1
|
Salehi Babadi P, Dayer D, Jafarinia M, Forouzanfar M. Do human adipose stem cell-derived artificial insulin-producing cells develop tumorigenic characteristics throughout differentiation? Mol Biol Rep 2025; 52:404. [PMID: 40253677 DOI: 10.1007/s11033-025-10432-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/11/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND Artificial insulin-producing cells (IPCs) used to treat diabetes mellitus type 1 (DMT1) are naturally hampered by their carcinogenicity. This in vitro study aimed to examine the carcinogenic potential of IPCs produced by the differentiation of human adipose tissue-derived mesenchymal stem cells (hADSCs). METHODS AND RESULTS hADSCs were transformed into IPCs by administering insulin-transferrin, selenium (ITS), and nicotinamide in a 14-day differentiation protocol. The cells were transfected with 20 μg of pure Pdx1-pIRES recombinant vector on the tenth day of differentiation. The successful transfection was confirmed by Pdx1 overexpression and GFP fluorescence activity. The differentiated cells' capacity to release insulin and glucose-dependent C-peptide was used to evaluate their functionality. Gene expression was assessed using real-time PCR. Meanwhile, protein expression was investigated using western blotting. The transfected cells exhibited fluorescence activity and Pdx1 overexpression. The differentiated IPCs were able to secrete C-peptide and insulin. The artificial IPCs showed significantly reduced Oct4 and Nanog expression. However, the differentiation process induced a noticeable elevation in tPA expression. The artificial IPCs expressed much lower c-MYC expression compared to undifferentiated hADSCs. The differentiated cells exhibited a significant elevation in Glut2, MMP-2, CD24, P16, and P21 expression. CONCLUSIONS The differentiation technique used in this work produced functional beta-like cells devoid of typical markers of stem cells. The synthetic IPCs displayed characteristics of newly generated β-like cells. The artificial IPCs showed no signs of expressing tumor-associated markers. The findings imply that the artificial IPC cells lack tumor characteristics in vitro.
Collapse
Affiliation(s)
| | - Dian Dayer
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Mojtaba Jafarinia
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Mohsen Forouzanfar
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| |
Collapse
|
2
|
Generali M, Kehl D, Meier D, Zorndt D, Atrott K, Saito H, Emmert MY, Hoerstrup SP. Generation and purification of iPSC-derived cardiomyocytes for clinical applications. Stem Cell Res Ther 2025; 16:189. [PMID: 40251664 PMCID: PMC12008852 DOI: 10.1186/s13287-025-04319-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/07/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Over the past decade, the field of cell therapy has rapidly expanded with the aim to replace and repair damaged cells and/or tissue. Depending on the disease many different cell types can be used as part of such a therapy. Here we focused on the potential treatment of myocardial infarction, where currently available treatment options are not able to regenerate the loss of healthy heart tissue. METHOD We generated good manufacturing practice (GMP)-compatible cardiomyocytes (iCMs) from transgene- and xenofree induced pluripotent stem cells (iPSCs) that can be seamless adapted for clinical applications. Further protocols were established for replating and freezing/thawing iCMs under xenofree conditions. RESULTS iCMs showed a cardiac phenotype, with the expression of specific cardiac markers and absence of pluripotency markers at RNA and protein level. To ensure a pure iCMs population for in vivo applications, we minimized risks of iPSC contamination using RNA-switch technology to ensure safety. CONCLUSION We describe the generation and further processing of xeno- and transgene-free iCMs. The use of GMP-compliant differentiation protocols ab initio facilitates the clinical translation of this project in later stages.
Collapse
Affiliation(s)
- M Generali
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.
| | - D Kehl
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - D Meier
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - D Zorndt
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - K Atrott
- Center for Surgical Research, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | - H Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, 113-0032, Japan
| | - M Y Emmert
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - S P Hoerstrup
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Wyss Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
3
|
Cotta GC, Teixeira dos Santos RC, Costa GMJ, Lacerda SMDSN. Reporter Alleles in hiPSCs: Visual Cues on Development and Disease. Int J Mol Sci 2024; 25:11009. [PMID: 39456792 PMCID: PMC11507014 DOI: 10.3390/ijms252011009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Reporter alleles are essential for advancing research with human induced pluripotent stem cells (hiPSCs), notably in developmental biology and disease modeling. This study investigates the state-of-the-art gene-editing techniques tailored for generating reporter alleles in hiPSCs, emphasizing their effectiveness in investigating cellular dynamics and disease mechanisms. Various methodologies, including the application of CRISPR/Cas9 technology, are discussed for accurately integrating reporter genes into the specific genomic loci. The synthesis of findings from the studies utilizing these reporter alleles reveals insights into developmental processes, genetic disorder modeling, and therapeutic screening, consolidating the existing knowledge. These hiPSC-derived models demonstrate remarkable versatility in replicating human diseases and evaluating drug efficacy, thereby accelerating translational research. Furthermore, this review addresses challenges and future directions in refining the reporter allele design and application to bolster their reliability and relevance in biomedical research. Overall, this investigation offers a comprehensive perspective on the methodologies, applications, and implications of reporter alleles in hiPSC-based studies, underscoring their essential role in advancing both fundamental scientific understanding and clinical practice.
Collapse
Affiliation(s)
| | | | | | - Samyra Maria dos Santos Nassif Lacerda
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, 31270-901 Belo Horizonte, Brazil; (G.C.C.); (R.C.T.d.S.); (G.M.J.C.)
| |
Collapse
|
4
|
Liang Z, Hu Y, Li CY, Yau WL, Tan K, Kuang Y. VPg-based bidirectional synthetic mRNA circuits enable orthogonal protein regulation for high-resolution cell separation. Chem Commun (Camb) 2024; 60:5427-5430. [PMID: 38685869 DOI: 10.1039/d4cc01725k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Synthetic mRNA circuits commonly sense input to produce binary output signals for cell separation. Based on virus-origin cap-independent translation initiation machinery and RBP-aptamer interaction, we designed smart synthetic mRNA-based circuits that sense single input molecules to bidirectionally tune output signals in an orthogonal manner, enabling high-resolution separation of cell populations.
Collapse
Affiliation(s)
- Zhenghua Liang
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Room 5578, Academic Bldg, Clear Water Bay, Kowloon, Hong Kong.
| | - Yaxin Hu
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Room 5578, Academic Bldg, Clear Water Bay, Kowloon, Hong Kong.
| | - Cheuk Yin Li
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Room 5578, Academic Bldg, Clear Water Bay, Kowloon, Hong Kong.
| | - Wai Laam Yau
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Room 5578, Academic Bldg, Clear Water Bay, Kowloon, Hong Kong.
| | - Kaixin Tan
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Room 5578, Academic Bldg, Clear Water Bay, Kowloon, Hong Kong.
| | - Yi Kuang
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Room 5578, Academic Bldg, Clear Water Bay, Kowloon, Hong Kong.
| |
Collapse
|
5
|
Mao Y, Wang S, Yu J, Li W. Engineering pluripotent stem cells with synthetic biology for regenerative medicine. MEDICAL REVIEW (2021) 2024; 4:90-109. [PMID: 38680679 PMCID: PMC11046572 DOI: 10.1515/mr-2023-0050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/14/2024] [Indexed: 05/01/2024]
Abstract
Pluripotent stem cells (PSCs), characterized by self-renewal and capacity of differentiating into three germ layers, are the programmable building blocks of life. PSC-derived cells and multicellular systems, particularly organoids, exhibit great potential for regenerative medicine. However, this field is still in its infancy, partly due to limited strategies to robustly and precisely control stem cell behaviors, which are tightly regulated by inner gene regulatory networks in response to stimuli from the extracellular environment. Synthetic receptors and genetic circuits are powerful tools to customize the cellular sense-and-response process, suggesting their underlying roles in precise control of cell fate decision and function reconstruction. Herein, we review the progress and challenges needed to be overcome in the fields of PSC-based cell therapy and multicellular system generation, respectively. Furthermore, we summarize several well-established synthetic biology tools and their applications in PSC engineering. Finally, we highlight the challenges and perspectives of harnessing synthetic biology to PSC engineering for regenerative medicine.
Collapse
Affiliation(s)
- Yihuan Mao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Siqi Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jiazhen Yu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
6
|
Chen SJ, Hashimoto K, Fujio K, Hayashi K, Paul SK, Yuzuriha A, Qiu WY, Nakamura E, Kanashiro MA, Kabata M, Nakamura S, Sugimoto N, Kaneda A, Yamamoto T, Saito H, Takayama N, Eto K. A let-7 microRNA-RALB axis links the immune properties of iPSC-derived megakaryocytes with platelet producibility. Nat Commun 2024; 15:2588. [PMID: 38519457 PMCID: PMC10960040 DOI: 10.1038/s41467-024-46605-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 03/04/2024] [Indexed: 03/25/2024] Open
Abstract
We recently achieved the first-in-human transfusion of induced pluripotent stem cell-derived platelets (iPSC-PLTs) as an alternative to standard transfusions, which are dependent on donors and therefore variable in supply. However, heterogeneity characterized by thrombopoiesis-biased or immune-biased megakaryocytes (MKs) continues to pose a bottleneck against the standardization of iPSC-PLT manufacturing. To address this problem, here we employ microRNA (miRNA) switch biotechnology to distinguish subpopulations of imMKCLs, the MK cell lines producing iPSC-PLTs. Upon miRNA switch-based screening, we find imMKCLs with lower let-7 activity exhibit an immune-skewed transcriptional signature. Notably, the low activity of let-7a-5p results in the upregulation of RAS like proto-oncogene B (RALB) expression, which is crucial for the lineage determination of immune-biased imMKCL subpopulations and leads to the activation of interferon-dependent signaling. The dysregulation of immune properties/subpopulations, along with the secretion of inflammatory cytokines, contributes to a decline in the quality of the whole imMKCL population.
Collapse
Affiliation(s)
- Si Jing Chen
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazuya Hashimoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Kosuke Fujio
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Karin Hayashi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Sudip Kumar Paul
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Akinori Yuzuriha
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Wei-Yin Qiu
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Emiri Nakamura
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | | | - Mio Kabata
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Sou Nakamura
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Naoshi Sugimoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Atsushi Kaneda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Medical-risk Avoidance Based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Hirohide Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.
| | - Naoya Takayama
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Koji Eto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|
7
|
Hu Y, Li CY, Lu Q, Kuang Y. Multiplex miRNA reporting platform for real-time profiling of living cells. Cell Chem Biol 2024; 31:150-162.e7. [PMID: 38035883 DOI: 10.1016/j.chembiol.2023.11.002] [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: 06/20/2023] [Revised: 09/15/2023] [Accepted: 11/03/2023] [Indexed: 12/02/2023]
Abstract
Accurately characterizing cell types within complex cell structures provides invaluable information for comprehending the cellular status during biological processes. In this study, we have developed an miRNA-switch cocktail platform capable of reporting and tracking the activities of multiple miRNAs (microRNAs) at the single-cell level, while minimizing disruption to the cell culture. Drawing on the principles of traditional miRNA-sensing mRNA switches, our platform incorporates subcellular tags and employs intelligent engineering to segment three subcellular regions using two fluorescent proteins. These designs enable the quantification of multiple miRNAs within the same cell. Through our experiments, we have demonstrated the platform's ability to track marker miRNA levels during cell differentiation and provide spatial information of heterogeneity on outlier cells exhibiting extreme miRNA levels. Importantly, this platform offers real-time and in situ miRNA reporting, allowing for multidimensional evaluation of cell profile and paving the way for a comprehensive understanding of cellular events during biological processes.
Collapse
Affiliation(s)
- Yaxin Hu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Cheuk Yin Li
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Qiuyu Lu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Yi Kuang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China.
| |
Collapse
|
8
|
Tan K, Hu Y, Liang Z, Li CY, Yau WL, Kuang Y. Dual Input-Controlled Synthetic mRNA Circuit for Bidirectional Protein Expression Regulation. ACS Synth Biol 2023; 12:2516-2523. [PMID: 37652441 PMCID: PMC10510700 DOI: 10.1021/acssynbio.3c00144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Indexed: 09/02/2023]
Abstract
Synthetic mRNA circuits manipulate cell fate by controlling output protein expression via cell-specific input molecule detection. Most current circuits either repress or enhance output production upon input binding. Such binary input-output mechanisms restrict the fine-tuning of protein expression to control complex cellular events. Here we designed mRNA circuits using enhancer/repressor modules that were independently controlled by different input molecules, resulting in bidirectional output regulation; the maximal enhancement over maximal repression was 57 fold. The circuit either enhances or represses protein production in different cells based on the difference in the expression of two microRNAs. This study examined novel bidirectional circuit designs capable of fine-tuning protein production by sensing multiple input molecules. It also broadened the scope of cell manipulation by synthetic mRNA circuits, facilitating the development of mRNA circuits for precise cell manipulation and providing cell-based solutions to biomedical problems.
Collapse
Affiliation(s)
- Kaixin Tan
- Department
of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Room 5578, Academic Building, Clear
Water Bay, Kowloon, Hong Kong
| | - Yaxin Hu
- Department
of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Room 5578, Academic Building, Clear
Water Bay, Kowloon, Hong Kong
| | - Zhenghua Liang
- Department
of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Room 5578, Academic Building, Clear
Water Bay, Kowloon, Hong Kong
| | - Cheuk Yin Li
- Department
of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Room 5578, Academic Building, Clear
Water Bay, Kowloon, Hong Kong
| | - Wai Laam Yau
- Department
of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Room 5578, Academic Building, Clear
Water Bay, Kowloon, Hong Kong
| | - Yi Kuang
- Department
of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Room 5578, Academic Building, Clear
Water Bay, Kowloon, Hong Kong
| |
Collapse
|
9
|
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
|
10
|
Kishino Y, Fukuda K. Unlocking the Pragmatic Potential of Regenerative Therapies in Heart Failure with Next-Generation Treatments. Biomedicines 2023; 11:biomedicines11030915. [PMID: 36979894 PMCID: PMC10046277 DOI: 10.3390/biomedicines11030915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/28/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
Patients with chronic heart failure (HF) have a poor prognosis due to irreversible impairment of left ventricular function, with 5-year survival rates <60%. Despite advances in conventional medicines for HF, prognosis remains poor, and there is a need to improve treatment further. Cell-based therapies to restore the myocardium offer a pragmatic approach that provides hope for the treatment of HF. Although first-generation cell-based therapies using multipotent cells (bone marrow-derived mononuclear cells, mesenchymal stem cells, adipose-derived regenerative cells, and c-kit-positive cardiac cells) demonstrated safety in preclinical models of HF, poor engraftment rates, and a limited ability to form mature cardiomyocytes (CMs) and to couple electrically with existing CMs, meant that improvements in cardiac function in double-blind clinical trials were limited and largely attributable to paracrine effects. The next generation of stem cell therapies uses CMs derived from human embryonic stem cells or, increasingly, from human-induced pluripotent stem cells (hiPSCs). These cell therapies have shown the ability to engraft more successfully and improve electromechanical function of the heart in preclinical studies, including in non-human primates. Advances in cell culture and delivery techniques promise to further improve the engraftment and integration of hiPSC-derived CMs (hiPSC-CMs), while the use of metabolic selection to eliminate undifferentiated cells will help minimize the risk of teratomas. Clinical trials of allogeneic hiPSC-CMs in HF are now ongoing, providing hope for vast numbers of patients with few other options available.
Collapse
Affiliation(s)
| | - Keiichi Fukuda
- Correspondence: ; Tel.: +81-3-5363-3874; Fax: +81-3-5363-3875
| |
Collapse
|
11
|
Alexanova A, Raitoharju E, Valtonen J, Aalto-Setälä K, Viiri LE. Coronary artery disease patient-derived iPSC-hepatocytes have distinct miRNA profile that may alter lipid metabolism. Sci Rep 2023; 13:1706. [PMID: 36717592 PMCID: PMC9886909 DOI: 10.1038/s41598-023-28981-7] [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: 09/02/2022] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Metabolic dysfunction, partly driven by altered liver function, predisposes to coronary artery disease (CAD), but the role of liver in vulnerable atherosclerotic plaque development remains unclear. Here we produced hepatocyte-like cells (HLCs) from 27 induced pluripotent stem cell (iPSC) lines derived from 15 study subjects with stable CAD (n = 5), acute CAD (n = 5) or healthy controls (n = 5). We performed a miRNA microarray screening throughout the differentiation, as well as compared iPSC-HLCs miRNA profiles of the patient groups to identify miRNAs involved in the development of CAD. MicroRNA profile changed during differentiation and started to resemble that of the primary human hepatocytes. In the microarray, 35 and 87 miRNAs were statistically significantly deregulated in the acute and stable CAD patients, respectively, compared to controls. Down-regulation of miR-149-5p, -92a-3p and -221-3p, and up-regulation of miR-122-5p was verified in the stable CAD patients when compared to other groups. The predicted targets of deregulated miRNAs were enriched in pathways connected to insulin signalling, inflammation and lipid metabolism. The iPSC-HLCs derived from stable CAD patients with extensive lesions had a distinct genetic miRNA profile possibly linked to metabolic dysfunction, potentially explaining the susceptibility to developing CAD. The iPSC-HLCs from acute CAD patients with only the acute rupture in otherwise healthy coronaries did not present a distinct miRNA profile, suggesting that hepatic miRNAs do not explain susceptibility to plaque rupture.
Collapse
Affiliation(s)
- Anna Alexanova
- The Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520, Tampere, Finland
| | - Emma Raitoharju
- The Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520, Tampere, Finland
- Molecular Epidemiology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere University Hospital, Tampere, Finland
| | - Joona Valtonen
- The Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520, Tampere, Finland
| | - Katriina Aalto-Setälä
- The Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520, Tampere, Finland
| | - Leena E Viiri
- The Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520, Tampere, Finland.
| |
Collapse
|
12
|
Free K, Nakanishi H, Itaka K. Development of Synthetic mRNAs Encoding Split Cytotoxic Proteins for Selective Cell Elimination Based on Specific Protein Detection. Pharmaceutics 2023; 15:pharmaceutics15010213. [PMID: 36678842 PMCID: PMC9867180 DOI: 10.3390/pharmaceutics15010213] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/25/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
For the selective elimination of deleterious cells (e.g., cancer cells and virus-infected cells), the use of a cytotoxic gene is a promising approach. DNA-based systems have achieved selective cell elimination but risk insertional mutagenesis. Here, we developed a synthetic mRNA-based system to selectively eliminate cells expressing a specific target protein. The synthetic mRNAs used in the system are designed to express an engineered protein pair that are based on a cytotoxic protein, Barnase. Each engineered protein is composed of an N- or C-terminal fragment of Barnase, a target protein binding domain, and an intein that aids in reconstituting full-length Barnase from the two fragments. When the mRNAs are transfected to cells expressing the target protein, both N- and C-terminal Barnase fragments bind to the target protein, causing the intein to excise itself and reconstitute cytotoxic full-length Barnase. In contrast, when the target protein is not present, the reconstitution of full-length Barnase is not induced. Four candidate constructs containing split Barnase were evaluated for the ability to selectively eliminate target protein-expressing cells. One of the candidate sets demonstrated highly selective cell death. This system will be a useful therapeutic tool to selectively eliminate deleterious cells.
Collapse
Affiliation(s)
| | - Hideyuki Nakanishi
- Correspondence: (H.N.); (K.I.); Tel.: +81-3-5280-8087 (H.N. & K.I.); Fax: +81-3-5280-8088 (H.N. & K.I.)
| | - Keiji Itaka
- Correspondence: (H.N.); (K.I.); Tel.: +81-3-5280-8087 (H.N. & K.I.); Fax: +81-3-5280-8088 (H.N. & K.I.)
| |
Collapse
|
13
|
Ono H, Saito H. Sensing intracellular signatures with synthetic mRNAs. RNA Biol 2023; 20:588-602. [PMID: 37582192 PMCID: PMC10431736 DOI: 10.1080/15476286.2023.2244791] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/30/2023] [Accepted: 07/31/2023] [Indexed: 08/17/2023] Open
Abstract
The bottom-up assembly of biological components in synthetic biology has contributed to a better understanding of natural phenomena and the development of new technologies for practical applications. Over the past few decades, basic RNA research has unveiled the regulatory roles of RNAs underlying gene regulatory networks; while advances in RNA biology, in turn, have highlighted the potential of a wide variety of RNA elements as building blocks to construct artificial systems. In particular, synthetic mRNA-based translational regulators, which respond to signals in cells and regulate the production of encoded output proteins, are gaining attention with the recent rise of mRNA therapeutics. In this Review, we discuss recent progress in RNA synthetic biology, mainly focusing on emerging technologies for sensing intracellular protein and RNA molecules and controlling translation.
Collapse
Affiliation(s)
- Hiroki Ono
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Sakyo-Ku, Japan
| | - Hirohide Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Sakyo-Ku, Japan
| |
Collapse
|
14
|
Prochazka L, Michaels YS, Lau C, Jones RD, Siu M, Yin T, Wu D, Jang E, Vázquez‐Cantú M, Gilbert PM, Kaul H, Benenson Y, Zandstra PW. Synthetic gene circuits for cell state detection and protein tuning in human pluripotent stem cells. Mol Syst Biol 2022; 18:e10886. [PMID: 36366891 PMCID: PMC9650275 DOI: 10.15252/msb.202110886] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
During development, cell state transitions are coordinated through changes in the identity of molecular regulators in a cell type‐ and dose‐specific manner. The ability to rationally engineer such transitions in human pluripotent stem cells (hPSC) will enable numerous applications in regenerative medicine. Herein, we report the generation of synthetic gene circuits that can detect a desired cell state using AND‐like logic integration of endogenous miRNAs (classifiers) and, upon detection, produce fine‐tuned levels of output proteins using an miRNA‐mediated output fine‐tuning technology (miSFITs). Specifically, we created an “hPSC ON” circuit using a model‐guided miRNA selection and circuit optimization approach. The circuit demonstrates robust PSC‐specific detection and graded output protein production. Next, we used an empirical approach to create an “hPSC‐Off” circuit. This circuit was applied to regulate the secretion of endogenous BMP4 in a state‐specific and fine‐tuned manner to control the composition of differentiating hPSCs. Our work provides a platform for customized cell state‐specific control of desired physiological factors in hPSC, laying the foundation for programming cell compositions in hPSC‐derived tissues and beyond.
Collapse
Affiliation(s)
- Laura Prochazka
- Institute of Biomedical Engineering (BME) University of Toronto Toronto ON Canada
- Donnelly Centre for Cellular & Biomolecular Research University of Toronto Toronto ON Canada
| | - Yale S Michaels
- Michael Smith Laboratories University of British Columbia Vancouver BC Canada
- School of Biomedical Engineering University of British Columbia Vancouver BC Canada
| | - Charles Lau
- Institute of Biomedical Engineering (BME) University of Toronto Toronto ON Canada
- Donnelly Centre for Cellular & Biomolecular Research University of Toronto Toronto ON Canada
- Michael Smith Laboratories University of British Columbia Vancouver BC Canada
- School of Biomedical Engineering University of British Columbia Vancouver BC Canada
| | - Ross D Jones
- Michael Smith Laboratories University of British Columbia Vancouver BC Canada
- School of Biomedical Engineering University of British Columbia Vancouver BC Canada
| | - Mona Siu
- Michael Smith Laboratories University of British Columbia Vancouver BC Canada
- School of Biomedical Engineering University of British Columbia Vancouver BC Canada
| | - Ting Yin
- Institute of Biomedical Engineering (BME) University of Toronto Toronto ON Canada
- Donnelly Centre for Cellular & Biomolecular Research University of Toronto Toronto ON Canada
| | - Diana Wu
- Institute of Biomedical Engineering (BME) University of Toronto Toronto ON Canada
- Donnelly Centre for Cellular & Biomolecular Research University of Toronto Toronto ON Canada
| | - Esther Jang
- Institute of Biomedical Engineering (BME) University of Toronto Toronto ON Canada
- Donnelly Centre for Cellular & Biomolecular Research University of Toronto Toronto ON Canada
| | - Mercedes Vázquez‐Cantú
- Institute of Biomedical Engineering (BME) University of Toronto Toronto ON Canada
- Donnelly Centre for Cellular & Biomolecular Research University of Toronto Toronto ON Canada
- Swiss Federal Institute of Technology (ETH) Zürich, Department of Biosystems Science and Engineering (D‐BSSE) Basel Switzerland
| | - Penney M Gilbert
- Institute of Biomedical Engineering (BME) University of Toronto Toronto ON Canada
- Donnelly Centre for Cellular & Biomolecular Research University of Toronto Toronto ON Canada
- Department of Cell and Systems Biology University of Toronto Toronto ON Canada
| | - Himanshu Kaul
- School of Engineering University of Leicester Leicester UK
- Department of Respiratory Sciences University of Leicester Leicester UK
| | - Yaakov Benenson
- Swiss Federal Institute of Technology (ETH) Zürich, Department of Biosystems Science and Engineering (D‐BSSE) Basel Switzerland
| | - Peter W Zandstra
- Michael Smith Laboratories University of British Columbia Vancouver BC Canada
- School of Biomedical Engineering University of British Columbia Vancouver BC Canada
| |
Collapse
|
15
|
Barbulescu GI, Bojin FM, Ordodi VL, Goje ID, Barbulescu AS, Paunescu V. Decellularized Extracellular Matrix Scaffolds for Cardiovascular Tissue Engineering: Current Techniques and Challenges. Int J Mol Sci 2022; 23:13040. [PMID: 36361824 PMCID: PMC9658138 DOI: 10.3390/ijms232113040] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/18/2022] [Accepted: 10/26/2022] [Indexed: 08/13/2023] Open
Abstract
Cardiovascular diseases are the leading cause of global mortality. Over the past two decades, researchers have tried to provide novel solutions for end-stage heart failure to address cardiac transplantation hurdles such as donor organ shortage, chronic rejection, and life-long immunosuppression. Cardiac decellularized extracellular matrix (dECM) has been widely explored as a promising approach in tissue-regenerative medicine because of its remarkable similarity to the original tissue. Optimized decellularization protocols combining physical, chemical, and enzymatic agents have been developed to obtain the perfect balance between cell removal, ECM composition, and function maintenance. However, proper assessment of decellularized tissue composition is still needed before clinical translation. Recellularizing the acellular scaffold with organ-specific cells and evaluating the extent of cardiomyocyte repopulation is also challenging. This review aims to discuss the existing literature on decellularized cardiac scaffolds, especially on the advantages and methods of preparation, pointing out areas for improvement. Finally, an overview of the state of research regarding the application of cardiac dECM and future challenges in bioengineering a human heart suitable for transplantation is provided.
Collapse
Affiliation(s)
- Greta Ionela Barbulescu
- Immuno-Physiology and Biotechnologies Center (CIFBIOTEH), Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Department of Clinical Practical Skills, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Florina Maria Bojin
- Immuno-Physiology and Biotechnologies Center (CIFBIOTEH), Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Clinical Emergency County Hospital “Pius Brinzeu” Timisoara, Center for Gene and Cellular Therapies in the Treatment of Cancer Timisoara-OncoGen, No 156 Liviu Rebreanu, 300723 Timisoara, Romania
| | - Valentin Laurentiu Ordodi
- Clinical Emergency County Hospital “Pius Brinzeu” Timisoara, Center for Gene and Cellular Therapies in the Treatment of Cancer Timisoara-OncoGen, No 156 Liviu Rebreanu, 300723 Timisoara, Romania
- Faculty of Industrial Chemistry and Environmental Engineering, “Politehnica” University Timisoara, No 2 Victoriei Square, 300006 Timisoara, Romania
| | - Iacob Daniel Goje
- Department of Medical Semiology I, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Advanced Cardiology and Hemostaseology Research Center, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Andreea Severina Barbulescu
- Center for Advanced Research in Gastroenterology and Hepatology, Department of Internal Medicine II, Division of Gastroenterology and Hepatology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Virgil Paunescu
- Immuno-Physiology and Biotechnologies Center (CIFBIOTEH), Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Clinical Emergency County Hospital “Pius Brinzeu” Timisoara, Center for Gene and Cellular Therapies in the Treatment of Cancer Timisoara-OncoGen, No 156 Liviu Rebreanu, 300723 Timisoara, Romania
| |
Collapse
|
16
|
Li CY, Liang Z, Hu Y, Zhang H, Setiasabda KD, Li J, Ma S, Xia X, Kuang Y. Cytidine-containing tails robustly enhance and prolong protein production of synthetic mRNA in cell and in vivo. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 30:300-310. [PMID: 36320322 PMCID: PMC9614650 DOI: 10.1016/j.omtn.2022.10.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
Synthetic mRNAs are rising rapidly as alternative therapeutic agents for delivery of proteins. However, the practical use of synthetic mRNAs has been restricted by their low cellular stability as well as poor protein production efficiency. The key roles of poly(A) tail on mRNA biology inspire us to explore the optimization of tail sequence to overcome the aforementioned limitations. Here, the systematic substitution of non-A nucleotides in the tails revealed that cytidine-containing tails can substantially enhance the protein production rate and duration of synthetic mRNAs both in vitro and in vivo. Such C-containing tails shield synthetic mRNAs from deadenylase CCR4-NOT transcription complex, as the catalytic CNOT proteins, especially CNOT6L and CNOT7, have lower efficiency in trimming of cytidine. Consistently, these enhancement effects of C-containing tails were observed on all synthetic mRNAs tested and were independent of transfection reagents and cell types. As the C-containing tails can be used along with other mRNA enhancement technologies to synergically boost protein production, we believe that these tails can be broadly used on synthetic mRNAs to directly promote their clinical applications.
Collapse
Affiliation(s)
- Cheuk Yin Li
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Zhenghua Liang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Yaxin Hu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Hongxia Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Kharis Daniel Setiasabda
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Jiawei Li
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong 518057, China
| | - Shaohua Ma
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong 518057, China
| | - Xiaojun Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Yi Kuang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China,HKUST Shenzhen Research Institute, Shenzhen, Guangdong 518057, China,Corresponding author Yi Kuang, Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Hong Kong, Hong Kong.
| |
Collapse
|
17
|
Rust R, Weber RZ, Generali M, Kehl D, Bodenmann C, Uhr D, Wanner D, Zürcher KJ, Saito H, Hoerstrup SP, Nitsch RM, Tackenberg C. Xeno-free induced pluripotent stem cell-derived neural progenitor cells for in vivo applications. J Transl Med 2022; 20:421. [PMID: 36114512 PMCID: PMC9482172 DOI: 10.1186/s12967-022-03610-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/24/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Currently, there is no regenerative therapy for patients with neurological and neurodegenerative disorders. Cell-therapies have emerged as a potential treatment for numerous brain diseases. Despite recent advances in stem cell technology, major concerns have been raised regarding the feasibility and safety of cell therapies for clinical applications. METHODS We generated good manufacturing practice (GMP)-compatible neural progenitor cells (NPCs) from transgene- and xeno-free induced pluripotent stem cells (iPSCs) that can be smoothly adapted for clinical applications. NPCs were characterized in vitro for their differentiation potential and in vivo after transplantation into wild type as well as genetically immunosuppressed mice. RESULTS Generated NPCs had a stable gene-expression over at least 15 passages and could be scaled for up to 1018 cells per initially seeded 106 cells. After withdrawal of growth factors in vitro, cells adapted a neural fate and mainly differentiated into active neurons. To ensure a pure NPC population for in vivo applications, we reduced the risk of iPSC contamination by applying micro RNA-switch technology as a safety checkpoint. Using lentiviral transduction with a fluorescent and bioluminescent dual-reporter construct, combined with non-invasive in vivo bioluminescent imaging, we longitudinally tracked the grafted cells in healthy wild-type and genetically immunosuppressed mice as well as in a mouse model of ischemic stroke. Long term in-depth characterization revealed that transplanted NPCs have the capability to survive and spontaneously differentiate into functional and mature neurons throughout a time course of a month, while no residual pluripotent cells were detectable. CONCLUSION We describe the generation of transgene- and xeno-free NPCs. This simple differentiation protocol combined with the ability of in vivo cell tracking presents a valuable tool to develop safe and effective cell therapies for various brain injuries.
Collapse
Affiliation(s)
- Ruslan Rust
- Institute for Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland.
| | - Rebecca Z Weber
- Institute for Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Melanie Generali
- Institute for Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Debora Kehl
- Institute for Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Chantal Bodenmann
- Institute for Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Daniela Uhr
- Institute for Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Debora Wanner
- Institute for Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Kathrin J Zürcher
- Institute for Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Hirohide Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Simon P Hoerstrup
- Institute for Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
- Wyss Translational Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Christian Tackenberg
- Institute for Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland.
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
18
|
Nakashima Y, Yoshida S, Tsukahara M. Semi-3D cultures using Laminin 221 as a coating material for human induced pluripotent stem cells. Regen Biomater 2022; 9:rbac060. [PMID: 36176714 PMCID: PMC9514851 DOI: 10.1093/rb/rbac060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/09/2022] [Accepted: 08/21/2022] [Indexed: 11/19/2022] Open
Abstract
It was previously believed that human induced pluripotent stem cells (hiPSCs) did not show adhesion to the coating material Laminin 221, which is known to have specific affinity for cardiomyocytes. In this study, we report that human mononuclear cell-derived hiPSCs, established with Sendai virus vector, form peninsular-like colonies rather than embryonic stem cell-like colonies; these peninsular-like colonies can be passaged more than 10 times after establishment. Additionally, initialization-deficient cells with residual Sendai virus vector adhered to the coating material Laminin 511 but not to Laminin 221. Therefore, the expression of undifferentiated markers tended to be higher in hiPSCs established on Laminin 221 than on Laminin 511. On Laminin 221, hiPSCs15M66 showed a semi-floating colony morphology. The expression of various markers of cell polarity was significantly lower in hiPSCs cultured on Laminin 221 than in hiPSCs cultured on Laminin 511. Furthermore, 201B7 and 15M66 hiPSCs showed 3D cardiomyocyte differentiation on Laminin 221. Thus, the coating material Laminin 221 provides semi-floating culture conditions for the establishment, culture and induced differentiation of hiPSCs.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto 606-8397, Japan
| | - Shinsuke Yoshida
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto 606-8397, Japan
| | - Masayoshi Tsukahara
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto 606-8397, Japan
| |
Collapse
|
19
|
Lu Q, Hu Y, Yin Li C, Kuang Y. Aptamer-Array-Guided Protein Assembly Enhances Synthetic mRNA Switch Performance. Angew Chem Int Ed Engl 2022; 61:e202207319. [PMID: 35703374 PMCID: PMC9544043 DOI: 10.1002/anie.202207319] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Indexed: 11/17/2022]
Abstract
Synthetic messenger RNA (mRNA) switches are powerful synthetic biological tools that can sense cellular molecules to manipulate cell fate. However, their performances are limited by high output signal noise due to leaky output protein expression. Here, we designed a readout control module that disables protein leakage from generating signal. Aptamer array on the switch guides the inactive output protein to self-assemble into functional assemblies that generate output signal. Leaky protein expression fails to saturate the array, thus produces marginal signal. In this study, we demonstrated that switches with this module exhibit substantially lower signal noise and, consequently, higher input sensitivity and wider output range. Such switches are applicable for different types of input molecules and output proteins. The work here demonstrates a new type of spatially guided protein self-assembly, affording novel synthetic mRNA switches that promise accurate cell manipulation for biomedical applications.
Collapse
Affiliation(s)
- Qiuyu Lu
- Department of Chemical and Biological EngineeringThe Hong Kong University of Science and TechnologyClear Water Bay, Kowloon, Hong KongHong Kong
| | - Yaxin Hu
- Department of Chemical and Biological EngineeringThe Hong Kong University of Science and TechnologyClear Water Bay, Kowloon, Hong KongHong Kong
| | - Cheuk Yin Li
- Department of Chemical and Biological EngineeringThe Hong Kong University of Science and TechnologyClear Water Bay, Kowloon, Hong KongHong Kong
| | - Yi Kuang
- Department of Chemical and Biological EngineeringThe Hong Kong University of Science and TechnologyClear Water Bay, Kowloon, Hong KongHong Kong
- HKUST Shenzhen Research InstituteShenzhenGuangdongChina
| |
Collapse
|
20
|
Highly sensitive and non-disruptive detection of residual undifferentiated cells by measuring miRNAs in culture supernatant. Sci Rep 2022; 12:10351. [PMID: 35725891 PMCID: PMC9209417 DOI: 10.1038/s41598-022-14273-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 06/03/2022] [Indexed: 11/10/2022] Open
Abstract
The clinical usage of induced pluripotent stem cell (iPSC)-derived regenerative medicine products is limited by the possibility of residual undifferentiated cells forming tumours after transplantation. Most of the existing quality control tests involve crushing of cells. As a result, the cells to be transplanted cannot be directly tested, thereby increasing the cost of transplantation. Therefore, we tested a highly sensitive and non-disruptive quality-testing method that involves measuring microRNAs (miRNAs) in culture supernatants released by cells. By measuring miR-302b in the culture supernatant, residual iPSCs were detected with higher sensitivity than by measuring LIN28 (Lin-28 Homolog A) in the cells. To use this method, we also monitored the progression of differentiation. Our novel highly sensitive and non-disruptive method for detecting residual undifferentiated cells will contribute to reducing the manufacturing cost of iPSC-derived products and improving the safety of transplantation.
Collapse
|
21
|
Lu Q, Hu Y, Li CY, Kuang Y. Aptamer‐Array‐Guided Protein Assembly Enhances Synthetic mRNA Switch Performance. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202207319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Qiuyu Lu
- Hong Kong University of Science and Technology School of Engineering Chemical and Biological Engineering HONG KONG
| | - Yaxin Hu
- Hong Kong University of Science and Technology School of Engineering Chemical and Biological Engineering HONG KONG
| | - Cheuk Yin Li
- Hong Kong University of Science and Technology School of Engineering Chemical and Biological Engineering HONG KONG
| | - Yi Kuang
- Hong Kong University of Science and Technology Chemical and Biological Engineering Room 5578, Academic Bldg,Clear Water Bay 000000 Kowloon HONG KONG
| |
Collapse
|
22
|
Yasui R, Matsui A, Sekine K, Okamoto S, Taniguchi H. Highly Sensitive Detection of Human Pluripotent Stem Cells by Loop-Mediated Isothermal Amplification. Stem Cell Rev Rep 2022; 18:2995-3007. [PMID: 35661077 PMCID: PMC9622575 DOI: 10.1007/s12015-022-10402-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2022] [Indexed: 11/24/2022]
Abstract
For safe regenerative medicines, contaminated or remaining tumorigenic undifferentiated cells in cell-derived products must be rigorously assessed through sensitive assays. Although in vitro nucleic acid tests offer particularly sensitive tumorigenicity-associated assays, the human pluripotent stem cell (hPSC) detectability is partly constrained by the small input amount of RNA per test. To overcome this limitation, we developed reverse transcription loop-mediated isothermal amplification (RT-LAMP) assays that are highly gene specific and robust against interfering materials. LAMP could readily assay microgram order of input sample per test and detected an equivalent model of 0.00002% hiPSC contamination in a simple one-pot reaction. For the evaluation of cell-derived total RNA, RT-LAMP detected spiked-in hPSCs among hPSC-derived trilineage cells utilizing multiple pluripotency RNAs. We also developed multiplex RT-LAMP assays and further applied for in situ cell imaging, achieving specific co-staining of pluripotency proteins and RNAs. Our attempts uncovered the utility of RT-LAMP approaches for tumorigenicity-associated assays, supporting practical applications of regenerative medicine.
Collapse
Affiliation(s)
- Ryota Yasui
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, 236-0004, Japan
- Fundamental Research Laboratory, Eiken Chemical Co., Ltd., Nogi, Tochigi, 329-0114, Japan
| | - Atsuka Matsui
- Biochemical Research Laboratory, Eiken Chemical Co., Ltd., Ohtawara, Tochigi, 324-0036, Japan
| | - Keisuke Sekine
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, 236-0004, Japan.
- Laboratory of Cancer Cell Systems, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| | - Satoshi Okamoto
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, 236-0004, Japan.
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.
| |
Collapse
|
23
|
Morita Y, Kishino Y, Fukuda K, Tohyama S. Scalable manufacturing of clinical-grade differentiated cardiomyocytes derived from human-induced pluripotent stem cells for regenerative therapy. Cell Prolif 2022; 55:e13248. [PMID: 35534945 PMCID: PMC9357358 DOI: 10.1111/cpr.13248] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 12/17/2022] Open
Abstract
Basic research on human pluripotent stem cell (hPSC)‐derived cardiomyocytes (CMs) for cardiac regenerative therapy is one of the most active and complex fields to achieve this alternative to heart transplantation and requires the integration of medicine, science, and engineering. Mortality in patients with heart failure remains high worldwide. Although heart transplantation is the sole strategy for treating severe heart failure, the number of donors is limited. Therefore, hPSC‐derived CM (hPSC‐CM) transplantation is expected to replace heart transplantation. To achieve this goal, for basic research, various issues should be considered, including how to induce hPSC proliferation efficiently for cardiac differentiation, induce hPSC‐CMs, eliminate residual undifferentiated hPSCs and non‐CMs, and assess for the presence of residual undifferentiated hPSCs in vitro and in vivo. In this review, we discuss the current stage of resolving these issues and future directions for realizing hPSC‐based cardiac regenerative therapy.
Collapse
Affiliation(s)
- Yuika Morita
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Synthetic RNA-based post-transcriptional expression control methods and genetic circuits. Adv Drug Deliv Rev 2022; 184:114196. [PMID: 35288218 DOI: 10.1016/j.addr.2022.114196] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/27/2022] [Accepted: 03/08/2022] [Indexed: 12/19/2022]
Abstract
RNA-based synthetic genetic circuits provide an alternative for traditional transcription-based circuits in applications where genomic integration is to be avoided. Incorporating various post-transcriptional control methods into such circuits allows for controlling the behaviour of the circuit through the detection of certain biomolecular inputs or reconstituting defined circuit behaviours, thus manipulating cellular functions. In this review, recent developments of various types of post-transcriptional control methods in mammalian cells are discussed as well as auxiliary components that allow for the creation and development of mRNA-based switches. How such post-transcriptional switches are combined into synthetic circuits as well as their applications in biomedical and preclinical settings are also described. Finally, we examine the challenges that need to be surmounted before RNA-based synthetic circuits can be reliably deployed into clinical settings.
Collapse
|
25
|
Fujita Y, Hirosawa M, Hayashi K, Hatani T, Yoshida Y, Yamamoto T, Saito H. A versatile and robust cell purification system with an RNA-only circuit composed of microRNA-responsive ON and OFF switches. SCIENCE ADVANCES 2022; 8:eabj1793. [PMID: 34985961 PMCID: PMC8730616 DOI: 10.1126/sciadv.abj1793] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Human induced pluripotent stem cells (iPSCs) are promising cell resources for cell therapy and drug discovery. However, iPSC-derived differentiated cells are often heterogenous and need purification using a flow cytometer, which has high cost and time consumption for large-scale purification. MicroRNAs (miRNAs) can be used as cell selection markers, because their activity differs between cell types. Here, we show miRNA-responsive ON and OFF switch mRNAs for robust cell purification. The ON switch contains a miRNA-target sequence after the polyadenylate tail, triggering translational activation by sensing the target miRNA. By designing RNA-only circuits with miRNA-ON and -OFF switch mRNAs that encode a lethal ribonuclease, Barnase, and its inhibitor, Barstar, we efficiently purified specific cell types, including human iPSCs and differentiated cardiomyocytes, without flow cytometry. Synthetic mRNA circuits composed of ON and OFF switches provide a safe, versatile, and time-saving method to purify various cell types for biological and clinical applications.
Collapse
Affiliation(s)
- Yoshihiko Fujita
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
- Corresponding author. (H.S.); (Y.F.)
| | - Moe Hirosawa
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Karin Hayashi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takeshi Hatani
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yoshinori Yoshida
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501 Japan
- Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto 606-8507, Japan
| | - Hirohide Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
- Corresponding author. (H.S.); (Y.F.)
| |
Collapse
|
26
|
Nakanishi H, Itaka K. Synthetic mRNA for ex vivo therapeutic applications. Drug Metab Pharmacokinet 2022; 44:100447. [DOI: 10.1016/j.dmpk.2022.100447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 01/06/2023]
|
27
|
Soma Y, Morita Y, Kishino Y, Kanazawa H, Fukuda K, Tohyama S. The Present State and Future Perspectives of Cardiac Regenerative Therapy Using Human Pluripotent Stem Cells. Front Cardiovasc Med 2021; 8:774389. [PMID: 34957258 PMCID: PMC8692665 DOI: 10.3389/fcvm.2021.774389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 10/25/2021] [Indexed: 12/13/2022] Open
Abstract
The number of patients with heart failure (HF) is increasing with aging in our society worldwide. Patients with HF who are resistant to medication and device therapy are candidates for heart transplantation (HT). However, the shortage of donor hearts is a serious issue. As an alternative to HT, cardiac regenerative therapy using human pluripotent stem cells (hPSCs), such as human embryonic stem cells and induced pluripotent stem cells, is expected to be realized. Differentiation of hPSCs into cardiomyocytes (CMs) is facilitated by mimicking normal heart development. To prevent tumorigenesis after transplantation, it is important to eliminate non-CMs, including residual hPSCs, and select only CMs. Among many CM selection systems, metabolic selection based on the differences in metabolism between CMs and non-CMs is favorable in terms of cost and efficacy. Large-scale culture systems have been developed because a large number of hPSC-derived CMs (hPSC-CMs) are required for transplantation in clinical settings. In large animal models, hPSC-CMs transplanted into the myocardium improved cardiac function in a myocardial infarction model. Although post-transplantation arrhythmia and immune rejection remain problems, their mechanisms and solutions are under investigation. In this manner, the problems of cardiac regenerative therapy are being solved individually. Thus, cardiac regenerative therapy with hPSC-CMs is expected to become a safe and effective treatment for HF in the near future. In this review, we describe previous studies related to hPSC-CMs and discuss the future perspectives of cardiac regenerative therapy using hPSC-CMs.
Collapse
Affiliation(s)
- Yusuke Soma
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Yuika Morita
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshikazu Kishino
- 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
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
28
|
Tani H, Tohyama S, Kishino Y, Kanazawa H, Fukuda K. Production of functional cardiomyocytes and cardiac tissue from human induced pluripotent stem cells for regenerative therapy. J Mol Cell Cardiol 2021; 164:83-91. [PMID: 34822838 DOI: 10.1016/j.yjmcc.2021.11.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/02/2021] [Accepted: 11/17/2021] [Indexed: 12/28/2022]
Abstract
The emergence of human induced pluripotent stem cells (hiPSCs) has revealed the potential for curing end-stage heart failure. Indeed, transplantation of hiPSC-derived cardiomyocytes (hiPSC-CMs) may have applications as a replacement for heart transplantation and conventional regenerative therapies. However, there are several challenges that still must be overcome for clinical applications, including large-scale production of hiPSCs and hiPSC-CMs, elimination of residual hiPSCs, purification of hiPSC-CMs, maturation of hiPSC-CMs, efficient engraftment of transplanted hiPSC-CMs, development of an injection device, and avoidance of post-transplant arrhythmia and immunological rejection. Thus, we developed several technologies based on understanding of the metabolic profiles of hiPSCs and hiPSC derivatives. In this review, we outline how to overcome these hurdles to realize the transplantation of hiPSC-CMs in patients with heart failure and introduce cutting-edge findings and perspectives for future regenerative therapy.
Collapse
Affiliation(s)
- Hidenori Tani
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan; Department of Emergency and Critical Care Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan.
| | - Yoshikazu Kishino
- 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
|
29
|
Keller A, Spits C. The Impact of Acquired Genetic Abnormalities on the Clinical Translation of Human Pluripotent Stem Cells. Cells 2021; 10:cells10113246. [PMID: 34831467 PMCID: PMC8625075 DOI: 10.3390/cells10113246] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/07/2021] [Accepted: 11/17/2021] [Indexed: 12/23/2022] Open
Abstract
Human pluripotent stem cells (hPSC) are known to acquire chromosomal abnormalities, which range from point mutations to large copy number changes, including full chromosome aneuploidy. These aberrations have a wide-ranging influence on the state of cells, in both the undifferentiated and differentiated state. Currently, very little is known on how these abnormalities will impact the clinical translation of hPSC, and particularly their potential to prime cells for oncogenic transformation. A further complication is that many of these abnormalities exist in a mosaic state in culture, which complicates their detection with conventional karyotyping methods. In this review we discuss current knowledge on how these aberrations influence the cell state and how this may impact the future of research and the cells’ clinical potential.
Collapse
|
30
|
Hunkler HJ, Groß S, Thum T, Bär C. Non-coding RNAs: key regulators of reprogramming, pluripotency, and cardiac cell specification with therapeutic perspective for heart regeneration. Cardiovasc Res 2021; 118:3071-3084. [PMID: 34718448 PMCID: PMC9732524 DOI: 10.1093/cvr/cvab335] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/27/2021] [Indexed: 01/01/2023] Open
Abstract
Myocardial infarction causes a massive loss of cardiomyocytes (CMs), which can lead to heart failure accompanied by fibrosis, stiffening of the heart, and loss of function. Heart failure causes high mortality rates and is a huge socioeconomic burden, which, based on diets and lifestyle in the developed world, is expected to increase further in the next years. At present, the only curative treatment for heart failure is heart transplantation associated with a number of limitations such as donor organ availability and transplant rejection among others. Thus, the development of cellular reprogramming and defined differentiation protocols provide exciting new possibilities for cell therapy approaches and which opened up a new era in regenerative medicine. Consequently, tremendous research efforts were undertaken to gain a detailed molecular understanding of the reprogramming processes and the in vitro differentiation of pluripotent stem cells into functional CMs for transplantation into the patient's injured heart. In the last decade, non-coding RNAs, particularly microRNAs, long non-coding RNAs, and circular RNAs emerged as critical regulators of gene expression that were shown to fine-tune cellular processes both on the transcriptional and the post-transcriptional level. Unsurprisingly, also cellular reprogramming, pluripotency, and cardiac differentiation and maturation are regulated by non-coding RNAs. In here, we review the current knowledge on non-coding RNAs in these processes and highlight how their modulation may enhance the quality and quantity of stem cells and their derivatives for safe and efficient clinical application in patients with heart failure. In addition, we summarize the clinical cell therapy efforts undertaken thus far.
Collapse
Affiliation(s)
- Hannah J Hunkler
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Sonja Groß
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Thomas Thum
- Corresponding authors. Tel: +49 511 532 5272; fax: +49 511 532 5274, E-mail: (T.T.); Tel: +49 511 532 2883; fax: +49 511 532 5274, E-mail: (C.B.)
| | - Christian Bär
- Corresponding authors. Tel: +49 511 532 5272; fax: +49 511 532 5274, E-mail: (T.T.); Tel: +49 511 532 2883; fax: +49 511 532 5274, E-mail: (C.B.)
| |
Collapse
|
31
|
Purification of Specific Cell Populations Differentiated from Stem Cells Using MicroRNA-Responsive Synthetic Messenger RNAs. Methods Mol Biol 2021; 2312:73-86. [PMID: 34228285 DOI: 10.1007/978-1-0716-1441-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
Abstract
Pluripotent stem cells have the potential to differentiate into various cell types that can be used for basic biological studies, drug discovery, and regenerative medicine. To obtain reliable results using the differentiated cells, the contamination of nontarget cells should be avoided. microRNAs (miRNAs) can serve as indicators to distinguish target and nontarget cells, because the activities of miRNAs are different among cell types.In this chapter, we introduce a method to purify target cells using synthetic messenger RNAs (mRNAs) that respond to cell-specific miRNAs. The method is composed of five steps: mRNA sequence design, template DNA preparation by PCR, in vitro mRNA transcription, mRNA transfection into cells, and fluorescence-activated cell sorting. This synthetic mRNA-based cell purification method will advance various applications of pluripotent stem cells.
Collapse
|
32
|
Ray A, Joshi JM, Sundaravadivelu PK, Raina K, Lenka N, Kaveeshwar V, Thummer RP. An Overview on Promising Somatic Cell Sources Utilized for the Efficient Generation of Induced Pluripotent Stem Cells. Stem Cell Rev Rep 2021; 17:1954-1974. [PMID: 34100193 DOI: 10.1007/s12015-021-10200-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 01/19/2023]
Abstract
Human induced Pluripotent Stem Cells (iPSCs) have enormous potential in understanding developmental biology, disease modeling, drug discovery, and regenerative medicine. The initial human iPSC studies used fibroblasts as a starting cell source to reprogram them; however, it has been identified to be a less appealing somatic cell source by numerous studies due to various reasons. One of the important criteria to achieve efficient reprogramming is determining an appropriate starting somatic cell type to induce pluripotency since the cellular source has a major influence on the reprogramming efficiency, kinetics, and quality of iPSCs. Therefore, numerous groups have explored various somatic cell sources to identify the promising sources for reprogramming into iPSCs with different reprogramming factor combinations. This review provides an overview of promising easily accessible somatic cell sources isolated in non-invasive or minimally invasive manner such as keratinocytes, urine cells, and peripheral blood mononuclear cells used for the generation of human iPSCs derived from healthy and diseased subjects. Notably, iPSCs generated from one of these cell types derived from the patient will offer ethical and clinical advantages. In addition, these promising somatic cell sources have the potential to efficiently generate bona fide iPSCs with improved reprogramming efficiency and faster kinetics. This knowledge will help in establishing strategies for safe and efficient reprogramming and the generation of patient-specific iPSCs from these cell types.
Collapse
Affiliation(s)
- Arnab Ray
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Jahnavy Madhukar Joshi
- Central Research Laboratory, SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara University, Dharwad, 580009, Karnataka, India
| | - Pradeep Kumar Sundaravadivelu
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Khyati Raina
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Nibedita Lenka
- National Centre for Cell Science, S. P. Pune University Campus, Pune - 411007, Ganeshkhind, Maharashtra, India
| | - Vishwas Kaveeshwar
- Central Research Laboratory, SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara University, Dharwad, 580009, Karnataka, India.
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
33
|
Flahou C, Morishima T, Takizawa H, Sugimoto N. Fit-For-All iPSC-Derived Cell Therapies and Their Evaluation in Humanized Mice With NK Cell Immunity. Front Immunol 2021; 12:662360. [PMID: 33897711 PMCID: PMC8059435 DOI: 10.3389/fimmu.2021.662360] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Human induced pluripotent stem cells (iPSCs) can be limitlessly expanded and differentiated into almost all cell types. Moreover, they are amenable to gene manipulation and, because they are established from somatic cells, can be established from essentially any person. Based on these characteristics, iPSCs have been extensively studied as cell sources for tissue grafts, blood transfusions and cancer immunotherapies, and related clinical trials have started. From an immune-matching perspective, autologous iPSCs are perfectly compatible in principle, but also require a prolonged time for reaching the final products, have high cost, and person-to-person variation hindering their common use. Therefore, certified iPSCs with reduced immunogenicity are expected to become off-the-shelf sources, such as those made from human leukocyte antigen (HLA)-homozygous individuals or genetically modified for HLA depletion. Preclinical tests using immunodeficient mice reconstituted with a human immune system (HIS) serve as an important tool to assess the human alloresponse against iPSC-derived cells. Especially, HIS mice reconstituted with not only human T cells but also human natural killer (NK) cells are considered crucial. NK cells attack so-called “missing self” cells that do not express self HLA class I, which include HLA-homozygous cells that express only one allele type and HLA-depleted cells. However, conventional HIS mice lack enough reconstituted human NK cells for these tests. Several measures have been developed to overcome this issue including the administration of cytokines that enhance NK cell expansion, such as IL-2 and IL-15, the administration of vectors that express those cytokines, and genetic manipulation to express the cytokines or to enhance the reconstitution of human myeloid cells that express IL15R-alpha. Using such HIS mice with enhanced human NK cell reconstitution, alloresponses against HLA-homozygous and HLA-depleted cells have been studied. However, most studies used HLA-downregulated tumor cells as the target cells and tested in vitro after purifying human cells from HIS mice. In this review, we give an overview of the current state of iPSCs in cell therapies, strategies to lessen their immunogenic potential, and then expound on the development of HIS mice with reconstituted NK cells, followed by their utilization in evaluating future universal HLA-engineered iPSC-derived cells.
Collapse
Affiliation(s)
- Charlotte Flahou
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Tatsuya Morishima
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan.,Laboratory of Hematopoietic Stem Cell Engineering, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Hitoshi Takizawa
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Naoshi Sugimoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
34
|
Salicylic diamines selectively eliminate residual undifferentiated cells from pluripotent stem cell-derived cardiomyocyte preparations. Sci Rep 2021; 11:2391. [PMID: 33504837 PMCID: PMC7841182 DOI: 10.1038/s41598-021-81351-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
Abstract
Clinical translation of pluripotent stem cell (PSC) derivatives is hindered by the tumorigenic risk from residual undifferentiated cells. Here, we identified salicylic diamines as potent agents exhibiting toxicity to murine and human PSCs but not to cardiomyocytes (CMs) derived from them. Half maximal inhibitory concentrations (IC50) of small molecules SM2 and SM6 were, respectively, 9- and 18-fold higher for human than murine PSCs, while the IC50 of SM8 was comparable for both PSC groups. Treatment of murine embryoid bodies in suspension differentiation cultures with the most effective small molecule SM6 significantly reduced PSC and non-PSC contamination and enriched CM populations that would otherwise be eliminated in genetic selection approaches. All tested salicylic diamines exerted their toxicity by inhibiting the oxygen consumption rate (OCR) in PSCs. No or only minimal and reversible effects on OCR, sarcomeric integrity, DNA stability, apoptosis rate, ROS levels or beating frequency were observed in PSC-CMs, although effects on human PSC-CMs seemed to be more deleterious at higher SM-concentrations. Teratoma formation from SM6-treated murine PSC-CMs was abolished or delayed compared to untreated cells. We conclude that salicylic diamines represent promising compounds for PSC removal and enrichment of CMs without the need for other selection strategies.
Collapse
|
35
|
Nakanishi H, Yoshii T, Kawasaki S, Hayashi K, Tsutsui K, Oki C, Tsukiji S, Saito H. Light-controllable RNA-protein devices for translational regulation of synthetic mRNAs in mammalian cells. Cell Chem Biol 2021; 28:662-674.e5. [PMID: 33508227 DOI: 10.1016/j.chembiol.2021.01.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/26/2020] [Accepted: 01/04/2021] [Indexed: 12/20/2022]
Abstract
The photo-regulation of transgene expression is one effective approach in mammalian synthetic biology due to its high spatial and temporal resolution. While DNAs are mainly used as vectors, modified RNAs (modRNAs) are also useful for medical applications of synthetic biology, because they can avoid insertional mutagenesis and immunogenicity. However, the optogenetic control of modRNA-delivered transgenes is much more difficult than that of DNA-delivered transgenes. Here, we develop two types of photo-controllable translational activation systems that are compatible with modRNAs. One is composed of a heterodimerization domain-fused split translational activator protein and a photocaged heterodimerizer. The other is composed of a destabilizing domain-fused translational activator protein and a photocaged stabilizer. The destabilized type can be used for not only translational activation but also translational repression of the modRNAs. These photo-controllable translation systems will expand the application of mammalian synthetic biology research.
Collapse
Affiliation(s)
- Hideyuki Nakanishi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Tatsuyuki Yoshii
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan; PRESTO, Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Shunsuke Kawasaki
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Karin Hayashi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Keita Tsutsui
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
| | - Choji Oki
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
| | - Shinya Tsukiji
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan; Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
| | - Hirohide Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
36
|
Ford E, Pearlman J, Ruan T, Manion J, Waller M, Neely GG, Caron L. Human Pluripotent Stem Cells-Based Therapies for Neurodegenerative Diseases: Current Status and Challenges. Cells 2020; 9:E2517. [PMID: 33233861 PMCID: PMC7699962 DOI: 10.3390/cells9112517] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases are characterized by irreversible cell damage, loss of neuronal cells and limited regeneration potential of the adult nervous system. Pluripotent stem cells are capable of differentiating into the multitude of cell types that compose the central and peripheral nervous systems and so have become the major focus of cell replacement therapies for the treatment of neurological disorders. Human embryonic stem cell (hESC) and human induced pluripotent stem cell (hiPSC)-derived cells have both been extensively studied as cell therapies in a wide range of neurodegenerative disease models in rodents and non-human primates, including Parkinson's disease, stroke, epilepsy, spinal cord injury, Alzheimer's disease, multiple sclerosis and pain. In this review, we discuss the latest progress made with stem cell therapies targeting these pathologies. We also evaluate the challenges in clinical application of human pluripotent stem cell (hPSC)-based therapies including risk of oncogenesis and tumor formation, immune rejection and difficulty in regeneration of the heterogeneous cell types composing the central nervous system.
Collapse
Affiliation(s)
- Elizabeth Ford
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW 2006, Australia; (E.F.); (J.P.); (T.R.); (J.M.); (M.W.); (G.G.N.)
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW 2006, Australia
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK
| | - Jodie Pearlman
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW 2006, Australia; (E.F.); (J.P.); (T.R.); (J.M.); (M.W.); (G.G.N.)
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW 2006, Australia
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK
| | - Travis Ruan
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW 2006, Australia; (E.F.); (J.P.); (T.R.); (J.M.); (M.W.); (G.G.N.)
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW 2006, Australia
| | - John Manion
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW 2006, Australia; (E.F.); (J.P.); (T.R.); (J.M.); (M.W.); (G.G.N.)
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW 2006, Australia
- Department of Urology, Boston Children’s Hospital, Boston, MA 02115, USA
- Departments of Surgery and Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Matthew Waller
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW 2006, Australia; (E.F.); (J.P.); (T.R.); (J.M.); (M.W.); (G.G.N.)
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW 2006, Australia
| | - Gregory G. Neely
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW 2006, Australia; (E.F.); (J.P.); (T.R.); (J.M.); (M.W.); (G.G.N.)
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW 2006, Australia
| | - Leslie Caron
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW 2006, Australia; (E.F.); (J.P.); (T.R.); (J.M.); (M.W.); (G.G.N.)
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
37
|
Chandy M, Rhee JW, Ozen MO, Williams DR, Pepic L, Liu C, Zhang H, Malisa J, Lau E, Demirci U, Wu JC. Atlas of Exosomal microRNAs Secreted From Human iPSC-Derived Cardiac Cell Types. Circulation 2020; 142:1794-1796. [PMID: 33136510 DOI: 10.1161/circulationaha.120.048364] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Mark Chandy
- Stanford Cardiovascular Institute (M.C., J-W.R., D.R.W., L.P., C.L., H.Z., J.M., E.L., J.C.W.), Stanford University School of Medicine, CA
| | - June-Wha Rhee
- Stanford Cardiovascular Institute (M.C., J-W.R., D.R.W., L.P., C.L., H.Z., J.M., E.L., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (J-W.R., J.C.W.), Stanford University School of Medicine, CA
| | - Mehmet O Ozen
- Department of Radiology (M.O.O., U.D., J.C.W.), Stanford University School of Medicine, CA
| | - Damon R Williams
- Stanford Cardiovascular Institute (M.C., J-W.R., D.R.W., L.P., C.L., H.Z., J.M., E.L., J.C.W.), Stanford University School of Medicine, CA
| | - Lejla Pepic
- Stanford Cardiovascular Institute (M.C., J-W.R., D.R.W., L.P., C.L., H.Z., J.M., E.L., J.C.W.), Stanford University School of Medicine, CA
| | - Chun Liu
- Stanford Cardiovascular Institute (M.C., J-W.R., D.R.W., L.P., C.L., H.Z., J.M., E.L., J.C.W.), Stanford University School of Medicine, CA
| | - Hao Zhang
- Stanford Cardiovascular Institute (M.C., J-W.R., D.R.W., L.P., C.L., H.Z., J.M., E.L., J.C.W.), Stanford University School of Medicine, CA
| | - Jessica Malisa
- Stanford Cardiovascular Institute (M.C., J-W.R., D.R.W., L.P., C.L., H.Z., J.M., E.L., J.C.W.), Stanford University School of Medicine, CA
| | - Edward Lau
- Stanford Cardiovascular Institute (M.C., J-W.R., D.R.W., L.P., C.L., H.Z., J.M., E.L., J.C.W.), Stanford University School of Medicine, CA
| | - Utkan Demirci
- Department of Radiology (M.O.O., U.D., J.C.W.), Stanford University School of Medicine, CA
| | - Joseph C Wu
- Stanford Cardiovascular Institute (M.C., J-W.R., D.R.W., L.P., C.L., H.Z., J.M., E.L., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (J-W.R., J.C.W.), Stanford University School of Medicine, CA.,Department of Radiology (M.O.O., U.D., J.C.W.), Stanford University School of Medicine, CA
| |
Collapse
|
38
|
Robust detection of undifferentiated iPSC among differentiated cells. Sci Rep 2020; 10:10293. [PMID: 32581272 PMCID: PMC7314783 DOI: 10.1038/s41598-020-66845-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 05/28/2020] [Indexed: 12/24/2022] Open
Abstract
Recent progress in human induced pluripotent stem cells (iPSC) technologies suggest that iPSC application in regenerative medicine is a closer reality. Numerous challenges prevent iPSC application in the development of numerous tissues and for the treatment of various diseases. A key concern in therapeutic applications is the safety of the cell products to be transplanted into patients. Here, we present novel method for detecting residual undifferentiated iPSCs amongst directed differentiated cells of all three germ lineages. Marker genes, which are expressed specifically and highly in undifferentiated iPSC, were selected from single cell RNA sequence data to perform robust and sensitive detection of residual undifferentiated cells in differentiated cell products. ESRG (Embryonic Stem Cell Related), CNMD (Chondromodulin), and SFRP2 (Secreted Frizzled Related Protein 2) were well-correlated with the actual amounts of residual undifferentiated cells and could be used to detect residual cells in a highly sensitive manner using qPCR. In addition, such markers could be used to detect residual undifferentiated cells from various differentiated cells, including hepatic cells and pancreatic cells for the endodermal lineage, endothelial cells and mesenchymal cells for the mesodermal lineage, and neural cells for the ectodermal lineage. Our method facilitates robust validation and could enhance the safety of the cell products through the exclusion of undifferentiated iPSC.
Collapse
|
39
|
Haramoto Y, Onuma Y, Mawaribuchi S, Nakajima Y, Aiki Y, Higuchi K, Shimizu M, Tateno H, Hirabayashi J, Ito Y. A technique for removing tumourigenic pluripotent stem cells using rBC2LCN lectin. Regen Ther 2020; 14:306-314. [PMID: 32462059 PMCID: PMC7240284 DOI: 10.1016/j.reth.2020.03.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/27/2020] [Accepted: 03/11/2020] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION Tumourigenesis attributed to residual undifferentiated cells in a graft is considered to be a significant issue in cell therapy using human pluripotent stem cells. To ensure the safety of regenerative medicine derived from pluripotent stem cells, residual undifferentiated cells must be eliminated in the manufacturing process. We previously described the lectin probe rBC2LCN, which binds harmlessly and specifically to the cell surface of human pluripotent stem cells. We report here a technique using rBC2LCN to remove pluripotent cells from a heterogenous population to reduce the chance of teratoma formation. METHODS We demonstrate a method for separating residual tumourigenic cells using rBC2LCN-bound magnetic beads. This technology is a novel use of their previous discovery that rBC2LCN is a lectin that selectively binds to pluripotent cells. We optimize and validate a method to remove hPSCs from a mixture with human fibroblasts using rBC2LCN-conjugated magnetic beads. RESULTS Cells with the potential to form teratoma could be effectively eliminated from a heterogeneous cell population with biotin-labelled rBC2LCN and streptavidin-bound magnetic beads. The efficiency was measured by FACS, ddPCR, and animal transplantation, suggesting that magnetic cell separation using rBC2LCN is quite efficient for eliminating hPSCs from mixed cell populations. CONCLUSIONS The removal of residual tumourigenic cells based on rBC2LCN could be a practical option for laboratory use and industrialisation of regenerative medicine using human pluripotent stem cells.
Collapse
Affiliation(s)
- Yoshikazu Haramoto
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Yasuko Onuma
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Shuuji Mawaribuchi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Yoshiro Nakajima
- Division of Developmental Biology, Department of Anatomy, Kyoto Prefectural University of Medicine, Kawaramachi, Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yasuhiko Aiki
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Kumiko Higuchi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Madoka Shimizu
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Hiroaki Tateno
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 2, 1-1-1, Umezono, Tsukuba, Ibaraki, 305-8568, Japan
| | - Jun Hirabayashi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 2, 1-1-1, Umezono, Tsukuba, Ibaraki, 305-8568, Japan
| | - Yuzuru Ito
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| |
Collapse
|
40
|
Sugawara T, Miura T, Kawasaki T, Umezawa A, Akutsu H. The hsa-miR-302 cluster controls ectodermal differentiation of human pluripotent stem cell via repression of DAZAP2. Regen Ther 2020; 15:1-9. [PMID: 32490061 PMCID: PMC7251312 DOI: 10.1016/j.reth.2020.03.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/03/2020] [Accepted: 03/11/2020] [Indexed: 12/27/2022] Open
Abstract
Introduction Recent studies have revealed that microRNAs (miRNAs, miRs) are important for self-renewal, differentiation, and cellular reprogramming of somatic cells into induced pluripotent stem cells (iPSC); however, their functional roles and target genes that are regulated by human PSC-specific miRs including hsa-miR-302 clusters remain largely unknown. Analysis of their target gene will give us the opportunity to understand the functional roles of such miRs. Methods We analyzed the expression profiles of miRs in 4 somatic cell lines, 8 human iPSC lines derived from 4 different cell types, 3 human ESC lines, and embryoid bodies differentiated from the human ESCs to identify human PSC-specific miRs. We also analyzed the simultaneous expression profiles of miRs and mRNAs to identify candidate targets of human PSC-specific miRs. Then, we constructed a vector for overexpressing one of the target gene to dissect the functions of human PSC-specific miR in maintenance of self-renew and differentiation. Results We focused on hsa-miR-302 cluster as a human PSC-specific miR and identified 22 candidate targets of hsa-miR-302 cluster that were moderately expressed in undifferentiated human PSCs and up-regulated in differentiated cells. Deleted in azoospermia-associated protein 2 (DAZAP2), one such target, was directly repressed by hsa-miR-302a, -302b, -302c and -302d, but not by hsa-miR-367. Overexpression of DAZAP2 caused a decrease in cell proliferation of undifferentiated human iPSCs, although morphology and undifferentiated marker gene expression was not affected. In addition, neural differentiation was suppressed in DAZAP2-overexpressing human iPSCs. Conclusion Our study revealed that hsa-miR-302 cluster controls the cell proliferation of human PSCs and the neural differentiation of human PSCs by repression of DAZAP2, thereby highlighting an additional function of human PSC-specific miRs in maintaining pluripotency.
Collapse
Affiliation(s)
| | | | | | | | - Hidenori Akutsu
- Corresponding author. Department of Reproductive Medicine, National Center for Child Health and Developmen, Okura 2-10-1, Setagaya-ku, Tokyo, 157-8535, Japan. Fax: +81-3-5494-7048.
| |
Collapse
|
41
|
Parr CJC, Wada S, Kotake K, Kameda S, Matsuura S, Sakashita S, Park S, Sugiyama H, Kuang Y, Saito H. N 1-Methylpseudouridine substitution enhances the performance of synthetic mRNA switches in cells. Nucleic Acids Res 2020; 48:e35. [PMID: 32090264 PMCID: PMC7102939 DOI: 10.1093/nar/gkaa070] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 12/26/2019] [Accepted: 02/12/2020] [Indexed: 01/04/2023] Open
Abstract
Synthetic messenger RNA (mRNA) tools often use pseudouridine and 5-methyl cytidine as substitutions for uridine and cytidine to avoid the immune response and cytotoxicity induced by introducing mRNA into cells. However, the influence of base modifications on the functionality of the RNA tools is poorly understood. Here we show that synthetic mRNA switches containing N1-methylpseudouridine (m1Ψ) as a substitution of uridine substantially out-performed all other modified bases studied, exhibiting enhanced microRNA and protein sensitivity, better cell-type separation ability, and comparably low immune stimulation. We found that the observed phenomena stem from the high protein expression from m1Ψ containing mRNA and efficient translational repression in the presence of target microRNAs or proteins. In addition, synthetic gene circuits with m1Ψ significantly improve performance in cells. These findings indicate that synthetic mRNAs with m1Ψ modification have enormous potentials in the research and application of biofunctional RNA tools.
Collapse
Affiliation(s)
- Callum J C Parr
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53, Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shunsuke Wada
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53, Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kenjiro Kotake
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53, Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shigetoshi Kameda
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53, Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Satoshi Matsuura
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53, Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Souhei Sakashita
- Department of Chemistry, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwakecho, Sakyo-Ku, Kyoto 606-8502, Japan
| | - Soyoung Park
- Department of Chemistry, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwakecho, Sakyo-Ku, Kyoto 606-8502, Japan
| | - Hiroshi Sugiyama
- Department of Chemistry, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwakecho, Sakyo-Ku, Kyoto 606-8502, Japan
| | - Yi Kuang
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Room 5578, Academic Bldg, Clear Water Bay, Kowloon, Hong Kong
| | - Hirohide Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53, Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
42
|
Caliciviral protein-based artificial translational activator for mammalian gene circuits with RNA-only delivery. Nat Commun 2020; 11:1297. [PMID: 32157083 PMCID: PMC7064597 DOI: 10.1038/s41467-020-15061-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 02/12/2020] [Indexed: 12/13/2022] Open
Abstract
Synthetic RNA-based gene circuits enable sophisticated gene regulation without the risk of insertional mutagenesis. While various RNA binding proteins have been used for translational repression in gene circuits, the direct translational activation of synthetic mRNAs has not been achieved. Here we develop Caliciviral VPg-based Translational activator (CaVT), which activates the translation of synthetic mRNAs without the canonical 5'-cap. The level of translation can be modulated by changing the locations, sequences, and modified nucleosides of CaVT-binding motifs in the target mRNAs, enabling the simultaneous translational activation and repression of different mRNAs with RNA-only delivery. We demonstrate the efficient regulation of apoptosis and genome editing by tuning translation levels with CaVT. In addition, we design programmable CaVT that responds to endogenous microRNAs or small molecules, achieving both cell-state-specific and conditional translational activation from synthetic mRNAs. CaVT will become an important tool in synthetic biology for both biological studies and future therapeutic applications.
Collapse
|
43
|
Ohno H, Akamine S, Saito H. Synthetic mRNA-Based Systems in Mammalian Cells. ACTA ACUST UNITED AC 2020; 4:e1900247. [PMID: 32402126 DOI: 10.1002/adbi.201900247] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 02/02/2023]
Abstract
Living organisms are programmed to perform multiple functions by sensing intra- and extra-cellular environments and by controlling gene expressions. Synthetic biologists aim to program cells by mimicking, designing, and constructing genetic circuits. Synthetic mRNA-based genetic switches and circuits have attracted attention for future therapeutic applications because of their safety and functional diversity. Here, the mRNA-based switches and circuits that detect specific microRNAs or proteins expressed in a target cell to control transgene expression and cell fate are reviewed. Future perspectives of artificial RNA systems for cell engineering will also be addressed.
Collapse
Affiliation(s)
- Hirohisa Ohno
- 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Sae Akamine
- 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hirohide Saito
- 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
44
|
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
|
45
|
Sunohara T, Morizane A, Matsuura S, Miyamoto S, Saito H, Takahashi J. MicroRNA-Based Separation of Cortico-Fugal Projection Neuron-Like Cells Derived From Embryonic Stem Cells. Front Neurosci 2019; 13:1141. [PMID: 31708734 PMCID: PMC6819314 DOI: 10.3389/fnins.2019.01141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 10/10/2019] [Indexed: 12/26/2022] Open
Abstract
The purification of pluripotent stem cell-derived cortico-fugal projection neurons (PSC-CFuPNs) is useful for disease modeling and cell therapies related to the dysfunction of cortical motor neurons, such as amyotrophic lateral sclerosis (ALS) or stroke. However, no CFuPN-specific surface markers for the purification are known. Recently, microRNAs (miRNAs) have been reported as alternatives to surface markers. Here, we investigated this possibility by applying the miRNA switch, an mRNA technology, to enrich PSC-CFuPNs. An array study of miRNAs in mouse fetal brain tissue revealed that CFuPNs highly express miRNA-124-3p at E14.5 and E16.5. In response, we designed a miRNA switched that responds to miRNA-124-3p and applied it to mouse embryonic stem cell (ESC)-derived cortical neurons. Flow cytometry and quantitative polymerase chain reaction (qPCR) analyses showed the miRNA-124-3p switch enriched CFuPN-like cells from this population. Immunocytechemical analysis confirmed vGlut1/Emx1/Bcl11b triple positive CFuPN-like cells were increased from 6.5 to 42%. Thus, our miRNA-124-3p switch can uniquely enrich live CFuPN-like cells from mouse ESC-derived cortical neurons.
Collapse
Affiliation(s)
- Tadashi Sunohara
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Asuka Morizane
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Satoshi Matsuura
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Susumu Miyamoto
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hirohide Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
46
|
Elovic E, Etzion S, Cohen S. MiR-499 Responsive Lethal Construct for Removal of Human Embryonic Stem Cells after Cardiac Differentiation. Sci Rep 2019; 9:14490. [PMID: 31601830 PMCID: PMC6787023 DOI: 10.1038/s41598-019-50899-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/19/2019] [Indexed: 01/26/2023] Open
Abstract
Deriving cell populations from human embryonic stem cells (hESCs) for cell-based therapy is considered a promising strategy to achieve functional cells, yet its translation to clinical practice depends on achieving fully defined differentiated cells. In this work, we generated a miRNA-responsive lethal mRNA construct that selectively induces rapid apoptosis in hESCs by expressing a mutant (S184del) Bax variant. Insertion of miR-499 target sites in the construct enabled to enrich hESC-derived cardiomyocytes (CMs) in culture. A deterministic non-linear model was developed and validated with experimental data, to predict the outcome for each treatment cycle and the number of treatment cycle repetitions required to achieve completely purified cTNT-positive cells. The enriched hESC-CMs displayed physiological sarcomere orientation, functional calcium handling and after transplantation into SCID-NOD mice did not form teratomas. The modular miRNA responsive lethal mRNA construct could be employed in additional directed differentiation protocols, by adjusting the miRNA to the specific cells of choice.
Collapse
Affiliation(s)
- Edan Elovic
- The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Sharon Etzion
- Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Smadar Cohen
- The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel. .,Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel. .,The Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel.
| |
Collapse
|
47
|
Nakanishi H, Saito H. Mammalian gene circuits with biomolecule-responsive RNA devices. Curr Opin Chem Biol 2019; 52:16-22. [DOI: 10.1016/j.cbpa.2019.04.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/05/2019] [Accepted: 04/15/2019] [Indexed: 02/06/2023]
|
48
|
Tan HL, Tan BZ, Goh WXT, Cua S, Choo A. In vivo surveillance and elimination of teratoma-forming human embryonic stem cells with monoclonal antibody 2448 targeting annexin A2. Biotechnol Bioeng 2019; 116:2996-3005. [PMID: 31388993 PMCID: PMC6790577 DOI: 10.1002/bit.27135] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/25/2019] [Accepted: 07/29/2019] [Indexed: 12/14/2022]
Abstract
This study describes the use of a previously reported chimerised monoclonal antibody (mAb), ch2448, to kill human embryonic stem cells (hESCs) in vivo and prevent or delay the formation of teratomas. ch2448 was raised against hESCs and was previously shown to effectively kill ovarian and breast cancer cells in vitro and in vivo. The antigen target was subsequently found to be Annexin A2, an oncofetal antigen expressed on both embryonic cells and cancer cells. Against cancer cells, ch2448 binds and kills via antibody‐dependent cell‐mediated cytotoxicity (ADCC) and/or antibody‐drug conjugate (ADC) routes. Here, we investigate if the use of ch2448 can be extended to hESC. ch2448 was found to bind specifically to undifferentiated hESC but not differentiated progenitors. Similar to previous study using cancer cells, ch2448 kills hESC in vivo either indirectly by eliciting ADCC or directly as an ADC. The treatment with ch2448 post‐transplantation eliminated the in vivo circulating undifferentiated cells and prevented or delayed the formation of teratomas. This surveillance role of ch2448 adds an additional layer of safeguard to enhance the safety and efficacious use of pluripotent stem cell‐derived products in regenerative medicine. Thereby, translating the use of ch2448 in the treatment of cancers to a proof of concept study in hESC (or pluripotent stem cell [PSC]), we show that mAbs can also be used to eliminate teratoma forming cells in vivo during PSC‐derived cell therapies. We propose to use this strategy to complement existing methods to eliminate teratoma‐forming cells in vitro. Residual undifferentiated cells may escape in vitro removal methods and be introduced into patients together with the differentiated cells.
Collapse
Affiliation(s)
- Heng Liang Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Bao Zhu Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Winfred Xi Tai Goh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Simeon Cua
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Andre Choo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| |
Collapse
|
49
|
Hirosawa M, Fujita Y, Saito H. Cell-Type-Specific CRISPR Activation with MicroRNA-Responsive AcrllA4 Switch. ACS Synth Biol 2019; 8:1575-1582. [PMID: 31268303 DOI: 10.1021/acssynbio.9b00073] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Anti-CRISPR proteins have the potential to regulate CRISPR-Cas systems in a cell-type-specific manner. To selectively edit the genome in target cells, we controlled the expression of AcrllA4, a Streptococcus pyogenes Cas9 inhibitor, based on endogenous microRNA (miRNA) activity. We designed a miRNA-responsive AcrllA4 switch, which is a synthetic mRNA that contains a completely complementary sequence to an arbitrary miRNA at the 5'-UTR region and encodes AcrllA4. Together with the Cas9- or dCas9-VPR-guide RNA complex, this switch functions as a cell-specific Cas9 or dCas9-VPR activator that induces gene knockout or activation depending on the target miRNA. By sensing intracellular miRNAs, the conditional CRISPR-Cas9 ON system that we report could provide a powerful tool for future therapeutic applications and genome engineering.
Collapse
Affiliation(s)
- Moe Hirosawa
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA) , Kyoto University , Kyoto 606-8501 , Japan
- Graduate School of Medicine , Kyoto University , Kyoto 606-8501 , Japan
| | - Yoshihiko Fujita
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA) , Kyoto University , Kyoto 606-8501 , Japan
| | - Hirohide Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA) , Kyoto University , Kyoto 606-8501 , Japan
| |
Collapse
|
50
|
Recent Updates on Induced Pluripotent Stem Cells in Hematological Disorders. Stem Cells Int 2019; 2019:5171032. [PMID: 31191673 PMCID: PMC6525795 DOI: 10.1155/2019/5171032] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/31/2019] [Indexed: 02/07/2023] Open
Abstract
Over the past decade, enormous progress has been made in the field of induced pluripotent stem cells (iPSCs). Patients' somatic cells such as skin fibroblasts or blood cells can be used to generate disease-specific pluripotent stem cells, which have unlimited proliferation and can differentiate into all cell types of the body. Human iPSCs offer great promises and opportunities for treatments of degenerative diseases and studying disease pathology and drug screening. So far, many iPSC-derived disease models have led to the discovery of novel pathological mechanisms as well as new drugs in the pipeline that have been tested in the iPSC-derived cells for efficacy and potential toxicities. Furthermore, recent advances in genome editing technology in combination with the iPSC technology have provided a versatile platform for studying stem cell biology and regenerative medicine. In this review, an overview of iPSCs, patient-specific iPSCs for disease modeling and drug screening, applications of iPSCs and genome editing technology in hematological disorders, remaining challenges, and future perspectives of iPSCs in hematological diseases will be discussed.
Collapse
|