1
|
Lv X, Liu B, Su X, Tian X, Wang H. Unlocking cardioprotection: iPSC exosomes deliver Nec-1 to target PARP1/AIFM1 axis, alleviating HF oxidative stress and mitochondrial dysfunction. J Transl Med 2024; 22:681. [PMID: 39061056 PMCID: PMC11282728 DOI: 10.1186/s12967-024-05204-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 04/15/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Heart failure (HF) is characterized by oxidative stress and mitochondrial dysfunction. This study investigates the therapeutic potential of Necrostatin-1 (Nec-1) delivered through exosomes derived from induced pluripotent stem cells (iPSCs) to address these pathologies in HF. METHODS An HF rat model was established, and comprehensive assessments were performed using echocardiography, hemodynamics, and ventricular mass index measurements. iPSCs were used to isolate exosomes, loaded with Nec-1, and characterized for efficient delivery into cardiomyocytes. The interaction between Nec-1-loaded exosomes (Nec-1-Exos), poly (ADP-ribose) polymerase 1 (PARP1), and apoptosis-inducing factor mitochondria-associated 1 (AIFM1) was explored. Gain-of-function experiments assessed changes in cardiomyocyte parameters, and histological analyses were conducted on myocardial tissues. RESULTS Cardiomyocytes successfully internalized Nec-1-loaded exosomes, leading to downregulation of PARP1, inhibition of AIFM1 nuclear translocation, increased ATP and superoxide dismutase levels, reduced reactive oxygen species and malonaldehyde levels, and restored mitochondrial membrane potential. Histological examinations confirmed the modulation of the PARP1/AIFM1 axis by Nec-1, mitigating HF. CONCLUSIONS iPSC-derived exosomes carrying Nec-1 attenuate oxidative stress and mitochondrial dysfunction in HF by targeting the PARP1/AIFM1 axis. This study proposes a promising therapeutic strategy for HF management and highlights the potential of exosome-mediated drug delivery.
Collapse
Affiliation(s)
- Xiaobing Lv
- Department of Cardiology, Jinan Central Hospital, Shandong University, No.105 Jiefang Road, Lixia District, Jinan, Shandong Province, 250013, P. R. China
- Department of Cardiology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, P.R. China
| | - Boqin Liu
- Department of Cardiology, Qingdao Municipal Hospital (West Yard), Qingdao, 266000, P.R. China
| | - Xiaoting Su
- Department of Obstetric, the Affiliated Hospital of Qingdao University, Qingdao, 266000, P.R. China
| | - Xintao Tian
- Department of Emergency Internal Medicine, the Affiliated Hospital of Qingdao University, Qingdao, 266000, P.R. China
| | - Huating Wang
- Department of Cardiology, Jinan Central Hospital, Shandong University, No.105 Jiefang Road, Lixia District, Jinan, Shandong Province, 250013, P. R. China.
| |
Collapse
|
2
|
Goto T, Nakamura Y, Ito Y, Miyagawa S. Regenerative medicine in cardiovascular disease. Regen Ther 2024; 26:859-866. [PMID: 39430582 PMCID: PMC11490749 DOI: 10.1016/j.reth.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 10/22/2024] Open
Abstract
Owing to the rapid increase in the number of people with severe heart failure, regenerative medicine is anticipated to play a role in overcoming the limitations inherent in existing surgical interventions. There are essentially two types of cardiac regenerative therapies for a failing heart. Cellular regenerative therapies using various stem cells improve the functional recovery of the heart mainly by cytokine paracrine effects. The implantation of induced pluripotent stem cell-derived cardiomyocytes can contribute not only to the inhibition of adverse heart remodeling by paracrine effects but also to the supply of newly born functional myocytes with the recipient myocardium as "mechanically working cells." Cell transplantation, including autologous myoblast transplantation, reduces heart failure exacerbations and benefits patients without the need for other treatment options. Although cellular therapy is currently the mainstream approach, it requires an in-house cell-processing center with an aseptic environment. In addition, these stem cells are usually introduced via several invasive delivery methods, including intracoronary administration, and cellular sheet implantation. Simplifying the culture methods for these cells is a crucial problem that needs to be resolved. Drug-induced regenerative therapy is another option that enhances self-endogenous regenerative systems in the human body and does not require invasive methods or cell cultures. Therefore, drug-induced regenerative therapies may overcome the disadvantages of these cellular therapies. The purpose of this report is to summarize cell transplantation therapy in the cardiovascular system and regenerative therapy for heart failure using an autologous endogenous regenerative system.
Collapse
Affiliation(s)
- Takasumi Goto
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Cardiovascular Surgery, Toyonaka Municipal Hospital, Osaka, Japan
| | - Yuki Nakamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshito Ito
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
3
|
He X, Liang J, Paul C, Huang W, Dutta S, Wang Y. Advances in Cellular Reprogramming-Based Approaches for Heart Regenerative Repair. Cells 2022; 11:3914. [PMID: 36497171 PMCID: PMC9740402 DOI: 10.3390/cells11233914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/18/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Continuous loss of cardiomyocytes (CMs) is one of the fundamental characteristics of many heart diseases, which eventually can lead to heart failure. Due to the limited proliferation ability of human adult CMs, treatment efficacy has been limited in terms of fully repairing damaged hearts. It has been shown that cell lineage conversion can be achieved by using cell reprogramming approaches, including human induced pluripotent stem cells (hiPSCs), providing a promising therapeutic for regenerative heart medicine. Recent studies using advanced cellular reprogramming-based techniques have also contributed some new strategies for regenerative heart repair. In this review, hiPSC-derived cell therapeutic methods are introduced, and the clinical setting challenges (maturation, engraftment, immune response, scalability, and tumorigenicity), with potential solutions, are discussed. Inspired by the iPSC reprogramming, the approaches of direct cell lineage conversion are merging, such as induced cardiomyocyte-like cells (iCMs) and induced cardiac progenitor cells (iCPCs) derived from fibroblasts, without induction of pluripotency. The studies of cellular and molecular pathways also reveal that epigenetic resetting is the essential mechanism of reprogramming and lineage conversion. Therefore, CRISPR techniques that can be repurposed for genomic or epigenetic editing become attractive approaches for cellular reprogramming. In addition, viral and non-viral delivery strategies that are utilized to achieve CM reprogramming will be introduced, and the therapeutic effects of iCMs or iCPCs on myocardial infarction will be compared. After the improvement of reprogramming efficiency by developing new techniques, reprogrammed iCPCs or iCMs will provide an alternative to hiPSC-based approaches for regenerative heart therapies, heart disease modeling, and new drug screening.
Collapse
Affiliation(s)
- Xingyu He
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jialiang Liang
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Christian Paul
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Wei Huang
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Suchandrima Dutta
- Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Yigang Wang
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
4
|
Yoshida S, Miyagawa S, Matsuzaki T, Ishii Y, Fukuda-Kawaguchi E, Kawamura T, Kawamura A, Nakamura Y, Toda K, Sawa Y. Chimerism through the activation of invariant natural killer T cells prolongs graft survival after transplantation of induced pluripotent stem cell–derived allogeneic cardiomyocytes. PLoS One 2022; 17:e0264317. [PMID: 35235568 PMCID: PMC8890721 DOI: 10.1371/journal.pone.0264317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 02/08/2022] [Indexed: 11/21/2022] Open
Abstract
The loss of functional cells through immunological rejection after transplantation reduces the efficacy of regenerative therapies for cardiac failure that use allogeneic induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). Recently, mixed-chimera mice with donor-specific immunotolerance have been established using the RGI-2001 (liposomal formulation of α-galactosyl ceramide) ligand, which activates invariant natural killer T (iNKT) cells. The present study aimed to investigate whether mixed chimerism, established using RGI-2001, prolongs graft survival in allogeneic iPSC-CM transplantation. Mixed-chimera mice were established via combinatorial treatment with RGI-2001 and anti-CD154 antibodies in an irradiated murine bone marrow transplant model. Luciferase-expressing allogeneic iPSC-CMs were transplanted into mixed-chimera and untreated mice, followed by in vivo imaging. RGI-2001 enhanced iNKT cell activation in mice, and mixed chimerism was successfully established. In vivo imaging revealed that while the allografts were completely obliterated within 2 weeks when transplanted to untreated mice, their survivals were not affected in the mixed-chimera mice. Furthermore, numerous CD3+ cells infiltrated allografts in untreated mice, but fewer CD3+ cells were present in mixed-chimera mice. We conclude that mixed-chimera mice established using RGI-2001 showed prolonged graft survival after allogeneic iPSC-CM transplantation. This donor-specific immunotolerance might increase the efficacy of regenerative therapies for heart failure with allogeneic iPSC-CMs.
Collapse
Affiliation(s)
- Shohei Yoshida
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takashi Matsuzaki
- Department of DDS Pharmaceutical Development, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasuyuki Ishii
- REGiMMUNE Corp, Tokyo, Japan
- Department of Immunological Diagnosis, Juntendo University Graduate School of Medicine, Bunkyo City, Japan
| | | | - Takuji Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ai Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuki Nakamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koichi Toda
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- * E-mail:
| |
Collapse
|
5
|
Nakamura M, Yaku H, Ako J, Arai H, Asai T, Chikamori T, Daida H, Doi K, Fukui T, Ito T, Kadota K, Kobayashi J, Komiya T, Kozuma K, Nakagawa Y, Nakao K, Niinami H, Ohno T, Ozaki Y, Sata M, Takanashi S, Takemura H, Ueno T, Yasuda S, Yokoyama H, Fujita T, Kasai T, Kohsaka S, Kubo T, Manabe S, Matsumoto N, Miyagawa S, Mizuno T, Motomura N, Numata S, Nakajima H, Oda H, Otake H, Otsuka F, Sasaki KI, Shimada K, Shimokawa T, Shinke T, Suzuki T, Takahashi M, Tanaka N, Tsuneyoshi H, Tojo T, Une D, Wakasa S, Yamaguchi K, Akasaka T, Hirayama A, Kimura K, Kimura T, Matsui Y, Miyazaki S, Okamura Y, Ono M, Shiomi H, Tanemoto K. JCS 2018 Guideline on Revascularization of Stable Coronary Artery Disease. Circ J 2022; 86:477-588. [DOI: 10.1253/circj.cj-20-1282] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Masato Nakamura
- Division of Cardiovascular Medicine, Toho University Ohashi Medical Center
| | - Hitoshi Yaku
- Department of Cardiovascular Surgery, Kyoto Prefectural University of Medicine
| | - Junya Ako
- Department of Cardiovascular Medicine, Kitasato University Graduate School of Medical Sciences
| | - Hirokuni Arai
- Department of Cardiovascular Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Tohru Asai
- Department of Cardiovascular Surgery, Juntendo University Graduate School of Medicine
| | | | - Hiroyuki Daida
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine
| | - Kiyoshi Doi
- General and Cardiothoracic Surgery, Gifu University Graduate School of Medicine
| | - Toshihiro Fukui
- Department of Cardiovascular Surgery, Graduate School of Medical Sciences, Kumamoto University
| | - Toshiaki Ito
- Department of Cardiovascular Surgery, Japanese Red Cross Nagoya Daiichi Hospital
| | | | - Junjiro Kobayashi
- Department of Cardiovascular Surgery, National Cerebral and Cardiovascular Center
| | - Tatsuhiko Komiya
- Department of Cardiovascular Surgery, Kurashiki Central Hospital
| | - Ken Kozuma
- Department of Internal Medicine, Teikyo University Faculty of Medicine
| | - Yoshihisa Nakagawa
- Department of Cardiovascular Medicine, Shiga University of Medical Science
| | - Koichi Nakao
- Division of Cardiology, Saiseikai Kumamoto Hospital Cardiovascular Center
| | - Hiroshi Niinami
- Department of Cardiovascular Surgery, Tokyo Women’s Medical University
| | - Takayuki Ohno
- Department of Cardiovascular Surgery, Mitsui Memorial Hospital
| | - Yukio Ozaki
- Department of Cardiology, Fujita Health University Hospital
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | | | - Hirofumi Takemura
- Department of Cardiovascular Surgery, Graduate School of Medical Sciences, Kanazawa University
| | | | - Satoshi Yasuda
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| | - Hitoshi Yokoyama
- Department of Cardiovascular Surgery, Fukushima Medical University
| | - Tomoyuki Fujita
- Department of Cardiovascular Surgery, National Cerebral and Cardiovascular Center
| | - Tokuo Kasai
- Department of Cardiology, Uonuma Institute of Community Medicine, Niigata University Uonuma Kikan Hospital
| | - Shun Kohsaka
- Department of Cardiology, Keio University School of Medicine
| | - Takashi Kubo
- Department of Cardiovascular Medicine, Wakayama Medical University
| | - Susumu Manabe
- Department of Cardiovascular Surgery, Tsuchiura Kyodo General Hospital
| | | | - Shigeru Miyagawa
- Frontier of Regenerative Medicine, Graduate School of Medicine, Osaka University
| | - Tomohiro Mizuno
- Department of Cardiovascular Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Noboru Motomura
- Department of Cardiovascular Surgery, Graduate School of Medicine, Toho University
| | - Satoshi Numata
- Department of Cardiovascular Surgery, Kyoto Prefectural University of Medicine
| | - Hiroyuki Nakajima
- Department of Cardiovascular Surgery, Saitama Medical University International Medical Center
| | - Hirotaka Oda
- Department of Cardiology, Niigata City General Hospital
| | - Hiromasa Otake
- Department of Cardiovascular Medicine, Kobe University Graduate School of Medicine
| | - Fumiyuki Otsuka
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| | - Ken-ichiro Sasaki
- Division of Cardiovascular Medicine, Kurume University School of Medicine
| | - Kazunori Shimada
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine
| | - Tomoki Shimokawa
- Department of Cardiovascular Surgery, Sakakibara Heart Institute
| | - Toshiro Shinke
- Division of Cardiology, Department of Medicine, Showa University School of Medicine
| | - Tomoaki Suzuki
- Department of Cardiovascular Surgery, Shiga University of Medical Science
| | - Masao Takahashi
- Department of Cardiovascular Surgery, Hiratsuka Kyosai Hospital
| | - Nobuhiro Tanaka
- Department of Cardiology, Tokyo Medical University Hachioji Medical Center
| | | | - Taiki Tojo
- Department of Cardiovascular Medicine, Kitasato University Graduate School of Medical Sciences
| | - Dai Une
- Department of Cardiovascular Surgery, Okayama Medical Center
| | - Satoru Wakasa
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Graduate School of Medicine
| | - Koji Yamaguchi
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Takashi Akasaka
- Department of Cardiovascular Medicine, Wakayama Medical University
| | | | - Kazuo Kimura
- Cardiovascular Center, Yokohama City University Medical Center
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University
| | - Yoshiro Matsui
- Department of Cardiovascular and Thoracic Surgery, Graduate School of Medicine, Hokkaido University
| | - Shunichi Miyazaki
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Kindai University
| | | | - Minoru Ono
- Department of Cardiac Surgery, Graduate School of Medicine, The University of Tokyo
| | - Hiroki Shiomi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University
| | - Kazuo Tanemoto
- Department of Cardiovascular Surgery, Kawasaki Medical School
| | | |
Collapse
|
6
|
Krog RT, de Miranda NFCC, Vahrmeijer AL, Kooreman NG. The Potential of Induced Pluripotent Stem Cells to Advance the Treatment of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13225789. [PMID: 34830945 PMCID: PMC8616212 DOI: 10.3390/cancers13225789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/12/2021] [Accepted: 11/13/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Despite improvements in the treatment of several cancer types, the extremely poor prognosis of pancreatic cancer patients has remained unchanged over the last decades. Therefore, new therapeutic regimens for pancreatic cancer are highly needed. In this review, we will discuss the potential of induced pluripotent stem cells (iPSCs) to generate representative pancreatic cancer models that can aid the development of novel diagnostics and therapeutic strategies. Furthermore, the potential of iPSCs as pancreatic cancer vaccines or as a basis for cellular therapies will be discussed. With promising preclinical results and ongoing clinical trials, the potential of iPSCs to further the treatment of pancreatic cancer is being explored and, in turn, will hopefully provide additional therapies to increase the poor survival rates of this patient population. Abstract Advances in the treatment of pancreatic ductal adenocarcinoma (PDAC) using neoadjuvant chemoradiotherapy, chemotherapy, and immunotherapy have had minimal impact on the overall survival of patients. A general lack of immunogenic features and a complex tumor microenvironment (TME) are likely culprits for therapy refractoriness in PDAC. Induced pluripotent stem cells (iPSCs) should be explored as a means to advance the treatment options for PDAC, by providing representative in vitro models of pancreatic cancer development. In addition, iPSCs could be used for tailor-made cellular immunotherapies or as a source of tumor-associated antigens in the context of vaccination.
Collapse
Affiliation(s)
- Ricki T. Krog
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (R.T.K.); (A.L.V.)
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | | | - Alexander L. Vahrmeijer
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (R.T.K.); (A.L.V.)
| | - Nigel G. Kooreman
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (R.T.K.); (A.L.V.)
- Correspondence:
| |
Collapse
|
7
|
Yamamoto R, Kino-Oka M. Design of suspension culture system with bubble sparging for human induced pluripotent stem cells in a plastic fluid. J Biosci Bioeng 2021; 132:190-197. [PMID: 34052116 DOI: 10.1016/j.jbiosc.2021.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 11/29/2022]
Abstract
Bubble sparging has been used to supply oxygen to large-scale bioreactor systems. However, sparged bubbles cause cell death by rupturing due to shear stress, and the foam layer carries a risk of contamination. Large-scale culture of human induced pluripotent stem cells (hiPSCs) is required for manufacturing, but hiPSCs show high sensitivity to shear stress, and also, aseptic processing is important for their expansion. In this study, a culture system with bubble sparging for hiPSC proliferation was designed using a plastic fluid as a culture medium. The rising bubble velocity in the plastic fluid decreased and was lower than that in a Newtonian fluid when the time interval between bubbles generation, Δt, was greater than 0.14 s. Under this condition, aggregate distribution in the plastic fluid was maintained without liquid flow. Although large aeration induced aggregate coalescence and growth inhibition, the apparent specific growth rate at Δt > 0.14 s increased with an increase in the aeration rate, and the maximum value was similar to that of the conventional suspension culture in a stirred bioreactor system. The gas hold-up in the plastic fluid was higher than that in a Newtonian fluid because of the lower rising bubble velocity, which leads to the suppression of bubble sparging. Therefore, our results indicated that using a plastic fluid leads to a more efficient oxygen supply without agitation in a spatial-temporal phase-transition culture system.
Collapse
Affiliation(s)
- Riku Yamamoto
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
8
|
Goto H, Komae H, Sekine H, Homma J, Lee S, Yokota T, Matsuura K, Someya T, Ono M, Shimizu T. Continuous measurement of surface electrical potentials from transplanted cardiomyocyte tissue derived from human-induced pluripotent stem cells under physiological conditions in vivo. Heart Vessels 2021; 36:899-909. [PMID: 33683408 DOI: 10.1007/s00380-021-01824-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/26/2021] [Indexed: 10/22/2022]
Abstract
Recording the electrical potentials of bioengineered cardiac tissue after transplantation would help to monitor the maturation of the tissue and detect adverse events such as arrhythmia. However, a few studies have reported the measurement of myocardial tissue potentials in vivo under physiological conditions. In this study, human-induced pluripotent stem cell-derived cardiomyocyte (hiPSCM) sheets were stacked and ectopically transplanted into the subcutaneous tissue of rats for culture in vivo. Three months after transplantation, a flexible nanomesh sensor was implanted onto the hiPSCM tissue to record its surface electrical potentials under physiological conditions, i.e., without the need for anesthetic agents that might adversely affect cardiomyocyte function. The nanomesh sensor was able to record electrical potentials in non-sedated, ambulating animals for up to 48 h. When compared with recordings made with conventional needle electrodes in anesthetized animals, the waveforms obtained with the nanomesh sensor showed less dispersion of waveform interval and waveform duration. However, waveform amplitude tended to show greater dispersion for the nanomesh sensor than for the needle electrodes, possibly due to motion artifacts produced by movements of the animal or local tissue changes in response to surgical implantation of the sensor. The implantable nanomesh sensor utilized in this study potentially could be used for long-term monitoring of bioengineered myocardial tissue in vivo under physiological conditions.
Collapse
Affiliation(s)
- Hiroshi Goto
- Department of Cardiac Surgery, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.,Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Hyoe Komae
- Department of Cardiac Surgery, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan. .,Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
| | - Hidekazu Sekine
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
| | - Jun Homma
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Sunghoon Lee
- Department of Electrical Engineering and Information Systems, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Tomoyuki Yokota
- Department of Electrical Engineering and Information Systems, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Katsuhisa Matsuura
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Takao Someya
- Department of Electrical Engineering and Information Systems, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Minoru Ono
- Department of Cardiac Surgery, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| |
Collapse
|
9
|
Bui TVA, Hwang JW, Lee JH, Park HJ, Ban K. Challenges and Limitations of Strategies to Promote Therapeutic Potential of Human Mesenchymal Stem Cells for Cell-Based Cardiac Repair. Korean Circ J 2021; 51:97-113. [PMID: 33525065 PMCID: PMC7853896 DOI: 10.4070/kcj.2020.0518] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) represent a population of adult stem cells residing in many tissues, mainly bone marrow, adipose tissue, and umbilical cord. Due to the safety and availability of standard procedures and protocols for isolation, culturing, and characterization of these cells, MSCs have emerged as one of the most promising sources for cell-based cardiac regenerative therapy. Once transplanted into a damaged heart, MSCs release paracrine factors that nurture the injured area, prevent further adverse cardiac remodeling, and mediate tissue repair along with vasculature. Numerous preclinical studies applying MSCs have provided significant benefits following myocardial infarction. Despite promising results from preclinical studies using animal models, MSCs are not up to the mark for human clinical trials. As a result, various approaches have been considered to promote the therapeutic potency of MSCs, such as genetic engineering, physical treatments, growth factor, and pharmacological agents. Each strategy has targeted one or multi-potentials of MSCs. In this review, we will describe diverse approaches that have been developed to promote the therapeutic potential of MSCs for cardiac regenerative therapy. Particularly, we will discuss major characteristics of individual strategy to enhance therapeutic efficacy of MSCs including scientific principles, advantages, limitations, and improving factors. This article also will briefly introduce recent novel approaches that MSCs enhanced therapeutic potentials of other cells for cardiac repair.
Collapse
Affiliation(s)
- Thi Van Anh Bui
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Ji Won Hwang
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Korea.,Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Jung Hoon Lee
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Hun Jun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Korea.,Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.,Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
10
|
Samura T, Miyagawa S, Kawamura T, Fukushima S, Yokoyama JY, Takeda M, Harada A, Ohashi F, Sato-Nishiuchi R, Toyofuku T, Toda K, Sekiguchi K, Sawa Y. Laminin-221 Enhances Therapeutic Effects of Human-Induced Pluripotent Stem Cell-Derived 3-Dimensional Engineered Cardiac Tissue Transplantation in a Rat Ischemic Cardiomyopathy Model. J Am Heart Assoc 2020; 9:e015841. [PMID: 32783519 PMCID: PMC7660810 DOI: 10.1161/jaha.119.015841] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background Extracellular matrix, especially laminin‐221, may play crucial roles in viability and survival of human‐induced pluripotent stem cell‐derived cardiomyocytes (hiPS‐CMs) after in vivo transplant. Then, we hypothesized laminin‐221 may have an adjuvant effect on therapeutic efficacy by enhancing cell viability and survival after transplantation of 3‐dimensional engineered cardiac tissue (ECT) to a rat model of myocardial infarction. Methods and Results In vitro study indicates the impacts of laminin‐221 on hiPS‐CMs were analyzed on the basis of mechanical function, mitochondrial function, and tolerance to hypoxia. We constructed 3‐dimensional ECT containing hiPS‐CMs and fibrin gel conjugated with laminin‐221. Heart function and in vivo behavior were assessed after engraftment of 3‐dimensional ECT (laminin‐conjugated ECT, n=10; ECT, n=10; control, n=10) in a rat model of myocardial infarction. In vitro assessment indicated that laminin‐221 improves systolic velocity, diastolic velocity, and maximum capacity of oxidative metabolism of hiPS‐CMs. Cell viability and lactate dehydrogenase production revealed that laminin‐221 improved tolerance to hypoxia. Furthermore, analysis of mRNA expression revealed that antiapoptotic genes were upregulated in the laminin group under hypoxic conditions. Left ventricular ejection fraction of the laminin‐conjugated ECT group was significantly better than that of other groups 4 weeks after transplantation. Laminin‐conjugated ECT transplantation was associated with significant improvements in expression levels of rat vascular endothelial growth factor. In early assessments, cell survival was also improved in laminin‐conjugated ECTs compared with ECT transplantation without laminin‐221. Conclusions In vitro laminin‐221 enhanced mechanical and metabolic function of hiPS‐CMs and improved the therapeutic impact of 3‐dimensional ECT in a rat ischemic cardiomyopathy model. These findings suggest that adjuvant laminin‐221 may provide a clinical benefit to hiPS‐CM constructs.
Collapse
Affiliation(s)
- Takaaki Samura
- Department of Cardiovascular Surgery Osaka University Graduate School of Medicine Osaka Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery Osaka University Graduate School of Medicine Osaka Japan
| | - Takuji Kawamura
- Department of Cardiovascular Surgery Osaka University Graduate School of Medicine Osaka Japan
| | - Satsuki Fukushima
- Department of Cardiovascular Surgery Osaka University Graduate School of Medicine Osaka Japan
| | - Jun-Ya Yokoyama
- Department of Cardiovascular Surgery Osaka University Graduate School of Medicine Osaka Japan
| | - Maki Takeda
- Department of Cardiovascular Surgery Osaka University Graduate School of Medicine Osaka Japan
| | - Akima Harada
- Department of Cardiovascular Surgery Osaka University Graduate School of Medicine Osaka Japan
| | - Fumiya Ohashi
- Department of Cardiovascular Surgery Osaka University Graduate School of Medicine Osaka Japan
| | - Ryoko Sato-Nishiuchi
- Division of Matrixome Research and Application Institute for Protein Research Osaka University Osaka Japan
| | - Toshihiko Toyofuku
- Department of Immunology and Regenerative Medicine Osaka University Graduate School of Medicine Osaka Japan
| | - Koichi Toda
- Department of Cardiovascular Surgery Osaka University Graduate School of Medicine Osaka Japan
| | - Kiyotoshi Sekiguchi
- Division of Matrixome Research and Application Institute for Protein Research Osaka University Osaka Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery Osaka University Graduate School of Medicine Osaka Japan
| |
Collapse
|
11
|
Song SY, Kim H, Yoo J, Kwon SP, Park BW, Kim JJ, Ban K, Char K, Park HJ, Kim BS. Prevascularized, multiple-layered cell sheets of direct cardiac reprogrammed cells for cardiac repair. Biomater Sci 2020; 8:4508-4520. [DOI: 10.1039/d0bm00701c] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We developed cardiac-reprogrammed cell sheets via cardiac-mimetic cell culture system with biodegradable PLGA membrane. The prevascularized, multiple-layered cell sheets prevented heart failure after myocardial infarction.
Collapse
Affiliation(s)
- Seuk Young Song
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul
- Republic of Korea
| | - Hyeok Kim
- Department of Medical Life Science
- College of Medicine
- The Catholic University of Korea
- Seoul
- Republic of Korea
| | - Jin Yoo
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul
- Republic of Korea
| | - Sung Pil Kwon
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul
- Republic of Korea
| | - Bong Woo Park
- Department of Medical Life Science
- College of Medicine
- The Catholic University of Korea
- Seoul
- Republic of Korea
| | - Jin-ju Kim
- Department of Medical Life Science
- College of Medicine
- The Catholic University of Korea
- Seoul
- Republic of Korea
| | - Kiwon Ban
- Department of Biomedical Sciences
- City University of Hong Kong
- Kowloon Tong
- Hong Kong
| | - Kookheon Char
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul
- Republic of Korea
| | - Hun-Jun Park
- Department of Medical Life Science
- College of Medicine
- The Catholic University of Korea
- Seoul
- Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul
- Republic of Korea
- Institute of Chemical Processes
| |
Collapse
|
12
|
van der Ende MY, Said MA, van Veldhuisen DJ, Verweij N, van der Harst P. Genome-wide studies of heart failure and endophenotypes: lessons learned and future directions. Cardiovasc Res 2019; 114:1209-1225. [PMID: 29912321 DOI: 10.1093/cvr/cvy083] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 04/16/2018] [Indexed: 12/28/2022] Open
Abstract
Heart failure (HF) is a complex clinical syndrome resulting from structural or functional impairments of ventricular filling or ejection of blood. HF has a poor prognosis and the burden to society remains tremendous. The unfulfilled expectation is that expanding our knowledge of the genetic architecture of HF will help to quickly advance the quality of risk assessment, diagnoses, and treatment. To date, genome-wide association studies (GWAS) of HF have led to disappointing results with only limited progress in our understanding and tempering the earlier expectations. However, the analyses of traits closely related to HF (also called 'endophenotypes') have led to promising and novel findings. For example, GWAS of NT-proBNP levels not only identified variants in the NNPA-NPPB locus but also substantiated data suggesting that natriuretic peptides in itself are associated with a lower risk of hypertension and HF. Many other genetic associates currently await experimental follow-up in which genes are prioritized based on bioinformatic analyses and various model organisms are employed to obtain functional insights. Promising genes with identified function could later be used in personalized medicine. Also, targeting specific pathogenic gene mutations is promising to protect future generations from HF, such as recently done in human embryos carrying the cardiomyopathy-associated MYBPC3 mutation. This review discusses the current status of GWAS of HF and its endophenotypes. In addition, future directions such as functional follow-up and application of GWAS results are discussed.
Collapse
Affiliation(s)
- Maaike Yldau van der Ende
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, RB Groningen, The Netherlands
| | - Mir Abdullah Said
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, RB Groningen, The Netherlands
| | - Dirk Jan van Veldhuisen
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, RB Groningen, The Netherlands
| | - Niek Verweij
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, RB Groningen, The Netherlands
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, RB Groningen, The Netherlands
| |
Collapse
|
13
|
Miyagawa S, Sawa Y. Building a new strategy for treating heart failure using Induced Pluripotent Stem Cells. J Cardiol 2018; 72:445-448. [PMID: 30172684 DOI: 10.1016/j.jjcc.2018.05.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 05/01/2018] [Indexed: 12/31/2022]
Abstract
Although cell therapy using myoblasts, bone marrow cells, or other stem cells appears to improve functional recovery of the failing heart, mainly by cytokine paracrine effects, its effectiveness in severely damaged myocardium is limited, probably because there are too few residual myocytes to promote cytokine-induced angiogenesis. Recently, cardiogenic stem cells, such as c-kit-positive cells, were reported to generate cardiomyogenic lineages, and basic research experiments showed that implanting these cells, which can differentiate into cardiomyocytes, improves heart function. However, this functional recovery may have also mainly depended on cytokine paracrine effects, because the differentiation to cardiomyocytes in vivo was poor. In contrast, while Induced Pluripotent Stem Cell-derived cardiomyocytes have paracrine effects, they also have the potential to supply newly born myocytes that can function synchronously with the recipient myocardium as "mechanically working cells" in severely damaged myocardium. Thus, they could represent a "true" myocardial regeneration therapy that can actually regenerate severely damaged myocardium. In addition, iPS cells, especially disease-specific iPS cells, have other applications in regenerative medicine such as in drug screening. In this report, we present the state of basic research in the field of cardiac iPS cells.
Collapse
Affiliation(s)
- Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
14
|
Jiang Y, Park P, Hong SM, Ban K. Maturation of Cardiomyocytes Derived from Human Pluripotent Stem Cells: Current Strategies and Limitations. Mol Cells 2018; 41:613-621. [PMID: 29890820 PMCID: PMC6078855 DOI: 10.14348/molcells.2018.0143] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 01/06/2023] Open
Abstract
The capacity of differentiation of human pluripotent stem cells (hPSCs), which include both embryonic stem cells and induced pluripotent stem cells, into cardiomyocytes (CMs) in vitro provides an unlimited resource for human CMs for a wide range of applications such as cell based cardiac repair, cardiac drug toxicology screening, and human cardiac disease modeling. However, their applicability is significantly limited by immature phenotypes. It has been well known that currently available CMs derived from hPSCs (hPSC-CMs) represent immature embryonic or fetal stage CMs and are functionally and structurally different from mature human CMs. To overcome this critical issue, several new approaches aiming to generate more mature hPSC-CMs have been developed. This review describes recent approaches to generate more mature hPSC-CMs including their scientific principles, advantages, and limitations.
Collapse
Affiliation(s)
- Yanqing Jiang
- University of Toronto, Hospital of Sick Children, Toronto,
Canada
| | - Peter Park
- Emory University, Department of Biology, Atlanta, Georgia,
USA
| | - Sang-Min Hong
- Department of Physical Education, Dongguk University Seoul, Seoul 04620,
Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong,
Hong Kong
| |
Collapse
|
15
|
Sougawa N, Miyagawa S, Fukushima S, Kawamura A, Yokoyama J, Ito E, Harada A, Okimoto K, Mochizuki-Oda N, Saito A, Sawa Y. Immunologic targeting of CD30 eliminates tumourigenic human pluripotent stem cells, allowing safer clinical application of hiPSC-based cell therapy. Sci Rep 2018; 8:3726. [PMID: 29487310 PMCID: PMC5829260 DOI: 10.1038/s41598-018-21923-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 02/13/2018] [Indexed: 01/14/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) are promising candidate cells for cardiomyogenesis in the failing heart. However, teratoma/tumour formation originating from undifferentiated iPSCs contaminating the graft is a critical concern for clinical application. Here, we hypothesized that brentuximab vedotin, which targets CD30, induces apoptosis in tumourigenic cells, thus increasing the safety of iPSC therapy for heart failure. Flow cytometry analysis identified consistent expression of CD30 in undifferentiated human iPSCs. Addition of brentuximab vedotin in vitro for 72 h efficiently induced cell death in human iPSCs, associated with a significant increase in G2/M phase cells. Brentuximab vedotin significantly reduced Lin28 expression in cardiomyogenically differentiated human iPSCs. Transplantation of human iPSC-derived cardiomyocytes (CMs) without treatment into NOG mice consistently induced teratoma/tumour formation, with a substantial number of Ki-67-positive cells in the graft at 4 months post-transplant, whereas iPSC-derived CMs treated with brentuximab vedotin prior to the transplantation did not show teratoma/tumour formation, which was associated with absence of Ki-67-positive cells in the graft over the same period. These findings suggest that in vitro treatment with brentuximab vedotin, targeting the CD30-positive iPSC fraction, reduced tumourigenicity in human iPSC-derived CMs, potentially providing enhanced safety for iPSC-based cardiomyogenesis therapy in clinical scenarios.
Collapse
Affiliation(s)
- Nagako Sougawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Satsuki Fukushima
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ai Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Junya Yokoyama
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Emiko Ito
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Akima Harada
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kaori Okimoto
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Noriko Mochizuki-Oda
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Atsuhiro Saito
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| |
Collapse
|
16
|
Ban K, Bae S, Yoon YS. Current Strategies and Challenges for Purification of Cardiomyocytes Derived from Human Pluripotent Stem Cells. Theranostics 2017. [PMID: 28638487 PMCID: PMC5479288 DOI: 10.7150/thno.19427] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Cardiomyocytes (CMs) derived from human pluripotent stem cells (hPSCs) are considered a most promising option for cell-based cardiac repair. Hence, various protocols have been developed for differentiating hPSCs into CMs. Despite remarkable improvement in the generation of hPSC-CMs, without purification, these protocols can only generate mixed cell populations including undifferentiated hPSCs or non-CMs, which may elicit adverse outcomes. Therefore, one of the major challenges for clinical use of hPSC-CMs is the development of efficient isolation techniques that allow enrichment of hPSC-CMs. In this review, we will discuss diverse strategies that have been developed to enrich hPSC-CMs. We will describe major characteristics of individual hPSC-CM purification methods including their scientific principles, advantages, limitations, and needed improvements. Development of a comprehensive system which can enrich hPSC-CMs will be ultimately useful for cell therapy for diseased hearts, human cardiac disease modeling, cardiac toxicity screening, and cardiac tissue engineering.
Collapse
|
17
|
Doetschman T, Georgieva T. Gene Editing With CRISPR/Cas9 RNA-Directed Nuclease. Circ Res 2017; 120:876-894. [DOI: 10.1161/circresaha.116.309727] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 01/30/2017] [Accepted: 02/06/2017] [Indexed: 12/22/2022]
Abstract
Genetic engineering of model organisms and cultured cells has for decades provided important insights into the mechanisms underlying cardiovascular development and disease. In the past few years the development of several nuclease systems has broadened the range of model/cell systems that can be engineered. Of these, the CRISPR (clustered regularly interspersed short palindromic repeats)/Cas9 (CRISPR-associated protein 9) system has become the favorite for its ease of application. Here we will review this RNA-guided nuclease system for gene editing with respect to its usefulness for cardiovascular studies and with an eye toward potential therapy. Studies on its off-target activity, along with approaches to minimize this activity will be given. The advantages of gene editing versus gene targeting in embryonic stem cells, including the breadth of species and cell types to which it is applicable, will be discussed. We will also cover its use in iPSC for research and possible therapeutic purposes; and we will review its use in muscular dystrophy studies where considerable progress has been made toward dystrophin correction in mice. The CRISPR/Ca9s system is also being used for high-throughput screening of genes, gene regulatory regions, and long noncoding RNAs. In addition, the CRISPR system is being used for nongene-editing purposes such as activation and inhibition of gene expression, as well as for fluorescence tagging of chromosomal regions and individual mRNAs to track their cellular location. Finally, an approach to circumvent the inability of post-mitotic cells to support homologous recombination-based gene editing will be presented. In conclusion, applications of the CRISPR/Cas system are expanding at a breath-taking pace and are revolutionizing approaches to gain a better understanding of human diseases.
Collapse
Affiliation(s)
- Thomas Doetschman
- From the BIO5 Institute (T.D., T.G.) and Department of Cellular and Molecular Medicine (T.D.), University of Arizona, Tucson
| | - Teodora Georgieva
- From the BIO5 Institute (T.D., T.G.) and Department of Cellular and Molecular Medicine (T.D.), University of Arizona, Tucson
| |
Collapse
|
18
|
Chen HY, Strappe PM, Wang LX. Stem Cell Therapies for Cardiovascular Diseases: What Does the Future Hold? Heart Lung Circ 2017; 26:205-208. [DOI: 10.1016/j.hlc.2016.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
19
|
Pasquier J, Gupta R, Rioult D, Hoarau-Véchot J, Courjaret R, Machaca K, Al Suwaidi J, Stanley EG, Rafii S, Elliott DA, Abi Khalil C, Rafii A. Coculturing with endothelial cells promotes in vitro maturation and electrical coupling of human embryonic stem cell-derived cardiomyocytes. J Heart Lung Transplant 2017; 36:684-693. [PMID: 28169114 DOI: 10.1016/j.healun.2017.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 12/22/2016] [Accepted: 01/04/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Pluripotent human embryonic stem cells (hESC) are a promising source of repopulating cardiomyocytes. We hypothesized that we could improve maturation of cardiomyocytes and facilitate electrical interconnections by creating a model that more closely resembles heart tissue; that is, containing both endothelial cells (ECs) and cardiomyocytes. METHODS We induced cardiomyocyte differentiation in the coculture of an hESC line expressing the cardiac reporter NKX2.5-green fluorescent protein (GFP), and an Akt-activated EC line (E4+ECs). We quantified spontaneous beating rates, synchrony, and coordination between different cardiomyocyte clusters using confocal imaging of Fura Red-detected calcium transients and computer-assisted image analysis. RESULTS After 8 days in culture, 94% ± 6% of the NKX2-5GFP+ cells were beating when hESCs embryonic bodies were plated on E4+ECs compared with 34% ± 12.9% for controls consisting of hESCs cultured on BD Matrigel (BD Biosciences) without ECs at Day 11 in culture. The spatial organization of beating areas in cocultures was different. The GFP+ cardiomyocytes were close to the E4+ECs. The average beats/min of the cardiomyocytes in coculture was faster and closer to physiologic heart rates compared with controls (50 ± 14 [n = 13] vs 25 ± 9 [n = 8]; p < 0.05). The coculture with ECs led to synchronized beating relying on the endothelial network, as illustrated by the loss of synchronization upon the disruption of endothelial bridges. CONCLUSIONS The coculturing of differentiating cardiomyocytes with Akt-activated ECs but not EC-conditioned media results in (1) improved efficiency of the cardiomyocyte differentiation protocol and (2) increased maturity leading to better intercellular coupling with improved chronotropy and synchrony.
Collapse
Affiliation(s)
- Jennifer Pasquier
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Doha, Qatar; Department of Genetic Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Renuka Gupta
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Damien Rioult
- UMR_I 02 INERIS-URCA-ULH Unité Stress Environnementaux et BIOsurveillance des milieux aquatiques, Université de Reims, Reims, France
| | - Jessica Hoarau-Véchot
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Doha, Qatar; Cardiovascular Epigenetics Laboratory, Department of Genetic Medicine, Doha, Qatar
| | - Raphael Courjaret
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Khaled Machaca
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar
| | | | - Edouard G Stanley
- Department of Medicine, Weill Cornell Medicine, New York, New York; Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Shahin Rafii
- Department of Medicine, Weill Cornell Medicine, New York, New York
| | - David A Elliott
- Department of Medicine, Weill Cornell Medicine, New York, New York; Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Charbel Abi Khalil
- Department of Genetic Medicine, Weill Cornell Medicine, New York, New York, USA; Cardiovascular Epigenetics Laboratory, Department of Genetic Medicine, Doha, Qatar; Heart Hospital, Hamad Medical Corporation, Doha, Qatar; Department of Medicine, Weill Cornell Medicine, New York, New York, USA.
| | - Arash Rafii
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Doha, Qatar; Department of Genetic Medicine, Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
20
|
|
21
|
Editorial Commentary: Reprogramming autologous mesenchymal stem cells to regenerate the lost myocardium in chronic heart failure: Reboot and restore? Trends Cardiovasc Med 2016; 26:405-6. [PMID: 26952041 DOI: 10.1016/j.tcm.2016.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/02/2016] [Indexed: 11/23/2022]
|