumbar region of rats. Subcutaneous pockets were created by blunt dissection around the incision into which the inert bodies were inserted (1 or 2 per rat). The incisions were closed with silk sutures, and the animals were allowed to recover. In case of the polydextran gel slurry 5 mL of the slurry was injected in the subcutaneous space using an 18 gauge needle. After implanting the foreign bodies a newly regenerated encapsulating tissue developed around the foreign bodies. The tissues were harvested after 4-42 d of implantation and studied by gross examination, histology, and histochemistry for organization, vascularity, and presence of mesenchymal stem cells (MSCs) (CD271+CD34+ cells).
RESULTS: Implanting a large cylindrically shaped polyvinyl tube resulted in a large flat sheet of tissue that could be tailored to a specific size and shape for use as a tissue patch for repairing large organs. Implanting a smaller sized polyvinyl tube yielded a cylindrical tissue that could be useful for repairing nerves and blood vessels. This type of patch could be obtained in different lengths by varying the length of the implanted tube. Implanting a suspension of inert polydextran suspension gave rise to a bladder-like tissue that could be potentially used for repairing heart valves. Histologically, the three different types of tissue patches generated were organized similarly, consisting of three layers, increasing in thickness until day 14. The inner layer in contact with the inert material was avascular; a middle layer that was highly vascular and filled with matrix, and an outer layer consisting of loose connective tissue. MSCs identified as CD271+CD34+ cells were present in the medial layer and around major blood vessels at day 4 but absent at later time points. The early-harvested tissues, endowed with MSCs, could be used for tissue repair, while the later-harvested tissues, being less vascular but thicker and tougher, could be used as filler tissue for cosmetic purposes.
CONCLUSION: An autologous, vascularized tissue patch of desired shape and size can be created in the subcutaneous space by implanting different types of inert bodies.
Collapse
|
23
|
Nakajima K, Fujita J, Matsui M, Tohyama S, Tamura N, Kanazawa H, Seki T, Kishino Y, Hirano A, Okada M, Tabei R, Sano M, Goto S, Tabata Y, Fukuda K. Gelatin Hydrogel Enhances the Engraftment of Transplanted Cardiomyocytes and Angiogenesis to Ameliorate Cardiac Function after Myocardial Infarction. PLoS One 2015; 10:e0133308. [PMID: 26186362 PMCID: PMC4505846 DOI: 10.1371/journal.pone.0133308] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 06/25/2015] [Indexed: 11/18/2022] Open
Abstract
Cell transplantation therapy will mean a breakthrough in resolving the donor shortage in cardiac transplantation. Cardiomyocyte (CM) transplantation, however, has been relatively inefficient in restoring cardiac function after myocardial infarction (MI) due to low engraftment of transplanted CM. In order to ameliorate engraftment of CM, the novel transplantation strategy must be invented. Gelatin hydrogel (GH) is a biodegradable water-soluble polymer gel. Gelatin is made of collagen. Although we observed that collagen strongly induced the aggregation of platelets to potentially cause coronary microembolization, GH did not enhance thrombogenicity. Therefore, GH is a suitable biomaterial in the cell therapy after heart failure. To assess the effect of GH on the improvement of cardiac function, fetal rat CM (5×10(6) or 1x10(6) cells) were transplanted with GH (10 mg/ml) to infarcted hearts. We compared this group with sham operated rats, CM in phosphate buffered saline (PBS), only PBS, and only GH-transplanted groups. Three weeks after transplantation, cardiac function was evaluated by echocardiography. The echocardiography confirmed that transplantation of 5×10(6) CM with GH significantly improved cardiac systolic function, compared with the CM+PBS group (fractional area change: 75.1±3.4% vs. 60.7±5.9%, p<0.05), only PBS, and only GH groups (60.1±6.5%, 65.0±2.8%, p<0.05). Pathological analyses demonstrated that in the CM+GH group, CM were efficiently engrafted in infarcted myocardium (p<0.01) and angiogenesis was significantly enhanced (p<0.05) in both central and peripheral areas of the scar. Moreover, quantitative RT-PCR revealed that angiogenic cytokines, such as basic fibroblast growth factor, vascular endothelial growth factor, and hepatocyte growth factor, were significantly enriched in the CM+GH group (p<0.05). Here, we report that GH confined the CM effectively in infarcted myocardium after transplantation, and that CM transplanted with GH improved cardiac function with a direct contraction effect and enhanced angiogenesis.
Collapse
Affiliation(s)
- Kazuaki Nakajima
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, Japan
| | - Jun Fujita
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, Japan
- * E-mail:
| | - Makoto Matsui
- Department of Biomaterials, Field of Tissue Engineering, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho Shogoin, Sakyo-ku, Kyoto, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, Japan
- Japan Society for the Promotion of Science, Tokyo, Japan
| | - Noriko Tamura
- Department of Medicine (Cardiology), Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, Japan
| | - Hideaki Kanazawa
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, Japan
| | - Tomohisa Seki
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, Japan
| | - Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, Japan
| | - Akinori Hirano
- Department of Cardiovascular Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, Japan
| | - Marina Okada
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, Japan
| | - Ryota Tabei
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, Japan
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, Japan
| | - Shinya Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, Japan
| | - Yasuhiko Tabata
- Department of Biomaterials, Field of Tissue Engineering, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho Shogoin, Sakyo-ku, Kyoto, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
24
|
Abstract
OPINION STATEMENT Cell therapy can be administered via injections delivered directly into the myocardium or as engineered cardiac tissue patches, which are the subject of this review. Engineered cardiac patches can be created from sheets of interconnected cells or by suspending the cells in a scaffold of material that is designed to mimic the native extracellular matrix. The sheet-based approach produces patches with well-aligned and electronically coupled cardiomyocytes, but cell-containing scaffolds are more readily vascularized by the host's circulatory system and, consequently, are currently more suitable for applications that require a thicker patch. Cell patches can also be modified for the co-delivery of peptides that may promote cell survival and activate endogenous repair mechanisms; nevertheless, techniques for controlling inflammation, limiting apoptosis, and improving vascular growth need continue to be developed to make it a therapeutic modality for patients with myocardial infarction.
Collapse
Affiliation(s)
- Jianyi Zhang
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, 55455, USA,
| |
Collapse
|
25
|
Lee H, Cho Y. An Innovative Strategy for the Fabrication of Functional Cell Sheets Using an Electroactive Conducting Polymer. Theranostics 2015; 5:1021-9. [PMID: 26155317 PMCID: PMC4493539 DOI: 10.7150/thno.12389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 05/09/2015] [Indexed: 12/18/2022] Open
Abstract
Here, we report the development of an electric field-assisted methodology for constructing 3D C2C12 cell sheets with the potential for cell surface modification. In this method, a conducting polymer, polypyrrole (Ppy), is electrodeposited via biotin doping, and then chemical conjugation of biotinylated bone morphogenetic protein 2 (BMP2) is achieved using a biotin-streptavidin cross-linker. Subsequently, C2C12 cells are cultured on BMP2-immobilized Ppy surfaces to induce interactions between cell surface receptors and bound BMP2 ligands. Following these procedures, layers of BMP2-immobilized cells can be easily detached from the Ppy surface by applying an electrical potential. This novel method results in high affinity, ligand-bound cell sheets, which exhibit homogeneous coverage with membrane-bound proteins and signal activation that occurs via maximal receptor accessibility. Using this strategy to engineer the cell surface with desirable ligands results in structures that mimic in vivo tissues; thus, the method reported here has potential applications in regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
| | - Youngnam Cho
- New Experimental Therapeutic Branch, National Cancer Center, 111 Jungbalsan-ro, Ilsandong-gu, Goyang, Gyeonggi-do 410-769, South Korea
| |
Collapse
|
26
|
Seki T, Fukuda K. Methods of induced pluripotent stem cells for clinical application. World J Stem Cells 2015; 7:116-125. [PMID: 25621111 PMCID: PMC4300922 DOI: 10.4252/wjsc.v7.i1.116] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 09/18/2014] [Accepted: 10/27/2014] [Indexed: 02/06/2023] Open
Abstract
Reprograming somatic cells using exogenetic gene expression represents a groundbreaking step in regenerative medicine. Induced pluripotent stem cells (iPSCs) are expected to yield novel therapies with the potential to solve many issues involving incurable diseases. In particular, applying iPSCs clinically holds the promise of addressing the problems of immune rejection and ethics that have hampered the clinical applications of embryonic stem cells. However, as iPSC research has progressed, new problems have emerged that need to be solved before the routine clinical application of iPSCs can become established. In this review, we discuss the current technologies and future problems of human iPSC generation methods for clinical use.
Collapse
|
27
|
Fujita J, Fukuda K. Future Prospects for Regenerated Heart Using Induced Pluripotent Stem Cells. J Pharmacol Sci 2014; 125:1-5. [DOI: 10.1254/jphs.14r01cp] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
28
|
Stanley WC, Keehan KH. Update on innovative initiatives for the American Journal of Physiology-Heart and Circulatory Physiology. Am J Physiol Heart Circ Physiol 2013; 304:H1045-9. [PMID: 23457015 DOI: 10.1152/ajpheart.00082.2013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
29
|
Xue J, Feng B, Zheng R, Lu Y, Zhou G, Liu W, Cao Y, Zhang Y, Zhang WJ. Engineering ear-shaped cartilage using electrospun fibrous membranes of gelatin/polycaprolactone. Biomaterials 2013; 34:2624-31. [PMID: 23352044 DOI: 10.1016/j.biomaterials.2012.12.011] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 12/14/2012] [Indexed: 11/29/2022]
Abstract
Tissue engineering approach continuously requires for emerging strategies to improve the efficacy in repairing and regeneration of tissue defects. Previously, we developed a sandwich model strategy for cartilage engineering, using the combination of acellular cartilage sheets (ACSs) and chondrocytes. However, the process for the preparation of ACSs is complicated, and it is also difficult to obtain large ACSs. The aim of this study was to engineer cartilage with precise three-dimensional (3-D) structures by applying electrospun fibrous membranes of gelatin/polycaprolactone (GT/PCL). We first prepared the electrospun GT/PCL membranes into rounded shape, and then seeded chondrocytes in the sandwich model. After in vitro and in vivo cultivation, the newly formed cartilage-like tissues were harvested. Macroscopic observations and histological analysis confirmed that the engineering of cartilage using the electrospun GT/PCL membranes was feasible. An ear-shaped cartilage was then constructed in the sandwich model, with the help of an ear-shaped titanium alloy mold. After 2 weeks of culture in vitro and 6 weeks of subcutaneous incubation in vivo, the ear-shaped cartilage largely maintained their original shape, with a shape similarity up to 91.41% of the titanium mold. In addition, the engineered cartilage showed good elasticity and impressive mechanical strength. These results demonstrated that the engineering of 3-D cartilage in a sandwich model using electrospun fibrous membranes was a facile and effective approach, which has the potential to be applied for the engineering of other tissues with complicated 3-D structures.
Collapse
Affiliation(s)
- Jixin Xue
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, Shanghai 200011, China
| | | | | | | | | | | | | | | | | |
Collapse
|