1
|
Puspita L, Juwono VB, Shim JW. Advances in human pluripotent stem cell reporter systems. iScience 2024; 27:110856. [PMID: 39290832 PMCID: PMC11407076 DOI: 10.1016/j.isci.2024.110856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
The capability of human pluripotent stem cells (hPSCs) to self-renew and differentiate into any cell type has greatly contributed to the advancement of biomedicine. Reporter lines derived from hPSCs have played a crucial role in elucidating the mechanisms underlying human development and diseases by acting as an alternative reporter system that cannot be used in living humans. To bring hPSCs closer to clinical application in transplantation, scientists have generated reporter lines for isolating the desired cell populations, as well as improving graft quality and treatment outcomes. This review presents an overview of the applications of hPSC reporter lines and the important variables in designing a reporter system, including options for gene delivery and editing tools, design of reporter constructs, and selection of reporter genes. It also provides insights into the prospects of hPSC reporter lines and the challenges that must be overcome to maximize the potential of hPSC reporter lines.
Collapse
Affiliation(s)
- Lesly Puspita
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea
| | - Virginia Blessy Juwono
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si 31151, Korea
| | - Jae-Won Shim
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si 31151, Korea
| |
Collapse
|
2
|
Jovanovic VM, Mesch KT, Tristan CA. hPSC-Derived Astrocytes at the Forefront of Translational Applications in Neurological Disorders. Cells 2024; 13:903. [PMID: 38891034 PMCID: PMC11172187 DOI: 10.3390/cells13110903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Abstract
Astrocytes, the most abundant glial cell type in the brain, play crucial roles in maintaining homeostasis within the central nervous system (CNS). Impairment or abnormalities of typical astrocyte functions in the CNS serve as a causative or contributing factor in numerous neurodevelopmental, neurodegenerative, and neuropsychiatric disorders. Currently, disease-modeling and drug-screening approaches, primarily focused on human astrocytes, rely on human pluripotent stem cell (hPSC)-derived astrocytes. However, it is important to acknowledge that these hPSC-derived astrocytes exhibit notable differences across studies and when compared to their in vivo counterparts. These differences may potentially compromise translational outcomes if not carefully accounted for. This review aims to explore state-of-the-art in vitro models of human astrocyte development, focusing on the developmental processes, functional maturity, and technical aspects of various hPSC-derived astrocyte differentiation protocols. Additionally, it summarizes their successful application in modeling neurological disorders. The discussion extends to recent advancements in the large-scale production of human astrocytes and their application in developing high-throughput assays conducive to therapeutic drug discovery.
Collapse
Affiliation(s)
- Vukasin M. Jovanovic
- Stem Cell Translation Laboratory (SCTL), Division of Preclinical Innovation (DPI), National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850, USA (C.A.T.)
| | | | | |
Collapse
|
3
|
Saito Y, Nose N, Iida T, Akazawa K, Kanno T, Fujimoto Y, Sasaki T, Akehi M, Higuchi T, Akagi S, Yoshida M, Miyoshi T, Ito H, Nakamura K. In vivo tracking transplanted cardiomyocytes derived from human induced pluripotent stem cells using nuclear medicine imaging. Front Cardiovasc Med 2023; 10:1261330. [PMID: 37745108 PMCID: PMC10512708 DOI: 10.3389/fcvm.2023.1261330] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction Transplantation of human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) is a promising treatment for heart failure. Information on long-term cell engraftment after transplantation is clinically important. However, clinically applicable evaluation methods have not yet been established. Methods In this study, to noninvasively assess transplanted cell engraftment, human SLC5A5, which encodes a sodium/iodide symporter (NIS) that transports radioactive tracers such as 125I, 18F-tetrafluoroborate (TFB), and 99mTc-pertechnetate (99mTcO4-), was transduced into human induced pluripotent stem cells (iPSCs), and nuclear medicine imaging was used to track engrafted human iPSC-CMs. Results To evaluate the pluripotency of NIS-expressing human iPSCs, they were subcutaneously transplanted into immunodeficient rats. Teratomas were detected by 99mTcO4- single photon emission computed tomography (SPECT/CT) imaging. NIS expression and the uptake ability of 125I were maintained in purified human iPSC-CMs. NIS-expressing human iPSC-CMs transplanted into immunodeficient rats could be detected over time using 99mTcO4- SPECT/CT imaging. Unexpectedly, NIS expression affected cell proliferation of human iPSCs and iPSC-derived cells. Discussion Such functionally designed iPSC-CMs have potential clinical applications as a noninvasive method of grafted cell evaluation, but further studies are needed to determine the effects of NIS transduction on cellular characteristics and functions.
Collapse
Affiliation(s)
- Yukihiro Saito
- Department of Cardiovascular Medicine, Okayama University Hospital, Okayama, Japan
| | - Naoko Nose
- Molecular Imaging Project of RECTOR Program, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toshihiro Iida
- Department of Cardiovascular Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kaoru Akazawa
- Department of Cardiovascular Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takayuki Kanno
- Molecular Imaging Project of RECTOR Program, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yuki Fujimoto
- Molecular Imaging Project of RECTOR Program, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takanori Sasaki
- Okayama Medical Innovation Center, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masaru Akehi
- Okayama Medical Innovation Center, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takahiro Higuchi
- Molecular Imaging Project of RECTOR Program, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Satoshi Akagi
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masashi Yoshida
- Department of Chronic Kidney Disease and Cardiovascular Disease, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toru Miyoshi
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hiroshi Ito
- Department of General Internal Medicine 3, Kawasaki Medical School, Okayama, Japan
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
4
|
Saleem A, Abbas MK, Wang Y, Lan F. hPSC gene editing for cardiac disease therapy. Pflugers Arch 2022; 474:1123-1132. [PMID: 36163402 DOI: 10.1007/s00424-022-02751-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/09/2022] [Accepted: 09/18/2022] [Indexed: 11/26/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality worldwide. However, the lack of human cardiomyocytes with proper genetic backgrounds limits the study of disease mechanisms. Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have significantly advanced the study of these conditions. Moreover, hPSC-CMs made it easy to study CVDs using genome-editing techniques. This article discusses the applications of these techniques in hPSC for studying CVDs. Recently, several genome-editing systems have been used to modify hPSCs, including zinc finger nucleases, transcription activator-like effector nucleases, and clustered regularly interspaced short palindromic repeat-associated protein 9 (CRISPR/Cas9). We focused on the recent advancement of genome editing in hPSCs, which dramatically improved the efficiency of the cell-based mechanism study and therapy for cardiac diseases.
Collapse
Affiliation(s)
- Amina Saleem
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, MOE Key Laboratory of Remodeling Related Cardiovascular Disease, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Research Institute Building, Beijinj Anzhen Hospital, Capital Medical University, Room 319, 2 Anzhen Road, Chaoyang District, Beijing, Beijing, 100029, China
| | - Muhammad Khawar Abbas
- BHMS Department, University College of Conventional Medicine, Faculty of Medicine and Allied Health Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Yongming Wang
- The State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, China
- The Key Lab of Reproduction Regulation of NPFPC in SIPPR, Institute of Reproduction & Development in Obstetrics & Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Feng Lan
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, MOE Key Laboratory of Remodeling Related Cardiovascular Disease, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Research Institute Building, Beijinj Anzhen Hospital, Capital Medical University, Room 319, 2 Anzhen Road, Chaoyang District, Beijing, Beijing, 100029, China.
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen Key Laboratory of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Beijing, 100029, China.
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central-China Branch of National Center for Cardiovascular Diseases, Zhengzhou, Beijing, 100037, China.
| |
Collapse
|
5
|
Shalaby N, Kelly J, Martinez F, Fox M, Qi Q, Thiessen J, Hicks J, Scholl TJ, Ronald JA. A Human-derived Dual MRI/PET Reporter Gene System with High Translational Potential for Cell Tracking. Mol Imaging Biol 2022; 24:341-351. [PMID: 35146614 PMCID: PMC9235057 DOI: 10.1007/s11307-021-01697-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Reporter gene imaging has been extensively used to longitudinally report on whole-body distribution and viability of transplanted engineered cells. Multi-modal cell tracking can provide complementary information on cell fate. Typical multi-modal reporter gene systems often combine clinical and preclinical modalities. A multi-modal reporter gene system for magnetic resonance imaging (MRI) and positron emission tomography (PET), two clinical modalities, would be advantageous by combining the sensitivity of PET with the high-resolution morphology and non-ionizing nature of MRI. PROCEDURES We developed and evaluated a dual MRI/PET reporter gene system composed of two human-derived reporter genes that utilize clinical reporter probes for engineered cell detection. As a proof-of-concept, breast cancer cells were engineered to co-express the human organic anion transporter polypeptide 1B3 (OATP1B3) that uptakes the clinical MRI contrast agent gadolinium ethoxybenzyl-diethylenetriaminepentaacetic acid (Gd-EOB-DTPA), and the human sodium iodide symporter (NIS) which uptakes the PET tracer, [18F] tetrafluoroborate ([18F] TFB). RESULTS T1-weighted MRI results in mice exhibited significantly higher MRI signals in reporter-gene-engineered mammary fat pad tumors versus contralateral naïve tumors (p < 0.05). No differences in contrast enhancement were observed at 5 h after Gd-EOB-DTPA administration using either intravenous or intraperitoneal injection. We also found significantly higher standard uptake values (SUV) in engineered tumors in comparison to the naïve tumors in [18F]TFB PET images (p < 0.001). Intratumoral heterogeneity in signal enhancement was more conspicuous in relatively higher resolution MR images compared to PET images. CONCLUSIONS Our study demonstrates the ability to noninvasively track cells engineered with our human-derived dual MRI/PET reporter system, enabling a more comprehensive evaluation of transplanted cells. Future work is focused on applying this tool to track therapeutic cells, which may one day enable the broader application of cell tracking within the healthcare system.
Collapse
Affiliation(s)
- Nourhan Shalaby
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada.
| | - John Kelly
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Francisco Martinez
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Mathew Fox
- Lawson Health Research Institute, London, Canada
- Saint Joseph's Health Care, Toronto, Canada
| | - Qi Qi
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Jonathan Thiessen
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Saint Joseph's Health Care, Toronto, Canada
- Lawson Cyclotron and Radiochemistry Facility, London, Canada
| | - Justin Hicks
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Lawson Health Research Institute, London, Canada
- Lawson Cyclotron and Radiochemistry Facility, London, Canada
| | - Timothy J Scholl
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Ontario Institute for Cancer Research, Toronto, Canada
| | - John A Ronald
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Lawson Health Research Institute, London, Canada
| |
Collapse
|
6
|
Saito Y, Nakamura K, Yoshida M, Sugiyama H, Akagi S, Miyoshi T, Morita H, Ito H. Enhancement of pacing function by HCN4 overexpression in human pluripotent stem cell-derived cardiomyocytes. Stem Cell Res Ther 2022; 13:141. [PMID: 35365232 PMCID: PMC8973792 DOI: 10.1186/s13287-022-02818-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/20/2022] [Indexed: 11/10/2022] Open
Abstract
Background The number of patients with bradyarrhythmia and the number of patients with cardiac pacemakers are increasing with the aging population and the increase in the number of patients with heart diseases. Some patients in whom a cardiac pacemaker has been implanted experience problems such as pacemaker infection and inconvenience due to electromagnetic interference. We have reported that overexpression of HCN channels producing a pacemaker current in mouse embryonic stem cell-derived cardiomyocytes showed enhanced pacing function in vitro and in vivo. The aim of this study was to determine whether HCN4 overexpression in human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) can strengthen the pacing function of the cells. Methods Human HCN4 was transduced in the AAVS1 locus of human induced pluripotent stem cells by nucleofection and HCN4-overexpressing iPSC-CMs were generated. Gene expression profiles, frequencies of spontaneous contraction and pacing abilities of HCN4-overexpressing and non-overexpressing iPSC-CMs in vitro were compared. Results HCN4-overexpressing iPSC-CMs showed higher spontaneous contraction rates than those of non-overexpressing iPSC-CMs. They responded to an HCN channel blocker and β adrenergic stimulation. The pacing rates against parent iPSC line-derived cardiomyocytes were also higher in HCN4-overexpressing iPSC-CMs than in non-overexpressing iPSC-CMs. Conclusions Overexpression of HCN4 showed enhancement of If current, spontaneous firing and pacing function in iPSC-CMs. These data suggest this transgenic cell line may be useful as a cardiac pacemaker. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02818-y.
Collapse
Affiliation(s)
- Yukihiro Saito
- Department of Cardiovascular Medicine, Okayama University Hospital, Okayama, Japan.
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, 700-8558, Kita-ku, Okayama, Japan.
| | - Masashi Yoshida
- Department of Chronic Kidney Disease and Cardiovascular Disease, Dentistry, and Pharmaceutical Science, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Hiroki Sugiyama
- Department of Internal Medicine, Okayama Saiseikai General Hospital, Okayama, Japan
| | - Satoshi Akagi
- Department of Cardiovascular Medicine, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, 700-8558, Kita-ku, Okayama, Japan
| | - Toru Miyoshi
- Department of Cardiovascular Medicine, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, 700-8558, Kita-ku, Okayama, Japan
| | - Hiroshi Morita
- Department of Cardiovascular Therapeutics, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Hiroshi Ito
- Department of Cardiovascular Medicine, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, 700-8558, Kita-ku, Okayama, Japan
| |
Collapse
|
7
|
Mullen MMS, Wu JC. Utilization of induced pluripotent stem cells to model the molecular network regulating congenital heart disease. Cardiovasc Res 2022; 118:664-666. [PMID: 34971365 PMCID: PMC8859623 DOI: 10.1093/cvr/cvab373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Indexed: 01/02/2023] Open
Affiliation(s)
- McKay M S Mullen
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Room 3200, Palo Alto, CA, 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Room 3200, Palo Alto, CA, 94305, USA
| |
Collapse
|
8
|
Wang S, Mao H, Hou L, Hu Z, Wang Y, Qi T, Tao C, Yang Y, Zhang C, Li M, Liu H, Hu S, Chai R, Wang Y. Compact SchCas9 Recognizes the Simple NNGR PAM. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104789. [PMID: 34874112 PMCID: PMC8811835 DOI: 10.1002/advs.202104789] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/11/2021] [Indexed: 05/20/2023]
Abstract
Clustered regularly interspaced short palindromic repeat (CRISPR)/SaCas9 is the most popular tool for in vivo genome editing due to its high efficiency and small genome. The authors previously developed four SaCas9 orthologs as genome-editing tools. Here, to expand the targeting scope, they investigate the diversity of protospacer adjacent motifs (PAMs) by screening a list of 16 SaCas9 orthologs, twelve of which display editing activity in mammalian cells. They recognize five types of PAMs: NNGRRT, NNGRRR, NNGRC, NNGA, and NNGR. Importantly, SchCas9 recognizes the simple NNGR PAM, representing the most relaxed PAM preference of compact Cas9s to date. It is further demonstrated that SchCas9 enables efficient genome editing in multiple human cell lines. Altogether, these compact Cas9 tools offer a new option for both basic research and clinical applications.
Collapse
Affiliation(s)
- Shuai Wang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Huilin Mao
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Linghui Hou
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Ziying Hu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Yao Wang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Tao Qi
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Chen Tao
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Yuan Yang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Chengdong Zhang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Miaomiao Li
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Huihui Liu
- Experimental Center of Forestry in North ChinaChinese Academy of ForestryBeijing102300China
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular ScienceCollaborative Innovation Center of HematologyState Key Laboratory of Radiation Medicine and ProtectionMedical CollegeSoochow UniversitySuzhou215000China
| | - Renjie Chai
- State Key Laboratory of BioelectronicsSchool of Life Sciences and TechnologyJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjing210096China
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantong226001China
| | - Yongming Wang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
- Shanghai Engineering Research Center of Industrial MicroorganismsShanghai200438China
| |
Collapse
|
9
|
Hu Z, Zhang C, Wang D, Gao S, Ong SG, Wang Y, Zheng WV. A Highly Sensitive GFP Activation Assay for Detection of DNA Cleavage in Cells. Front Cell Dev Biol 2021; 9:771248. [PMID: 34869366 PMCID: PMC8636026 DOI: 10.3389/fcell.2021.771248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/21/2021] [Indexed: 12/02/2022] Open
Abstract
CRISPR/Cas9 nucleases hold great potential for gene therapy, but they frequently induce unwanted off-target cleavage. We previously developed a GFP activation assay for detection of DNA cleavage in cells. Here, we demonstrate two novel applications of this assay. First, we use this assay to confirm off-target cleavage that cannot be detected by targeted deep sequencing in cells before. Second, we use this approach to detect multiple alternative PAMs recognized by SpCas9. These noncanonical PAMs are associated with low cleavage activity, but targets associated with these PAMs must be considered as potential off-target sites. Taken together, the GFP activation assay is a powerful platform for DNA cleavage detection in cells.
Collapse
Affiliation(s)
- Ziying Hu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China.,Centre for Assisted Reproduction, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chengdong Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Daqi Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Siqi Gao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sang-Ging Ong
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL, United States.,Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL, United States
| | - Yongming Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Industrial Microorganisms, Shanghai, China
| | - Wei V Zheng
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
10
|
Low BE, Christianson GJ, Lowell E, Qin W, Wiles MV. Functional humanization of immunoglobulin heavy constant gamma 1 Fc domain human FCGRT transgenic mice. MAbs 2021; 12:1829334. [PMID: 33025844 PMCID: PMC7577234 DOI: 10.1080/19420862.2020.1829334] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
A major asset of many monoclonal antibody (mAb)-based biologics is their persistence in circulation. The MHC class I family Fc receptor, FCGRT, is primarily responsible for this extended pharmacokinetic behavior. Engagement of FCGRT with the crystallizable fragment (Fc) domain protects IgG from catabolic elimination, thereby extending the persistence and bioavailability of IgG and related Fc-based biologics. There is a need for reliable in vivo models to facilitate the preclinical development of novel IgG-based biologics. FcRn-humanized mice have been widely accepted as translationally relevant surrogates for IgG-based biologics evaluations. Although such FCGRT-humanized mice, especially the mouse strain, B6.Cg-Fcgrttm1Dcr Tg(FCGRT)32Dcr (abbreviated Tg32), have been substantially validated for modeling humanized IgG-based biologics, there is a recognized caveat – they lack an endogenous source of human IgG that typifies the human competitive condition. Here, we used CRISPR/Cas9-mediated homology-directed repair to equip the hFCGRT Tg32 strain with a human IGHG1 Fc domain. This replacement now results in mice that produce human IgG1 Fc-mouse IgG Fab2 chimeric antibodies at physiologically relevant levels, which can be further heightened by immunization. This endogenous chimeric IgG1 significantly dampens the serum half-life of administered humanized mAbs in an hFCGRT-dependent manner. Thus, such IgG1-Fc humanized mice may provide a more physiologically relevant competitive hFCGRT-humanized mouse model for the preclinical development of human IgG-based biologics.
Collapse
Affiliation(s)
| | | | - Emily Lowell
- Previously at the Jackson Laboratory , Bar Harbor, ME, USA
| | - Wenning Qin
- Previously at the Jackson Laboratory , Bar Harbor, ME, USA
| | | |
Collapse
|
11
|
Ramasubramanian A, Muckom R, Sugnaux C, Fuentes C, Ekerdt BL, Clark DS, Healy KE, Schaffer DV. High-Throughput Discovery of Targeted, Minimally Complex Peptide Surfaces for Human Pluripotent Stem Cell Culture. ACS Biomater Sci Eng 2021; 7:1344-1360. [PMID: 33750112 DOI: 10.1021/acsbiomaterials.0c01462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human pluripotent stem cells harbor an unlimited capacity to generate therapeutically relevant cells for applications in regenerative medicine. However, to utilize these cells in the clinic, scalable culture systems that activate defined receptors and signaling pathways to sustain stem cell self-renewal are required; and synthetic materials offer considerable promise to meet these needs. De novo development of materials that target novel pathways has been stymied by a limited understanding of critical receptor interactions maintaining pluripotency. Here, we identify peptide agonists for the human pluripotent stem cell (hPSC) laminin receptor and pluripotency regulator, α6-integrin, through unbiased, library-based panning strategies. Biophysical characterization of adhesion suggests that identified peptides bind hPSCs through α6-integrin with sub-μM dissociation constants similar to laminin. By harnessing a high-throughput microculture platform, we developed predictive guidelines for presenting these integrin-targeting peptides alongside canonical binding motifs at optimal stoichiometries to generate nascent culture surfaces. Finally, when presented as self-assembled monolayers, predicted peptide combinations supported hPSC expansion, highlighting how unbiased screens can accelerate the discovery of targeted biomaterials.
Collapse
Affiliation(s)
- Anusuya Ramasubramanian
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Riya Muckom
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Caroline Sugnaux
- Department of Materials Science and Engineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Christina Fuentes
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Barbara L Ekerdt
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Douglas S Clark
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Kevin E Healy
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Materials Science and Engineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - David V Schaffer
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, United States
| |
Collapse
|
12
|
Mullen M, Zhang A, Lui GK, Romfh AW, Rhee JW, Wu JC. Race and Genetics in Congenital Heart Disease: Application of iPSCs, Omics, and Machine Learning Technologies. Front Cardiovasc Med 2021; 8:635280. [PMID: 33681306 PMCID: PMC7925393 DOI: 10.3389/fcvm.2021.635280] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/13/2021] [Indexed: 11/13/2022] Open
Abstract
Congenital heart disease (CHD) is a multifaceted cardiovascular anomaly that occurs when there are structural abnormalities in the heart before birth. Although various risk factors are known to influence the development of this disease, a full comprehension of the etiology and treatment for different patient populations remains elusive. For instance, racial minorities are disproportionally affected by this disease and typically have worse prognosis, possibly due to environmental and genetic disparities. Although research into CHD has highlighted a wide range of causal factors, the reasons for these differences seen in different patient populations are not fully known. Cardiovascular disease modeling using induced pluripotent stem cells (iPSCs) is a novel approach for investigating possible genetic variants in CHD that may be race specific, making it a valuable tool to help solve the mystery of higher incidence and mortality rates among minorities. Herein, we first review the prevalence, risk factors, and genetics of CHD and then discuss the use of iPSCs, omics, and machine learning technologies to investigate the etiology of CHD and its connection to racial disparities. We also explore the translational potential of iPSC-based disease modeling combined with genome editing and high throughput drug screening platforms.
Collapse
Affiliation(s)
- McKay Mullen
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Angela Zhang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Department of Genetics, Stanford School of Medicine, Stanford University, Stanford, CA, United States
| | - George K. Lui
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, United States
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford University, Stanford, CA, United States
| | - Anitra W. Romfh
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Department of Genetics, Stanford School of Medicine, Stanford University, Stanford, CA, United States
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford University, Stanford, CA, United States
| | - June-Wha Rhee
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Department of Genetics, Stanford School of Medicine, Stanford University, Stanford, CA, United States
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Department of Genetics, Stanford School of Medicine, Stanford University, Stanford, CA, United States
- Department of Radiology, Stanford University, Stanford, CA, United States
| |
Collapse
|
13
|
Kelly JJ, Saee-Marand M, Nyström NN, Evans MM, Chen Y, Martinez FM, Hamilton AM, Ronald JA. Safe harbor-targeted CRISPR-Cas9 homology-independent targeted integration for multimodality reporter gene-based cell tracking. SCIENCE ADVANCES 2021; 7:eabc3791. [PMID: 33523917 PMCID: PMC7817109 DOI: 10.1126/sciadv.abc3791] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 11/25/2020] [Indexed: 05/12/2023]
Abstract
Imaging reporter genes provides longitudinal information on the biodistribution, growth, and survival of engineered cells in vivo. A translational bottleneck to using reporter genes is the necessity to engineer cells with randomly integrating vectors. Here, we built homology-independent targeted integration (HITI) CRISPR-Cas9 minicircle donors for precise safe harbor-targeted knock-in of fluorescence, bioluminescence, and MRI (Oatp1a1) reporter genes. Our results showed greater knock-in efficiency using HITI vectors compared to homology-directed repair vectors. HITI clones demonstrated functional fluorescence and bioluminescence reporter activity as well as significant Oatp1a1-mediated uptake of the clinically approved MRI agent gadolinium ethoxybenzyl diethylenetriamine pentaacetic acid. Contrast-enhanced MRI improved the conspicuity of both subcutaneous and metastatic Oatp1a1-expressing tumors before they became palpable or even readily visible on precontrast images. Our work demonstrates the first CRISPR-Cas9 HITI system for knock-in of large DNA donor constructs at a safe harbor locus, enabling multimodal longitudinal in vivo imaging of cells.
Collapse
Affiliation(s)
- John J Kelly
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Moe Saee-Marand
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Nivin N Nyström
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Melissa M Evans
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Yuanxin Chen
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Francisco M Martinez
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Amanda M Hamilton
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - John A Ronald
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
14
|
Gene-Editing Technologies and Applications for Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00061-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
15
|
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
|
16
|
Gao Y, Wu S, Pan J, Zhang K, Li X, Xu Y, Jin C, He X, Shi J, Ma L, Wu F, Yao Y, Wang P, He Q, Lan F, Zhang H, Tian M. CRISPR/Cas9-edited triple-fusion reporter gene imaging of dynamics and function of transplanted human urinary-induced pluripotent stem cell-derived cardiomyocytes. Eur J Nucl Med Mol Imaging 2020; 48:708-720. [PMID: 33216174 DOI: 10.1007/s00259-020-05087-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/20/2020] [Indexed: 12/26/2022]
Abstract
PURPOSE To investigate the post-transplantation behaviour and therapeutic efficacy of human urinary-induced pluripotent stem cell-derived cardiomyocytes (hUiCMs) in infarcted heart. METHODS We used clustered regularly interspaced short palindromic repeats/CRISPR-associated nuclease 9 (CRISPR/Cas9) technology to integrate a triple-fusion (TF) reporter gene into the AAVS1 locus in human urine-derived hiPSCs (hUiPSCs) to generate TF-hUiPSCs that stably expressed monomeric red fluorescent protein for fluorescence imaging, firefly luciferase for bioluminescence imaging (BLI) and herpes simplex virus thymidine kinase for positron emission tomography (PET) imaging. RESULTS Transplanted cardiomyocytes derived from TF-hUiPSCs (TF-hUiCMs) engrafted and proliferated in the infarcted heart as monitored by both BLI and PET imaging and significantly improved cardiac function. Under ischaemic conditions, TF-hUiCMs enhanced cardiomyocyte (CM) glucose metabolism and promoted angiogenic activity. CONCLUSION This study established a CRISPR/Cas9-mediated multimodality reporter gene imaging system that can determine the dynamics and function of TF-hUiCMs in myocardial infarction, which is helpful for investigating the application of stem cell therapy.
Collapse
Affiliation(s)
- Yuanxue Gao
- Department of Nuclear Medicine and PET-CT Center, The Second Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Zhejiang, 310009, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Shuang Wu
- Department of Nuclear Medicine and PET-CT Center, The Second Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Zhejiang, 310009, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Jiayue Pan
- Department of Nuclear Medicine and PET-CT Center, The Second Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Zhejiang, 310009, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Kai Zhang
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, 650-0047, Japan
| | - Xiaoyi Li
- Department of Nuclear Medicine and PET-CT Center, The Second Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Zhejiang, 310009, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Yangyang Xu
- College of Chemical & Biological Engineering, Zhejiang University, Zhejiang, 310027, Hangzhou, China
| | - Chentao Jin
- Department of Nuclear Medicine and PET-CT Center, The Second Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Zhejiang, 310009, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Xiao He
- Department of Nuclear Medicine and PET-CT Center, The Second Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Zhejiang, 310009, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Jingjing Shi
- Department of Nuclear Medicine and PET-CT Center, The Second Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Zhejiang, 310009, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Lijuan Ma
- Department of Nuclear Medicine and PET-CT Center, The Second Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Zhejiang, 310009, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Fujian Wu
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Yao Yao
- Department of Nuclear Medicine and PET-CT Center, The Second Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Zhejiang, 310009, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Ping Wang
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Zhejiang, 310027, Hangzhou, China.,College of Biomedical Engineering and Instrument Science of Zhejiang University, Zhejiang, 310027, Hangzhou, China
| | - Qinggang He
- College of Chemical & Biological Engineering, Zhejiang University, Zhejiang, 310027, Hangzhou, China
| | - Feng Lan
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
| | - Hong Zhang
- Department of Nuclear Medicine and PET-CT Center, The Second Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China. .,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Zhejiang, 310009, Hangzhou, China. .,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Zhejiang, 310009, Hangzhou, China. .,Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Zhejiang, 310027, Hangzhou, China. .,College of Biomedical Engineering and Instrument Science of Zhejiang University, Zhejiang, 310027, Hangzhou, China.
| | - Mei Tian
- Department of Nuclear Medicine and PET-CT Center, The Second Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China. .,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Zhejiang, 310009, Hangzhou, China. .,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Zhejiang, 310009, Hangzhou, China. .,Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Zhejiang, 310027, Hangzhou, China.
| |
Collapse
|
17
|
Gao N, Zhang C, Hu Z, Li M, Wei J, Wang Y, Liu H. Characterization of Brevibacillus laterosporus Cas9 (BlatCas9) for Mammalian Genome Editing. Front Cell Dev Biol 2020; 8:583164. [PMID: 33195228 PMCID: PMC7604293 DOI: 10.3389/fcell.2020.583164] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/24/2020] [Indexed: 11/30/2022] Open
Abstract
Compact CRISPR/Cas9 systems that can be delivered by AAV for in vivo genome editing hold great promise for clinical applications. Brevibacillus laterosporus Cas9 (BlatCas9) is a compact Cas9 nuclease that has been identified for plant genome editing. Here, we characterize BlatCas9 as an alternative tool for mammalian genome editing. We demonstrate that BlatCas9 prefers a N4CNAA protospacer adjacent motif (PAM), but N4C PAM is also editable in mammalian cells. We next demonstrate that BlatCas9 enables genome editing in a variety of cell types. Furthermore, BlatCas9 can be packaged into AAV for genome editing. Finally, we characterize the specificity of BlatCas9. In summary, BlatCas9 offers an alternative tool for both basic research and clinical applications.
Collapse
Affiliation(s)
- Ning Gao
- Experimental Center of Forestry in North China, Chinese Academy of Forestry, Beijing, China.,State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Chengdong Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Ziying Hu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Miaomiao Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jingjing Wei
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yongming Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Industrial Microorganisms, Shanghai, China
| | - Huihui Liu
- Experimental Center of Forestry in North China, Chinese Academy of Forestry, Beijing, China
| |
Collapse
|
18
|
Qi T, Wu F, Xie Y, Gao S, Li M, Pu J, Li D, Lan F, Wang Y. Base Editing Mediated Generation of Point Mutations Into Human Pluripotent Stem Cells for Modeling Disease. Front Cell Dev Biol 2020. [PMID: 33102492 DOI: 10.3389/fcell.2020.590581.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are a powerful platform for disease modeling and drug discovery. However, the introduction of known pathogenic mutations into hPSCs is a time-consuming and labor-intensive process. Base editing is a newly developed technology that enables facile introduction of point mutations into specific loci within the genome of living cells. Here, we design an all-in-one episomal vector that expresses a single guide RNA (sgRNA) with an adenine base editor (ABE) or a cytosine base editor (CBE). Both ABE and CBE can efficiently introduce mutations into cells, A-to-G and C-to-T, respectively. We introduce disease-specific mutations of long QT syndrome into hPSCs to model LQT1, LQT2, and LQT3. Electrophysiological analysis of hPSC-derived cardiomyocytes (hPSC-CMs) using multi-electrode arrays (MEAs) reveals that edited hPSC-CMs display significant increases in duration of the action potential. Finally, we introduce the novel Brugada syndrome-associated mutation into hPSCs, demonstrating that this mutation can cause abnormal electrophysiology. Our study demonstrates that episomal encoded base editors (epi-BEs) can efficiently generate mutation-specific disease hPSC models.
Collapse
Affiliation(s)
- Tao Qi
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fujian Wu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuquan Xie
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Siqi Gao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Miaomiao Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Feng Lan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongming Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Industrial Microorganisms, Shanghai, China
| |
Collapse
|
19
|
Qi T, Wu F, Xie Y, Gao S, Li M, Pu J, Li D, Lan F, Wang Y. Base Editing Mediated Generation of Point Mutations Into Human Pluripotent Stem Cells for Modeling Disease. Front Cell Dev Biol 2020; 8:590581. [PMID: 33102492 PMCID: PMC7546412 DOI: 10.3389/fcell.2020.590581] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 09/08/2020] [Indexed: 12/19/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are a powerful platform for disease modeling and drug discovery. However, the introduction of known pathogenic mutations into hPSCs is a time-consuming and labor-intensive process. Base editing is a newly developed technology that enables facile introduction of point mutations into specific loci within the genome of living cells. Here, we design an all-in-one episomal vector that expresses a single guide RNA (sgRNA) with an adenine base editor (ABE) or a cytosine base editor (CBE). Both ABE and CBE can efficiently introduce mutations into cells, A-to-G and C-to-T, respectively. We introduce disease-specific mutations of long QT syndrome into hPSCs to model LQT1, LQT2, and LQT3. Electrophysiological analysis of hPSC-derived cardiomyocytes (hPSC-CMs) using multi-electrode arrays (MEAs) reveals that edited hPSC-CMs display significant increases in duration of the action potential. Finally, we introduce the novel Brugada syndrome-associated mutation into hPSCs, demonstrating that this mutation can cause abnormal electrophysiology. Our study demonstrates that episomal encoded base editors (epi-BEs) can efficiently generate mutation-specific disease hPSC models.
Collapse
Affiliation(s)
- Tao Qi
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fujian Wu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuquan Xie
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Siqi Gao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Miaomiao Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Feng Lan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongming Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Industrial Microorganisms, Shanghai, China
| |
Collapse
|
20
|
Mehrjardi NZ, Molcanyi M, Hatay FF, Timmer M, Shahbazi E, Ackermann JP, Herms S, Heilmann-Heimbach S, Wunderlich TF, Prochnow N, Haghikia A, Lampert A, Hescheler J, Neugebauer EAM, Baharvand H, Šarić T. Acquisition of chromosome 1q duplication in parental and genome-edited human-induced pluripotent stem cell-derived neural stem cells results in their higher proliferation rate in vitro and in vivo. Cell Prolif 2020; 53:e12892. [PMID: 32918782 PMCID: PMC7574866 DOI: 10.1111/cpr.12892] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/16/2020] [Accepted: 07/18/2020] [Indexed: 02/06/2023] Open
Abstract
Objectives Genetic engineering of human‐induced pluripotent stem cell‐derived neural stem cells (hiPSC‐NSC) may increase the risk of genomic aberrations. Therefore, we asked whether genetic modification of hiPSC‐NSCs exacerbates chromosomal abnormalities that may occur during passaging and whether they may cause any functional perturbations in NSCs in vitro and in vivo. Materials and Methods The transgenic cassette was inserted into the AAVS1 locus, and the genetic integrity of zinc‐finger nuclease (ZFN)‐modified hiPSC‐NSCs was assessed by the SNP‐based karyotyping. The hiPSC‐NSC proliferation was assessed in vitro by the EdU incorporation assay and in vivo by staining of brain slices with Ki‐67 antibody at 2 and 8 weeks after transplantation of ZFN‐NSCs with and without chromosomal aberration into the striatum of immunodeficient rats. Results During early passages, no chromosomal abnormalities were detected in unmodified or ZFN‐modified hiPSC‐NSCs. However, at higher passages both cell populations acquired duplication of the entire long arm of chromosome 1, dup(1)q. ZNF‐NSCs carrying dup(1)q exhibited higher proliferation rate than karyotypically intact cells, which was partly mediated by increased expression of AKT3 located on Chr1q. Compared to karyotypically normal ZNF‐NSCs, cells with dup(1)q also exhibited increased proliferation in vivo 2 weeks, but not 2 months, after transplantation. Conclusions These results demonstrate that, independently of ZFN‐editing, hiPSC‐NSCs have a propensity for acquiring dup(1)q and this aberration results in increased proliferation which might compromise downstream hiPSC‐NSC applications.
Collapse
Affiliation(s)
- Narges Zare Mehrjardi
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Marek Molcanyi
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Firuze Fulya Hatay
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Marco Timmer
- Department of Neurosurgery, University Hospital Cologne, Cologne, Germany
| | - Ebrahim Shahbazi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Justus P Ackermann
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Stefan Herms
- Department of Genomics, Life & Brain Center, Institute for Human Genetics, University of Bonn, Bonn, Germany.,Department of Biomedicine, Medical Genetics, Research Group Genomics, University Hospital Basel, Basel, Switzerland
| | - Stefanie Heilmann-Heimbach
- Department of Genomics, Life & Brain Center, Institute for Human Genetics, University of Bonn, Bonn, Germany
| | - Thomas F Wunderlich
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Max Planck Institute for Metabolism Research and Institute for Genetics, University of Cologne, Cologne, Germany.,Cologne Cluster of Excellence in Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Nora Prochnow
- Clinic for Neurology, St. Josef-Hospital, Clinic of the Ruhr-University Bochum, Bochum, Germany
| | - Aiden Haghikia
- Clinic for Neurology, St. Josef-Hospital, Clinic of the Ruhr-University Bochum, Bochum, Germany
| | - Angelika Lampert
- Institute of Physiology, Uniklinik, RWTH Aachen University, Aachen, Germany
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Edmund A M Neugebauer
- Medizinische Hochschule Brandenburg Theodor Fontane, Campus Neuruppin, Neuruppin, Germany
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Tomo Šarić
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| |
Collapse
|
21
|
Choi JS, Jeong IS, Park YJ, Kim SW. HGF and IL-10 expressing ALB::GFP reporter cells generated from iPSCs show robust anti-fibrotic property in acute fibrotic liver model. Stem Cell Res Ther 2020; 11:332. [PMID: 32746905 PMCID: PMC7398392 DOI: 10.1186/s13287-020-01745-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/05/2020] [Accepted: 05/25/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cell therapy using hepatocytes derived from stem cells has been regarded as a promising alternate to liver transplantation. However, the heterogeneity of these hepatocytes makes them unsuitable for therapeutic use. To overcome this limitation, we generated homogenous hepatocyte like induced hepatocyte-like (iHep) cells. METHODS iHep cells were generated from induced pluripotent stem cells (iPSCs) integrated with the albumin (ALB) reporter gene. The therapeutic properties of these iHep cells were investigated after transplantation in fibrotic liver tissues of a mouse model. RESULTS The iHep cells expressed hepatocyte specific genes and proteins, and exhibited high levels of hepatocyte growth factor (HGF) and interleukin (IL)-10 expressions. Transplantation of iHep cells significantly decreased thioacetamide (TAA)-induced liver fibrosis, apoptotic cells in the liver, and ameliorated abnormal liver function. Liver tissues engrafted with iHep cells exhibited decreased expression of pro-inflammatory factors such as transforming growth factor (TGF)-β, IL-6, and monocyte chemo attractant protein (MCP)-1. Furthermore, an increased number of proliferating hepatocytes and human albumin-expressing iHep cells were detected in mice liver. CONCLUSIONS This study has investigated and proven the liver regeneration potential of genome-edited iHep cells and promises to be a strong foundation for further studies exploring cell therapy as an alternative therapeutic option for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Ja Sung Choi
- Department of Internal Medicine, Catholic Kwandong University College of Medicine, International St. Mary's Hospital, Incheon, Republic of Korea
| | - In Sil Jeong
- Department Medicine, Catholic Kwandong University College of Medicine, Gangneung, Republic of Korea
| | - Young-Jin Park
- Department of Family Medicine, Dong-A University College of Medicine, Dong-A University Medical Center, Busan, Republic of Korea
| | - Sung-Whan Kim
- Department Medicine, Catholic Kwandong University College of Medicine, Gangneung, Republic of Korea. .,International St. Mary's Hospital, 25, Simgok-ro 100 beon-gil, Seo-gu, Incheon, 404-190, South Korea.
| |
Collapse
|
22
|
Ashmore-Harris C, Iafrate M, Saleem A, Fruhwirth GO. Non-invasive Reporter Gene Imaging of Cell Therapies, including T Cells and Stem Cells. Mol Ther 2020; 28:1392-1416. [PMID: 32243834 PMCID: PMC7264441 DOI: 10.1016/j.ymthe.2020.03.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/15/2020] [Accepted: 03/18/2020] [Indexed: 12/14/2022] Open
Abstract
Cell therapies represent a rapidly emerging class of new therapeutics. They are intended and developed for the treatment of some of the most prevalent human diseases, including cancer, diabetes, and for regenerative medicine. Currently, they are largely developed without precise assessment of their in vivo distribution, efficacy, or survival either clinically or preclinically. However, it would be highly beneficial for both preclinical cell therapy development and subsequent clinical use to assess these parameters in situ to enable enhancements in efficacy, applicability, and safety. Molecular imaging can be exploited to track cells non-invasively on the whole-body level and can enable monitoring for prolonged periods in a manner compatible with rapidly expanding cell types. In this review, we explain how in vivo imaging can aid the development and clinical translation of cell-based therapeutics. We describe the underlying principles governing non-invasive in vivo long-term cell tracking in the preclinical and clinical settings, including available imaging technologies, reporter genes, and imaging agents as well as pitfalls related to experimental design. Our emphasis is on adoptively transferred T cell and stem cell therapies.
Collapse
Affiliation(s)
- Candice Ashmore-Harris
- Imaging Therapy and Cancer Group, Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK; Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, King's College London, London SE1 9RT, UK
| | - Madeleine Iafrate
- Imaging Therapy and Cancer Group, Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Adeel Saleem
- Imaging Therapy and Cancer Group, Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK; Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London SE1 9RT, UK; Department of Haematological Medicine, King's College Hospital, London SE5 9RS, UK
| | - Gilbert O Fruhwirth
- Imaging Therapy and Cancer Group, Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK.
| |
Collapse
|
23
|
Wang B, Wang Z, Wang D, Zhang B, Ong SG, Li M, Yu W, Wang Y. krCRISPR: an easy and efficient strategy for generating conditional knockout of essential genes in cells. J Biol Eng 2019; 13:35. [PMID: 31049076 PMCID: PMC6480908 DOI: 10.1186/s13036-019-0150-y] [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] [Received: 12/27/2018] [Accepted: 02/15/2019] [Indexed: 02/06/2023] Open
Abstract
Background CRISPR/Cas9 system is a powerful tool for knocking out genes in cells. However, genes essential for cell survival cannot be directly knocked out. Traditionally, generation of conditional knockout cells requires multiple steps. Results In this study, we developed an easy and efficient strategy to generate conditional knockout cells by using double episomal vectors – one which expresses gRNA and Cas9 nuclease, and the other expresses an inducible rescue gene. Using this system which we named “krCRISPR” (knockout-rescue CRISPR), we showed that essential genes, HDAC3 and DNMT1, can be efficiently knocked out. When cells reach a desired confluency, the exogenous rescue genes can be silenced by the addition of doxycycline. Furthermore, the krCRISPR system enabled us to study the effects of the essential gene mutations on cells. We showed that the P507L mutation in DNMT1 led to downregulation of global DNA methylation in cells, indicating that it is a disease-causing mutation. Conclusions The krCRISPR system offers an easy and efficient platform that facilitates the study of essential genes’ function. Electronic supplementary material The online version of this article (10.1186/s13036-019-0150-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bei Wang
- 1MOE Key Laboratory of Contemporary Anthropology at School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai, 200438 China
| | - Zishi Wang
- 1MOE Key Laboratory of Contemporary Anthropology at School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai, 200438 China
| | - Daqi Wang
- 1MOE Key Laboratory of Contemporary Anthropology at School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai, 200438 China
| | - Baolong Zhang
- 2Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508 China
| | - Sang-Ging Ong
- 3Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612 USA.,4Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL 60612 USA
| | - Mingqing Li
- 5The Key Lab of Reproduction Regulation of NPFPC in SIPPR, Institute of Reproduction & Development in Obstetrics & Gynecology Hospital, Fudan University, Shanghai, 200011 China
| | - Wenqiang Yu
- 2Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508 China
| | - Yongming Wang
- 1MOE Key Laboratory of Contemporary Anthropology at School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai, 200438 China
| |
Collapse
|
24
|
Affiliation(s)
- Timon Seeger
- From the Stanford Cardiovascular Institute, CA (T.S., J.C.W.); Department of Medicine, Division of Cardiology (T.S., J.S.W.); Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine (M.P.); and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.P., J.C.W.)
| | - Matthew Porteus
- From the Stanford Cardiovascular Institute, CA (T.S., J.C.W.); Department of Medicine, Division of Cardiology (T.S., J.S.W.); Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine (M.P.); and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.P., J.C.W.)
| | - Joseph C Wu
- From the Stanford Cardiovascular Institute, CA (T.S., J.C.W.); Department of Medicine, Division of Cardiology (T.S., J.S.W.); Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine (M.P.); and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.P., J.C.W.).
| |
Collapse
|
25
|
Dubois VP, Zotova D, Parkins KM, Swick C, Hamilton AM, Kelly JJ, Ronald JA. Safe Harbor Targeted CRISPR-Cas9 Tools for Molecular-Genetic Imaging of Cells in Living Subjects. CRISPR J 2018; 1:440-449. [PMID: 31021241 DOI: 10.1089/crispr.2018.0030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Noninvasive molecular-genetic imaging of cells expressing imaging reporter genes is an invaluable approach for longitudinal monitoring of the biodistribution and viability of cancer cells and cell-based therapies in preclinical models and patients. However, labeling cells with reporter genes often relies on using gene transfer methods that randomly integrate the reporter genes into the genome, which may cause unwanted and serious detrimental effects. To overcome this, we have developed CRISPR-Cas9 tools to edit cells at the adeno-associated virus site 1 (AAVS1) safe harbour with a large donor construct (∼6.3 kilobases) encoding an antibiotic resistance gene and reporter genes for bioluminescence (BLI) and fluorescence imaging. HEK293T cells were transfected with a dual plasmid system encoding the Cas9 endonuclease and an AAVS1-targeted guide RNA in one plasmid, and a donor plasmid encoding a puromycin resistance gene, tdTomato and firefly luciferase flanked by AAVS1 homology arms. Puromycin-resistant clonal cells were isolated and AAVS1 integration was confirmed via PCR and sequencing of the PCR product. In vitro BLI signal correlated well to cell number (R2 = 0.9988; p < 0.05) and was stable over multiple passages. Engineered cells (2.5 × 106) were injected into the left hind flank of nude mice and in vivo BLI was performed on days 0, 7, 14, 21, and 28. BLI signal trended down from day 0 to day 7, but significantly increased by day 28 due to cell growth (p < 0.05). This describes the first CRISPR-Cas9 system for AAVS1 integration of large gene constructs for molecular-genetic imaging of cells in vivo. With further development, including improving editing efficiency, use of clinically relevant reporters, and evaluation in other cell populations that can be readily expanded in culture (e.g., immortalized cells or T cells), this CRISPR-Cas9 reporter gene system could be broadly applied to a number of in vivo cell tracking studies.
Collapse
Affiliation(s)
- Veronica P Dubois
- 1 Department of Medical Biophysics, Western University , London, Ontario, Canada.,2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada
| | - Darya Zotova
- 2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada
| | - Katie M Parkins
- 1 Department of Medical Biophysics, Western University , London, Ontario, Canada
| | - Connor Swick
- 2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada
| | - Amanda M Hamilton
- 2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada
| | - John J Kelly
- 2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada
| | - John A Ronald
- 1 Department of Medical Biophysics, Western University , London, Ontario, Canada.,2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada.,3 Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
26
|
Hamilton AM, Foster PJ, Ronald JA. Evaluating Nonintegrating Lentiviruses as Safe Vectors for Noninvasive Reporter-Based Molecular Imaging of Multipotent Mesenchymal Stem Cells. Hum Gene Ther 2018; 29:1213-1225. [DOI: 10.1089/hum.2018.111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Amanda M. Hamilton
- Imaging Research Laboratories, Robarts Research Institute, London, Canada
| | - Paula J. Foster
- Imaging Research Laboratories, Robarts Research Institute, London, Canada
- Medical Biophysics, University of Western Ontario, London, Canada
| | - John A. Ronald
- Imaging Research Laboratories, Robarts Research Institute, London, Canada
- Medical Biophysics, University of Western Ontario, London, Canada
- Lawson Health Research Institute, London, Canada
| |
Collapse
|
27
|
Nguyen PK, Neofytou E, Rhee JW, Wu JC. Potential Strategies to Address the Major Clinical Barriers Facing Stem Cell Regenerative Therapy for Cardiovascular Disease: A Review. JAMA Cardiol 2018; 1:953-962. [PMID: 27579998 DOI: 10.1001/jamacardio.2016.2750] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Importance Although progress continues to be made in the field of stem cell regenerative medicine for the treatment of cardiovascular disease, significant barriers to clinical implementation still exist. Objectives To summarize the current barriers to the clinical implementation of stem cell therapy in patients with cardiovascular disease and to discuss potential strategies to overcome them. Evidence Review Information for this review was obtained through a search of PubMed and the Cochrane database for English-language studies published between January 1, 2000, and July 25, 2016. Ten randomized clinical trials and 8 systematic reviews were included. Findings One of the major clinical barriers facing the routine implementation of stem cell therapy in patients with cardiovascular disease is the limited and inconsistent benefit observed thus far. Reasons for this finding are unclear but may be owing to poor cell retention and survival, as suggested by numerous preclinical studies and a small number of human studies incorporating imaging to determine cell fate. Additional studies in humans using imaging to determine cell fate are needed to understand how these factors contribute to the limited efficacy of stem cell therapy. Treatment strategies to address poor cell retention and survival are under investigation and include the following: coadministration of immunosuppressive and prosurvival agents, delivery of cardioprotective factors packaged in exosomes rather than the cells themselves, and use of tissue-engineering strategies to provide structural support for cells. If larger grafts are achieved using these strategies, it will be imperative to carefully monitor for the potential risks of tumorigenicity, immunogenicity, and arrhythmogenicity. Conclusions and Relevance Despite important achievements to date, stem cell therapy is not yet ready for routine clinical implementation. Significant research is still needed to address the clinical barriers outlined herein before the next wave of large clinical trials is under way.
Collapse
Affiliation(s)
- Patricia K Nguyen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California2Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California3Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Evgenios Neofytou
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California2Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - June-Wha Rhee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California2Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California2Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California4Department of Radiology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
28
|
Hu S, Zhao MT, Jahanbani F, Shao NY, Lee WH, Chen H, Snyder MP, Wu JC. Effects of cellular origin on differentiation of human induced pluripotent stem cell-derived endothelial cells. JCI Insight 2018; 1:85558. [PMID: 27398408 DOI: 10.1172/jci.insight.85558] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Human induced pluripotent stem cells (iPSCs) can be derived from various types of somatic cells by transient overexpression of 4 Yamanaka factors (OCT4, SOX2, C-MYC, and KLF4). Patient-specific iPSC derivatives (e.g., neuronal, cardiac, hepatic, muscular, and endothelial cells [ECs]) hold great promise in drug discovery and regenerative medicine. In this study, we aimed to evaluate whether the cellular origin can affect the differentiation, in vivo behavior, and single-cell gene expression signatures of human iPSC-derived ECs. We derived human iPSCs from 3 types of somatic cells of the same individuals: fibroblasts (FB-iPSCs), ECs (EC-iPSCs), and cardiac progenitor cells (CPC-iPSCs). We then differentiated them into ECs by sequential administration of Activin, BMP4, bFGF, and VEGF. EC-iPSCs at early passage (10 < P < 20) showed higher EC differentiation propensity and gene expression of EC-specific markers (PECAM1 and NOS3) than FB-iPSCs and CPC-iPSCs. In vivo transplanted EC-iPSC-ECs were recovered with a higher percentage of CD31+ population and expressed higher EC-specific gene expression markers (PECAM1, KDR, and ICAM) as revealed by microfluidic single-cell quantitative PCR (qPCR). In vitro EC-iPSC-ECs maintained a higher CD31+ population than FB-iPSC-ECs and CPC-iPSC-ECs with long-term culturing and passaging. These results indicate that cellular origin may influence lineage differentiation propensity of human iPSCs; hence, the somatic memory carried by early passage iPSCs should be carefully considered before clinical translation.
Collapse
Affiliation(s)
- Shijun Hu
- Stanford Cardiovascular Institute.,Department of Medicine, Division of Cardiology, and.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA.,Institute for Cardiovascular Science, Soochow University & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou, Jiangsu, China
| | - Ming-Tao Zhao
- Stanford Cardiovascular Institute.,Department of Medicine, Division of Cardiology, and.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Fereshteh Jahanbani
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Ning-Yi Shao
- Stanford Cardiovascular Institute.,Department of Medicine, Division of Cardiology, and.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Won Hee Lee
- Stanford Cardiovascular Institute.,Department of Medicine, Division of Cardiology, and.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Haodong Chen
- Stanford Cardiovascular Institute.,Department of Medicine, Division of Cardiology, and.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute.,Department of Medicine, Division of Cardiology, and.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
29
|
Miyagawa S, Sawa Y. Building a new strategy for treating heart failure using Induced Pluripotent Stem Cells. J Cardiol 2018; 72:445-448. [PMID: 30172684 DOI: 10.1016/j.jjcc.2018.05.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 05/01/2018] [Indexed: 12/31/2022]
Abstract
Although cell therapy using myoblasts, bone marrow cells, or other stem cells appears to improve functional recovery of the failing heart, mainly by cytokine paracrine effects, its effectiveness in severely damaged myocardium is limited, probably because there are too few residual myocytes to promote cytokine-induced angiogenesis. Recently, cardiogenic stem cells, such as c-kit-positive cells, were reported to generate cardiomyogenic lineages, and basic research experiments showed that implanting these cells, which can differentiate into cardiomyocytes, improves heart function. However, this functional recovery may have also mainly depended on cytokine paracrine effects, because the differentiation to cardiomyocytes in vivo was poor. In contrast, while Induced Pluripotent Stem Cell-derived cardiomyocytes have paracrine effects, they also have the potential to supply newly born myocytes that can function synchronously with the recipient myocardium as "mechanically working cells" in severely damaged myocardium. Thus, they could represent a "true" myocardial regeneration therapy that can actually regenerate severely damaged myocardium. In addition, iPS cells, especially disease-specific iPS cells, have other applications in regenerative medicine such as in drug screening. In this report, we present the state of basic research in the field of cardiac iPS cells.
Collapse
Affiliation(s)
- Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
30
|
Neyrinck K, Breuls N, Holvoet B, Oosterlinck W, Wolfs E, Vanbilloen H, Gheysens O, Duelen R, Gsell W, Lambrichts I, Himmelreich U, Verfaillie CM, Sampaolesi M, Deroose CM. The human somatostatin receptor type 2 as an imaging and suicide reporter gene for pluripotent stem cell-derived therapy of myocardial infarction. Am J Cancer Res 2018; 8:2799-2813. [PMID: 29774076 PMCID: PMC5957010 DOI: 10.7150/thno.22980] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 02/28/2018] [Indexed: 12/12/2022] Open
Abstract
Rationale: Pluripotent stem cells (PSCs) are being investigated as a cell source for regenerative medicine since they provide an infinitive pool of cells that are able to differentiate towards every cell type of the body. One possible therapeutic application involves the use of these cells to treat myocardial infarction (MI), a condition where billions of cardiomyocytes (CMs) are lost. Although several protocols have been developed to differentiate PSCs towards CMs, none of these provide a completely pure population, thereby still posing a risk for neoplastic teratoma formation. Therefore, we developed a strategy to (i) monitor cell behavior noninvasively via site-specific integration of firefly luciferase (Fluc) and the human positron emission tomography (PET) imaging reporter genes, sodium iodide symporter (hNIS) and somatostatin receptor type 2 (hSSTr2), and (ii) perform hSSTr2-mediated suicide gene therapy via the clinically used radiopharmacon 177Lu-DOTATATE. Methods: Human embryonic stem cells (ESCs) were gene-edited via zinc finger nucleases to express Fluc and either hNIS or hSSTr2 in the safe harbor locus, adeno-associated virus integration site 1. Firstly, these cells were exposed to 4.8 MBq 177Lu-DOTATATE in vitro and cell survival was monitored via bioluminescence imaging (BLI). Afterwards, hNIS+ and hSSTr2+ ESCs were transplanted subcutaneously and teratomas were allowed to form. At day 59, baseline 124I and 68Ga-DOTATATE PET and BLI scans were performed. The day after, animals received either saline or 55 MBq 177Lu-DOTATATE. Weekly BLI scans were performed, accompanied by 124I and 68Ga-DOTATATE PET scans at days 87 and 88, respectively. Finally, hSSTr2+ ESCs were differentiated towards CMs and transplanted intramyocardially in the border zone of an infarct that was induced by left anterior descending coronary artery ligation. After transplantation, the animals were monitored via BLI and PET, while global cardiac function was evaluated using cardiac magnetic resonance imaging. Results: Teratoma growth of both hNIS+ and hSSTr2+ ESCs could be followed noninvasively over time by both PET and BLI. After 177Lu-DOTATATE administration, successful cell killing of the hSSTr2+ ESCs was achieved both in vitro and in vivo, indicated by reductions in total tracer lesion uptake, BLI signal and teratoma volume. As undifferentiated hSSTr2+ ESCs are not therapeutically relevant, they were differentiated towards CMs and injected in immune-deficient mice with a MI. Long-term cell survival could be monitored without uncontrolled cell proliferation. However, no improvement in the left ventricular ejection fraction was observed. Conclusion: We developed isogenic hSSTr2-expressing ESCs that allow noninvasive cell monitoring in the context of PSC-derived regenerative therapy. Furthermore, we are the first to use the hSSTr2 not only as an imaging reporter gene, but also as a suicide mechanism for radionuclide therapy in the setting of PSC-derived cell treatment.
Collapse
|
31
|
Novel and innovative approaches for treatment of β-thalassemia. PEDIATRIC HEMATOLOGY ONCOLOGY JOURNAL 2017. [DOI: 10.1016/j.phoj.2017.11.153] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
32
|
Lin Y, Gil CH, Yoder MC. Differentiation, Evaluation, and Application of Human Induced Pluripotent Stem Cell-Derived Endothelial Cells. Arterioscler Thromb Vasc Biol 2017; 37:2014-2025. [PMID: 29025705 DOI: 10.1161/atvbaha.117.309962] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
The emergence of induced pluripotent stem cell (iPSC) technology paves the way to generate large numbers of patient-specific endothelial cells (ECs) that can be potentially delivered for regenerative medicine in patients with cardiovascular disease. In the last decade, numerous protocols that differentiate EC from iPSC have been developed by many groups. In this review, we will discuss several common strategies that have been optimized for human iPSC-EC differentiation and subsequent studies that have evaluated the potential of human iPSC-EC as a cell therapy or as a tool in disease modeling. In addition, we will emphasize the importance of using in vivo vessel-forming ability and in vitro clonogenic colony-forming potential as a gold standard with which to evaluate the quality of human iPSC-EC derived from various protocols.
Collapse
Affiliation(s)
- Yang Lin
- From the Department of Pediatrics, Herman B. Wells Center for Pediatric Research (Y.L., C.-H.G., M.C.Y.) and Department of Biochemistry and Molecular Biology (Y.L., M.C.Y.), Indiana University School of Medicine, Indianapolis
| | - Chang-Hyun Gil
- From the Department of Pediatrics, Herman B. Wells Center for Pediatric Research (Y.L., C.-H.G., M.C.Y.) and Department of Biochemistry and Molecular Biology (Y.L., M.C.Y.), Indiana University School of Medicine, Indianapolis
| | - Mervin C Yoder
- From the Department of Pediatrics, Herman B. Wells Center for Pediatric Research (Y.L., C.-H.G., M.C.Y.) and Department of Biochemistry and Molecular Biology (Y.L., M.C.Y.), Indiana University School of Medicine, Indianapolis.
| |
Collapse
|
33
|
Caddeo S, Boffito M, Sartori S. Tissue Engineering Approaches in the Design of Healthy and Pathological In Vitro Tissue Models. Front Bioeng Biotechnol 2017; 5:40. [PMID: 28798911 PMCID: PMC5526851 DOI: 10.3389/fbioe.2017.00040] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 06/26/2017] [Indexed: 12/16/2022] Open
Abstract
In the tissue engineering (TE) paradigm, engineering and life sciences tools are combined to develop bioartificial substitutes for organs and tissues, which can in turn be applied in regenerative medicine, pharmaceutical, diagnostic, and basic research to elucidate fundamental aspects of cell functions in vivo or to identify mechanisms involved in aging processes and disease onset and progression. The complex three-dimensional (3D) microenvironment in which cells are organized in vivo allows the interaction between different cell types and between cells and the extracellular matrix, the composition of which varies as a function of the tissue, the degree of maturation, and health conditions. In this context, 3D in vitro models can more realistically reproduce a tissue or organ than two-dimensional (2D) models. Moreover, they can overcome the limitations of animal models and reduce the need for in vivo tests, according to the "3Rs" guiding principles for a more ethical research. The design of 3D engineered tissue models is currently in its development stage, showing high potential in overcoming the limitations of already available models. However, many issues are still opened, concerning the identification of the optimal scaffold-forming materials, cell source and biofabrication technology, and the best cell culture conditions (biochemical and physical cues) to finely replicate the native tissue and the surrounding environment. In the near future, 3D tissue-engineered models are expected to become useful tools in the preliminary testing and screening of drugs and therapies and in the investigation of the molecular mechanisms underpinning disease onset and progression. In this review, the application of TE principles to the design of in vitro 3D models will be surveyed, with a focus on the strengths and weaknesses of this emerging approach. In addition, a brief overview on the development of in vitro models of healthy and pathological bone, heart, pancreas, and liver will be presented.
Collapse
Affiliation(s)
- Silvia Caddeo
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam, Amsterdam, Netherlands
| | - Monica Boffito
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Susanna Sartori
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| |
Collapse
|
34
|
Tachibana A, Santoso MR, Mahmoudi M, Shukla P, Wang L, Bennett M, Goldstone AB, Wang M, Fukushi M, Ebert AD, Woo YJ, Rulifson E, Yang PC. Paracrine Effects of the Pluripotent Stem Cell-Derived Cardiac Myocytes Salvage the Injured Myocardium. Circ Res 2017; 121:e22-e36. [PMID: 28743804 DOI: 10.1161/circresaha.117.310803] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 07/20/2017] [Accepted: 07/24/2017] [Indexed: 01/06/2023]
Abstract
RATIONALE Cardiac myocytes derived from pluripotent stem cells have demonstrated the potential to mitigate damage of the infarcted myocardium and improve left ventricular ejection fraction. However, the mechanism underlying the functional benefit is unclear. OBJECTIVE To evaluate whether the transplantation of cardiac-lineage differentiated derivatives enhance myocardial viability and restore left ventricular ejection fraction more effectively than undifferentiated pluripotent stem cells after a myocardial injury. Herein, we utilize novel multimodality evaluation of human embryonic stem cells (hESCs), hESC-derived cardiac myocytes (hCMs), human induced pluripotent stem cells (iPSCs), and iPSC-derived cardiac myocytes (iCMs) in a murine myocardial injury model. METHODS AND RESULTS Permanent ligation of the left anterior descending coronary artery was induced in immunosuppressed mice. Intramyocardial injection was performed with (1) hESCs (n=9), (2) iPSCs (n=8), (3) hCMs (n=9), (4) iCMs (n=14), and (5) PBS control (n=10). Left ventricular ejection fraction and myocardial viability, measured by cardiac magnetic resonance imaging and manganese-enhanced magnetic resonance imaging, respectively, was significantly improved in hCM- and iCM-treated mice compared with pluripotent stem cell- or control-treated mice. Bioluminescence imaging revealed limited cell engraftment in all treated groups, suggesting that the cell secretions may underlie the repair mechanism. To determine the paracrine effects of the transplanted cells, cytokines from supernatants from all groups were assessed in vitro. Gene expression and immunohistochemistry analyses of the murine myocardium demonstrated significant upregulation of the promigratory, proangiogenic, and antiapoptotic targets in groups treated with cardiac lineage cells compared with pluripotent stem cell and control groups. CONCLUSIONS This study demonstrates that the cardiac phenotype of hCMs and iCMs salvages the injured myocardium effectively than undifferentiated stem cells through their differential paracrine effects.
Collapse
Affiliation(s)
- Atsushi Tachibana
- From the Division of Cardiovascular Medicine (A.T., M.R.S., M.M., P.S., L.W., M.W., A.D.E., E.R., P.C.Y.), Division of Neonatal and Developmental Medicine (M.B.), and Department of Cardiothoracic Surgery (A.B.G., Y.J.W.), Stanford University, CA; Department of Radiological Sciences, Tokyo Metropolitan University, Japan (A.T., M.F.); Department of Critical Care Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, China (L.W.); Department of Cardiology and Pneumonology, Göttingen University Medical Center, Germany (A.D.E.); and German Center for Cardiovascular Research, Partner Site Göttingen, Germany (A.D.E.)
| | - Michelle R Santoso
- From the Division of Cardiovascular Medicine (A.T., M.R.S., M.M., P.S., L.W., M.W., A.D.E., E.R., P.C.Y.), Division of Neonatal and Developmental Medicine (M.B.), and Department of Cardiothoracic Surgery (A.B.G., Y.J.W.), Stanford University, CA; Department of Radiological Sciences, Tokyo Metropolitan University, Japan (A.T., M.F.); Department of Critical Care Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, China (L.W.); Department of Cardiology and Pneumonology, Göttingen University Medical Center, Germany (A.D.E.); and German Center for Cardiovascular Research, Partner Site Göttingen, Germany (A.D.E.)
| | - Morteza Mahmoudi
- From the Division of Cardiovascular Medicine (A.T., M.R.S., M.M., P.S., L.W., M.W., A.D.E., E.R., P.C.Y.), Division of Neonatal and Developmental Medicine (M.B.), and Department of Cardiothoracic Surgery (A.B.G., Y.J.W.), Stanford University, CA; Department of Radiological Sciences, Tokyo Metropolitan University, Japan (A.T., M.F.); Department of Critical Care Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, China (L.W.); Department of Cardiology and Pneumonology, Göttingen University Medical Center, Germany (A.D.E.); and German Center for Cardiovascular Research, Partner Site Göttingen, Germany (A.D.E.)
| | - Praveen Shukla
- From the Division of Cardiovascular Medicine (A.T., M.R.S., M.M., P.S., L.W., M.W., A.D.E., E.R., P.C.Y.), Division of Neonatal and Developmental Medicine (M.B.), and Department of Cardiothoracic Surgery (A.B.G., Y.J.W.), Stanford University, CA; Department of Radiological Sciences, Tokyo Metropolitan University, Japan (A.T., M.F.); Department of Critical Care Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, China (L.W.); Department of Cardiology and Pneumonology, Göttingen University Medical Center, Germany (A.D.E.); and German Center for Cardiovascular Research, Partner Site Göttingen, Germany (A.D.E.)
| | - Lei Wang
- From the Division of Cardiovascular Medicine (A.T., M.R.S., M.M., P.S., L.W., M.W., A.D.E., E.R., P.C.Y.), Division of Neonatal and Developmental Medicine (M.B.), and Department of Cardiothoracic Surgery (A.B.G., Y.J.W.), Stanford University, CA; Department of Radiological Sciences, Tokyo Metropolitan University, Japan (A.T., M.F.); Department of Critical Care Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, China (L.W.); Department of Cardiology and Pneumonology, Göttingen University Medical Center, Germany (A.D.E.); and German Center for Cardiovascular Research, Partner Site Göttingen, Germany (A.D.E.)
| | - Mihoko Bennett
- From the Division of Cardiovascular Medicine (A.T., M.R.S., M.M., P.S., L.W., M.W., A.D.E., E.R., P.C.Y.), Division of Neonatal and Developmental Medicine (M.B.), and Department of Cardiothoracic Surgery (A.B.G., Y.J.W.), Stanford University, CA; Department of Radiological Sciences, Tokyo Metropolitan University, Japan (A.T., M.F.); Department of Critical Care Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, China (L.W.); Department of Cardiology and Pneumonology, Göttingen University Medical Center, Germany (A.D.E.); and German Center for Cardiovascular Research, Partner Site Göttingen, Germany (A.D.E.)
| | - Andrew B Goldstone
- From the Division of Cardiovascular Medicine (A.T., M.R.S., M.M., P.S., L.W., M.W., A.D.E., E.R., P.C.Y.), Division of Neonatal and Developmental Medicine (M.B.), and Department of Cardiothoracic Surgery (A.B.G., Y.J.W.), Stanford University, CA; Department of Radiological Sciences, Tokyo Metropolitan University, Japan (A.T., M.F.); Department of Critical Care Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, China (L.W.); Department of Cardiology and Pneumonology, Göttingen University Medical Center, Germany (A.D.E.); and German Center for Cardiovascular Research, Partner Site Göttingen, Germany (A.D.E.)
| | - Mouer Wang
- From the Division of Cardiovascular Medicine (A.T., M.R.S., M.M., P.S., L.W., M.W., A.D.E., E.R., P.C.Y.), Division of Neonatal and Developmental Medicine (M.B.), and Department of Cardiothoracic Surgery (A.B.G., Y.J.W.), Stanford University, CA; Department of Radiological Sciences, Tokyo Metropolitan University, Japan (A.T., M.F.); Department of Critical Care Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, China (L.W.); Department of Cardiology and Pneumonology, Göttingen University Medical Center, Germany (A.D.E.); and German Center for Cardiovascular Research, Partner Site Göttingen, Germany (A.D.E.)
| | - Masahiro Fukushi
- From the Division of Cardiovascular Medicine (A.T., M.R.S., M.M., P.S., L.W., M.W., A.D.E., E.R., P.C.Y.), Division of Neonatal and Developmental Medicine (M.B.), and Department of Cardiothoracic Surgery (A.B.G., Y.J.W.), Stanford University, CA; Department of Radiological Sciences, Tokyo Metropolitan University, Japan (A.T., M.F.); Department of Critical Care Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, China (L.W.); Department of Cardiology and Pneumonology, Göttingen University Medical Center, Germany (A.D.E.); and German Center for Cardiovascular Research, Partner Site Göttingen, Germany (A.D.E.)
| | - Antje D Ebert
- From the Division of Cardiovascular Medicine (A.T., M.R.S., M.M., P.S., L.W., M.W., A.D.E., E.R., P.C.Y.), Division of Neonatal and Developmental Medicine (M.B.), and Department of Cardiothoracic Surgery (A.B.G., Y.J.W.), Stanford University, CA; Department of Radiological Sciences, Tokyo Metropolitan University, Japan (A.T., M.F.); Department of Critical Care Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, China (L.W.); Department of Cardiology and Pneumonology, Göttingen University Medical Center, Germany (A.D.E.); and German Center for Cardiovascular Research, Partner Site Göttingen, Germany (A.D.E.)
| | - Y Joseph Woo
- From the Division of Cardiovascular Medicine (A.T., M.R.S., M.M., P.S., L.W., M.W., A.D.E., E.R., P.C.Y.), Division of Neonatal and Developmental Medicine (M.B.), and Department of Cardiothoracic Surgery (A.B.G., Y.J.W.), Stanford University, CA; Department of Radiological Sciences, Tokyo Metropolitan University, Japan (A.T., M.F.); Department of Critical Care Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, China (L.W.); Department of Cardiology and Pneumonology, Göttingen University Medical Center, Germany (A.D.E.); and German Center for Cardiovascular Research, Partner Site Göttingen, Germany (A.D.E.)
| | - Eric Rulifson
- From the Division of Cardiovascular Medicine (A.T., M.R.S., M.M., P.S., L.W., M.W., A.D.E., E.R., P.C.Y.), Division of Neonatal and Developmental Medicine (M.B.), and Department of Cardiothoracic Surgery (A.B.G., Y.J.W.), Stanford University, CA; Department of Radiological Sciences, Tokyo Metropolitan University, Japan (A.T., M.F.); Department of Critical Care Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, China (L.W.); Department of Cardiology and Pneumonology, Göttingen University Medical Center, Germany (A.D.E.); and German Center for Cardiovascular Research, Partner Site Göttingen, Germany (A.D.E.)
| | - Phillip C Yang
- From the Division of Cardiovascular Medicine (A.T., M.R.S., M.M., P.S., L.W., M.W., A.D.E., E.R., P.C.Y.), Division of Neonatal and Developmental Medicine (M.B.), and Department of Cardiothoracic Surgery (A.B.G., Y.J.W.), Stanford University, CA; Department of Radiological Sciences, Tokyo Metropolitan University, Japan (A.T., M.F.); Department of Critical Care Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, China (L.W.); Department of Cardiology and Pneumonology, Göttingen University Medical Center, Germany (A.D.E.); and German Center for Cardiovascular Research, Partner Site Göttingen, Germany (A.D.E.).
| |
Collapse
|
35
|
Genetically encoded iron-associated proteins as MRI reporters for molecular and cellular imaging. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 10. [DOI: 10.1002/wnan.1482] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 04/18/2017] [Accepted: 05/04/2017] [Indexed: 02/06/2023]
|
36
|
Wolfs E, Holvoet B, Ordovas L, Breuls N, Helsen N, Schönberger M, Raitano S, Struys T, Vanbilloen B, Casteels C, Sampaolesi M, Van Laere K, Lambrichts I, Verfaillie CM, Deroose CM. Molecular Imaging of Human Embryonic Stem Cells Stably Expressing Human PET Reporter Genes After Zinc Finger Nuclease–Mediated Genome Editing. J Nucl Med 2017; 58:1659-1665. [DOI: 10.2967/jnumed.117.189779] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 05/01/2017] [Indexed: 11/16/2022] Open
|
37
|
Xie Y, Wang D, Lan F, Wei G, Ni T, Chai R, Liu D, Hu S, Li M, Li D, Wang H, Wang Y. An episomal vector-based CRISPR/Cas9 system for highly efficient gene knockout in human pluripotent stem cells. Sci Rep 2017; 7:2320. [PMID: 28539611 PMCID: PMC5443789 DOI: 10.1038/s41598-017-02456-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/10/2017] [Indexed: 01/28/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) represent a unique opportunity for understanding the molecular mechanisms underlying complex traits and diseases. CRISPR/Cas9 is a powerful tool to introduce genetic mutations into the hPSCs for loss-of-function studies. Here, we developed an episomal vector-based CRISPR/Cas9 system, which we called epiCRISPR, for highly efficient gene knockout in hPSCs. The epiCRISPR system enables generation of up to 100% Insertion/Deletion (indel) rates. In addition, the epiCRISPR system enables efficient double-gene knockout and genomic deletion. To minimize off-target cleavage, we combined the episomal vector technology with double-nicking strategy and recent developed high fidelity Cas9. Thus the epiCRISPR system offers a highly efficient platform for genetic analysis in hPSCs.
Collapse
Affiliation(s)
- Yifang Xie
- Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Daqi Wang
- The State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Feng Lan
- Beijing Anzhen Hospital, Beijing Insitute of Heart Lung and Blood Vessel Disease, Capital Medical University, Beijing, 100029, China
| | - Gang Wei
- The State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Ting Ni
- The State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Renjie Chai
- Co-innovation Center of Neuro regeneration, Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Dong Liu
- Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Shijun Hu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Soochow University, Soochow, 215007, China
| | - Mingqing Li
- The Key Lab of Reproduction Regulation of NPFPC in SIPPR, Institute of Reproduction & Development in Obstetrics & Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Dajin Li
- The Key Lab of Reproduction Regulation of NPFPC in SIPPR, Institute of Reproduction & Development in Obstetrics & Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Hongyan Wang
- The Key Lab of Reproduction Regulation of NPFPC in SIPPR, Institute of Reproduction & Development in Obstetrics & Gynecology Hospital, Fudan University, Shanghai, 200011, China. .,Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China.
| | - Yongming Wang
- The State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, China. .,The Key Lab of Reproduction Regulation of NPFPC in SIPPR, Institute of Reproduction & Development in Obstetrics & Gynecology Hospital, Fudan University, Shanghai, 200011, China.
| |
Collapse
|
38
|
Yang F, Liu C, Chen D, Tu M, Xie H, Sun H, Ge X, Tang L, Li J, Zheng J, Song Z, Qu J, Gu F. CRISPR/Cas9-loxP-Mediated Gene Editing as a Novel Site-Specific Genetic Manipulation Tool. MOLECULAR THERAPY-NUCLEIC ACIDS 2017. [PMID: 28624213 PMCID: PMC5429228 DOI: 10.1016/j.omtn.2017.04.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cre-loxP, as one of the site-specific genetic manipulation tools, offers a method to study the spatial and temporal regulation of gene expression/inactivation in order to decipher gene function. CRISPR/Cas9-mediated targeted genome engineering technologies are sparking a new revolution in biological research. Whether the traditional site-specific genetic manipulation tool and CRISPR/Cas9 could be combined to create a novel genetic tool for highly specific gene editing is not clear. Here, we successfully generated a CRISPR/Cas9-loxP system to perform gene editing in human cells, providing the proof of principle that these two technologies can be used together for the first time. We also showed that distinct non-homologous end-joining (NHEJ) patterns from CRISPR/Cas9-mediated gene editing of the targeting sequence locates at the level of plasmids (episomal) and chromosomes. Specially, the CRISPR/Cas9-mediated NHEJ pattern in the nuclear genome favors deletions (64%–68% at the human AAVS1 locus versus 4%–28% plasmid DNA). CRISPR/Cas9-loxP, a novel site-specific genetic manipulation tool, offers a platform for the dissection of gene function and molecular insights into DNA-repair pathways.
Collapse
Affiliation(s)
- Fayu Yang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Changbao Liu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Ding Chen
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Mengjun Tu
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Haihua Xie
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Huihui Sun
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Xianglian Ge
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Lianchao Tang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Jin Li
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Jiayong Zheng
- Department of Gynecology and Obstetrics, People's Hospital of Wenzhou, Wenzhou, Zhejiang 325000, China
| | - Zongming Song
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Jia Qu
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Feng Gu
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China.
| |
Collapse
|
39
|
May I Cut in? Gene Editing Approaches in Human Induced Pluripotent Stem Cells. Cells 2017; 6:cells6010005. [PMID: 28178187 PMCID: PMC5371870 DOI: 10.3390/cells6010005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 01/20/2017] [Accepted: 01/30/2017] [Indexed: 12/16/2022] Open
Abstract
In the decade since Yamanaka and colleagues described methods to reprogram somatic cells into a pluripotent state, human induced pluripotent stem cells (hiPSCs) have demonstrated tremendous promise in numerous disease modeling, drug discovery, and regenerative medicine applications. More recently, the development and refinement of advanced gene transduction and editing technologies have further accelerated the potential of hiPSCs. In this review, we discuss the various gene editing technologies that are being implemented with hiPSCs. Specifically, we describe the emergence of technologies including zinc-finger nuclease (ZFN), transcription activator-like effector nuclease (TALEN), and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 that can be used to edit the genome at precise locations, and discuss the strengths and weaknesses of each of these technologies. In addition, we present the current applications of these technologies in elucidating the mechanisms of human development and disease, developing novel and effective therapeutic molecules, and engineering cell-based therapies. Finally, we discuss the emerging technological advances in targeted gene editing methods.
Collapse
|
40
|
Abstract
Genome-editing tools, which include zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated 9 (Cas9) systems, have emerged as an invaluable technology to achieve somatic and germline genomic manipulation in cells and model organisms for multiple applications, including the creation of knockout alleles, introducing desired mutations into genomic DNA, and inserting novel transgenes. Genome editing is being rapidly adopted into all fields of biomedical research, including the cardiovascular field, where it has facilitated a greater understanding of lipid metabolism, electrophysiology, cardiomyopathies, and other cardiovascular disorders, has helped to create a wider variety of cellular and animal models, and has opened the door to a new class of therapies. In this Review, we discuss the applications of genome-editing technology throughout cardiovascular disease research and the prospect of in vivo genome-editing therapies in the future. We also describe some of the existing limitations of genome-editing tools that will need to be addressed if cardiovascular genome editing is to achieve its full scientific and therapeutic potential.
Collapse
|
41
|
Anisotropic engineered heart tissue made from laser-cut decellularized myocardium. Sci Rep 2016; 6:32068. [PMID: 27572147 PMCID: PMC5004193 DOI: 10.1038/srep32068] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/02/2016] [Indexed: 12/13/2022] Open
Abstract
We have developed an engineered heart tissue (EHT) system that uses laser-cut sheets of decellularized myocardium as scaffolds. This material enables formation of thin muscle strips whose biomechanical characteristics are easily measured and manipulated. To create EHTs, sections of porcine myocardium were laser-cut into ribbon-like shapes, decellularized, and mounted in specialized clips for seeding and culture. Scaffolds were first tested by seeding with neonatal rat ventricular myocytes. EHTs beat synchronously by day five and exhibited robust length-dependent activation by day 21. Fiber orientation within the scaffold affected peak twitch stress, demonstrating its ability to guide cells toward physiologic contractile anisotropy. Scaffold anisotropy also made it possible to probe cellular responses to stretch as a function of fiber angle. Stretch that was aligned with the fiber direction increased expression of brain natriuretic peptide, but off-axis stretches (causing fiber shear) did not. The method also produced robust EHTs from cardiomyocytes derived from human embryonic stem cells and induced pluripotent stem cells (hiPSC). hiPSC-EHTs achieved maximum peak stress of 6.5 mN/mm2 and twitch kinetics approaching reported values from adult human trabeculae. We conclude that laser-cut EHTs are a viable platform for novel mechanotransduction experiments and characterizing the biomechanical function of patient-derived cardiomyoctyes.
Collapse
|
42
|
Samuel R, Duda DG, Fukumura D, Jain RK. Vascular diseases await translation of blood vessels engineered from stem cells. Sci Transl Med 2016; 7:309rv6. [PMID: 26468328 DOI: 10.1126/scitranslmed.aaa1805] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The discovery of human induced pluripotent stem cells (hiPSCs) might pave the way toward a long-sought solution for obtaining sufficient numbers of autologous cells for tissue engineering. Several methods exist for generating endothelial cells or perivascular cells from hiPSCs in vitro for use in the building of vascular tissue. We discuss current developments in the generation of vascular progenitor cells from hiPSCs and the assessment of their functional capacity in vivo, opportunities and challenges for the clinical translation of engineered vascular tissue, and modeling of vascular diseases using hiPSC-derived vascular progenitor cells.
Collapse
Affiliation(s)
- Rekha Samuel
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA. Centre for Stem Cell Research, Christian Medical College, Bagayam, Vellore 632002, Tamil Nadu, India
| | - Dan G Duda
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
43
|
Bartulos O, Zhuang ZW, Huang Y, Mikush N, Suh C, Bregasi A, Wang L, Chang W, Krause DS, Young LH, Pober JS, Qyang Y. ISL1 cardiovascular progenitor cells for cardiac repair after myocardial infarction. JCI Insight 2016; 1:80920. [PMID: 27525311 DOI: 10.1172/jci.insight.80920] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular progenitor cells (CPCs) expressing the ISL1-LIM-homeodomain transcription factor contribute developmentally to cardiomyocytes in all 4 chambers of the heart. Here, we show that ISL1-CPCs can be applied to myocardial regeneration following injury. We used a rapid 3D methylcellulose approach to form murine and human ISL1-CPC spheroids that engrafted after myocardial infarction in murine hearts, where they differentiated into cardiomyocytes and endothelial cells, integrating into the myocardium and forming new blood vessels. ISL1-CPC spheroid-treated mice exhibited reduced infarct area and increased blood vessel formation compared with control animals. Moreover, left ventricular (LV) contractile function was significantly better in mice transplanted with ISL1-CPCs 4 weeks after injury than that in control animals. These results provide proof-of-concept of a cardiac repair strategy employing ISL1-CPCs that, based on our previous lineage-tracing studies, are committed to forming heart tissue, in combination with a robust methylcellulose spheroid-based delivery approach.
Collapse
Affiliation(s)
- Oscar Bartulos
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine.,Yale Stem Cell Center
| | - Zhen Wu Zhuang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine
| | - Yan Huang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine
| | - Nicole Mikush
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine
| | - Carol Suh
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine.,Yale Stem Cell Center
| | - Alda Bregasi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine
| | - Lin Wang
- Yale Stem Cell Center.,Department of Laboratory Medicine
| | - William Chang
- Department of Internal Medicine, Section of Nephrology
| | - Diane S Krause
- Yale Stem Cell Center.,Department of Laboratory Medicine.,Department of Cell Biology.,Pathology
| | - Lawrence H Young
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine.,Cellular and Molecular Physiology
| | - Jordan S Pober
- Pathology.,Immunobiology, and.,Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA
| | - Yibing Qyang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine.,Yale Stem Cell Center.,Pathology.,Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
44
|
Site-Specific Genome Engineering in Human Pluripotent Stem Cells. Int J Mol Sci 2016; 17:ijms17071000. [PMID: 27347935 PMCID: PMC4964376 DOI: 10.3390/ijms17071000] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/16/2016] [Accepted: 06/20/2016] [Indexed: 12/21/2022] Open
Abstract
The possibility to generate patient-specific induced pluripotent stem cells (iPSCs) offers an unprecedented potential of applications in clinical therapy and medical research. Human iPSCs and their differentiated derivatives are tools for diseases modelling, drug discovery, safety pharmacology, and toxicology. Moreover, they allow for the engineering of bioartificial tissue and are promising candidates for cellular therapies. For many of these applications, the ability to genetically modify pluripotent stem cells (PSCs) is indispensable, but efficient site-specific and safe technologies for genetic engineering of PSCs were developed only recently. By now, customized engineered nucleases provide excellent tools for targeted genome editing, opening new perspectives for biomedical research and cellular therapies.
Collapse
|
45
|
Affiliation(s)
- Ian Y Chen
- From Stanford Cardiovascular Institute (I.Y.C., J.C.W.), Division of Cardiovascular Medicine, Department of Medicine (I.Y.C., J.C.W.), and Department of Radiology (J.C.W.), Stanford University School of Medicine, CA
| | - Joseph C Wu
- From Stanford Cardiovascular Institute (I.Y.C., J.C.W.), Division of Cardiovascular Medicine, Department of Medicine (I.Y.C., J.C.W.), and Department of Radiology (J.C.W.), Stanford University School of Medicine, CA.
| |
Collapse
|
46
|
Yin C, Zhang T, Li F, Yang F, Putatunda R, Young WB, Khalili K, Hu W, Zhang Y. Functional screening of guide RNAs targeting the regulatory and structural HIV-1 viral genome for a cure of AIDS. AIDS 2016; 30:1163-74. [PMID: 26990633 PMCID: PMC4851589 DOI: 10.1097/qad.0000000000001079] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE There is an urgent need for the development of HIV-1 genome eradication strategies that lead to a permanent cure for HIV-1/AIDS. We previously reported that four guide RNAs (gRNAs) targeting HIV-1 long terminal repeats (LTR) effectively eradicated the entire HIV-1 genome. In this study, we sought to identify the best gRNAs targeting HIV-1 LTR and viral structural region and optimize gRNA pairing that can efficiently eradicate the HIV-1 genome. DESIGN Highly specific gRNAs were designed using bioinformatics tools, and their capacity of guiding CRISPR-associated system 9 to cleave HIV-1 proviral DNA was evaluated using high-throughput HIV-1 luciferase reporter assay and rapid Direct-PCR genotyping. METHODS The target seed sequences for each gRNA were cloned into lentiviral vectors. HEK293T cells were cotransfected with a pEcoHIV-NL4-3-firefly-luciferase reporter vector (1 : 20) over lentiviral vectors carrying CRISPR-associated system 9 and single gRNA or various combinations of gRNAs. The EcoHIV DNA cleaving efficiency was evaluated by Direct-PCR genotyping, and the EcoHIV transcription/replication activity was examined by a luciferase reporter assay. RESULTS Most of the designed gRNAs are effective to eliminate the predicted HIV-1 genome sequence between the selected two target sites. This is evidenced by the presence of PCR genotypic deletion/insertion and the decrease of luciferase reporter activity. In particular, a combination of viral structural gRNAs with LTR gRNAs provided a higher efficiency of genome eradication and an easier approach for PCR genotyping. CONCLUSION Our screening strategy can specifically and effectively identify gRNAs targeting HIV-1 LTR and structural region to excise proviral HIV-1 from the host genome.
Collapse
Affiliation(s)
- Chaoran Yin
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Ting Zhang
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Fang Li
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Fan Yang
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Raj Putatunda
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Won-Bin Young
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Wenhui Hu
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| | - Yonggang Zhang
- Department of Neuroscience, Center for Neurovirology and The Comprehensive NeuroAIDS Center, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140
| |
Collapse
|
47
|
Increased Understanding of Stem Cell Behavior in Neurodegenerative and Neuromuscular Disorders by Use of Noninvasive Cell Imaging. Stem Cells Int 2016; 2016:6235687. [PMID: 26997958 PMCID: PMC4779824 DOI: 10.1155/2016/6235687] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 12/13/2022] Open
Abstract
Numerous neurodegenerative and neuromuscular disorders are associated with cell-specific depletion in the human body. This imbalance in tissue homeostasis is in healthy individuals repaired by the presence of endogenous stem cells that can replace the lost cell type. However, in most disorders, a genetic origin or limited presence or exhaustion of stem cells impairs correct cell replacement. During the last 30 years, methods to readily isolate and expand stem cells have been developed and this resulted in a major change in the regenerative medicine field as it generates sufficient amount of cells for human transplantation applications. Furthermore, stem cells have been shown to release cytokines with beneficial effects for several diseases. At present however, clinical stem cell transplantations studies are struggling to demonstrate clinical efficacy despite promising preclinical results. Therefore, to allow stem cell therapy to achieve its full potential, more insight in their in vivo behavior has to be achieved. Different methods to noninvasively monitor these cells have been developed and are discussed. In some cases, stem cell monitoring even reached the clinical setting. We anticipate that by further exploring these imaging possibilities and unraveling their in vivo behavior further improvement in stem cell transplantations will be achieved.
Collapse
|
48
|
Merkert S, Martin U. Targeted genome engineering using designer nucleases: State of the art and practical guidance for application in human pluripotent stem cells. Stem Cell Res 2016; 16:377-86. [PMID: 26921872 DOI: 10.1016/j.scr.2016.02.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/14/2016] [Accepted: 02/09/2016] [Indexed: 12/26/2022] Open
Abstract
Within the last years numerous publications successfully applied sequence specific designer nucleases for genome editing in human PSCs. However, despite this abundance of reports together with the rapid development and improvement accomplished with the technology, it is still difficult to choose the optimal methodology for a specific application of interest. With focus on the most suitable approach for specific applications, we present a practical guidance for successful gene editing in human PSCs using designer nucleases. We discuss experimental considerations, limitations and critical aspects which will guide the investigator for successful implementation of this technology.
Collapse
Affiliation(s)
- Sylvia Merkert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; REBIRTH - Cluster of Excellence, Hannover Medical School, Germany; Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; REBIRTH - Cluster of Excellence, Hannover Medical School, Germany; Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Germany.
| |
Collapse
|
49
|
Sackett SD, Brown ME, Tremmel DM, Ellis T, Burlingham WJ, Odorico JS. Modulation of human allogeneic and syngeneic pluripotent stem cells and immunological implications for transplantation. Transplant Rev (Orlando) 2016; 30:61-70. [PMID: 26970668 DOI: 10.1016/j.trre.2016.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/05/2016] [Indexed: 01/20/2023]
Abstract
Tissues derived from induced pluripotent stem cells (iPSCs) are a promising source of cells for building various regenerative medicine therapies; from simply transplanting cells to reseeding decellularized organs to reconstructing multicellular tissues. Although reprogramming strategies for producing iPSCs have improved, the clinical use of iPSCs is limited by the presence of unique human leukocyte antigen (HLA) genes, the main immunologic barrier to transplantation. In order to overcome the immunological hurdles associated with allogeneic tissues and organs, the generation of patient-histocompatible iPSCs (autologous or HLA-matched cells) provides an attractive platform for personalized medicine. However, concerns have been raised as to the fitness, safety and immunogenicity of iPSC derivatives because of variable differentiation potential of different lines and the identification of genetic and epigenetic aberrations that can occur during the reprogramming process. In addition, significant cost and regulatory barriers may deter commercialization of patient specific therapies in the short-term. Nonetheless, recent studies provide some evidence of immunological benefit for using autologous iPSCs. Yet, more studies are needed to evaluate the immunogenicity of various autologous and allogeneic human iPSC-derived cell types as well as test various methods to abrogate rejection. Here, we present perspectives of using allogeneic vs. autologous iPSCs for transplantation therapies and the advantages and disadvantages of each related to differentiation potential, immunogenicity, genetic stability and tumorigenicity. We also review the current literature on the immunogenicity of syngeneic iPSCs and discuss evidence that questions the feasibility of HLA-matched iPSC banks. Finally, we will discuss emerging methods of abrogating or reducing host immune responses to PSC derivatives.
Collapse
Affiliation(s)
- S D Sackett
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - M E Brown
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - D M Tremmel
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - T Ellis
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - W J Burlingham
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - J S Odorico
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
50
|
In vitro cardiac tissue models: Current status and future prospects. Adv Drug Deliv Rev 2016; 96:203-13. [PMID: 26428618 DOI: 10.1016/j.addr.2015.09.011] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/14/2015] [Accepted: 09/21/2015] [Indexed: 01/15/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide. Achieving the next phase of potential treatment strategies and better prognostic tools will require a concerted effort from interdisciplinary fields. Biomaterials-based cardiac tissue models are revolutionizing the area of preclinical research and translational applications. The goal of in vitro cardiac tissue modeling is to create physiological functional models of the human myocardium, which is a difficult task due to the complex structure and function of the human heart. This review describes the advances made in area of in vitro cardiac models using biomaterials and bioinspired platforms. The field has progressed extensively in the past decade, and we envision its applications in the areas of drug screening, disease modeling, and precision medicine.
Collapse
|