51
|
Parent AV, Faleo G, Chavez J, Saxton M, Berrios DI, Kerper NR, Tang Q, Hebrok M. Selective deletion of human leukocyte antigens protects stem cell-derived islets from immune rejection. Cell Rep 2021; 36:109538. [PMID: 34407395 DOI: 10.1016/j.celrep.2021.109538] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 04/23/2021] [Accepted: 07/26/2021] [Indexed: 11/21/2022] Open
Abstract
Stem cell-based replacement therapies hold the promise to restore function of damaged or degenerated tissue such as the pancreatic islets in people with type 1 diabetes. Wide application of these therapies requires overcoming the fundamental roadblock of immune rejection. To address this issue, we use genetic engineering to create human pluripotent stem cells (hPSCs) in which the majority of the polymorphic human leukocyte antigens (HLAs), the main drivers of allogeneic rejection, are deleted. We retain the common HLA class I allele HLA-A2 and less polymorphic HLA-E/F/G to allow immune surveillance and inhibition of natural killer (NK) cells. We employ a combination of in vitro assays and humanized mouse models to demonstrate that these gene manipulations significantly reduce NK cell activity and T-cell-mediated alloimmune response against hPSC-derived islet cells. In summary, our approach produces hypoimmunogenic hPSCs that can be readily matched with recipients to avoid alloimmune rejection.
Collapse
Affiliation(s)
- Audrey V Parent
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Gaetano Faleo
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jessica Chavez
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael Saxton
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David I Berrios
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Natanya R Kerper
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Qizhi Tang
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
52
|
Cobb H, Aparicio-Domingo S, Canto-Soler MV. Transitioning into GMP-Compliance: Alternative Methods for Producing Retinal Organoids for Transplantation. Transl Vis Sci Technol 2021; 10:9. [PMID: 34383873 PMCID: PMC8362632 DOI: 10.1167/tvst.10.10.9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Three-dimensional retinal organoids derived from human induced pluripotent stem cells (hiPSCs) are gaining much attention as a possible source for cell transplantation to treat retinal degenerative conditions. However, the protocol for producing retinal organoids is time and labor intensive, involving a sequence of precise steps, and thus has yet to be successfully translated into a Good Manufacturing Practice (GMP)-compliant procedure. This review seeks to define the progress that has already been made in the pursuit of designing a GMP-compliant, streamlined, and automated protocol for retinal organoid production for optimal clinical success. The reviewed literature compares various approaches for cell culture automation, appropriate xeno-free conditions, and cell sources for iPSC line generation; yet, there are still important gaps for these three key considerations that remain to be addressed. Thus, the authors also discuss further potential strategies to successfully achieve GMP-compliant production of retinal organoids for eventual safe and efficient use in clinical trials.
Collapse
Affiliation(s)
- Hannah Cobb
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, School of Medicine, Aurora, CO, USA
| | - Silvia Aparicio-Domingo
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, School of Medicine, Aurora, CO, USA
| | - M Valeria Canto-Soler
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, School of Medicine, Aurora, CO, USA.,Charles C. Gates Center for Regenerative Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
53
|
Zha S, Tay JCK, Zhu S, Li Z, Du Z, Wang S. Generation of Mesenchymal Stromal Cells with Low Immunogenicity from Human PBMC-Derived β2 Microglobulin Knockout Induced Pluripotent Stem Cells. Cell Transplant 2021; 29:963689720965529. [PMID: 33172291 PMCID: PMC7784598 DOI: 10.1177/0963689720965529] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are viewed as immune-privileged cells and have been broadly applied in allogeneic adoptive cell transfer for regenerative medicine or immune-suppressing purpose. However, the surface expression of human leukocyte antigen (HLA) class I molecules on MSCs could still possibly induce the rejection of allogeneic MSCs from the recipients. Here, we disrupted the β2 microglobulin (B2M) gene in human peripheral blood mononuclear cell-derived induced pluripotent stem cells (iPSCs) with two clustered regulatory interspaced short palindromic repeat (CRISPR)-associated Cas9 endonuclease-based methods. The B2M knockout iPSCs did not express HLA class I molecules but maintained their pluripotency and genome stability. Subsequently, MSCs were derived from the HLA-negative iPSCs (iMSCs). We demonstrated that B2M knockout did not affect iMSC phenotype, multipotency, and immune suppressive characteristics and, most importantly, reduced iMSC immunogenicity to allogeneic peripheral blood mononuclear cells. Thus, B2M knockout iPSCs could serve as unlimited off-the-shelf cell resources in adoptive cell transfer, while the derived iMSCs hold great potential as universal grafts in allogeneic MSC transplantation.
Collapse
Affiliation(s)
- Shijun Zha
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Johan Chin-Kang Tay
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Sumin Zhu
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Zhendong Li
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Zhicheng Du
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Shu Wang
- Department of Biological Sciences, National University of Singapore, Singapore
| |
Collapse
|
54
|
Li C, Mei H, Hu Y. Applications and explorations of CRISPR/Cas9 in CAR T-cell therapy. Brief Funct Genomics 2021; 19:175-182. [PMID: 31950135 PMCID: PMC7239310 DOI: 10.1093/bfgp/elz042] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/22/2019] [Accepted: 12/10/2019] [Indexed: 12/14/2022] Open
Abstract
Chimeric antigen receptor(CAR) T-cell therapy has shown remarkable effects and promising prospects in patients with refractory or relapsed malignancies, pending further progress in the next-generation CAR T cells with more optimized structure, enhanced efficacy and reduced toxicities. The clustered regulatory interspaced short palindromic repeat/CRISPR-associated protein 9 (CRISPR/Cas9) technology holds immense promise for advancing the field owing to its flexibility, simplicity, high efficiency and multiplexing in precise genome editing. Herein, we review the applications and explorations of CRISPR/Cas9 technology in constructing allogenic universal CAR T cells, disrupting inhibitory signaling to enhance potency and exploration of safer and more controllable novel CAR T cells.
Collapse
Affiliation(s)
| | | | - Yu Hu
- Corresponding author: Heng Mei, Hubei clinical medical center of cell therapy for neoplastic disease, Wuhan 430022, Republic of China. Tel: +86-27-85726007, Fax: +86-27-85726387; E-mail:
| |
Collapse
|
55
|
Afolabi LO, Afolabi MO, Sani MM, Okunowo WO, Yan D, Chen L, Zhang Y, Wan X. Exploiting the CRISPR-Cas9 gene-editing system for human cancers and immunotherapy. Clin Transl Immunology 2021; 10:e1286. [PMID: 34188916 PMCID: PMC8219901 DOI: 10.1002/cti2.1286] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/23/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022] Open
Abstract
The discovery of clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9 (CRISPR-Cas9) technology has brought advances in the genetic manipulation of eukaryotic cells, which has revolutionised cancer research and treatment options. It is increasingly being used in cancer immunotherapy, including adoptive T and natural killer (NK) cell transfer, secretion of antibodies, cytokine stimulation and overcoming immune checkpoints. CRISPR-Cas9 technology is used in autologous T cells and NK cells to express various innovative antigen designs and combinations of chimeric antigen receptors (CARs) targeted at specific antigens for haematological and solid tumors. Additionally, advanced engineering in immune cells to enhance their sensing circuits with sophisticated functionality is now possible. Intensive research on the CRISPR-Cas9 system has provided scientists with the ability to overcome the hostile tumor microenvironment and generate more products for future clinical use, especially off-the-shelf, universal cellular products, bringing exciting milestones for immunotherapy. This review discussed the application and challenges of CRISPR technology in cancer research and immunotherapy, its advances and prospects for promoting new cell-based therapeutic beyond immune oncology.
Collapse
Affiliation(s)
- Lukman O Afolabi
- Guangdong Immune Cell therapy Engineering and Technology research CenterCenter for Protein and Cell‐based DrugsInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- University of Chinese Academy of SciencesBeijingChina
- Department of BiochemistryFaculty of ScienceFederal University DutseDutseNigeria
| | - Mariam O Afolabi
- Open FIESTA CenterTsinghua UniversityShenzhenChina
- State Key Laboratory of Chemical OncogenomicsGraduate School at ShenzhenTsinghua UniversityShenzhenChina
| | - Musbahu M Sani
- Department of BiochemistryFaculty of ScienceFederal University DutseDutseNigeria
| | - Wahab O Okunowo
- Department of BiochemistryCollege of MedicineUniversity of LagosLagosNigeria
| | - Dehong Yan
- Guangdong Immune Cell therapy Engineering and Technology research CenterCenter for Protein and Cell‐based DrugsInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- University of Chinese Academy of SciencesBeijingChina
| | - Liang Chen
- Guangdong Immune Cell therapy Engineering and Technology research CenterCenter for Protein and Cell‐based DrugsInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yaou Zhang
- Open FIESTA CenterTsinghua UniversityShenzhenChina
- State Key Laboratory of Chemical OncogenomicsGraduate School at ShenzhenTsinghua UniversityShenzhenChina
- School of Life SciencesTsinghua UniversityBeijingChina
| | - Xiaochun Wan
- Guangdong Immune Cell therapy Engineering and Technology research CenterCenter for Protein and Cell‐based DrugsInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
56
|
Kwon D, Ahn HJ, Han MJ, Ji M, Ahn J, Seo KW, Kang KS. Human Leukocyte Antigen Class I Pseudo-Homozygous Mesenchymal Stem Cells Derived from Human Induced Pluripotent Stem Cells. Stem Cell Rev Rep 2021; 16:792-808. [PMID: 32712868 DOI: 10.1007/s12015-020-09990-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cells (MSC) are an important type of cell that are highly recognized for their safety and efficacy as a cell therapy agent. In order to obtain MSC, primary tissues (adipose tissue, bone marrow, and umbilical cord blood) must be used; however, these tissues, especially umbilical cord blood, are difficult to obtain due to various reasons, such as the low birth rate trend. In addition, to maximize the safety and efficacy of MSC as allogenic cell therapeutic agents, it is desirable to minimize the possibility of an immune rejection reaction after in vivo transplantation. This study tried to establish a novel method for producing induced pluripotent stem cells (iPSC)-derived MSC in which the human leukocyte antigen (HLA)-class I gene is knocked out. To do so, dermal fibroblast originated iPSC generation using Yamanaka 4-factor, HLA class I gene edited iPSC generation using CRISPR/Cas9, and differentiation from iPSC to MSC using MSC culture medium was utilized. Through this, HLA-A, B, and C pseudo-homozygous iPSC-derived MSC (KO iMSC) were produced by monoallelically knocking out the polymorphic HLA-A, B, and C genes, which are the major causes of immune rejection during allogenic cell transplantation. Produced KO iMSC possesses multipotency and it was safe in vivo to be able to be differentiated to cartilage. In addition, it was not attacked by natural killer cells unlike HLA class I null cells. In conclusion, KO iMSC that do not induce immune rejection during allogenic cell transplantation can be produced. In the future, KO iMSC can be successfully utilized as allogenic cell therapeutic agents for many recipients through HLA screening.
Collapse
Affiliation(s)
- Daekee Kwon
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul, 08826, South Korea
| | - Hee-Jin Ahn
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul, 08826, South Korea
| | - Mi-Jung Han
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul, 08826, South Korea
| | - Minjun Ji
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul, 08826, South Korea
| | - Jongchan Ahn
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul, 08826, South Korea
| | - Kwang-Won Seo
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul, 08826, South Korea
| | - Kyung-Sun Kang
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul, 08826, South Korea. .,Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
57
|
HLA DR Genome Editing with TALENs in Human iPSCs Produced Immune-Tolerant Dendritic Cells. Stem Cells Int 2021; 2021:8873383. [PMID: 34093711 PMCID: PMC8163544 DOI: 10.1155/2021/8873383] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 03/30/2021] [Accepted: 04/24/2021] [Indexed: 11/17/2022] Open
Abstract
Although human induced pluripotent stem cells (iPSCs) can serve as a universal cell source for regenerative medicine, the use of iPSCs in clinical applications is limited by prohibitive costs and prolonged generation time. Moreover, allogeneic iPSC transplantation requires preclusion of mismatches between the donor and recipient human leukocyte antigen (HLA). We, therefore, generated universally compatible immune nonresponsive human iPSCs by gene editing. Transcription activator-like effector nucleases (TALENs) were designed for selective elimination of HLA DR expression. The engineered nucleases completely disrupted the expression of HLA DR on human dermal fibroblast cells (HDF) that did not express HLA DR even after stimulation with IFN-γ. Teratomas formed by HLA DR knockout iPSCs did not express HLA DR, and dendritic cells differentiated from HLA DR knockout iPSCs reduced CD4+ T cell activation. These engineered iPSCs might provide a novel translational approach to treat multiple recipients from a limited number of cell donors.
Collapse
|
58
|
Petrash CC, Palestine AG, Canto-Soler MV. Immunologic Rejection of Transplanted Retinal Pigmented Epithelium: Mechanisms and Strategies for Prevention. Front Immunol 2021; 12:621007. [PMID: 34054796 PMCID: PMC8153373 DOI: 10.3389/fimmu.2021.621007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/28/2021] [Indexed: 12/15/2022] Open
Abstract
Replacement of dysfunctional retinal pigmented epithelium (RPE) with grafts derived from stem cells has the potential to improve vision for patients with retinal disorders. In fact, the potential is such that a great number of groups are attempting to realize this therapy through individual strategies with a variety of stem cell products, hosts, immunomodulatory regimen, and techniques to assess the success of their design. Comparing the findings of different investigators is complicated by a number of factors. The immune response varies greatly between xenogeneic and allogeneic transplantation. A unique immunologic environment is created in the subretinal space, the target of RPE grafts. Both functional assessment and imaging techniques used to evaluate transplants are susceptible to erroneous conclusions. Lastly, the pharmacologic regimens used in RPE transplant trials are as numerous and variable as the trials themselves, making it difficult to determine useful results. This review will discuss the causes of these complicating factors, digest the strategies and results from clinical and preclinical studies, and suggest places for improvement in the design of future transplants and investigations.
Collapse
Affiliation(s)
- Carson C Petrash
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States.,Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Alan G Palestine
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - M Valeria Canto-Soler
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States.,Charles C. Gates Center for Regenerative Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
59
|
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: 5.0] [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
|
60
|
Iwamoto Y, Seki Y, Taya K, Tanaka M, Iriguchi S, Miyake Y, Nakayama EE, Miura T, Shioda T, Akari H, Takaori-Kondo A, Kaneko S. Generation of macrophages with altered viral sensitivity from genome-edited rhesus macaque iPSCs to model human disease. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:262-273. [PMID: 33869654 PMCID: PMC8039773 DOI: 10.1016/j.omtm.2021.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 03/11/2021] [Indexed: 01/14/2023]
Abstract
Because of their close biological similarity to humans, non-human primate (NHP) models are very useful for the development of induced pluripotent stem cell (iPSC)-based cell and regenerative organ transplantation therapies. However, knowledge on the establishment, differentiation, and genetic modification of NHP-iPSCs, especially rhesus macaque iPSCs, is limited. We succeeded in establishing iPSCs from the peripheral blood of rhesus macaques (Rh-iPSCs) by combining the Yamanaka reprograming factors and two inhibitors (GSK-3 inhibitor [CHIR 99021] and MEK1/2 inhibitor [PD0325901]) and differentiated the cells into functional macrophages through hematopoietic progenitor cells. To confirm feasibility of the Rh-iPSC-derived macrophages as a platform for bioassays to model diseases, we knocked out TRIM5 gene in Rh-iPSCs by CRISPR-Cas9, which is a species-specific HIV resistance factor. TRIM5 knockout (KO) iPSCs had the same differentiation potential to macrophages as did Rh-iPSCs, but the differentiated macrophages showed a gain of sensitivity to HIV infection in vitro. Our reprogramming, gene editing, and differentiation protocols used to obtain Rh-iPSC-derived macrophages can be applied to other gene mutations, expanding the number of NHP gene therapy models.
Collapse
Affiliation(s)
- Yoshihiro Iwamoto
- Shin Kaneko Laboratory, Department of Cell Growth and Development, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yohei Seki
- Center for Human Evolution Modeling Research, Primate Research Institute, Kyoto University, Kyoto, Japan
| | - Kahoru Taya
- Shin Kaneko Laboratory, Department of Cell Growth and Development, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Masahiro Tanaka
- Shin Kaneko Laboratory, Department of Cell Growth and Development, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Shoichi Iriguchi
- Shin Kaneko Laboratory, Department of Cell Growth and Development, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yasuyuki Miyake
- Shin Kaneko Laboratory, Department of Cell Growth and Development, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Emi E Nakayama
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tomoyuki Miura
- Laboratory of Primate Model, Research Center for Infectious Diseases, Institute for Frontier Life and Medical Science, Kyoto University, Kyoto, Japan
| | - Tatsuo Shioda
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Hirofumi Akari
- Center for Human Evolution Modeling Research, Primate Research Institute, Kyoto University, Kyoto, Japan.,Laboratory of Infectious Disease Model, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shin Kaneko
- Shin Kaneko Laboratory, Department of Cell Growth and Development, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
61
|
Sung TC, Jiang YP, Hsu JY, Ling QD, Chen H, Kumar SS, Chang Y, Hsu ST, Ye Q, Higuchi A. Transient characteristics of universal cells on human-induced pluripotent stem cells and their differentiated cells derived from foetal stem cells with mixed donor sources. Cell Prolif 2021; 54:e12995. [PMID: 33522648 PMCID: PMC7941237 DOI: 10.1111/cpr.12995] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/02/2021] [Accepted: 01/02/2021] [Indexed: 12/14/2022] Open
Abstract
Introduction It is important to prepare ‘hypoimmunogenic’ or ‘universal’ human pluripotent stem cells (hPSCs) with gene‐editing technology by knocking out or in immune‐related genes, because only a few hypoimmunogenic or universal hPSC lines would be sufficient to store for their off‐the‐shelf use. However, these hypoimmunogenic or universal hPSCs prepared previously were all genetically edited, which makes laborious processes to check and evaluate no abnormal gene editing of hPSCs. Methods Universal human‐induced pluripotent stem cells (hiPSCs) were generated without gene editing, which were reprogrammed from foetal stem cells (human amniotic fluid stem cells) with mixing 2‐5 allogenic donors but not with single donor. We evaluated human leucocyte antigen (HLA)‐expressing class Ia and class II of our hiPSCs and their differentiated cells into embryoid bodies, cardiomyocytes and mesenchymal stem cells. We further evaluated immunogenic response of transient universal hiPSCs with allogenic mononuclear cells from survival rate and cytokine production, which were generated by the cells due to immunogenic reactions. Results Our universal hiPSCs during passages 10‐25 did not have immunogenic reaction from allogenic mononuclear cells even after differentiation into cardiomyocytes, embryoid bodies and mesenchymal stem cells. Furthermore, the cells including the differentiated cells did not express HLA class Ia and class II. Cardiomyocytes differentiated from transient universal hiPSCs at passage 21‐22 survived and continued beating even after treatment with allogenic mononuclear cells.
Collapse
Affiliation(s)
- Tzu-Cheng Sung
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan
| | - Yi-Peng Jiang
- Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan
| | - Jhe-Yu Hsu
- Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan
| | - Qing-Dong Ling
- Cathay Medical Research Institute, Cathay General Hospital, Taipei, Taiwan
| | - Hao Chen
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Suresh S Kumar
- Department of Biotechnology, Bharath Institute of Higher Education and Research, Chennai, India
| | - Yung Chang
- Department of Chemical Engineering and R&D Center for Membrane Technology, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Shih-Tien Hsu
- Department of Internal Medicine, Taiwan Landseed Hospital, Pingjen City, Taiwan
| | - Qingsong Ye
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Hubei, China.,School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Department of Oral Maxillofacial Surgery, Skeletal Biology Research Center, Massachusetts General Hospital, Harvard School of Dental Medicine, Boston, MA, USA
| | - Akon Higuchi
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan.,Department of Chemical Engineering and R&D Center for Membrane Technology, Chung Yuan Christian University, Taoyuan, Taiwan.,Wenzhou Institute, University of Chinese Academy of Science, Wenzhou, China.,Nano Medical Engineering Laboratory, Riken Cluster for Pioneering Research, Riken, Japan
| |
Collapse
|
62
|
Verhoeff K, Henschke SJ, Marfil-Garza BA, Dadheech N, Shapiro AMJ. Inducible Pluripotent Stem Cells as a Potential Cure for Diabetes. Cells 2021; 10:cells10020278. [PMID: 33573247 PMCID: PMC7911560 DOI: 10.3390/cells10020278] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 02/07/2023] Open
Abstract
Over the last century, diabetes has been treated with subcutaneous insulin, a discovery that enabled patients to forego death from hyperglycemia. Despite novel insulin formulations, patients with diabetes continue to suffer morbidity and mortality with unsustainable costs to the health care system. Continuous glucose monitoring, wearable insulin pumps, and closed-loop artificial pancreas systems represent an advance, but still fail to recreate physiologic euglycemia and are not universally available. Islet cell transplantation has evolved into a successful modality for treating a subset of patients with ‘brittle’ diabetes but is limited by organ donor supply and immunosuppression requirements. A novel approach involves generating autologous or immune-protected islet cells for transplant from inducible pluripotent stem cells to eliminate detrimental immune responses and organ supply limitations. In this review, we briefly discuss novel mechanisms for subcutaneous insulin delivery and define their shortfalls. We describe embryological development and physiology of islets to better understand their role in glycemic control and, finally, discuss cell-based therapies for diabetes and barriers to widespread use. In response to these barriers, we present the promise of stem cell therapy, and review the current gaps requiring solutions to enable widespread use of stem cells as a potential cure for diabetes.
Collapse
Affiliation(s)
- Kevin Verhoeff
- Department of Surgery, University of Alberta, Edmonton, AB T6G 2B7, Canada;
- Correspondence: ; Tel.: +1-780-984-1836
| | - Sarah J. Henschke
- Department of Emergency Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada;
| | | | - Nidheesh Dadheech
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada;
| | - Andrew Mark James Shapiro
- FRCS (Eng) FRCSC MSM FCAHS, Clinical Islet Transplant Program, Alberta Diabetes Institute, Department of Surgery, Canadian National Transplant Research Program, Edmonton, AB T6G 2B7, Canada;
| |
Collapse
|
63
|
Hoerster K, Uhrberg M, Wiek C, Horn PA, Hanenberg H, Heinrichs S. HLA Class I Knockout Converts Allogeneic Primary NK Cells Into Suitable Effectors for "Off-the-Shelf" Immunotherapy. Front Immunol 2021; 11:586168. [PMID: 33584651 PMCID: PMC7878547 DOI: 10.3389/fimmu.2020.586168] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/04/2020] [Indexed: 11/13/2022] Open
Abstract
Cellular immunotherapy using chimeric antigen receptors (CARs) so far has almost exclusively used autologous peripheral blood-derived T cells as immune effector cells. However, harvesting sufficient numbers of T cells is often challenging in heavily pre-treated patients with malignancies and perturbed hematopoiesis and perturbed hematopoiesis. Also, such a CAR product will always be specific for the individual patient. In contrast, NK cell infusions can be performed in non-HLA-matched settings due to the absence of alloreactivity of these innate immune cells. Still, the infused NK cells are subject to recognition and rejection by the patient's immune system, thereby limiting their life-span in vivo and undermining the possibility for multiple infusions. Here, we designed genome editing and advanced lentiviral transduction protocols to render primary human NK cells unsusceptible/resistant to an allogeneic response by the recipient's CD8+ T cells. After knocking-out surface expression of HLA class I molecules by targeting the B2M gene via CRISPR/Cas9, we also co-expressed a single-chain HLA-E molecule, thereby preventing NK cell fratricide of B2M-knockout (KO) cells via "missing self"-induced lysis. Importantly, these genetically engineered NK cells were functionally indistinguishable from their unmodified counterparts with regard to their phenotype and their natural cytotoxicity towards different AML cell lines. In co-culture assays, B2M-KO NK cells neither induced immune responses of allogeneic T cells nor re-activated allogeneic T cells which had been expanded/primed using irradiated PBMNCs of the respective NK cell donor. Our study demonstrates the feasibility of genome editing in primary allogeneic NK cells to diminish their recognition and killing by mismatched T cells and is an important prerequisite for using non-HLA-matched primary human NK cells as readily available, "off-the-shelf" immune effectors for a variety of immunotherapy indications in human cancer.
Collapse
Affiliation(s)
- Keven Hoerster
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Markus Uhrberg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine University, Düsseldorf, Germany
| | - Constanze Wiek
- Department of Otorhinolaryngology & Head/Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Peter A. Horn
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Essen, Germany
| | - Helmut Hanenberg
- Department of Otorhinolaryngology & Head/Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Department of Pediatrics III, University Children’s Hospital of Essen, University Duisburg-Essen, Essen, Germany
| | - Stefan Heinrichs
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Essen, Germany
| |
Collapse
|
64
|
Kim A, Lee KG, Kwon Y, Lee KI, Yang HM, Habib O, Kim J, Kim ST, Kim SJ, Kim JS, Hwang DY. Off-the-Shelf, Immune-Compatible Human Embryonic Stem Cells Generated Via CRISPR-Mediated Genome Editing. Stem Cell Rev Rep 2021; 17:1053-1067. [PMID: 33423156 PMCID: PMC8166669 DOI: 10.1007/s12015-020-10113-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2020] [Indexed: 10/27/2022]
Abstract
Human embryonic stem cells (hESCs) hold promise in regenerative medicine but allogeneic immune rejections caused by highly polymorphic human leukocyte antigens (HLAs) remain a barrier to their clinical applications. Here, we used a CRISPR/Cas9-mediated HLA-editing strategy to generate a variety of HLA homozygous-like hESC lines from pre-established hESC lines. We edited four pre-established HLA-heterozygous hESC lines and created a mini library of 14 HLA-edited hESC lines in which single HLA-A and HLA-B alleles and both HLA-DR alleles are disrupted. The HLA-edited hESC derivatives elicited both low T cell- and low NK cell-mediated immune responses. Our library would cover about 40% of the Asian-Pacific population. We estimate that HLA-editing of only 19 pre-established hESC lines would give rise to 46 different hESC lines to cover 90% of the Asian-Pacific population. This study offers an opportunity to generate an off-the-shelf HLA-compatible hESC bank, available for immune-compatible cell transplantation, without embryo destruction. Graphical Abstract.
Collapse
Affiliation(s)
- Annie Kim
- Center for Genome Engineering, Institute for Basic Science, Seoul, Republic of Korea.,Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Kun-Gu Lee
- Department of Biomedical Science, Graduate School of CHA University, Seongnam, South Korea
| | - Yeongbeen Kwon
- Samsung Advanced Institute for Health Sciences & Technology(SAIHST), Graduate School, Department of Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea.,Transplantation Research Center, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Kang-In Lee
- Department of Biomedical Science, Graduate School of CHA University, Seongnam, South Korea.,ToolGen, Inc., Seoul, South Korea
| | - Heung-Mo Yang
- Transplantation Research Center, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Republic of Korea.,GenNbio Inc., Seoul, South Korea.,Department of Medicine, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Omer Habib
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea.,Department of Chemistry, Hanyang University, Seoul, Republic of Korea
| | | | - Sang-Tae Kim
- Center for Genome Engineering, Institute for Basic Science, Seoul, Republic of Korea.,Department of Life Sciences, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, South Korea
| | - Sung Joo Kim
- Transplantation Research Center, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Republic of Korea.,GenNbio Inc., Seoul, South Korea.,Department of Medicine, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Jin-Soo Kim
- Center for Genome Engineering, Institute for Basic Science, Seoul, Republic of Korea. .,Department of Chemistry, Seoul National University, Seoul, Republic of Korea.
| | - Dong-Youn Hwang
- Department of Biomedical Science, Graduate School of CHA University, Seongnam, South Korea.
| |
Collapse
|
65
|
Romitti M, Eski SE, Fonseca BF, Gillotay P, Singh SP, Costagliola S. Single-Cell Trajectory Inference Guided Enhancement of Thyroid Maturation In Vitro Using TGF-Beta Inhibition. Front Endocrinol (Lausanne) 2021; 12:657195. [PMID: 34135860 PMCID: PMC8202408 DOI: 10.3389/fendo.2021.657195] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/10/2021] [Indexed: 12/30/2022] Open
Abstract
The thyroid gland regulates metabolism and growth via secretion of thyroid hormones by thyroid follicular cells (TFCs). Loss of TFCs, by cellular dysfunction, autoimmune destruction or surgical resection, underlies hypothyroidism. Recovery of thyroid hormone levels by transplantation of mature TFCs derived from stem cells in vitro holds great therapeutic promise. However, the utilization of in vitro derived tissue for regenerative medicine is restricted by the efficiency of differentiation protocols to generate mature organoids. Here, to improve the differentiation efficiency for thyroid organoids, we utilized single-cell RNA-Seq to chart the molecular steps undertaken by individual cells during the in vitro transformation of mouse embryonic stem cells to TFCs. Our single-cell atlas of mouse organoid systematically and comprehensively identifies, for the first time, the cell types generated during production of thyroid organoids. Using pseudotime analysis, we identify TGF-beta as a negative regulator of thyroid maturation in vitro. Using pharmacological inhibition of TGF-beta pathway, we improve the level of thyroid maturation, in particular the induction of Nis expression. This in turn, leads to an enhancement of iodide organification in vitro, suggesting functional improvement of the thyroid organoid. Our study highlights the potential of single-cell molecular characterization in understanding and improving thyroid maturation and paves the way for identification of therapeutic targets against thyroid disorders.
Collapse
Affiliation(s)
- Mírian Romitti
- *Correspondence: Mírian Romitti, ; Sumeet Pal Singh, ; Sabine Costagliola,
| | | | | | | | - Sumeet Pal Singh
- *Correspondence: Mírian Romitti, ; Sumeet Pal Singh, ; Sabine Costagliola,
| | - Sabine Costagliola
- *Correspondence: Mírian Romitti, ; Sumeet Pal Singh, ; Sabine Costagliola,
| |
Collapse
|
66
|
Kong M, Zhou D. Establishment of universal human embryonic stem cell lines. Immunol Lett 2020; 230:59-62. [PMID: 33309828 DOI: 10.1016/j.imlet.2020.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/19/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022]
Abstract
The potential application of human embryonic stem cells in regenerative medicine using cell, tissue or organ transplantation has aroused great interest. However, HLA incompatibility between donor cells or tissues and the recipient is a primary obstacle to the use of unmatched human embryonic stem cells and their derivatives as donor 'grafts' for patient treatment without some form of immunosuppressive therapy. This is because, for most tissues, which express HLA Class I antigens, the recipient patient's immune system will recognize the difference between their and the donor's HLA types, leading to graft rejection in the absence of immunosuppressive therapy. One approach to overcoming this obstacle and enabling the use of a single or limited range of suitably selected human embryonic stem cells and their derivatives without needing extensive HLA matching is to use gene-editing technology to establish a universally or widely HLA compatible human embryonic stem cell line, thereby providing a potentially unlimited source of cells for future cell, tissue or organ transplantation. This article reviews current strategies and methods for establishing such universal or near universally HLA compatible human embryonic stem cell lines.
Collapse
Affiliation(s)
- MingYue Kong
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China; Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China.
| | - Di Zhou
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China; National Engineering and Research Center of Human Stem Cells, Changsha, China; Key Laboratory of Stem Cells and Reproductive Engineering, Ministry of Health, Changsha, China; Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China.
| |
Collapse
|
67
|
Jackson AO, Rahman GA, Yin K, Long S. Enhancing Matured Stem-Cardiac Cell Generation and Transplantation: A Novel Strategy for Heart Failure Therapy. J Cardiovasc Transl Res 2020; 14:556-572. [PMID: 33258081 DOI: 10.1007/s12265-020-10085-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/10/2020] [Indexed: 12/25/2022]
Abstract
Heart failure (HF) remains one of the major causes of morbidity and mortality worldwide. Recent studies have shown that stem cells (SCs) including bone marrow mesenchymal stem (BMSC), embryonic bodies (EB), embryonic stem (ESC), human induced pluripotent stem (hiPSC)-derived cardiac cells generation, and transplantation treated myocardial infarction (MI) in vivo and in human. However, the immature phenotypes compromise their clinical application requiring immediate intervention to improve stem-derived cardiac cell (S-CCs) maturation. Recently, an unbiased multi-omic analysis involving genomics, transcriptomics, epigenomics, proteomics, and metabolomics identified specific strategies for the generation of matured S-CCs that may enhance patients' recovery processes upon transplantation. However, these strategies still remain undisclosed. Here, we summarize the recently discovered strategies for the matured S-CC generation. In addition, cardiac patch formation and transplantation that accelerated HF recuperation in clinical trials are discussed. A better understanding of this work may lead to efficient generation of matured S-CCs for regenerative medicine. Graphical abstract.
Collapse
Affiliation(s)
- Ampadu O Jackson
- Department of Biochemistry and Molecular Biology, University of South China, Hengyang, 421001, Hunan Province, China.,International College, University of South China, Hengyang, 421001, Hunan Province, China.,Cape Coast Teaching Hospital, Cape Coast, Department of Surgery, School of Medical Science, University of Cape Coast, Cape Coast, Ghana
| | - Ganiyu A Rahman
- Cape Coast Teaching Hospital, Cape Coast, Department of Surgery, School of Medical Science, University of Cape Coast, Cape Coast, Ghana
| | - Kai Yin
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, China
| | - Shiyin Long
- Department of Biochemistry and Molecular Biology, University of South China, Hengyang, 421001, Hunan Province, China.
| |
Collapse
|
68
|
Antao AM, Karapurkar JK, Lee DR, Kim KS, Ramakrishna S. Disease modeling and stem cell immunoengineering in regenerative medicine using CRISPR/Cas9 systems. Comput Struct Biotechnol J 2020; 18:3649-3665. [PMID: 33304462 PMCID: PMC7710510 DOI: 10.1016/j.csbj.2020.11.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 12/14/2022] Open
Abstract
CRISPR/Cas systems are popular genome editing tools that belong to a class of programmable nucleases and have enabled tremendous progress in the field of regenerative medicine. We here outline the structural and molecular frameworks of the well-characterized type II CRISPR system and several computational tools intended to facilitate experimental designs. The use of CRISPR tools to generate disease models has advanced research into the molecular aspects of disease conditions, including unraveling the molecular basis of immune rejection. Advances in regenerative medicine have been hindered by major histocompatibility complex-human leukocyte antigen (HLA) genes, which pose a major barrier to cell- or tissue-based transplantation. Based on progress in CRISPR, including in recent clinical trials, we hypothesize that the generation of universal donor immune-engineered stem cells is now a realistic approach to tackling a multitude of disease conditions.
Collapse
Affiliation(s)
- Ainsley Mike Antao
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | | | - Dong Ryul Lee
- Department of Biomedical Science, College of Life Science, CHA University, Seoul, South Korea
- CHA Stem Cell Institute, CHA University, Seoul, South Korea
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
- College of Medicine, Hanyang University, Seoul, South Korea
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
- College of Medicine, Hanyang University, Seoul, South Korea
| |
Collapse
|
69
|
Ye Q, Sung TC, Yang JM, Ling QD, He Y, Higuchi A. Generation of universal and hypoimmunogenic human pluripotent stem cells. Cell Prolif 2020; 53:e12946. [PMID: 33174655 PMCID: PMC7705897 DOI: 10.1111/cpr.12946] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
There is a need to store very large numbers of conventional human pluripotent stem cell (hPSC) lines for their off‐the‐shelf usage in stem cell therapy. Therefore, it is valuable to generate “universal” or “hypoimmunogenic” hPSCs with gene‐editing technology by knocking out or in immune‐related genes. A few universal or hypoimmunogenic hPSC lines should be enough to store for their off‐the‐shelf usage. Here, we overview and discuss how to prepare universal or hypoimmunogenic hPSCs and their disadvantages. β2‐Microglobulin‐knockout hPSCs did not harbour human leukocyte antigen (HLA)‐expressing class I cells but rather activated natural killer (NK) cells. To avoid NK cell and macrophage activities, homozygous hPSCs expressing a single allele of an HLA class I molecule, such as HLA‐C, were developed. Major HLA class I molecules were knocked out, and PD‐L1, HLA‐G and CD47 were knocked in hPSCs using CRISPR/Cas9 gene editing. These cells escaped activation of not only T cells but also NK cells and macrophages, generating universal hPSCs.
Collapse
Affiliation(s)
- Qingsong Ye
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, China.,Skeletal Biology Research Center, Department of Oral Maxillofacial Surgery, Massachusetts General Hospital & Harvard School of Dental Medicine, Boston, MA, USA
| | - Tzu-Cheng Sung
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan
| | - Jen-Ming Yang
- Department of Chemical and Materials Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Qing-Dong Ling
- Cathay Medical Research Institute, Cathay General Hospital, Taipei, Taiwan
| | - Yan He
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Akon Higuchi
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan.,Wenzhou Institute, University of Chinese Academy of Science, Wenzhou, China.,Department of Chemical Engineering and R&D Center for Membrane Technology, Chung Yuan Christian University, Taoyuan, Taiwan.,Center for Emergent Matter Science, Riken, Saitama, Japan
| |
Collapse
|
70
|
Sullivan S, Fairchild PJ, Marsh SGE, Müller CR, Turner ML, Song J, Turner D. Haplobanking induced pluripotent stem cells for clinical use. Stem Cell Res 2020; 49:102035. [PMID: 33221677 DOI: 10.1016/j.scr.2020.102035] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 07/20/2020] [Accepted: 10/05/2020] [Indexed: 02/08/2023] Open
Abstract
The development of induced pluripotent stem cells (iPSCs) by Shinya Yamanaka and colleagues in 2006 has led to a potential new paradigm in cellular therapeutics, including the possibility of producing patient-specific, disease-specific and immune matched allogeneic cell therapies. One can envisage two routes to immunologically compatible iPSC therapies: using genetic modification to generate a 'universal donor' with reduced expression of Human Leukocyte Antigens (HLA) and other immunological targets or developing a haplobank containing iPSC lines specifically selected to provide HLA matched products to large portions of the population. HLA matched lines can be stored in a designated physical or virtual global bank termed a 'haplobank'. The process of 'iPSC haplobanking' refers to the banking of iPSC cell lines, selected to be homozygous for different HLA haplotypes, from which therapeutic products can be derived and matched immunologically to patient populations. By matching iPSC and derived products to a patient's HLA class I and II molecules, one would hope to significantly reduce the risk of immune rejection and the use of immunosuppressive medication. Immunosuppressive drugs are used in several conditions (including autoimmune disease and in transplantation procedures) to reduce rejection of infused cells, or transplanted tissue and organs, due to major and minor histocompatibility differences between donor and recipient. Such regimens can lead to immune compromise and pathological consequences such as opportunistic infections or malignancies due to decreased cancer immune surveillance. In this article, we will discuss what is practically involved if one is developing and executing an iPSC haplobanking strategy.
Collapse
Affiliation(s)
- Stephen Sullivan
- Global Alliance for iPSC Therapies, Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK.
| | - Paul J Fairchild
- University of Oxford, Sir William Dunn School of Pathology, South Parks Road, Oxford OX1 3RE, UK
| | - Steven G E Marsh
- HLA Informatics Group, Anthony Nolan Research Institute, Royal Free Campus, London, UK; UCL Cancer Institute, University College London, London, UK
| | - Carlheinz R Müller
- Zentrales Knochenmarkspender-Register Deutschland (ZKRD), Helmholtzstraße, 1089081 Ulm, Germany
| | - Marc L Turner
- Global Alliance for iPSC Therapies, Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK; Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh, UK
| | - Jihwan Song
- Global Alliance for iPSC Therapies, Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK; Department of Biomedical Science, CHA Stem Cell Institute, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - David Turner
- Global Alliance for iPSC Therapies, Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK; Histocompatibility and Immunogenetics Laboratory, Royal Infirmary of Edinburgh, Edinburgh, UK
| |
Collapse
|
71
|
Majid QA, Fricker ATR, Gregory DA, Davidenko N, Hernandez Cruz O, Jabbour RJ, Owen TJ, Basnett P, Lukasiewicz B, Stevens M, Best S, Cameron R, Sinha S, Harding SE, Roy I. Natural Biomaterials for Cardiac Tissue Engineering: A Highly Biocompatible Solution. Front Cardiovasc Med 2020; 7:554597. [PMID: 33195451 PMCID: PMC7644890 DOI: 10.3389/fcvm.2020.554597] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVD) constitute a major fraction of the current major global diseases and lead to about 30% of the deaths, i.e., 17.9 million deaths per year. CVD include coronary artery disease (CAD), myocardial infarction (MI), arrhythmias, heart failure, heart valve diseases, congenital heart disease, and cardiomyopathy. Cardiac Tissue Engineering (CTE) aims to address these conditions, the overall goal being the efficient regeneration of diseased cardiac tissue using an ideal combination of biomaterials and cells. Various cells have thus far been utilized in pre-clinical studies for CTE. These include adult stem cell populations (mesenchymal stem cells) and pluripotent stem cells (including autologous human induced pluripotent stem cells or allogenic human embryonic stem cells) with the latter undergoing differentiation to form functional cardiac cells. The ideal biomaterial for cardiac tissue engineering needs to have suitable material properties with the ability to support efficient attachment, growth, and differentiation of the cardiac cells, leading to the formation of functional cardiac tissue. In this review, we have focused on the use of biomaterials of natural origin for CTE. Natural biomaterials are generally known to be highly biocompatible and in addition are sustainable in nature. We have focused on those that have been widely explored in CTE and describe the original work and the current state of art. These include fibrinogen (in the context of Engineered Heart Tissue, EHT), collagen, alginate, silk, and Polyhydroxyalkanoates (PHAs). Amongst these, fibrinogen, collagen, alginate, and silk are isolated from natural sources whereas PHAs are produced via bacterial fermentation. Overall, these biomaterials have proven to be highly promising, displaying robust biocompatibility and, when combined with cells, an ability to enhance post-MI cardiac function in pre-clinical models. As such, CTE has great potential for future clinical solutions and hence can lead to a considerable reduction in mortality rates due to CVD.
Collapse
Affiliation(s)
- Qasim A. Majid
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Annabelle T. R. Fricker
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - David A. Gregory
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Natalia Davidenko
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Olivia Hernandez Cruz
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Bioengineering, Department of Materials, IBME, Faculty of Engineering, Imperial College London, United Kingdom
| | - Richard J. Jabbour
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Thomas J. Owen
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Pooja Basnett
- Applied Biotechnology Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | - Barbara Lukasiewicz
- Applied Biotechnology Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | - Molly Stevens
- Department of Bioengineering, Department of Materials, IBME, Faculty of Engineering, Imperial College London, United Kingdom
| | - Serena Best
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Ruth Cameron
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Sian E. Harding
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ipsita Roy
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
72
|
Jarrell DK, Vanderslice EJ, VeDepo MC, Jacot JG. Engineering Myocardium for Heart Regeneration-Advancements, Considerations, and Future Directions. Front Cardiovasc Med 2020; 7:586261. [PMID: 33195474 PMCID: PMC7588355 DOI: 10.3389/fcvm.2020.586261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/31/2020] [Indexed: 12/28/2022] Open
Abstract
Heart disease is the leading cause of death in the United States among both adults and infants. In adults, 5-year survival after a heart attack is <60%, and congenital heart defects are the top killer of liveborn infants. Problematically, the regenerative capacity of the heart is extremely limited, even in newborns. Furthermore, suitable donor hearts for transplant cannot meet the demand and require recipients to use immunosuppressants for life. Tissue engineered myocardium has the potential to replace dead or fibrotic heart tissue in adults and could also be used to permanently repair congenital heart defects in infants. In addition, engineering functional myocardium could facilitate the development of a whole bioartificial heart. Here, we review and compare in vitro and in situ myocardial tissue engineering strategies. In the context of this comparison, we consider three challenges that must be addressed in the engineering of myocardial tissue: recapitulation of myocardial architecture, vascularization of the tissue, and modulation of the immune system. In addition to reviewing and analyzing current progress, we recommend specific strategies for the generation of tissue engineered myocardial patches for heart regeneration and repair.
Collapse
Affiliation(s)
- Dillon K Jarrell
- Jacot Laboratory for Pediatric Regenerative Medicine, Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Ethan J Vanderslice
- Jacot Laboratory for Pediatric Regenerative Medicine, Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Mitchell C VeDepo
- Jacot Laboratory for Pediatric Regenerative Medicine, Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jeffrey G Jacot
- Jacot Laboratory for Pediatric Regenerative Medicine, Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
73
|
Koga K, Wang B, Kaneko S. Current status and future perspectives of HLA-edited induced pluripotent stem cells. Inflamm Regen 2020; 40:23. [PMID: 33014207 PMCID: PMC7528263 DOI: 10.1186/s41232-020-00132-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 07/01/2020] [Indexed: 12/22/2022] Open
Abstract
In 2007, Human-induced pluripotent stem cells (iPSCs) were generated by transducing four genes (Oct3/4, Sox2, Klf4, c-Myc). Because iPSCs can differentiate into any types of cells in the body and have fewer ethical issues compared to embryonic stem (ES) cells, application of iPSCs for regenerative medicine has been actively examined. In fact, iPSCs have already been used for clinical applications, but at present, only autologous iPSC-derived grafts or HLA homozygous iPSC-derived grafts are being transplanted into patients following HLA matching. HLA is an important molecule that enables the immune system differentiates between self and non-self-components; thus, HLA mismatch is a major hurdle in the transplantation of iPSCs. To deliver inexpensive off-the-shelf iPSC-derived regenerative medicine products to more patients, it is necessary to generate universal iPSCs that can be transplanted regardless of the HLA haplotypes. The current strategy to generate universal iPSCs has two broad aims: deleting HLA expression and avoiding attacks from NK cells, which are caused by HLA deletion. Deletion of B2M and CIITA genes using the CRISPR/Cas9 system has been reported to suppress the expression of HLA class I and class II, respectively. Transduction of NK inhibitory ligands, such as HLA-E and CD47, has been used to avoid NK cell attacks. Most recently, the HLA-C retaining method has been used to generate semi-universal iPSCs. Twelve haplotypes of HLA-C retaining iPSCs can cover 95% of the global population. In future, studying which types of universal iPSCs are most effective for engraftment in various physiological conditions is necessary.
Collapse
Affiliation(s)
- Keiko Koga
- Takeda-CiRA Joint Program (T-CiRA), 2-26-1, Muraoka-Higashi, Fujisawa, Kanagawa 251-8555 Japan.,T-CiRA discovery, Takeda Pharmaceutical Company, 2-26-1, Muraoka-Higashi, Fujisawa, Kanagawa 251-8555 Japan
| | - Bo Wang
- Takeda-CiRA Joint Program (T-CiRA), 2-26-1, Muraoka-Higashi, Fujisawa, Kanagawa 251-8555 Japan.,Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS cell research (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Shin Kaneko
- Takeda-CiRA Joint Program (T-CiRA), 2-26-1, Muraoka-Higashi, Fujisawa, Kanagawa 251-8555 Japan.,Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS cell research (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507 Japan
| |
Collapse
|
74
|
Pavlovic K, Tristán-Manzano M, Maldonado-Pérez N, Cortijo-Gutierrez M, Sánchez-Hernández S, Justicia-Lirio P, Carmona MD, Herrera C, Martin F, Benabdellah K. Using Gene Editing Approaches to Fine-Tune the Immune System. Front Immunol 2020; 11:570672. [PMID: 33117361 PMCID: PMC7553077 DOI: 10.3389/fimmu.2020.570672] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/20/2020] [Indexed: 12/26/2022] Open
Abstract
Genome editing technologies not only provide unprecedented opportunities to study basic cellular system functionality but also improve the outcomes of several clinical applications. In this review, we analyze various gene editing techniques used to fine-tune immune systems from a basic research and clinical perspective. We discuss recent advances in the development of programmable nucleases, such as zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeat (CRISPR)-Cas-associated nucleases. We also discuss the use of programmable nucleases and their derivative reagents such as base editing tools to engineer immune cells via gene disruption, insertion, and rewriting of T cells and other immune components, such natural killers (NKs) and hematopoietic stem and progenitor cells (HSPCs). In addition, with regard to chimeric antigen receptors (CARs), we describe how different gene editing tools enable healthy donor cells to be used in CAR T therapy instead of autologous cells without risking graft-versus-host disease or rejection, leading to reduced adoptive cell therapy costs and instant treatment availability for patients. We pay particular attention to the delivery of therapeutic transgenes, such as CARs, to endogenous loci which prevents collateral damage and increases therapeutic effectiveness. Finally, we review creative innovations, including immune system repurposing, that facilitate safe and efficient genome surgery within the framework of clinical cancer immunotherapies.
Collapse
Affiliation(s)
- Kristina Pavlovic
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Cellular Therapy Unit, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - María Tristán-Manzano
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - Noelia Maldonado-Pérez
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - Marina Cortijo-Gutierrez
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - Sabina Sánchez-Hernández
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - Pedro Justicia-Lirio
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
- LentiStem Biotech, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - M. Dolores Carmona
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Cellular Therapy Unit, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Concha Herrera
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Cellular Therapy Unit, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
- Department of Hematology, Reina Sofía University Hospital, Córdoba, Spain
| | - Francisco Martin
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - Karim Benabdellah
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| |
Collapse
|
75
|
Protze SI, Lee JH, Keller GM. Human Pluripotent Stem Cell-Derived Cardiovascular Cells: From Developmental Biology to Therapeutic Applications. Cell Stem Cell 2020; 25:311-327. [PMID: 31491395 DOI: 10.1016/j.stem.2019.07.010] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Advances in our understanding of cardiovascular development have provided a roadmap for the directed differentiation of human pluripotent stem cells (hPSCs) to the major cell types found in the heart. In this Perspective, we review the state of the field in generating and maturing cardiovascular cells from hPSCs based on our fundamental understanding of heart development. We then highlight their applications for studying human heart development, modeling disease-performing drug screening, and cell replacement therapy. With the advancements highlighted here, the promise that hPSCs will deliver new treatments for degenerative and debilitating diseases may soon be fulfilled.
Collapse
Affiliation(s)
- Stephanie I Protze
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Jee Hoon Lee
- BlueRock Therapeutics ULC, Toronto, ON M5G 1L7, Canada
| | - Gordon M Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7, Canada.
| |
Collapse
|
76
|
Maeda T, Nagano S, Kashima S, Terada K, Agata Y, Ichise H, Ohtaka M, Nakanishi M, Fujiki F, Sugiyama H, Kitawaki T, Kadowaki N, Takaori-Kondo A, Masuda K, Kawamoto H. Regeneration of Tumor-Antigen-Specific Cytotoxic T Lymphocytes from iPSCs Transduced with Exogenous TCR Genes. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 19:250-260. [PMID: 33102617 PMCID: PMC7566080 DOI: 10.1016/j.omtm.2020.09.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 09/16/2020] [Indexed: 12/18/2022]
Abstract
In the current adoptive T cell therapy, T cells from a patient are given back to that patient after ex vivo activation, expansion, or genetic manipulation. However, such strategy depends on the quality of the patient’s T cells, sometimes leading to treatment failure. It would therefore be ideal to use allogeneic T cells as “off-the-shelf” T cells. To this aim, we have been developing a strategy where potent tumor-antigen-specific cytotoxic T lymphocytes (CTLs) are regenerated from T-cell-derived induced pluripotent stem cells (T-iPSCs). However, certain issues still remain that make it difficult to establish highly potent T-iPSCs: poor reprogramming efficiency of T cells into iPSCs and high variability in the differentiation capability of each T-iPSC clone. To expand the versatility of this approach, we thought of a method to produce iPSCs equivalent to T-iPSCs, namely, iPSCs transduced with exogenous T cell receptor (TCR) genes (TCR-iPSCs). To test this idea, we first cloned TCR genes from WT1-specific CTLs regenerated from T-iPSCs and then established WT1-TCR-iPSCs. We show that the regenerated CTLs from TCR-iPSCs exerted cytotoxic activity comparable to those from T-iPSCs against WT1 peptide-loaded cell line in in vitro model. These results collectively demonstrate the feasibility of the TCR-iPSC strategy.
Collapse
Affiliation(s)
- Takuya Maeda
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan.,Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Seiji Nagano
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan.,Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Soki Kashima
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan.,Department of Urology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Koji Terada
- Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Yasutoshi Agata
- Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Hiroshi Ichise
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Manami Ohtaka
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki 305-8568, Japan
| | - Mahito Nakanishi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki 305-8568, Japan
| | - Fumihiro Fujiki
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Toshio Kitawaki
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Norimitsu Kadowaki
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Kyoko Masuda
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Hiroshi Kawamoto
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
77
|
Luciani M, Gritti A, Meneghini V. Human iPSC-Based Models for the Development of Therapeutics Targeting Neurodegenerative Lysosomal Storage Diseases. Front Mol Biosci 2020; 7:224. [PMID: 33062642 PMCID: PMC7530250 DOI: 10.3389/fmolb.2020.00224] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/10/2020] [Indexed: 01/30/2023] Open
Abstract
Lysosomal storage diseases (LSDs) are a group of rare genetic conditions. The absence or deficiency of lysosomal proteins leads to excessive storage of undigested materials and drives secondary pathological mechanisms including autophagy, calcium homeostasis, ER stress, and mitochondrial abnormalities. A large number of LSDs display mild to severe central nervous system (CNS) involvement. Animal disease models and post-mortem tissues partially recapitulate the disease or represent the final stage of CNS pathology, respectively. In the last decades, human models based on induced pluripotent stem cells (hiPSCs) have been extensively applied to investigate LSD pathology in several tissues and organs, including the CNS. Neural stem/progenitor cells (NSCs) derived from patient-specific hiPSCs (hiPS-NSCs) are a promising tool to define the effects of the pathological storage on neurodevelopment, survival and function of neurons and glial cells in neurodegenerative LSDs. Additionally, the development of novel 2D co-culture systems and 3D hiPSC-based models is fostering the investigation of neuron-glia functional and dysfunctional interactions, also contributing to define the role of neurodevelopment and neuroinflammation in the onset and progression of the disease, with important implications in terms of timing and efficacy of treatments. Here, we discuss the advantages and limits of the application of hiPS-NSC-based models in the study and treatment of CNS pathology in different LSDs. Additionally, we review the state-of-the-art and the prospective applications of NSC-based therapy, highlighting the potential exploitation of hiPS-NSCs for gene and cell therapy approaches in the treatment of neurodegenerative LSDs.
Collapse
Affiliation(s)
- Marco Luciani
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
78
|
Abstract
Regenerative therapies, including both gene and cellular therapies, aim to induce regeneration of cells, tissues and organs and restore their functions. In this short Spotlight, we summarize the latest advances in cellular therapies using pluripotent stem cells (PSCs), highlighting the current status of clinical trials using induced (i)PSC-derived cells. We also discuss the different cellular products that might be used in clinical studies, and consider safety issues and other challenges in iPSC-based cell therapy.
Collapse
Affiliation(s)
- Hideyuki Okano
- Keio University School of Medicine, Department of Physiology, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan .,Keio University Global Research Institute, 2-15-45 Mita, Minato-ku, Tokyo 108-8345, Japan
| | - Doug Sipp
- Keio University School of Medicine, Department of Physiology, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan.,Keio University Global Research Institute, 2-15-45 Mita, Minato-ku, Tokyo 108-8345, Japan.,RIKEN Center for Developmental Biology, 2-2-3 Minatojima Minamimachi, Chuo-ku, Kobe 650-0047, Japan.,RIKEN Center for Advanced Intelligence Project, Nihonbashi 1-chome Mitsui Building, 15th floor, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan
| |
Collapse
|
79
|
Shi L, Li W, Liu Y, Chen Z, Hui Y, Hao P, Xu X, Zhang S, Feng H, Zhang B, Zhou S, Li N, Xiao L, Liu L, Ma L, Zhang X. Generation of hypoimmunogenic human pluripotent stem cells via expression of membrane-bound and secreted β2m-HLA-G fusion proteins. Stem Cells 2020; 38:1423-1437. [PMID: 32930470 DOI: 10.1002/stem.3269] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 07/24/2020] [Accepted: 08/11/2020] [Indexed: 01/16/2023]
Abstract
Allogeneic immune rejection is a major barrier for the application of human pluripotent stem cells (hPSCs) in regenerative medicine. A broad spectrum of immune cells, including T cells, natural killer (NK) cells, and antigen-presenting cells, which either cause direct cell killing or constitute an immunogenic environment, are involved in allograft immune rejection. A strategy to protect donor cells from cytotoxicity while decreasing the secretion of inflammatory cytokines of lymphocytes is still lacking. Here, we engineered hPSCs with no surface expression of classical human leukocyte antigen (HLA) class I proteins via beta-2 microglobulin (B2M) knockout or biallelic knockin of HLA-G1 within the frame of endogenous B2M loci. Elimination of the surface expression of HLA class I proteins protected the engineered hPSCs from cytotoxicity mediated by T cells. However, this lack of surface expression also resulted in missing-self response and NK cell activation, which were largely compromised by expression of β2m-HLA-G1 fusion proteins. We also proved that the engineered β2m-HLA-G5 fusion proteins were soluble, secretable, and capable of safeguarding low immunogenic environments by lowering inflammatory cytokines secretion in allografts. Our current study reveals a novel strategy that may offer unique advantages to construct hypoimmunogenic hPSCs via the expression of membrane-bound and secreted β2m-HLA-G fusion proteins. These engineered hPSCs are expected to serve as an unlimited cell source for generating universally compatible "off-the-shelf" cell grafts in the future.
Collapse
Affiliation(s)
- Lei Shi
- College of Animal Sciences, Zhejiang University, Hangzhou, People's Republic of China.,Brain and Spinal Cord Clinical Research Center, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Wenjing Li
- College of Animal Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Yang Liu
- Brain and Spinal Cord Clinical Research Center, Tongji University School of Medicine, Shanghai, People's Republic of China.,Key Laboratory of Neuroregeneration of Shanghai Universities, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Zhenyu Chen
- Brain and Spinal Cord Clinical Research Center, Tongji University School of Medicine, Shanghai, People's Republic of China.,Key Laboratory of Neuroregeneration of Shanghai Universities, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yi Hui
- Brain and Spinal Cord Clinical Research Center, Tongji University School of Medicine, Shanghai, People's Republic of China.,Key Laboratory of Neuroregeneration of Shanghai Universities, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Pengcheng Hao
- Brain and Spinal Cord Clinical Research Center, Tongji University School of Medicine, Shanghai, People's Republic of China.,Key Laboratory of Neuroregeneration of Shanghai Universities, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xiangjie Xu
- Brain and Spinal Cord Clinical Research Center, Tongji University School of Medicine, Shanghai, People's Republic of China.,Key Laboratory of Neuroregeneration of Shanghai Universities, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Shuwei Zhang
- Brain and Spinal Cord Clinical Research Center, Tongji University School of Medicine, Shanghai, People's Republic of China.,Key Laboratory of Neuroregeneration of Shanghai Universities, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Hexi Feng
- Brain and Spinal Cord Clinical Research Center, Tongji University School of Medicine, Shanghai, People's Republic of China.,Key Laboratory of Neuroregeneration of Shanghai Universities, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Bowen Zhang
- Brain and Spinal Cord Clinical Research Center, Tongji University School of Medicine, Shanghai, People's Republic of China.,Key Laboratory of Neuroregeneration of Shanghai Universities, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Shanshan Zhou
- Brain and Spinal Cord Clinical Research Center, Tongji University School of Medicine, Shanghai, People's Republic of China.,Key Laboratory of Neuroregeneration of Shanghai Universities, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Nan Li
- Brain and Spinal Cord Clinical Research Center, Tongji University School of Medicine, Shanghai, People's Republic of China.,Key Laboratory of Neuroregeneration of Shanghai Universities, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Lei Xiao
- College of Animal Sciences, Zhejiang University, Hangzhou, People's Republic of China.,Shanghai SiDanSai Biotechnology Limited Company, Shanghai, People's Republic of China
| | - Ling Liu
- Brain and Spinal Cord Clinical Research Center, Tongji University School of Medicine, Shanghai, People's Republic of China.,Key Laboratory of Neuroregeneration of Shanghai Universities, Tongji University School of Medicine, Shanghai, People's Republic of China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, People's Republic of China
| | - Lin Ma
- Brain and Spinal Cord Clinical Research Center, Tongji University School of Medicine, Shanghai, People's Republic of China.,Key Laboratory of Neuroregeneration of Shanghai Universities, Tongji University School of Medicine, Shanghai, People's Republic of China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, People's Republic of China
| | - Xiaoqing Zhang
- Brain and Spinal Cord Clinical Research Center, Tongji University School of Medicine, Shanghai, People's Republic of China.,Key Laboratory of Neuroregeneration of Shanghai Universities, Tongji University School of Medicine, Shanghai, People's Republic of China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, People's Republic of China.,Key Laboratory of Reconstruction and Regeneration of Spine and Spinal Cord Injury, Ministry of Education, Shanghai, People's Republic of China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, People's Republic of China
| |
Collapse
|
80
|
Selvakumar D, Clayton ZE, Chong JJH. Robust Cardiac Regeneration: Fulfilling the Promise of Cardiac Cell Therapy. Clin Ther 2020; 42:1857-1879. [PMID: 32943195 DOI: 10.1016/j.clinthera.2020.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE We review the history of cardiac cell therapy, highlighting lessons learned from initial adult stem cell (ASC) clinical trials. We present pluripotent stem cell-derived cardiomyocytes (PSC-CMs) as a leading candidate for robust regeneration of infarcted myocardium but identify several issues that must be addressed before successful clinical translation. METHODS We conducted an unstructured literature review of PubMed-listed articles, selecting the most comprehensive and relevant research articles, review articles, clinical trials, and basic or translation articles in the field of cardiac cell therapy. Articles were identified using the search terms adult stem cells, pluripotent stem cells, cardiac stem cell, and cardiac regeneration or from references of relevant articles, Articles were prioritized and selected based on their impact, originality, or potential clinical applicability. FINDINGS Since its inception, the ASC therapy field has been troubled by conflicting preclinical data, academic controversies, and inconsistent trial designs. These issues have damaged perceptions of cardiac cell therapy among investors, the academic community, health care professionals, and, importantly, patients. In hindsight, the key issue underpinning these problems was the inability of these cell types to differentiate directly into genuine cardiomyocytes, rendering them unable to replace damaged myocardium. Despite this, beneficial effects through indirect paracrine or immunomodulatory effects remain possible and continue to be investigated. However, in preclinical models, PSC-CMs have robustly remuscularized infarcted myocardium with functional, force-generating cardiomyocytes. Hence, PSC-CMs have now emerged as a leading candidate for cardiac regeneration, and unpublished reports of first-in-human delivery of these cells have recently surfaced. However, the cardiac cell therapy field's history should serve as a cautionary tale, and we identify several translational hurdles that still remain. Preclinical solutions to issues such as arrhythmogenicity, immunogenicity, and poor engraftment rates are needed, and next-generation clinical trials must draw on robust knowledge of mechanistic principles of the therapy. IMPLICATIONS The clinical transplantation of functional stem cell-derived heart tissue with seamless integration into native myocardium is a lofty goal. However, considerable advances have been made during the past 2 decades. Currently, PSC-CMs appear to be the best prospect to reach this goal, but several hurdles remain. The history of adult stem cell trials has taught us that shortcuts cannot be taken without dire consequences, and it is essential that progress not be hurried and that a worldwide, cross-disciplinary approach be used to ensure safe and effective clinical translation.
Collapse
Affiliation(s)
- Dinesh Selvakumar
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Zoe E Clayton
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - James J H Chong
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia.
| |
Collapse
|
81
|
Suh TC, Amanah AY, Gluck JM. Electrospun Scaffolds and Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Cardiac Tissue Engineering Applications. Bioengineering (Basel) 2020; 7:E105. [PMID: 32899986 PMCID: PMC7552723 DOI: 10.3390/bioengineering7030105] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 01/14/2023] Open
Abstract
Tissue engineering (TE) combines cells, scaffolds, and growth factors to assemble functional tissues for repair or replacement of tissues and organs. Cardiac TE is focused on developing cardiac cells, tissues, and structures-most notably the heart. This review presents the requirements, challenges, and research surrounding electrospun scaffolds and induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs) towards applications to TE hearts. Electrospinning is an attractive fabrication method for cardiac TE scaffolds because it produces fibers that demonstrate the optimal potential for mimicking the complex structure of the cardiac extracellular matrix (ECM). iPSCs theoretically offer the capacity to generate limitless numbers of CMs for use in TE hearts, however these iPSC-CMs are electrophysiologically, morphologically, mechanically, and metabolically immature compared to adult CMs. This presents a functional limitation to their use in cardiac TE, and research aiming to address this limitation is presented in this review.
Collapse
Affiliation(s)
- Taylor Cook Suh
- Textile Engineering, Chemistry and Science Department, Wilson College of Textiles, NC State University, Raleigh, NC 27695, USA
| | - Alaowei Y Amanah
- Textile Engineering, Chemistry and Science Department, Wilson College of Textiles, NC State University, Raleigh, NC 27695, USA
| | - Jessica M Gluck
- Textile Engineering, Chemistry and Science Department, Wilson College of Textiles, NC State University, Raleigh, NC 27695, USA
| |
Collapse
|
82
|
Cell-Based Therapeutic Approaches for Cystic Fibrosis. Int J Mol Sci 2020; 21:ijms21155219. [PMID: 32718005 PMCID: PMC7432606 DOI: 10.3390/ijms21155219] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/20/2020] [Accepted: 07/20/2020] [Indexed: 01/01/2023] Open
Abstract
Cystic Fibrosis (CF) is a chronic autosomal recessive disease caused by defects in the cystic fibrosis transmembrane conductance regulator gene (CFTR). Cystic Fibrosis affects multiple organs but progressive remodeling of the airways, mucus accumulation, and chronic inflammation in the lung, result in lung disease as the major cause of morbidity and mortality. While advances in management of CF symptoms have increased the life expectancy of this devastating disease, and there is tremendous excitement about the potential of new agents targeting the CFTR molecule itself, there is still no curative treatment. With the recent advances in the identification of endogenous airway progenitor cells and in directed differentiation of pluripotent cell sources, cell-based therapeutic approaches for CF have become a plausible treatment method with the potential to ultimately cure the disease. In this review, we highlight the current state of cell therapy in the CF field focusing on the relevant autologous and allogeneic cell populations under investigation and the challenges associated with their use. In addition, we present advances in induced pluripotent stem (iPS) cell approaches and emerging new genetic engineering methods, which have the capacity to overcome the current limitations hindering cell therapy approaches.
Collapse
|
83
|
Otsuka R, Wada H, Murata T, Seino KI. Immune reaction and regulation in transplantation based on pluripotent stem cell technology. Inflamm Regen 2020; 40:12. [PMID: 32636970 PMCID: PMC7329400 DOI: 10.1186/s41232-020-00125-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 06/08/2020] [Indexed: 12/21/2022] Open
Abstract
The development of pluripotent stem cell (PSC)-based technologies provides us a new therapeutic approach that generates grafts for transplantation. In order to minimize the risk of immune reaction, the banking of induced pluripotent stem cells (iPSCs) from donors with homozygous human leukocyte antigen (HLA) haplotype is planned in Japan. Even though pre-stocked and safety validated HLA-homozygous iPSCs are selected, immunological rejection may potentially occur because the causes of rejection are not always due to HLA mismatches. A couple of studies concerning such immunological issues have reported that genetic ablation of HLA molecules from PSC combined with gene transduction of several immunoregulatory molecules may be effective in avoiding immunological rejection. Also, our research group has recently proposed a concept that attempts to regulate recipient immune system by PSC-derived immunoregulatory cells, which results in prolonged survival of the same PSC-derived allografts. PSC-based technologies enable us to choose a new therapeutic option; however, considering its safety from an immunological point of view should be of great importance for safe clinical translation of this technology.
Collapse
Affiliation(s)
- Ryo Otsuka
- Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Sapporo, Hokkaido 060-0815 Japan
| | - Haruka Wada
- Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Sapporo, Hokkaido 060-0815 Japan
| | - Tomoki Murata
- Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Sapporo, Hokkaido 060-0815 Japan
| | - Ken-Ichiro Seino
- Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Sapporo, Hokkaido 060-0815 Japan
| |
Collapse
|
84
|
Generation and characterization of HLA-universal platelets derived from induced pluripotent stem cells. Sci Rep 2020; 10:8472. [PMID: 32439978 PMCID: PMC7242456 DOI: 10.1038/s41598-020-65577-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/22/2020] [Indexed: 12/11/2022] Open
Abstract
Platelet demand has increased around the world. However, the inadequacy of donors, the risk of transfusion-transmitted infections and associated reactions, and the refractory nature of platelet transfusions are among the limitations of allogeneic platelet transfusions. To alleviate these problems, we propose generating platelets in a laboratory that do not induce alloimmunity to human leukocyte antigen (HLA) class I, which is a major cause of immune reaction in platelet transfusion refractoriness. Induced pluripotent stem cells (iPSCs) were generated from peripheral blood mononuclear cells (PBMCs) of a healthy Thai woman. We then knocked out the β2-microglobulin (β2m) gene in the cells using paired CRISPR/Cas9 nickases and sequentially differentiated the cells into haematopoietic stem cells (HSCs), megakaryocytes (MKs) and platelets. Silencing of HLA class I expression was observed on the cell surface of β2m-knockout iPSCs, iPSC-derived HSCs, MKs and platelets. The HLA-universal iPSC-derived platelets were shown to be activated, and they aggregated after stimulation. In addition, our in vivo platelet survival experiments demonstrated that human platelets were detectable at 2 and 24 hours after injecting the β2m-KO MKs. In summary, we successfully generated functional iPSC-derived platelets in vitro without HLA class I expression by knocking out the β2m gene using paired CRISPR/Cas9 nickases.
Collapse
|
85
|
Strategies for Genetically Engineering Hypoimmunogenic Universal Pluripotent Stem Cells. iScience 2020; 23:101162. [PMID: 32502965 PMCID: PMC7270609 DOI: 10.1016/j.isci.2020.101162] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/07/2020] [Accepted: 05/11/2020] [Indexed: 01/18/2023] Open
Abstract
Despite progress in developing cell therapies, such as T cell or stem cell therapies to treat diseases, immunoincompatibility remains a major barrier to clinical application. Given the fact that a host's immune system may reject allogeneic transplanted cells, methods have been developed to genetically modify patients' primary cells. To advance beyond this time-consuming and costly approach, recent research efforts focus on generating universal pluripotent stem cells to benefit a broader spectrum of patients. In this review, we first summarize current achievements to harness immunosuppressive mechanisms in cells to reduce immunogenicity. Then, we discuss several recent studies demonstrating the feasibility of genetically modifying pluripotent stem cells to escape immune attack and summarize the methods to evaluate hypoimmunogenicity. Although challenges remain, progress to develop genetically engineered universal pluripotent stem cells holds the promise of expediting their use in future gene and cell therapeutics and regenerative medicine.
Collapse
|
86
|
Hong L, Zhang C, Jiang Y, Liu H, Huang H, Guo D. Therapeutic status and the prospect of CRISPR/Cas9 gene editing in multiple myeloma. Future Oncol 2020; 16:1125-1136. [PMID: 32338048 DOI: 10.2217/fon-2019-0822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In recent years, CRISPR/Cas9, a novel gene-editing technology, has shown considerable potential in the design of novel research methods and future options for treating multiple myeloma (MM). The use of CRISPR/Cas9 promises faster and more accurate identification and validation of target genes. In this review, we summarize the current research status of the application of CRISPR technology in MM, especially in detecting the expression of MM gene, exploring the mechanism of drug action, screening for drug-resistant genes, developing immunotherapy and screening for new drug targets. Given the tremendous progress that has been made, we believe that CRISPR/Cas9 possesses great potential in MM-related clinical practice.
Collapse
Affiliation(s)
- Lemin Hong
- Department of Hematology, The Affiliated Hospital of Nantong University, Jiangsu, PR China
| | - Chenlu Zhang
- Department of Hematology, The Affiliated Hospital of Nantong University, Jiangsu, PR China
| | - Yijing Jiang
- Department of Hematology, The Affiliated Hospital of Nantong University, Jiangsu, PR China
| | - Haiyan Liu
- Department of Hematology, The Affiliated Hospital of Nantong University, Jiangsu, PR China
| | - Hongming Huang
- Department of Hematology, The Affiliated Hospital of Nantong University, Jiangsu, PR China
| | - Dan Guo
- Department of Hematology, The Affiliated Hospital of Nantong University, Jiangsu, PR China
| |
Collapse
|
87
|
Haworth R, Sharpe M. Accept or Reject: The Role of Immune Tolerance in the Development of Stem Cell Therapies and Possible Future Approaches. Toxicol Pathol 2020; 49:1308-1316. [PMID: 32319357 DOI: 10.1177/0192623320918241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In 2011, Goldring and colleagues published a review article describing the potential safety issues of novel stem cell-derived treatments. Immunogenicity and immunotoxicity of the administered cell product were considered risks in the light of clinical experience of transplantation. The relative immunogenicity of mesenchymal stem cells, embryonic stem cells (ESCs), and induced pluripotent stem cells (iPSCs) was being addressed through in vitro and in vivo models. But the question arose as to whether the implanted cells needed to be identical to the recipient in every respect, including epigenetically, to evade immune recognition? If so, this set a high bar which may preclude use of many cells derived from iPSCs which have vestiges of a fetal phenotype and epigenetic memory of their cell of origin. However, for autologous iPSCs, the immunogenicity reduces once the surface antigen expression profile becomes close to that of the parent somatic cells. Therefore, a cell product containing incompletely differentiated cells could be more immunogenic. The properties of the administered cells, the immune privilege of the administration site, and the host immune status influence graft success or failure. In addition, the various approaches available to characterize potential immunogenicity of a cell therapy will be discussed.
Collapse
|
88
|
|
89
|
Raffin C, Vo LT, Bluestone JA. T reg cell-based therapies: challenges and perspectives. Nat Rev Immunol 2020; 20:158-172. [PMID: 31811270 PMCID: PMC7814338 DOI: 10.1038/s41577-019-0232-6] [Citation(s) in RCA: 408] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2019] [Indexed: 12/25/2022]
Abstract
Cellular therapies using regulatory T (Treg) cells are currently undergoing clinical trials for the treatment of autoimmune diseases, transplant rejection and graft-versus-host disease. In this Review, we discuss the biology of Treg cells and describe new efforts in Treg cell engineering to enhance specificity, stability, functional activity and delivery. Finally, we envision that the success of Treg cell therapy in autoimmunity and transplantation will encourage the clinical use of adoptive Treg cell therapy for non-immune diseases, such as neurological disorders and tissue repair.
Collapse
Affiliation(s)
- Caroline Raffin
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Linda T Vo
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey A Bluestone
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
90
|
Shao L, Zhang Y, Pan X, Liu B, Liang C, Zhang Y, Wang Y, Yan B, Xie W, Sun Y, Shen Z, Yu XY, Li Y. Knockout of beta-2 microglobulin enhances cardiac repair by modulating exosome imprinting and inhibiting stem cell-induced immune rejection. Cell Mol Life Sci 2020; 77:937-952. [PMID: 31312880 PMCID: PMC11104803 DOI: 10.1007/s00018-019-03220-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/26/2019] [Accepted: 07/05/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Allogeneic human umbilical mesenchymal stem cells (alloUMSC) are convenient cell source for stem cell-based therapy. However, immune rejection is a major obstacle for clinical application of alloUMSC for cardiac repair after myocardial infarction (MI). The immune rejection is due to the presence of human leukocyte antigen (HLA) class I molecule which is increased during MI. The aim of this study was to knockout HLA light chain β2-microglobulin (B2M) in UMSC to enhance stem cell engraftment and survival after transplantation. METHODS AND RESULTS We developed an innovative strategy using CRISPR/Cas9 to generate UMSC with B2M deletion (B2M-UMSC). AlloUMSC injection induced CD8+ T cell-mediated immune rejection in immune competent rats, whereas no CD8+ T cell-mediated killing against B2M-UMSC was observed even when the cells were treated with IFN-γ. Moreover, we demonstrate that UMSC-derived exosomes can inhibit cardiac fibrosis and restore cardiac function, and exosomes derived from B2M-UMSC are more efficient than those derived from UMSC, indicating that the beneficial effect of exosomes can be enhanced by modulating exosome's imprinting. Mechanistically, microRNA sequencing identifies miR-24 as a major component of the exosomes from B2M-UMSCs. Bioinformatics analysis identifies Bim as a putative target of miR-24. Loss-of-function studies at the cellular level and gain-of-function approaches in exosomes show that the beneficial effects of B2M-UMSCs are mediated by the exosome/miR-24/Bim pathway. CONCLUSION Our findings demonstrate that modulation of exosome's imprinting via B2M knockout is an efficient strategy to prevent the immune rejection of alloUMSCs. This study paved the way to the development of new strategies for tissue repair and regeneration without the need for HLA matching.
Collapse
Affiliation(s)
- Lianbo Shao
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Yu Zhang
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Xiangbin Pan
- Department of Cardiac Surgery, Fuwai Hospital, Beijing, 100037, People's Republic of China
| | - Bin Liu
- Department of Cardiology, The First Hospital of Jilin University, Changchun, 130041, Jilin, People's Republic of China
| | - Chun Liang
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | - Yuqing Zhang
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Yanli Wang
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Bing Yan
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Wenping Xie
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Yi Sun
- Fuwai Yunnan Cardiovascular Hospital, Kunming, 650302, Yunnan, People's Republic of China
| | - Zhenya Shen
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Xi-Yong Yu
- Guangzhou Medical University, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Yangxin Li
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China.
| |
Collapse
|
91
|
iPSC-Derived Platelets Depleted of HLA Class I Are Inert to Anti-HLA Class I and Natural Killer Cell Immunity. Stem Cell Reports 2019; 14:49-59. [PMID: 31883921 PMCID: PMC6962657 DOI: 10.1016/j.stemcr.2019.11.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 11/29/2019] [Accepted: 11/29/2019] [Indexed: 12/17/2022] Open
Abstract
The ex vivo production of platelets depleted of human leukocyte antigen class I (HLA-I) could serve as a universal measure to overcome platelet transfusion refractoriness caused by HLA-I incompatibility. Here, we developed human induced pluripotent cell-derived HLA-I-deficient platelets (HLA-KO iPLATs) in a clinically applicable imMKCL system by genetic manipulation and assessed their immunogenic properties including natural killer (NK) cells, which reject HLA-I downregulated cells. HLA-KO iPLATs were deficient for all HLA-I but did not elicit a cytotoxic response by NK cells in vitro and showed circulation equal to wild-type iPLATs upon transfusion in our newly established Hu-NK-MSTRG mice reconstituted with human NK cells. Additionally, HLA-KO iPLATs successfully circulated in an alloimmune platelet transfusion refractoriness model of Hu-NK-MISTRG mice. Mechanistically, the lack of NK cell-activating ligands on platelets may be responsible for evading the NK cell response. This study revealed the unique non-immunogenic property of platelets and provides a proof of concept for the clinical application of HLA-KO iPLATs. Clinically applicable iPSC-derived HLA class I knockout platelets (HLA-KO iPLATs) HLA-KO iPLATs do not elicit NK cell activation in vitro HLA-KO iPLATs circulate comparably with wild type in human NK cell-reconstituted mice HLA-KO iPLATs circulate competently in alloimmune PTR model mice
Collapse
|
92
|
Bilodeau C, Goltsis O, Rogers IM, Post M. Limitations of recellularized biological scaffolds for human transplantation. J Tissue Eng Regen Med 2019; 14:521-538. [PMID: 31826325 DOI: 10.1002/term.3004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022]
Abstract
A shortage of donor organs for transplantation and the dependence of the recipients on immunosuppressive therapy have motivated researchers to consider alternative regenerative approaches. The answer may reside in acellular scaffolds generated from cadaveric human and animal tissues. Acellular scaffolds are expected to preserve the architectural and mechanical properties of the original organ, permitting cell attachment, growth, and differentiation. Although theoretically, the use of acellular scaffolds for transplantation should pose no threat to the recipient's immune system, experimental data have revealed significant immune responses to allogeneic and xenogeneic transplanted scaffolds. Herein, we review the various factors of the scaffold that could trigger an inflammatory and/or immune response, thereby compromising its use for human transplant therapy. In addition, we provide an overview of the major cell types that have been considered for recellularization of the scaffold and their potential contribution to triggering an immune response.
Collapse
Affiliation(s)
- Claudia Bilodeau
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Olivia Goltsis
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Ian M Rogers
- Lunenfeld Research Institute, Mount Sinai Health, Toronto, Ontario, Canada
| | - Martin Post
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
93
|
Yoshimura Y, Yamanishi A, Kamitani T, Kim JS, Takeda J. Generation of targeted homozygosity in the genome of human induced pluripotent stem cells. PLoS One 2019; 14:e0225740. [PMID: 31805151 PMCID: PMC6894808 DOI: 10.1371/journal.pone.0225740] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/11/2019] [Indexed: 11/18/2022] Open
Abstract
When loss of heterozygosity (LOH) is correlated with loss or gain of a disease phenotype, it is often necessary to identify which gene or genes are involved. Here, we developed a region-specific LOH-inducing system based on mitotic crossover in human induced pluripotent stem cells (hiPSCs). We first tested our system on chromosome 19. To detect homozygous clones generated by LOH, a positive selection cassette was inserted at the AASV1 locus of chromosome 19. LOHs were generated by the combination of allele-specific double-stranded DNA breaks introduced by CRISPR/Cas9 and suppression of Bloom syndrome (BLM) gene expression by the Tet-Off system. The BLM protein inhibitor ML216 exhibited a similar crossover efficiency and distribution of crossover sites. We next applied this system to the short arm of chromosome 6, where human leukocyte antigen (HLA) loci are located. Genotyping and flow cytometric analysis demonstrated that LOHs associated with chromosomal crossover occurred at the expected positions. Although careful examination of HLA-homozygous hiPSCs generated from parental cells is needed for cancer predisposition and effectiveness of differentiation, they may help to mitigate the current shortcoming of hiPSC-based transplantation related to the immunological differences between the donor and host.
Collapse
Affiliation(s)
- Yasuhide Yoshimura
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- * E-mail: (JT); (YY)
| | - Ayako Yamanishi
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tomo Kamitani
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Jin-Soo Kim
- Center for Genome Engineering, Institute for Basic Science, Seoul, South Korea
| | - Junji Takeda
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- * E-mail: (JT); (YY)
| |
Collapse
|
94
|
Zhang Y, Wang Y, Shao L, Pan X, Liang C, Liu B, Zhang Y, Xie W, Yan B, Liu F, Yu XY, Li Y. Knockout of beta-2 microglobulin reduces stem cell-induced immune rejection and enhances ischaemic hindlimb repair via exosome/miR-24/Bim pathway. J Cell Mol Med 2019; 24:695-710. [PMID: 31729180 PMCID: PMC6933348 DOI: 10.1111/jcmm.14778] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/30/2019] [Accepted: 10/05/2019] [Indexed: 12/14/2022] Open
Abstract
Generating universal human umbilical mesenchymal stem cells (UMSCs) without immune rejection is desirable for clinical application. Here we developed an innovative strategy using CRISPR/Cas9 to generate B2M‐UMSCs in which human leucocyte antigen (HLA) light chain β2‐microglobulin (B2M) was deleted. The therapeutic potential of B2M‐UMSCs was examined in a mouse ischaemic hindlimb model. We show that B2M‐UMSCs facilitated perfusion recovery and enhanced running capability, without inducing immune rejection. The beneficial effect was mediated by exosomes. Mechanistically, microRNA (miR) sequencing identified miR‐24 as a major component of the exosomes originating from B2M‐UMSCs. We identified Bim as a potential target of miR‐24 through bioinformatics analysis, which was further confirmed by loss‐of‐function and gain‐of‐function approaches. Taken together, our data revealed that knockout of B2M is a convenient and efficient strategy to prevent UMSCs‐induced immune rejection, and it provides a universal clinical‐scale cell source for tissue repair and regeneration without the need for HLA matching in the future.
Collapse
Affiliation(s)
- Yuqing Zhang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanli Wang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lianbo Shao
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiangbin Pan
- Department of Cardiac Surgery, Fuwai Hospital, Beijing, China
| | - Chun Liang
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Bin Liu
- Department of Cardiology, Second Hospital of Jilin University, Jilin, China
| | - Yu Zhang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenping Xie
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bing Yan
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Feng Liu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xi-Yong Yu
- Guangzhou Medical University, Guangzhou, China
| | - Yangxin Li
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
95
|
Mattapally S, Pawlik KM, Fast VG, Zumaquero E, Lund FE, Randall TD, Townes TM, Zhang J. Human Leukocyte Antigen Class I and II Knockout Human Induced Pluripotent Stem Cell-Derived Cells: Universal Donor for Cell Therapy. J Am Heart Assoc 2019; 7:e010239. [PMID: 30488760 PMCID: PMC6405542 DOI: 10.1161/jaha.118.010239] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Background We aim to generate a line of “universal donor” human induced pluripotent stem cells (hiPSCs) that are nonimmunogenic and, therefore, can be used to derive cell products suitable for allogeneic transplantation. Methods and Results hiPSCs carrying knockout mutations for 2 key components (β2 microglobulin and class II major histocompatibility class transactivator) of major histocompatibility complexes I and II (ie, human leukocyte antigen [HLA] I/II knockout hiPSCs) were generated using the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR associated protein 9 (Cas9) gene‐editing system and differentiated into cardiomyocytes. Pluripotency‐gene expression and telomerase activity in wild‐type (WT) and HLAI/II knockout hiPSCs, cardiomyocyte marker expression in WT and HLAI/II knockout hiPSC‐derived cardiomyocytes, and assessments of electrophysiological properties (eg, conduction velocity, action‐potential and calcium transient half‐decay times, and calcium transient increase times) in spheroid‐fusions composed of WT and HLAI/II knockout cardiomyocytes, were similar. However, the rates of T‐cell activation before (≈21%) and after (≈24%) exposure to HLAI/II knockout hiPSC‐derived cardiomyocytes were nearly indistinguishable and dramatically lower than after exposure to WT hiPSC‐derived cardiomyocytes (≈75%), and when WT and HLAI/II knockout hiPSC‐derived cardiomyocyte spheroids were cultured with human peripheral blood mononuclear cells, the WT hiPSC‐derived cardiomyocyte spheroids were smaller and displayed contractile irregularities. Finally, expression of HLA‐E and HLA‐F was inhibited in HLAI/II knockout cardiomyocyte spheroids after coculture with human peripheral blood mononuclear cells, although HLA‐G was not inhibited; these results are consistent with the essential role of class II major histocompatibility class transactivator in transcriptional activation of the HLA‐E and HLA‐F genes, but not the HLA‐G gene. Expression of HLA‐G is known to inhibit natural killer cell recognition and killing of cells that lack other HLAs. Conclusions HLAI/II knockout hiPSCs can be differentiated into cardiomyocytes that induce little or no activity in human immune cells and, consequently, are suitable for allogeneic transplantation.
Collapse
Affiliation(s)
- Saidulu Mattapally
- 1 Department of Biomedical Engineering School of Medicine School of Engineering The University of Alabama at Birmingham AL
| | - Kevin M Pawlik
- 2 Department of Biochemistry and Molecular Genetics School of Medicine The University of Alabama at Birmingham AL
| | - Vladimir G Fast
- 1 Department of Biomedical Engineering School of Medicine School of Engineering The University of Alabama at Birmingham AL
| | - Esther Zumaquero
- 3 Department of Microbiology School of Medicine The University of Alabama at Birmingham AL
| | - Frances E Lund
- 3 Department of Microbiology School of Medicine The University of Alabama at Birmingham AL
| | - Troy D Randall
- 4 Department of Medicine/Rheumatology School of Medicine The University of Alabama at Birmingham AL
| | - Tim M Townes
- 2 Department of Biochemistry and Molecular Genetics School of Medicine The University of Alabama at Birmingham AL
| | - Jianyi Zhang
- 1 Department of Biomedical Engineering School of Medicine School of Engineering The University of Alabama at Birmingham AL
| |
Collapse
|
96
|
Bogomiakova ME, Eremeev AV, Lagarkova MA. At Home among Strangers: Is It Possible to Create Hypoimmunogenic Pluripotent Stem Cell Lines? Mol Biol 2019. [DOI: 10.1134/s0026893319050042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
97
|
Happle C, Lachmann N, Ackermann M, Mirenska A, Göhring G, Thomay K, Mucci A, Hetzel M, Glomb T, Suzuki T, Chalk C, Glage S, Dittrich-Breiholz O, Trapnell B, Moritz T, Hansen G. Pulmonary Transplantation of Human Induced Pluripotent Stem Cell-derived Macrophages Ameliorates Pulmonary Alveolar Proteinosis. Am J Respir Crit Care Med 2019; 198:350-360. [PMID: 29652170 DOI: 10.1164/rccm.201708-1562oc] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Although the transplantation of induced pluripotent stem cell (iPSC)-derived cells harbors enormous potential for the treatment of pulmonary diseases, in vivo data demonstrating clear therapeutic benefits of human iPSC-derived cells in lung disease models are missing. OBJECTIVES We have tested the therapeutic potential of iPSC-derived macrophages in a humanized disease model of hereditary pulmonary alveolar proteinosis (PAP). Hereditary PAP is caused by a genetic defect of the GM-CSF (granulocyte-macrophage colony-stimulating factor) receptor, which leads to disturbed macrophage differentiation and protein/surfactant degradation in the lungs, subsequently resulting in severe respiratory insufficiency. METHODS Macrophages derived from human iPSCs underwent intrapulmonary transplantation into humanized PAP mice, and engraftment, in vivo differentiation, and therapeutic efficacy of the transplanted cells were analyzed. MEASUREMENTS AND MAIN RESULTS On intratracheal application, iPSC-derived macrophages engrafted in the lungs of humanized PAP mice. After 2 months, transplanted cells displayed the typical morphology, surface markers, functionality, and transcription profile of primary human alveolar macrophages. Alveolar proteinosis was significantly reduced as demonstrated by diminished protein content and surfactant protein D levels, decreased turbidity of the BAL fluid, and reduced surfactant deposition in the lungs of transplanted mice. CONCLUSIONS We here demonstrate for the first time that pulmonary transplantation of human iPSC-derived macrophages leads to pulmonary engraftment, their in situ differentiation to an alveolar macrophage phenotype, and a reduction of alveolar proteinosis in a humanized PAP model. To our knowledge, this finding presents the first proof-of-concept for the therapeutic potential of human iPSC-derived cells in a pulmonary disease and may have profound implications beyond the rare disease of PAP.
Collapse
Affiliation(s)
- Christine Happle
- 1 Department of Pediatric Pneumology, Allergology and Neonatology.,2 Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL)
| | - Nico Lachmann
- 3 Junior Research Group (JRG) Translational Hematology of Congenital Diseases, Regenerative Biology and Reconstructive Therapies (REBIRTH) Cluster of Excellence.,4 Institute of Experimental Hematology
| | - Mania Ackermann
- 3 Junior Research Group (JRG) Translational Hematology of Congenital Diseases, Regenerative Biology and Reconstructive Therapies (REBIRTH) Cluster of Excellence.,4 Institute of Experimental Hematology
| | - Anja Mirenska
- 1 Department of Pediatric Pneumology, Allergology and Neonatology
| | | | | | - Adele Mucci
- 4 Institute of Experimental Hematology.,6 Research Group-Reprogramming and Gene Therapy, REBIRTH Cluster of Excellence
| | - Miriam Hetzel
- 4 Institute of Experimental Hematology.,6 Research Group-Reprogramming and Gene Therapy, REBIRTH Cluster of Excellence
| | - Torsten Glomb
- 7 Core Unit Transcriptomics, Institute for Physiological Chemistry, and
| | - Takuji Suzuki
- 8 Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Claudia Chalk
- 8 Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Silke Glage
- 9 Central Animal Facility, Hannover Medical School, Hannover, Germany; and
| | | | - Bruce Trapnell
- 8 Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Thomas Moritz
- 4 Institute of Experimental Hematology.,6 Research Group-Reprogramming and Gene Therapy, REBIRTH Cluster of Excellence
| | - Gesine Hansen
- 1 Department of Pediatric Pneumology, Allergology and Neonatology.,2 Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL)
| |
Collapse
|
98
|
Singh J, Zúñiga-Pflücker JC. Producing proT cells to promote immunotherapies. Int Immunol 2019; 30:541-550. [PMID: 30102361 DOI: 10.1093/intimm/dxy051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 08/08/2018] [Indexed: 12/17/2022] Open
Abstract
T lymphocytes are critical mediators of the adaptive immune system and they can be harnessed as therapeutic agents against pathogens and in cancer immunotherapy. T cells can be isolated and expanded from patients and potentially generated in vitro using clinically relevant systems. An ultimate goal for T-cell immunotherapy is to establish a safe, universal effector cell type capable of transcending allogeneic and histocompatibility barriers. To this end, human pluripotent stem cells offer an advantage in generating a boundless supply of T cells that can be readily genetically engineered. Here, we review emerging T-cell therapeutics, including tumor-infiltrating lymphocytes, chimeric antigen receptors and progenitor T cells (proT cells) as well as feeder cell-free in vitro systems for their generation. Furthermore, we explore their potential for adoption in the clinic and highlight the challenges that must be addressed to increase the therapeutic success of a universal immunotherapy.
Collapse
Affiliation(s)
- Jastaranpreet Singh
- Department of Immunology, University of Toronto, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | | |
Collapse
|
99
|
Sharma D, Ross D, Wang G, Jia W, Kirkpatrick SJ, Zhao F. Upgrading prevascularization in tissue engineering: A review of strategies for promoting highly organized microvascular network formation. Acta Biomater 2019; 95:112-130. [PMID: 30878450 DOI: 10.1016/j.actbio.2019.03.016] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/20/2019] [Accepted: 03/06/2019] [Indexed: 01/05/2023]
Abstract
Functional and perfusable vascular network formation is critical to ensure the long-term survival and functionality of engineered tissues after their transplantation. Although several vascularization strategies have been reviewed in past, the significance of microvessel organization in three-dimensional (3D) scaffolds has been largely ignored. Advances in high-resolution microscopy and image processing have revealed that the majority of tissues including cardiac, skeletal muscle, bone, and skin contain highly organized microvessels that orient themselves to align with tissue architecture for optimum molecular exchange and functional performance. Here, we review strategies to develop highly organized and mature vascular networks in engineered tissues, with a focus on electromechanical stimulation, surface topography, micro scaffolding, surface-patterning, microfluidics and 3D printing. This review will provide researchers with state of the art approaches to engineer vascularized functional tissues for diverse applications. STATEMENT OF SIGNIFICANCE: Vascularization is one of the critical challenges facing tissue engineering. Recent technological advances have enabled researchers to develop microvascular networks in engineered tissues. Although far from translational applications, current vascularization strategies have shown promising outcomes. This review emphasizes the most recent technological advances and future challenges for developing organized microvascular networks in vitro. The next critical step is to achieve highly perfusable, dense, mature and organized microvascular networks representative of native tissues.
Collapse
|
100
|
Chakravarti D, Caraballo LD, Weinberg BH, Wong WW. Inducible Gene Switches with Memory in Human T Cells for Cellular Immunotherapy. ACS Synth Biol 2019; 8:1744-1754. [PMID: 31268301 PMCID: PMC6703182 DOI: 10.1021/acssynbio.8b00512] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
![]()
Cell-based therapies that employ
engineered T cells—including
those modified to express chimeric antigen receptors (CARs)—to
target cancer cells have demonstrated promising responses in clinical
trials. However, engineered T cell responses must be regulated to
prevent severe side effects such as cytokine storms and off-target
responses. Here we present a class of recombinase-based gene circuits
that will enable inducible, one-time state switching in adoptive T
cell therapy using an FDA-approved drug, creating a generalizable
platform that can be used to control when and how strongly a gene
is expressed. These circuits exhibit memory such that induced T cells
will maintain any changes made even when the drug inducer is removed.
This memory feature avoids prolonged drug inducer exposure, thus reducing
the complexity and potential side effect associated with the drug
inducer. We have utilized these circuits to control the expression
of an anti-Her2-CAR, demonstrating the ability of these circuits to
regulate CAR expression and T cell activity. We envision this platform
can be extended to regulate other genes involved in T cell behavior
for applications in various adoptive T cell therapies.
Collapse
Affiliation(s)
- Deboki Chakravarti
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States
- Biological Design Center, Boston University, Boston, Massachusetts 02215, United States
| | - Leidy D. Caraballo
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States
- Biological Design Center, Boston University, Boston, Massachusetts 02215, United States
| | - Benjamin H. Weinberg
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States
- Biological Design Center, Boston University, Boston, Massachusetts 02215, United States
| | - Wilson W. Wong
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States
- Biological Design Center, Boston University, Boston, Massachusetts 02215, United States
| |
Collapse
|