1
|
Long P, Shi Y, Sun F, Wei Y, Wu B, Li Q, Jie Q, Ma Y. Establishment of a non‐integrated induced pluripotent stem cell line derived from human chorionic villi cells. J Clin Lab Anal 2022; 36:e24464. [PMID: 35527669 PMCID: PMC9169189 DOI: 10.1002/jcla.24464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background Few studies have investigated the generation of induced pluripotent stem cells (iPSCs) derived from human primary chorionic villi (CV) cells. The present study aimed to explore an optimal electroporation (EP) condition for generating non‐integrated iPSCs from CV cells (CV‐iPSCs). Methods The EGFP plasmid was transfected into CV cells under different EP conditions to evaluate cell adherence and the rate of EGFP positive cells. Subsequently, CV cells were transfected with the pEP4‐E02S‐ET2K and pCEP4‐miR‐302–367 plasmids under optimal EP conditions. Finally, CV‐iPSC pluripotency, karyotype analysis, and differentiation ability were investigated. Results Following EP for 48 h under different conditions, different confluency, and transfection efficiency were observed in CV cells. Higher cell density was observed in CV cells exposed to 200 V for 100 s, while higher transfection efficiency was obtained in cells electroporated at a pulse of 300 V for 300 s. To generate typical primitive iPSCs, CV cells were transfected with pEP4‐E02S‐ET2K and pCEP4‐miR‐302–367 plasmids using EP and were then cultured in induction medium for 20 days under selected conditions. Subsequently, monoclonal iPSCs were isolated and were evaluated pluripotency with AP positive staining, the expression of OCT4, SOX2, and NANOG in vitro and the formation of three germ layer teratomas in vivo. Conclusion CV‐iPSCs were successfully established under the conditions of 100 μl shock cup and EP pulse of 200 V for 300 s for two times. This may provide a novel strategy for investigating the pathogenesis of several diseases and gene therapy.
Collapse
Affiliation(s)
- Ping Long
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
- Guizhou Qiannan People's Hospital Guizhou China
| | - Yuechuan Shi
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
- Hainan Medical University Haikou Hainan China
| | - Fei Sun
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
- Department of Obstetrics and Gynecology of Nanfang Hospital Southern Medical University Guangzhou China
| | - Yunjian Wei
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
| | - Bangyong Wu
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
| | - Qi Li
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
| | - Qiuling Jie
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
| | - Yanlin Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
- Hainan Medical University Haikou Hainan China
| |
Collapse
|
2
|
Effects of Platelet Rich Plasma and Amniotic Cell Culture Medium on Wound Healing Following Experimental Animal Tracheal Injury Model: A Comparative Study. J Craniofac Surg 2021; 32:1937-1941. [PMID: 33741881 DOI: 10.1097/scs.0000000000007396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Prolonged inflammation after tracheal injury invariably results in a degree of stenosis. The topical application of platelet-rich plasma (PRP) and human amniotic fluid-derived cell culture medium (ACCM) have been shown to promote wound healing. The effects of PRP and amniotic cell culture medium (Gibco AmnioMAX - II ) were investigated in a rat model through morphometric, histological, and biochemical parameters. MATERIAL METHODS Thirty-two male Sprague Dawley rats were included in the study: 4 rats provided for the preparation of PRP. Three groups of 7 rats were divided into PRP and ACCM groups, a control and a sham group respectively. A transverse incision on the ventral aspect of the third trachea spanning half of the tracheal circumference was performed. The incision was repaired with 7/0 polypropylene in the sham group. In the control group, 0.5 ml saline solution was applied on to the repaired injury site. In the other two groups, 0.5 mL PRP or ACCM were applied topically on the tracheal repair. Tissue samples were harvested 30 days after surgery for morphometric measurements and biochemical analyses for oxidative stress markers, IL-1beta, IL-6, and VEGF. Connective tissue thickness was evaluated histologically. Statistical analysis included the Mann-Whitney U and Kruskal Wallis tests. RESULTS A notable difference was detected (P = 0,025) in cartilage segment length measurements of the trachea between the ACCM group and the sham and control groups (P < 0.03). A significant difference was found in the analysis of TAS, TOS, and OSI values between the study groups and the control and sham groups (P < 0.005). There were also differences in IL1-beta and IL-6 levels between ACCM and PRP groups (P < 0.05). For the same parameters, the differences were significant between the PRP and, sham and control groups (P = 0,004 and P = 0,002 respectively), and between the ACCM and, sham and control groups (P = 0,003 and P = 0,002 respectively).VEGF values demonstrated a significant difference between the PRP and sham group (P = 0,002), and between ACCM and sham/control groups (p=0,002 for both), the highest VEGF value was in ACCM group while the lowest value was in the sham group. In the histological assessment of connective tissue, a significant difference was observed between ACCM and the other groups. CONCLUSION Amniotic fluid-derived cell culture medium shows less oxidative stress status than the other applications. ACCM is more effective on inflammatory and angiogenetic processes. Connective tissue thickness results were consistent with those biochemical and morphologic results. Additionally, a significant difference was observed in histological data between ACCM and PRP. Overall, ACCM proved to be efficient on tracheal healing. These effects can be attributed to the abundance of growth factors in both PRP and amniotic fluid-derived cell culture medium (ACCM).
Collapse
|
3
|
Wattanapanitch M. Correction of Hemoglobin E/Beta-Thalassemia Patient-Derived iPSCs Using CRISPR/Cas9. Methods Mol Biol 2021; 2211:193-211. [PMID: 33336279 DOI: 10.1007/978-1-0716-0943-9_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
HbE/β-thalassemia is one of the most common thalassemic syndromes in Southeast Asia and Thailand. Patients have mutations in β hemoglobin (HBB) gene resulting in decreased and/or abnormal production of β hemoglobin. Here, we describe a protocol for CRISPR/Cas9-mediated gene correction of the mutated hemoglobin E from one allele of the HBB gene by homology-directed repair (HDR) in HbE/β-thalassemia patient-derived induced pluripotent stem cells (iPSCs) using a CRISPR/Cas9 plasmid-based transfection method and a single-stranded DNA oligonucleotide (ssODN) repair template harboring the correct nucleotides. Our strategy allows the seamless HbE gene correction with the editing efficiency (HDR) up to 3%, as confirmed by Sanger sequencing. This protocol provides a simple one-step genetic correction of HbE mutation in the patient-derived iPSCs. Further differentiation of the corrected iPSCs into hematopoietic stem/progenitor cells will provide an alternative renewable source of cells for the application in autologous transplantation in the future.
Collapse
Affiliation(s)
- Methichit Wattanapanitch
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
4
|
Jamal M, Bashir A, Al-Sayegh M, Huang GTJ. Oral tissues as sources for induced pluripotent stem cell derivation and their applications for neural, craniofacial, and dental tissue regeneration. CELL SOURCES FOR IPSCS 2021:71-106. [DOI: 10.1016/b978-0-12-822135-8.00007-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
5
|
Al Abbar A, Ngai SC, Nograles N, Alhaji SY, Abdullah S. Induced Pluripotent Stem Cells: Reprogramming Platforms and Applications in Cell Replacement Therapy. Biores Open Access 2020; 9:121-136. [PMID: 32368414 PMCID: PMC7194323 DOI: 10.1089/biores.2019.0046] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2020] [Indexed: 12/15/2022] Open
Abstract
The generation of induced pluripotent stem cells (iPSCs) from differentiated mature cells is one of the most promising technologies in the field of regenerative medicine. The ability to generate patient-specific iPSCs offers an invaluable reservoir of pluripotent cells, which could be genetically engineered and differentiated into target cells to treat various genetic and degenerative diseases once transplanted, hence counteracting the risk of graft versus host disease. In this context, we review the scientific research streams that lead to the emergence of iPSCs, the roles of reprogramming factors in reprogramming to pluripotency, and the reprogramming strategies. As iPSCs serve tremendous correction potentials for various diseases, we highlight the successes and challenges of iPSCs in cell replacement therapy and the synergy of iPSCs and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene editing tools in therapeutics research.
Collapse
Affiliation(s)
- Akram Al Abbar
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Siew Ching Ngai
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Malaysia
| | - Nadine Nograles
- Newcastle University Medicine Malaysia, Educity, Iskandar Puteri, Johor, Malaysia
| | - Suleiman Yusuf Alhaji
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Syahril Abdullah
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
6
|
Paes BCMF, Stabeli LCJR, Costa PNM, Orellana MD, Kashima S, Covas DT, Picanço-Castro V. Generation of hematopoietic stem/progenitor cells with sickle cell mutation from induced pluripotent stem cell in serum-free system. Hematol Transfus Cell Ther 2020; 43:156-164. [PMID: 32229108 PMCID: PMC8211631 DOI: 10.1016/j.htct.2020.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/13/2020] [Indexed: 01/13/2023] Open
Abstract
Introduction Sickle cell disease (SCD) is a monogenic disease and it is estimated that 300,000 infants are born annually with it. Most treatments available are only palliative, whereas the allogeneic hematopoietic stem cell transplantation offers the only potential cure for SCD. Objective Generation of human autologous cells, when coupled with induced pluripotent stem cell (iPSC) technology, is a promising approach for developing study models. In this study, we provide a simple and efficient model for generating hematopoietic cells using iPSCs derived from a sickle cell anemia patient and an inexpensive in-house-prepared medium. Method This study used iPSCs previously generated from peripheral blood mononuclear cells (PBMCs) from a patient with sickle cell anemia (iPSC_scd). Hematopoietic and erythroid differentiation was performed in two steps. Firstly, with the induction of hematopoietic differentiation through embryoid body formation, we evaluated the efficiency of two serum-free media; and secondly, the induction of hematopoietic stem/progenitor cells to erythroid progenitor cells was performed. Results The patient-specific cell line generated CD34+/CD45+ and CD45+/CD43+ hematopoietic stem/progenitor cells and erythroid progenitors, comprising CD36+, CD71+ and CD235a+ populations, as well as the formation of hematopoietic colonies, including erythroid colonies, in culture in a semi-solid medium. Conclusion In conjunction, our results described a simple serum-free platform to differentiate human the iPSCs into hematopoietic progenitor cells. This platform is an emerging application of iPSCs in vitro disease modeling, which can significantly improve the search for new pharmacological drugs for sickle cell disease.
Collapse
Affiliation(s)
- Bárbara C M F Paes
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, SP, Brazil; Universidade de São Paulo, Hemocentro de Ribeirão Preto, Centro de Terapia Celular, Ribeirão Preto, SP, Brazil
| | - Luiza C J R Stabeli
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, SP, Brazil; Universidade de São Paulo, Hemocentro de Ribeirão Preto, Centro de Terapia Celular, Ribeirão Preto, SP, Brazil
| | - Péricles N M Costa
- Universidade de São Paulo, Hemocentro de Ribeirão Preto, Centro de Terapia Celular, Ribeirão Preto, SP, Brazil
| | - Maristela Delgado Orellana
- Universidade de São Paulo, Hemocentro de Ribeirão Preto, Centro de Terapia Celular, Ribeirão Preto, SP, Brazil
| | - Simone Kashima
- Universidade de São Paulo, Hemocentro de Ribeirão Preto, Centro de Terapia Celular, Ribeirão Preto, SP, Brazil
| | - Dimas Tadeu Covas
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, SP, Brazil; Universidade de São Paulo, Hemocentro de Ribeirão Preto, Centro de Terapia Celular, Ribeirão Preto, SP, Brazil
| | - Virgínia Picanço-Castro
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, SP, Brazil; Universidade de São Paulo, Hemocentro de Ribeirão Preto, Centro de Terapia Celular, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
7
|
Shangaris P, Loukogeorgakis SP, Subramaniam S, Flouri C, Jackson LH, Wang W, Blundell MP, Liu S, Eaton S, Bakhamis N, Ramachandra DL, Maghsoudlou P, Urbani L, Waddington SN, Eddaoudi A, Archer J, Antoniou MN, Stuckey DJ, Schmidt M, Thrasher AJ, Ryan TM, De Coppi P, David AL. In Utero Gene Therapy (IUGT) Using GLOBE Lentiviral Vector Phenotypically Corrects the Heterozygous Humanised Mouse Model and Its Progress Can Be Monitored Using MRI Techniques. Sci Rep 2019; 9:11592. [PMID: 31406195 PMCID: PMC6690943 DOI: 10.1038/s41598-019-48078-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 07/29/2019] [Indexed: 02/06/2023] Open
Abstract
In utero gene therapy (IUGT) to the fetal hematopoietic compartment could be used to treat congenital blood disorders such as β-thalassemia. A humanised mouse model of β-thalassemia was used, in which heterozygous animals are anaemic with splenomegaly and extramedullary hematopoiesis. Intrahepatic in utero injections of a β globin-expressing lentiviral vector (GLOBE), were performed in fetuses at E13.5 of gestation. We analysed animals at 12 and 32 weeks of age, for vector copy number in bone marrow, peripheral blood liver and spleen and we performed integration site analysis. Compared to noninjected heterozygous animals IUGT normalised blood haemoglobin levels and spleen weight. Integration site analysis showed polyclonality. The left ventricular ejection fraction measured using magnetic resonance imaging (MRI) in treated heterozygous animals was similar to that of normal non-β-thalassemic mice but significantly higher than untreated heterozygous thalassemia mice suggesting that IUGT ameliorated poor cardiac function. GLOBE LV-mediated IUGT normalised the haematological and anatomical phenotype in a heterozygous humanised model of β-thalassemia.
Collapse
Affiliation(s)
- Panicos Shangaris
- Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK.
- UCL Institute of Child Health, UCL, London, United Kingdom.
| | | | | | - Christina Flouri
- Department of Medical and Molecular Genetics, KCL, London, United Kingdom
| | | | - Wei Wang
- Department of Translational Oncology, National Centre for Tumour Diseases, Heidelberg, Germany
| | | | - Shanrun Liu
- Biochemistry and Molecular Genetics, UAB, Birmingham, Alabama, United States
| | - Simon Eaton
- UCL Institute of Child Health, UCL, London, United Kingdom
| | - Nahla Bakhamis
- Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | | | | | - Luca Urbani
- UCL Institute of Child Health, UCL, London, United Kingdom
| | - Simon N Waddington
- Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ayad Eddaoudi
- UCL Institute of Child Health, UCL, London, United Kingdom
| | - Joy Archer
- Central Diagnostic Services, Queen's Vet School Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Michael N Antoniou
- Department of Medical and Molecular Genetics, KCL, London, United Kingdom
| | - Daniel J Stuckey
- Centre for Advanced Biomedical Imaging, UCL, London, United Kingdom
| | - Manfred Schmidt
- Department of Translational Oncology, National Centre for Tumour Diseases, Heidelberg, Germany
| | | | - Thomas M Ryan
- Biochemistry and Molecular Genetics, UAB, Birmingham, Alabama, United States
| | - Paolo De Coppi
- UCL Institute of Child Health, UCL, London, United Kingdom
| | - Anna L David
- Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| |
Collapse
|
8
|
Georgomanoli M, Papapetrou EP. Modeling blood diseases with human induced pluripotent stem cells. Dis Model Mech 2019; 12:12/6/dmm039321. [PMID: 31171568 PMCID: PMC6602313 DOI: 10.1242/dmm.039321] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are derived from somatic cells through a reprogramming process, which converts them to a pluripotent state, akin to that of embryonic stem cells. Over the past decade, iPSC models have found increasing applications in the study of human diseases, with blood disorders featuring prominently. Here, we discuss methodological aspects pertaining to iPSC generation, hematopoietic differentiation and gene editing, and provide an overview of uses of iPSCs in modeling the cell and gene therapy of inherited genetic blood disorders, as well as their more recent use as models of myeloid malignancies. We also discuss the strengths and limitations of iPSCs compared to model organisms and other cellular systems commonly used in hematology research.
Collapse
Affiliation(s)
- Maria Georgomanoli
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eirini P Papapetrou
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
9
|
Recent Updates on Induced Pluripotent Stem Cells in Hematological Disorders. Stem Cells Int 2019; 2019:5171032. [PMID: 31191673 PMCID: PMC6525795 DOI: 10.1155/2019/5171032] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/31/2019] [Indexed: 02/07/2023] Open
Abstract
Over the past decade, enormous progress has been made in the field of induced pluripotent stem cells (iPSCs). Patients' somatic cells such as skin fibroblasts or blood cells can be used to generate disease-specific pluripotent stem cells, which have unlimited proliferation and can differentiate into all cell types of the body. Human iPSCs offer great promises and opportunities for treatments of degenerative diseases and studying disease pathology and drug screening. So far, many iPSC-derived disease models have led to the discovery of novel pathological mechanisms as well as new drugs in the pipeline that have been tested in the iPSC-derived cells for efficacy and potential toxicities. Furthermore, recent advances in genome editing technology in combination with the iPSC technology have provided a versatile platform for studying stem cell biology and regenerative medicine. In this review, an overview of iPSCs, patient-specific iPSCs for disease modeling and drug screening, applications of iPSCs and genome editing technology in hematological disorders, remaining challenges, and future perspectives of iPSCs in hematological diseases will be discussed.
Collapse
|
10
|
Abstract
Stem-cell therapy is a promising method for treating patients with a wide range of diseases and injuries. Increasing government funding of scientific research has promoted rapid developments in stem-cell research in China, as evidenced by the substantial increase in the number and quality of publications in the past 5 years. Multiple high-quality studies have been performed in China that concern cell reprogramming, stem-cell homeostasis, gene modifications, and immunomodulation. The number of translation studies, including basic and preclinical investigations, has also increased. Around 100 stem-cell banks have been established in China, 10 stem-cell drugs are currently in the approval process, and >400 stem cell-based clinical trials are currently registered in China. With continued state funding, advanced biotechnical support, and the development of regulatory standards for the clinical application of stem cells, further innovations are expected that will lead to a boom in stem-cell therapies. This review highlights recent achievements in stem-cell research in China and discusses future prospects.
Collapse
Affiliation(s)
- Lei Hu
- 1 Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing, China
| | - Bin Zhao
- 1 Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing, China
| | - Songlin Wang
- 1 Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing, China.,2 Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences , Beijing, China
| |
Collapse
|
11
|
Dolatshad H, Tatwavedi D, Ahmed D, Tegethoff JF, Boultwood J, Pellagatti A. Application of induced pluripotent stem cell technology for the investigation of hematological disorders. Adv Biol Regul 2019; 71:19-33. [PMID: 30341008 DOI: 10.1016/j.jbior.2018.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 06/08/2023]
Abstract
Induced pluripotent stem cells (iPSCs) were first described over a decade ago and are currently used in various basic biology and clinical research fields. Recent advances in the field of human iPSCs have opened the way to a better understanding of the biology of human diseases. Disease-specific iPSCs provide an unparalleled opportunity to establish novel human cell-based disease models, with the potential to enhance our understanding of the molecular mechanisms underlying human malignancies, and to accelerate the identification of effective new drugs. When combined with genome editing technologies, iPSCs represent a new approach to study single or multiple disease-causing mutations and model specific diseases in vitro. In addition, genetically corrected patient-specific iPSCs could potentially be used for stem cell based therapy. Furthermore, the reprogrammed cells share patient-specific genetic background, offering a new platform to develop personalized therapy/medicine for patients. In this review we discuss the recent advances in iPSC research technology and their potential applications in hematological diseases. Somatic cell reprogramming has presented new routes for generating patient-derived iPSCs, which can be differentiated to hematopoietic stem cells and the various downstream hematopoietic lineages. iPSC technology shows promise in the modeling of both inherited and acquired hematological disorders. A direct reprogramming and differentiation strategy is able to recapitulate hematological disorder progression and capture the earliest molecular alterations that underlie the initiation of hematological malignancies.
Collapse
Affiliation(s)
- Hamid Dolatshad
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Dharamveer Tatwavedi
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Doaa Ahmed
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK; Clinical Pathology Department, Assiut University Hospitals, Faculty of Medicine, Assiut, Egypt
| | - Jana F Tegethoff
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Jacqueline Boultwood
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Andrea Pellagatti
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK.
| |
Collapse
|
12
|
Xu B, Magli A, Anugrah Y, Koester SJ, Perlingeiro RCR, Shen W. Nanotopography-responsive myotube alignment and orientation as a sensitive phenotypic biomarker for Duchenne Muscular Dystrophy. Biomaterials 2018; 183:54-66. [PMID: 30149230 PMCID: PMC6239205 DOI: 10.1016/j.biomaterials.2018.08.047] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/09/2018] [Accepted: 08/20/2018] [Indexed: 01/08/2023]
Abstract
Duchenne Muscular Dystrophy (DMD) is a fatal genetic disorder currently having no cure. Here we report that culture substrates patterned with nanogrooves and functionalized with Matrigel (or laminin) present an engineered cell microenvironment to allow myotubes derived from non-diseased, less-affected DMD, and severely-affected DMD human induced pluripotent stem cells (hiPSCs) to exhibit prominent differences in alignment and orientation, providing a sensitive phenotypic biomarker to potentially facilitate DMD drug development and early diagnosis. We discovered that myotubes differentiated from myogenic progenitors derived from non-diseased hiPSCs align nearly perpendicular to nanogrooves, a phenomenon not reported previously. We further found that myotubes derived from hiPSCs of a dystrophin-null DMD patient orient randomly, and those from hiPSCs of a patient carrying partially functional dystrophin align approximately 14° off the alignment direction of non-diseased myotubes. Substrates engineered with micron-scale grooves and/or cell adhesion molecules only interacting with integrins all guide parallel myotube alignment to grooves and lose the ability to distinguish different cell types. Disruption of the interaction between the Dystrophin-Associated-Protein-Complex (DAPC) and laminin by heparin or anti-α-dystroglycan antibody IIH6 disenables myotubes to align perpendicular to nanogrooves, suggesting that this phenotype is controlled by the DAPC-mediated cytoskeleton-extracellular matrix linkage.
Collapse
Affiliation(s)
- Bin Xu
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alessandro Magli
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Yoska Anugrah
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Steven J Koester
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Rita C R Perlingeiro
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA; Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Wei Shen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA; Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
13
|
Kunisaki SM. Amniotic Fluid Stem Cells for the Treatment of Surgical Disorders in the Fetus and Neonate. Stem Cells Transl Med 2018; 7:767-773. [PMID: 30085416 PMCID: PMC6216434 DOI: 10.1002/sctm.18-0018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 05/14/2018] [Accepted: 06/08/2018] [Indexed: 12/11/2022] Open
Abstract
Over the past decade, amniotic fluid‐derived stem cells have emerged as a novel experimental approach aimed at improving outcomes in children with congenital anomalies, including spina bifida, heart defects, and diaphragmatic hernia. Interest in these cells for the treatment of prenatally diagnosed diseases has arisen based on numerous studies demonstrating the relative ease of harvesting an abundant quantity of amniocytes from a small aliquot of fluid, the unique properties of amniocytes themselves, and the beneficial effects of amniotic fluid‐derived stem cells in experimental animal models. This report gives a brief overview of the rationale and current status of amniotic fluid stem cell‐based therapies, focusing on its relevance to birth defects affecting the fetus and neonate. The author proposes a roadmap for further study that would be required prior to clinical application of amniotic fluid stem cell technologies. stem cells translational medicine2018;7:767–773
Collapse
Affiliation(s)
- Shaun M Kunisaki
- Department of Surgery, Fetal Diagnosis and Treatment Center and Section of Pediatric Surgery, University of Michigan, C.S. Mott Children's and Von Voigtlander Women's Hospital, Ann Arbor, Michigan, USA
| |
Collapse
|
14
|
Chapin J, Giardina PJ. Thalassemia Syndromes. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00040-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
15
|
Fu S, Ding J, Liu D, Huang H, Li M, Liu Y, Tu L, Liu D. Generation of human-induced pluripotent stem cells from burn patient-derived skin fibroblasts using a non-integrative method. Int J Mol Med 2018; 41:87-94. [PMID: 29115387 PMCID: PMC5746323 DOI: 10.3892/ijmm.2017.3206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 10/19/2017] [Indexed: 11/23/2022] Open
Abstract
Patient specific induced pluripotent stem cells (iPSCs) have been recognized as a possible source of cells for skin tissue engineering. They have the potential to greatly benefit patients with large areas of burned skin or skin defects. However, the integration virus-based reprogramming method is associated with a high risk of genetic mutation and mouse embryonic fibroblast feeder-cells may be a pollutant. In the present study, human skin fibroblasts (HSFs) were successfully harvested from patients with burns and patient-specific iPSCs were generated using a non-integration method with a feeder-free approach. The octamer-binding transcription factor 4 (OCT4), sex-determining region Y box 2 (SOX2) and NANOG transcription factors were delivered using Sendai virus vectors. iPSCs exhibited representative human embryonic stem cell-like morphology and proliferation characteristics. They also expressed pluripotent markers, including OCT4, NANOG, SOX2, TRA181, stage-specific embryonic antigen 4 and TRA-160, and exhibited a normal karyotype. Teratoma and embryoid body formation revealed that iPSCs were able to differentiate into cells of all three germ layers in vitro and in vivo. The results of the present study demonstrate that HSFs derived from patients with burns, may be reprogrammed into stem cells with pluripotency, which provides a basis for cell‑based skin tissue engineering in the future.
Collapse
Affiliation(s)
- Shangfeng Fu
- Burns Institute, The First Affiliated Hospital of Nanchang University
| | - Jianwu Ding
- Department of Oncology, The Second Affiliated Hospital of Nanchang University
| | - Dewu Liu
- Burns Institute, The First Affiliated Hospital of Nanchang University
| | - Heping Huang
- Burns Institute, The First Affiliated Hospital of Nanchang University
- Department of Plastic and Aesthetic Surgery, Jingxi Maternal and Child Health Hospital
| | - Min Li
- Burns Institute, The First Affiliated Hospital of Nanchang University
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanchang University
| | - Yang Liu
- Burns Institute, The First Affiliated Hospital of Nanchang University
| | - Longxiang Tu
- Burns Institute, The First Affiliated Hospital of Nanchang University
| | - Deming Liu
- Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
16
|
Cai L, Bai H, Mahairaki V, Gao Y, He C, Wen Y, Jin YC, Wang Y, Pan RL, Qasba A, Ye Z, Cheng L. A Universal Approach to Correct Various HBB Gene Mutations in Human Stem Cells for Gene Therapy of Beta-Thalassemia and Sickle Cell Disease. Stem Cells Transl Med 2017; 7:87-97. [PMID: 29164808 PMCID: PMC5746148 DOI: 10.1002/sctm.17-0066] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/25/2017] [Indexed: 12/21/2022] Open
Abstract
Beta-thalassemia is one of the most common recessive genetic diseases, caused by mutations in the HBB gene. Over 200 different types of mutations in the HBB gene containing three exons have been identified in patients with β-thalassemia (β-thal) whereas a homozygous mutation in exon 1 causes sickle cell disease (SCD). Novel therapeutic strategies to permanently correct the HBB mutation in stem cells that are able to expand and differentiate into erythrocytes producing corrected HBB proteins are highly desirable. Genome editing aided by CRISPR/Cas9 and other site-specific engineered nucleases offers promise to precisely correct a genetic mutation in the native genome without alterations in other parts of the human genome. Although making a sequence-specific nuclease to enhance correction of a specific HBB mutation by homology-directed repair (HDR) is becoming straightforward, targeting various HBB mutations of β-thal is still challenging because individual guide RNA as well as a donor DNA template for HDR of each type of HBB gene mutation have to be selected and validated. Using human induced pluripotent stem cells (iPSCs) from two β-thal patients with different HBB gene mutations, we devised and tested a universal strategy to achieve targeted insertion of the HBB cDNA in exon 1 of HBB gene using Cas9 and two validated guide RNAs. We observed that HBB protein production was restored in erythrocytes derived from iPSCs of two patients. This strategy of restoring functional HBB gene expression will be able to correct most types of HBB gene mutations in β-thal and SCD. Stem Cells Translational Medicine 2018;7:87-97.
Collapse
Affiliation(s)
- Liuhong Cai
- Center for Reproductive Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.,Division of Hematology, Department of Medicine, Baltimore, Maryland, USA.,Stem Cell Program in the Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hao Bai
- Division of Hematology, Department of Medicine, Baltimore, Maryland, USA.,Stem Cell Program in the Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vasiliki Mahairaki
- Stem Cell Program in the Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yongxing Gao
- Division of Hematology, Department of Medicine, Baltimore, Maryland, USA.,Stem Cell Program in the Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chaoxia He
- Division of Hematology, Department of Medicine, Baltimore, Maryland, USA.,Stem Cell Program in the Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yanfei Wen
- Center for Reproductive Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.,Center for Reproductive Medicine, Jiangmen Hospital, Sun Yat-sen University, Jiangmen, People's Republic of China
| | - You-Chuan Jin
- Stem Cell Program in the Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
| | - You Wang
- Stem Cell Program in the Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Public Health Studies, Johns Hopkins University Krieger School of Art and Science, Baltimore, Maryland, USA
| | - Rachel L Pan
- Stem Cell Program in the Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Biology, Emory University, Atlanta, Georgia, USA
| | - Armaan Qasba
- Stem Cell Program in the Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Biology, McDaniel College, Westminster, Maryland, USA
| | - Zhaohui Ye
- Division of Hematology, Department of Medicine, Baltimore, Maryland, USA.,Division of Cellular and Gene Therapies, Gene Transfer and Immunogenicity Branch, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Linzhao Cheng
- Division of Hematology, Department of Medicine, Baltimore, Maryland, USA.,Stem Cell Program in the Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
17
|
Chau M, Deveau TC, Song M, Wei ZZ, Gu X, Yu SP, Wei L. Transplantation of iPS cell-derived neural progenitors overexpressing SDF-1α increases regeneration and functional recovery after ischemic stroke. Oncotarget 2017; 8:97537-97553. [PMID: 29228630 PMCID: PMC5722582 DOI: 10.18632/oncotarget.22180] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 08/07/2017] [Indexed: 02/07/2023] Open
Abstract
Ischemic stroke is a leading cause of human death and disability while clinical treatments are limited. The adult brain possesses endogenous regenerative activities that may benefit tissue repair after stroke. Trophic factors such as stromal cell-derived factor 1 alpha (SDF-1α) are upregulated in the ischemic brain, which promote endogenous regeneration. The regenerative response, however, is normally insufficient. Transplantation of exogenous cells has been explored as regenerative therapies. One promising cell type for transplantation is induced pluripotent stem (iPS) cells which are cells genetically reprogrammed from adult somatic cells. We hypothesized that transplanting neural progenitor cells derived from iPS cells (iPS-NPCs) could provide cell replacement and trophic support. The trophic factor SDF-1α was overexpressed in iPS-NPCs by lentiviral transduction to test if SDF-1α could increase regeneration in the ischemic brain. These SDF-1α-iPS-NPCs were differentiated in vitro to express mature neuronal and synaptic markers. Differentiated cells expressed functional Na+ and K+ channels, and fired action potentials. In the oxygen glucose deprivation (OGD) test, SDF-1α-iPS-NPCs survived significantly better compared to control iPS-NPCs. In mice subjected to focal cerebral ischemia in the sensorimotor cortex, iPS-NPCs and SDF-1α-iPS-NPCs were intracranially transplanted into the ischemic cortex 7 days after stroke. Neuronal differentiation of transplanted cells was identified using NeuN 14 days after transplantation. Mice that received SDF-1α-iPS-NPCs had greater numbers of NeuN/BrdU and Glut-1/BrdU co-labeled cells in the peri-infarct area and improved locomotion compared to the control iPS-NPC transplantation. Thus, SDF-1α upregulation in transplanted cells may be a therapeutic strategy to enhance endogenous neurovascular repair after ischemic stroke in adult mice.
Collapse
Affiliation(s)
- Monica Chau
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Todd C. Deveau
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mingke Song
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Zheng Z. Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
18
|
Rawat N, Singh MK. Induced pluripotent stem cell: A headway in reprogramming with promising approach in regenerative biology. Vet World 2017; 10:640-649. [PMID: 28717316 PMCID: PMC5499081 DOI: 10.14202/vetworld.2017.640-649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 04/26/2017] [Indexed: 12/17/2022] Open
Abstract
Since the embryonic stem cells have knocked the doorsteps, they have proved themselves in the field of science, research, and medicines, but the hovered restrictions confine their application in human welfare. Alternate approaches used to reprogram the cells to the pluripotent state were not up to par, but the innovation of induced pluripotent stem cells (iPSCs) paved a new hope for the researchers. Soon after the discovery, iPSCs technology is undergoing renaissance day by day, i.e., from the use of genetic material to recombinant proteins and now only chemicals are employed to convert somatic cells to iPSCs. Thus, this technique is moving straightforward and productive at an astonishing pace. Here, we provide a brief introduction to iPSCs, the mechanism and methods for their generation, their prevailing and prospective applications and the future opportunities that can be expected from them.
Collapse
Affiliation(s)
- N Rawat
- Embryo Biotechnology Lab, Animal Biotechnology Centre, ICAR - National Dairy Research Institute, Karnal - 132 001, Haryana, India
| | - M K Singh
- Embryo Biotechnology Lab, Animal Biotechnology Centre, ICAR - National Dairy Research Institute, Karnal - 132 001, Haryana, India
| |
Collapse
|
19
|
Abstract
The induced pluripotent stem cell (iPSC) was first described more than 10 years ago and is currently used in various basic science and clinical research fields. The aim of this report is to examine the trends in research using iPSCs over the last 10 years. The 2006-2016 PubMed database was searched using the MeSH term "induced pluripotent stem cells." Only original research articles were selected, with a total of 3323 articles. These were classified according to research theme into reprogramming, differentiation protocols for specific cells and/or tissues, pathophysiological research on diseases, and discovery of new drugs, and then the trends over the years were analyzed. We also focused on 232 research publications on the pathophysiological causes of diseases and drug discovery with impact factor (IF; Thomson Reuters) of six or more. The IF of each article was summed up by year, by main target disease, and by country, and the total IF score was expressed as trends of research. The trends of research activities of reprogramming and differentiation on specific cells and/or tissues reached maxima in 2013/2014. On the other hand, research on pathophysiology and drug discovery increased continuously. The 232 articles with IF ≥6 dealt with neurological, immunological/hematological, cardiovascular, and digestive tract diseases, in that order. The majority of articles were published from the United States, followed by Japan, Germany, and United Kingdom. In conclusion, iPSCs have become a general tool for pathophysiological research on disease and drug discovery.
Collapse
Affiliation(s)
- Takaharu Negoro
- Platform of Therapeutics for Rare Disease, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Hanayuki Okura
- Platform of Therapeutics for Rare Disease, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Akifumi Matsuyama
- Platform of Therapeutics for Rare Disease, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| |
Collapse
|
20
|
Kawamura F, Inaki M, Katafuchi A, Abe Y, Tsuyama N, Kurosu Y, Yanagi A, Higuchi M, Muto S, Yamaura T, Suzuki H, Noji H, Suzuki S, Yoshida MA, Sasatani M, Kamiya K, Onodera M, Sakai A. Establishment of induced pluripotent stem cells from normal B cells and inducing AID expression in their differentiation into hematopoietic progenitor cells. Sci Rep 2017; 7:1659. [PMID: 28490810 PMCID: PMC5431994 DOI: 10.1038/s41598-017-01627-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 03/27/2017] [Indexed: 12/27/2022] Open
Abstract
B cell derived induced pluripotent stem cells (BiPSCs) were recently established from peripheral blood B cells by the simultaneous transfection of Yamanaka factors (Oct3/4, Sox2, Klf4, c-Myc) and C/EBPα using a Sendai virus vector. Here, using a different method, we established BiPSCs with immunoglobulin heavy chain (IgH) gene rearrangement from normal B cells purified from lymph nodes. The critical points of our method are pre-stimulation of B cells with IL-21 and CD40-ligand (CD40L), followed by consecutive transfection of highly concentrated Yamanaka factors using a retroviral vector. Following each transfection the cells were centrifuged onto a retronectin coated plate and the activated by IL-4, IL-2, and CD40L. Furthermore, we established BiPSCs (BiPSC-A) in which activation-induced cytidine deaminase (AID) could be induced using the doxycycline-controlled. Both the parental BiPSC and BiPSC-A showed the capability of differentiating into hematopoietic progenitor cells (HPCs) based on confirmation of CD34 expression and colony-formation from CD34-positive cells. The findings that BiPSC-A can differentiate into HPCs suggest that there is a possibility that induction of AID expression would result in chromosomal translocations in the process of differentiation from BiPSCs, and therefore that these BiPSCs could be useful in elucidating the tumor origin of abnormal B cells in myelomagenesis.
Collapse
Affiliation(s)
- Fumihiko Kawamura
- Department of Radiation Life Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Makoto Inaki
- Department of Genetics, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Atsushi Katafuchi
- Department of Radiation Life Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yu Abe
- Department of Radiation Life Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Naohiro Tsuyama
- Department of Radiation Life Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yumiko Kurosu
- Department of Radiation Life Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Aki Yanagi
- Department of Radiation Life Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Mitsunori Higuchi
- Department of Regenerative Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Satoshi Muto
- Department of Regenerative Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Takumi Yamaura
- Department of Regenerative Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Hiroyuki Suzuki
- Department of Regenerative Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Hideyoshi Noji
- Department of Medical Oncology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shinichi Suzuki
- Department of Thyroid and Endocrinology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Mitsuaki A Yoshida
- Department of Radiation Biology, Institute of Radiation Emergency Medicine, Hirosaki University, Hirosaki, Japan
| | - Megumi Sasatani
- Department of Experimental Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Kenji Kamiya
- Department of Experimental Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Masafumi Onodera
- Department of Genetics, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Akira Sakai
- Department of Radiation Life Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan.
| |
Collapse
|
21
|
Bertin E, Piccoli M, Franzin C, Nagy A, Mileikovsky M, De Coppi P, Pozzobon M. The Production of Pluripotent Stem Cells from Mouse Amniotic Fluid Cells Using a Transposon System. J Vis Exp 2017. [PMID: 28287531 DOI: 10.3791/54598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Induced pluripotent stem (iPS) cells are generated from mouse and human somatic cells by forced expression of defined transcription factors using different methods. Here, we produced iPS cells from mouse amniotic fluid cells, using a non-viral-based transposon system. All obtained iPS cell lines exhibited characteristics of pluripotent cells, including the ability to differentiate toward derivatives of all three germ layers in vitro and in vivo. This strategy opens up the possibility of using cells from diseased fetuses to develop new therapies for birth defects.
Collapse
Affiliation(s)
- Enrica Bertin
- Stem Cell and Regenerative Medicine Laboratory, Fondazione Istituto di Ricerca Pediatrica Citta della Speranza
| | - Martina Piccoli
- Stem Cell and Regenerative Medicine Laboratory, Fondazione Istituto di Ricerca Pediatrica Citta della Speranza
| | - Chiara Franzin
- Stem Cell and Regenerative Medicine Laboratory, Fondazione Istituto di Ricerca Pediatrica Citta della Speranza
| | - Andras Nagy
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital
| | | | - Paolo De Coppi
- Stem Cells and Regenerative Medicine Section, Developmental Biology and Cancer Programme, UCL Institute of Child Health and Great Ormond Street Hospital
| | - Michela Pozzobon
- Stem Cell and Regenerative Medicine Laboratory, Fondazione Istituto di Ricerca Pediatrica Citta della Speranza;
| |
Collapse
|
22
|
Gene and Cell Therapy for β-Thalassemia and Sickle Cell Disease with Induced Pluripotent Stem Cells (iPSCs): The Next Frontier. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1013:219-240. [PMID: 29127683 DOI: 10.1007/978-1-4939-7299-9_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In recent years, breakthroughs in human pluripotent stem cell (hPSC) research, namely cellular reprogramming and the emergence of sophisticated genetic engineering technologies, have opened new frontiers for cell and gene therapy. The prospect of using hPSCs, either autologous or histocompatible, as targets of genetic modification and their differentiated progeny as cell products for transplantation, presents a new paradigm of regenerative medicine of potential tremendous value for the treatment of blood disorders, including beta-thalassemia (BT) and sickle cell disease (SCD). Despite advances at a remarkable pace and great promise, many roadblocks remain before clinical translation can be realistically considered. Here we discuss the theoretical advantages of cell therapies utilizing hPSC derivatives, recent proof-of-principle studies and the main challenges towards realizing the potential of hPSC therapies in the clinic.
Collapse
|
23
|
Dong AC, Rivella S. Gene Addition Strategies for β-Thalassemia and Sickle Cell Anemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1013:155-176. [PMID: 29127680 DOI: 10.1007/978-1-4939-7299-9_6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Beta-thalassemia and sickle cell anemia are two of the most common diseases related to the hemoglobin protein. In these diseases, the beta-globin gene is mutated, causing severe anemia and ineffective erythropoiesis. Patients can additionally present with a number of life-threatening co-morbidities, such as stroke or spontaneous fractures. Current treatment involves transfusion and iron chelation; allogeneic bone marrow transplant is the only curative option, but is limited by the availability of matching donors and graft-versus-host disease. As these two diseases are monogenic diseases, they make an attractive setting for gene therapy. Gene therapy aims to correct the mutated beta-globin gene or add back a functional copy of beta- or gamma-globin. Initial gene therapy work was done with oncoretroviral vectors, but has since shifted to lentiviral vectors. Currently, there are a few clinical trials underway to test the curative potential of some of these lentiviral vectors. This review will highlight the work done thus far, and present the challenges still facing gene therapy, such as genome toxicity concerns and achieving sufficient transgene expression to cure those with the most severe forms of thalassemia.
Collapse
Affiliation(s)
- Alisa C Dong
- Division of Hematology-Oncology, Department of Pediatrics, Weill Cornell Medical College, 515 E. 71st St., Room S-709, New York, NY, 10021, USA
| | - Stefano Rivella
- Division of Hematology-Oncology, Department of Pediatrics, Weill Cornell Medical College, 515 E. 71st St., S702, Box 284, New York, NY, 10021, USA.
| |
Collapse
|
24
|
Paes BCMF, Moço PD, Pereira CG, Porto GS, de Sousa Russo EM, Reis LCJ, Covas DT, Picanço-Castro V. Ten years of iPSC: clinical potential and advances in vitro hematopoietic differentiation. Cell Biol Toxicol 2016; 33:233-250. [PMID: 28039590 DOI: 10.1007/s10565-016-9377-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 12/18/2016] [Indexed: 01/19/2023]
Abstract
Ten years have passed since the first publication announcing the generation of induced pluripotent stem cells (iPSCs). Issues related to ethics, immune rejection, and cell availability seemed to be solved following this breakthrough. The development of iPSC technology allows advances in in vitro cell differentiation for cell therapy purpose and other clinical applications. This review provides a perspective on the iPSC potential for cell therapies, particularly for hematological applications. We discuss the advances in in vitro hematopoietic differentiation, the possibilities to employ iPSC in hematology studies, and their potential clinical application in hematologic diseases. The generation of red blood cells and functional T cells and the genome editing technology applied to mutation correction are also covered. We highlight some of the requirements and obstacles to be overcome before translating these cells from research to the clinic, for instance, iPSC variability, genotoxicity, the differentiation process, and engraftment. Also, we evaluate the patent landscape and compile the clinical trials in the field of pluripotent stem cells. Currently, we know much more about iPSC than in 2006, but there are still challenges that must be solved. A greater understanding of molecular mechanisms underlying the generation of hematopoietic stem cells is necessary to produce suitable and transplantable hematopoietic stem progenitor cells from iPSC.
Collapse
Affiliation(s)
- Bárbara Cristina Martins Fernandes Paes
- Ribeirão Preto Medical School and Center for Cell-based Therapy (CTC), University of São Paulo, São Paulo, Brazil
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, São Paulo, 14051-140, Brazil
| | - Pablo Diego Moço
- Ribeirão Preto Medical School and Center for Cell-based Therapy (CTC), University of São Paulo, São Paulo, Brazil
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, São Paulo, 14051-140, Brazil
| | - Cristiano Gonçalves Pereira
- School of Economics, Business Administration and Accounting at Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Geciane Silveira Porto
- School of Economics, Business Administration and Accounting at Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Elisa Maria de Sousa Russo
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, São Paulo, 14051-140, Brazil
- Ribeirão Preto Pharmaceutical Sciences School, University of São Paulo, São Paulo, Brazil
| | - Luiza Cunha Junqueira Reis
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, São Paulo, 14051-140, Brazil
- Ribeirão Preto Pharmaceutical Sciences School, University of São Paulo, São Paulo, Brazil
| | - Dimas Tadeu Covas
- Ribeirão Preto Medical School and Center for Cell-based Therapy (CTC), University of São Paulo, São Paulo, Brazil
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, São Paulo, 14051-140, Brazil
| | - Virginia Picanço-Castro
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, São Paulo, 14051-140, Brazil.
| |
Collapse
|
25
|
Finotti A, Borgatti M, Gambari R. Ground state naïve pluripotent stem cells and CRISPR/Cas9 gene correction for β-thalassemia. Stem Cell Investig 2016; 3:66. [PMID: 27868048 DOI: 10.21037/sci.2016.09.21] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 09/19/2016] [Indexed: 01/31/2023]
Affiliation(s)
- Alessia Finotti
- Department of Life Sciences and Biotechnology, Ferrara University, Ferrara, Italy
| | - Monica Borgatti
- Department of Life Sciences and Biotechnology, Ferrara University, Ferrara, Italy
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, Ferrara University, Ferrara, Italy; Biotechnology Center, Ferrara University, Ferrara, Italy
| |
Collapse
|
26
|
Osteogenic Differentiation of Human Amniotic Fluid Mesenchymal Stem Cells Is Determined by Epigenetic Changes. Stem Cells Int 2016; 2016:6465307. [PMID: 27818691 PMCID: PMC5080506 DOI: 10.1155/2016/6465307] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 09/19/2016] [Indexed: 12/30/2022] Open
Abstract
Osteogenic differentiation of human amniotic fluid derived mesenchymal stem cells (AF-MSCs) has been widely studied in vitro and in vivo as a potential tool for regenerative medicine and tissue engineering. While most of the studies analyze changes in transcriptional profile during differentiation to date there is not much information regarding epigenetic changes in AF-MSCs during differentiation. The aim of our study was to evaluate epigenetic changes during osteogenic differentiation of AF-MS cells. Isolated AF-MSCs were characterized morphologically and osteogenic differentiation was confirmed by cell staining and determining expression of alkaline phosphatase and osteopontin by RT-qPCR. Variation in gene expression levels of pluripotency markers and specific microRNAs were also evaluated. Analysis of epigenetic changes revealed that levels of chromatin modifying enzymes such as Polycomb repressive complex 2 (PRC2) proteins (EZH2 and SUZ12), DNMT1, HDAC1, and HDAC2 were reduced after osteogenic differentiation of AF-MSCs. We demonstrated that the level of specific histone markers keeping active state of chromatin (H3K4me3, H3K9Ac, and others) increased and markers of repressed state of chromatin (H3K27me3) decreased. Our results show that osteogenic differentiation of AF-MSCs is conducted by various epigenetic alterations resulting in global chromatin remodeling and provide insights for further epigenetic investigations in human AF-MSCs.
Collapse
|
27
|
Spitalieri P, Talarico RV, Botta A, Murdocca M, D'Apice MR, Orlandi A, Giardina E, Santoro M, Brancati F, Novelli G, Sangiuolo F. Generation of Human Induced Pluripotent Stem Cells from Extraembryonic Tissues of Fetuses Affected by Monogenic Diseases. Cell Reprogram 2016; 17:275-87. [PMID: 26474030 DOI: 10.1089/cell.2015.0003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The generation of human induced pluripotent stem cells (hiPSCs) derived from an autologous extraembryonic fetal source is an innovative personalized regenerative technology that can transform own-self cells into embryonic stem-like ones. These cells are regarded as a promising candidate for cell-based therapy, as well as an ideal target for disease modeling and drug discovery. Thus, hiPSCs enable researchers to undertake studies for treating diseases or for future applications of in utero therapy. We used a polycistronic lentiviral vector (hSTEMCCA-loxP) encoding OCT4, SOX2, KLF4, and cMYC genes and containing loxP sites, excisible by Cre recombinase, to reprogram patient-specific fetal cells derived from prenatal diagnosis for several genetic disorders, such as myotonic dystrophy type 1 (DM1), β-thalassemia (β-Thal), lymphedema-distichiasis syndrome (LDS), spinal muscular atrophy (SMA), cystic fibrosis (CF), as well as from wild-type (WT) fetal cells. Because cell types tested to create hiPSCs influence both the reprogramming process efficiency and the kinetics, we used chorionic villus (CV) and amniotic fluid (AF) cells, demonstrating how they represent an ideal cell resource for a more efficient generation of hiPSCs. The successful reprogramming of both CV and AF cells into hiPSCs was confirmed by specific morphological, molecular, and immunocytochemical markers and also by their teratogenic potential when inoculated in vivo. We further demonstrated the stability of reprogrammed cells over 10 and more passages and their capability to differentiate into the three embryonic germ layers, as well as into neural cells. These data suggest that hiPSCs-CV/AF can be considered a valid cellular model to accomplish pathogenesis studies and therapeutic applications.
Collapse
Affiliation(s)
- Paola Spitalieri
- 1 Department of Biomedicine and Prevention, Tor Vergata University of Rome , Rome, 00133, Italy
| | - Rosa V Talarico
- 1 Department of Biomedicine and Prevention, Tor Vergata University of Rome , Rome, 00133, Italy
| | - Annalisa Botta
- 1 Department of Biomedicine and Prevention, Tor Vergata University of Rome , Rome, 00133, Italy
| | - Michela Murdocca
- 1 Department of Biomedicine and Prevention, Tor Vergata University of Rome , Rome, 00133, Italy
| | | | - Augusto Orlandi
- 1 Department of Biomedicine and Prevention, Tor Vergata University of Rome , Rome, 00133, Italy
| | - Emiliano Giardina
- 1 Department of Biomedicine and Prevention, Tor Vergata University of Rome , Rome, 00133, Italy .,3 Molecular Genetics Laboratory UILDM , Santa Lucia Foundation, Rome, 00142, Italy
| | | | - Francesco Brancati
- 2 Department of Laboratory Medicine, Policlinic of Tor Vergata , Rome, 00133, Italy
| | - Giuseppe Novelli
- 1 Department of Biomedicine and Prevention, Tor Vergata University of Rome , Rome, 00133, Italy .,2 Department of Laboratory Medicine, Policlinic of Tor Vergata , Rome, 00133, Italy
| | - Federica Sangiuolo
- 1 Department of Biomedicine and Prevention, Tor Vergata University of Rome , Rome, 00133, Italy .,2 Department of Laboratory Medicine, Policlinic of Tor Vergata , Rome, 00133, Italy
| |
Collapse
|
28
|
Jiang G, Herron TJ, Di Bernardo J, Walker KA, O'Shea KS, Kunisaki SM. Human Cardiomyocytes Prior to Birth by Integration-Free Reprogramming of Amniotic Fluid Cells. Stem Cells Transl Med 2016; 5:1595-1606. [PMID: 27465073 DOI: 10.5966/sctm.2016-0016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 06/13/2016] [Indexed: 01/26/2023] Open
Abstract
: The establishment of an abundant source of autologous cardiac progenitor cells would represent a major advance toward eventual clinical translation of regenerative medicine strategies in children with prenatally diagnosed congenital heart disease. In support of this concept, we sought to examine whether functional, transgene-free human cardiomyocytes (CMs) with potential for patient-specific and autologous applications could be reliably generated following routine amniocentesis. Under institutional review board approval, amniotic fluid specimens (8-10 ml) at 20 weeks gestation were expanded and reprogrammed toward pluripotency using nonintegrating Sendai virus (SeV) expressing OCT4, SOX2, cMYC, and KLF4. Following exposure of these induced pluripotent stem cells to cardiogenic differentiation conditions, spontaneously beating amniotic fluid-derived cardiomyocytes (AF-CMs) were successfully generated with high efficiency. After 6 weeks, quantitative gene expression revealed a mixed population of differentiated atrial, ventricular, and nodal AF-CMs, as demonstrated by upregulation of multiple cardiac markers, including MYH6, MYL7, TNNT2, TTN, and HCN4, which were comparable to levels expressed by neonatal dermal fibroblast-derived CM controls. AF-CMs had a normal karyotype and demonstrated loss of NANOG, OCT4, and the SeV transgene. Functional characterization of SIRPA+ AF-CMs showed a higher spontaneous beat frequency in comparison with dermal fibroblast controls but revealed normal calcium transients and appropriate chronotropic responses after β-adrenergic agonist stimulation. Taken together, these data suggest that somatic cells present within human amniotic fluid can be used to generate a highly scalable source of functional, transgene-free, autologous CMs before a child is born. This approach may be ideally suited for patients with prenatally diagnosed cardiac anomalies. SIGNIFICANCE This study presents transgene-free human amniotic fluid-derived cardiomyocytes (AF-CMs) for potential therapy in tissue engineering and regenerative medicine applications. Using 8-10 ml of amniotic fluid harvested at 20 weeks gestation from normal pregnancies, a mixed population of atrial, ventricular, and nodal AF-CMs were reliably generated after Sendai virus reprogramming toward pluripotency. Functional characterization of purified populations of beating AF-CMs revealed normal calcium transients and appropriate chronotropic responses after β-adrenergic agonist stimulation in comparison with dermal fibroblast controls. Because AF-CMs can be generated in fewer than 16 weeks, this approach may be ideally suited for eventual clinical translation at birth in children with prenatally diagnosed cardiac anomalies.
Collapse
Affiliation(s)
- Guihua Jiang
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Pluripotent Stem Cell Laboratory, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Todd J Herron
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Julie Di Bernardo
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kendal A Walker
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Pluripotent Stem Cell Laboratory, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - K Sue O'Shea
- Pluripotent Stem Cell Laboratory, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Shaun M Kunisaki
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Pluripotent Stem Cell Laboratory, University of Michigan Medical School, Ann Arbor, Michigan, USA
- C.S. Mott Children's Hospital and Von Voigtlander Women's Hospital, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
29
|
Wang B, Jakus AE, Baptista PM, Soker S, Soto-Gutierrez A, Abecassis MM, Shah RN, Wertheim JA. Functional Maturation of Induced Pluripotent Stem Cell Hepatocytes in Extracellular Matrix-A Comparative Analysis of Bioartificial Liver Microenvironments. Stem Cells Transl Med 2016; 5:1257-67. [PMID: 27421950 PMCID: PMC4996436 DOI: 10.5966/sctm.2015-0235] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 03/07/2016] [Indexed: 01/12/2023] Open
Abstract
The ability of two three-dimensional bioscaffold systems to reverse the primary limitations of induced pluripotent stem cell (iPSC)-derived hepatocytes was compared. Proliferation and function of iPSC hepatocytes were significantly enhanced when cultured within scaffolds made from extracellular matrix (ECM). This ECM scaffold enhanced phenotypic maturation of iPSC hepatocytes compared with other platforms, likely owing to its biologically diverse makeup. Induced pluripotent stem cells (iPSCs) are new diagnostic and potentially therapeutic tools to model disease and assess the toxicity of pharmaceutical medications. A common limitation of cell lineages derived from iPSCs is a blunted phenotype compared with fully developed, endogenous cells. We examined the influence of novel three-dimensional bioartificial microenvironments on function and maturation of hepatocyte-like cells differentiated from iPSCs and grown within an acellular, liver-derived extracellular matrix (ECM) scaffold. In parallel, we also compared a bioplotted poly-l-lactic acid (PLLA) scaffold that allows for cell growth in three dimensions and formation of cell-cell contacts but is infused with type I collagen (PLLA-collagen scaffold) alone as a “deconstructed” control scaffold with narrowed biological diversity. iPSC-derived hepatocytes cultured within both scaffolds remained viable, became polarized, and formed bile canaliculi-like structures; however, cells grown within ECM scaffolds had significantly higher P450 (CYP2C9, CYP3A4, CYP1A2) mRNA levels and metabolic enzyme activity compared with iPSC hepatocytes grown in either bioplotted PLLA collagen or Matrigel sandwich control culture. Additionally, the rate of albumin synthesis approached the level of primary cryopreserved hepatocytes with lower transcription of fetal-specific genes, α-fetoprotein and CYP3A7, compared with either PLLA-collagen scaffolds or sandwich culture. These studies show that two acellular, three-dimensional culture systems increase the function of iPSC-derived hepatocytes. However, scaffolds derived from ECM alone induced further hepatocyte maturation compared with bioplotted PLLA-collagen scaffolds. This effect is likely mediated by the complex composition of ECM scaffolds in contrast to bioplotted scaffolds, suggesting their utility for in vitro hepatocyte assays or drug discovery. Significance Through the use of novel technology to develop three-dimensional (3D) scaffolds, the present study demonstrated that hepatocyte-like cells derived via induced pluripotent stem cell (iPSC) technology mature on 3D extracellular matrix scaffolds as a result of 3D matrix structure and scaffold biology. The result is an improved hepatic phenotype with increased synthetic and catalytic potency, an improvement on the blunted phenotype of iPSC-derived hepatocytes, a critical limitation of iPSC technology. These findings provide insight into the influence of 3D microenvironments on the viability, proliferation, and function of iPSC hepatocytes to yield a more mature population of cells for cell toxicity studies and disease modeling.
Collapse
Affiliation(s)
- Bo Wang
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Adam E Jakus
- Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, USA Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois, USA
| | - Pedro M Baptista
- Instituto de Investigación Sanitaria de Aragón, Centro de Investigación Biomédica de Aragón, Zaragoza, Spain Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas (CIBERehd), Zaragoza, Spain Fundacion ARAID, Zaragoza, Spain
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Alejandro Soto-Gutierrez
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA Department of Pathology, Thomas E. Starzl Transplantation Institute, and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael M Abecassis
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA Department of Surgery, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ramille N Shah
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, USA Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois, USA Department of Surgery, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA
| | - Jason A Wertheim
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, USA Department of Surgery, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA Department of Surgery, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA
| |
Collapse
|
30
|
Niu X, He W, Song B, Ou Z, Fan D, Chen Y, Fan Y, Sun X. Combining Single Strand Oligodeoxynucleotides and CRISPR/Cas9 to Correct Gene Mutations in β-Thalassemia-induced Pluripotent Stem Cells. J Biol Chem 2016; 291:16576-85. [PMID: 27288406 DOI: 10.1074/jbc.m116.719237] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Indexed: 01/01/2023] Open
Abstract
β-Thalassemia (β-Thal) is one of the most common genetic diseases in the world. The generation of patient-specific β-Thal-induced pluripotent stem cells (iPSCs), correction of the disease-causing mutations in those cells, and then differentiation into hematopoietic stem cells offers a new therapeutic strategy for this disease. Here, we designed a CRISPR/Cas9 to specifically target the Homo sapiens hemoglobin β (HBB) gene CD41/42(-CTTT) mutation. We demonstrated that the combination of single strand oligodeoxynucleotides with CRISPR/Cas9 was capable of correcting the HBB gene CD41/42 mutation in β-Thal iPSCs. After applying a correction-specific PCR assay to purify the corrected clones followed by sequencing to confirm mutation correction, we verified that the purified clones retained full pluripotency and exhibited normal karyotyping. Additionally, whole-exome sequencing showed that the mutation load to the exomes was minimal after CRISPR/Cas9 targeting. Furthermore, the corrected iPSCs were selected for erythroblast differentiation and restored the expression of HBB protein compared with the parental iPSCs. This method provides an efficient and safe strategy to correct the HBB gene mutation in β-Thal iPSCs.
Collapse
Affiliation(s)
- Xiaohua Niu
- From the Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Wenyin He
- From the Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Bing Song
- From the Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Zhanhui Ou
- From the Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Di Fan
- From the Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Yuchang Chen
- From the Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Yong Fan
- From the Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Xiaofang Sun
- From the Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| |
Collapse
|
31
|
Spitalieri P, Talarico VR, Murdocca M, Novelli G, Sangiuolo F. Human induced pluripotent stem cells for monogenic disease modelling and therapy. World J Stem Cells 2016; 8:118-35. [PMID: 27114745 PMCID: PMC4835672 DOI: 10.4252/wjsc.v8.i4.118] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 01/21/2016] [Accepted: 02/14/2016] [Indexed: 02/06/2023] Open
Abstract
Recent and advanced protocols are now available to derive human induced pluripotent stem cells (hiPSCs) from patients affected by genetic diseases. No curative treatments are available for many of these diseases; thus, hiPSCs represent a major impact on patient' health. hiPSCs represent a valid model for the in vitro study of monogenic diseases, together with a better comprehension of the pathogenic mechanisms of the pathology, for both cell and gene therapy protocol applications. Moreover, these pluripotent cells represent a good opportunity to test innovative pharmacological treatments focused on evaluating the efficacy and toxicity of novel drugs. Today, innovative gene therapy protocols, especially gene editing-based, are being developed, allowing the use of these cells not only as in vitro disease models but also as an unlimited source of cells useful for tissue regeneration and regenerative medicine, eluding ethical and immune rejection problems. In this review, we will provide an up-to-date of modelling monogenic disease by using hiPSCs and the ultimate applications of these in vitro models for cell therapy. We consider and summarize some peculiar aspects such as the type of parental cells used for reprogramming, the methods currently used to induce the transcription of the reprogramming factors, and the type of iPSC-derived differentiated cells, relating them to the genetic basis of diseases and to their inheritance model.
Collapse
Affiliation(s)
- Paola Spitalieri
- Paola Spitalieri, Valentina Rosa Talarico, Michela Murdocca, Giuseppe Novelli, Federica Sangiuolo, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Valentina Rosa Talarico
- Paola Spitalieri, Valentina Rosa Talarico, Michela Murdocca, Giuseppe Novelli, Federica Sangiuolo, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Michela Murdocca
- Paola Spitalieri, Valentina Rosa Talarico, Michela Murdocca, Giuseppe Novelli, Federica Sangiuolo, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Giuseppe Novelli
- Paola Spitalieri, Valentina Rosa Talarico, Michela Murdocca, Giuseppe Novelli, Federica Sangiuolo, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Federica Sangiuolo
- Paola Spitalieri, Valentina Rosa Talarico, Michela Murdocca, Giuseppe Novelli, Federica Sangiuolo, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| |
Collapse
|
32
|
Montserrat N, Garreta E, Izpisua Belmonte JC. Regenerative strategies for kidney engineering. FEBS J 2016; 283:3303-24. [DOI: 10.1111/febs.13704] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 01/22/2016] [Accepted: 03/01/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Nuria Montserrat
- Pluripotent Stem Cells and Activation of Endogenous Tissue Programs for Organ Regeneration (PR Lab) Institute for Bioengineering of Catalonia (IBEC) Barcelona Spain
- Networking Biomedical Research Center in Bioengineering Biomaterials and Nanomedicine (CIBER‐BBN) Madrid Spain
| | - Elena Garreta
- Pluripotent Stem Cells and Activation of Endogenous Tissue Programs for Organ Regeneration (PR Lab) Institute for Bioengineering of Catalonia (IBEC) Barcelona Spain
| | | |
Collapse
|
33
|
Qin M, Chen R, Li H, Liang H, Xue Q, Li F, Chen Y, Zhang X. Direct Reprogramming of Human Amniotic Fluid Stem Cells by OCT4 and Application in Repairing of Cerebral Ischemia Damage. Int J Biol Sci 2016; 12:558-68. [PMID: 27019637 PMCID: PMC4807416 DOI: 10.7150/ijbs.11051] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 02/09/2016] [Indexed: 02/02/2023] Open
Abstract
Amniotic fluid stem cells (AFSCs) are a type of fetal stem cell whose stemness encompasses both embryonic and adult stem cells, suggesting that they may be easily and efficiently reprogrammed into induced pluripotent stem cells (iPSCs). To further simplify the reprogramming process, the creation of AFSC-derived iPSCs using a single factor is desirable. Here we report the generation of one-factor human AFSC-iPSCs (AiPSCs) from human AFSCs by ectopic expression of the transcription factor OCT4. Just like human embryonic stem cells, AiPSCs exhibited similar epigenetic status, global gene expression profiles, teratoma formation and in vitro & in vivo pluripotency. Our results indicate that the OCT4 is necessary and sufficient to directly reprogram human AFSCs into pluripotent AiPSCs. Moreover, reflecting the similar memory characteristics of AFSCs and neural stem cells, we show that AiPSC membrane-derived vesicles (MVs) repair cerebral ischemia damage. We anticipate that the successful generation of one-factor AiPSCs will facilitate the creation of patient-specific pluripotent stem cells without the need for transgenic expression of oncogenes. Moreover, MVs from tissue-specific AiPSCs have potential in tissue repair, representing a novel application of iPSCs.
Collapse
|
34
|
Yan X, Xu N, Meng C, Wang B, Yuan J, Wang C, Li Y. Generation of induced pluripotent stem cells from human mesenchymal stem cells of parotid gland origin. Am J Transl Res 2016; 8:419-432. [PMID: 27158336 PMCID: PMC4846893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/01/2016] [Indexed: 06/05/2023]
Abstract
The technology to reprogram human somatic cells to pluripotent state allows the generation of patient-specific induced pluripotent stem cells (iPSCs) and holds a great promise for regenerative medicine and autologous transplantation. Here we, for the first time, identified mesenchymal stem cells isolated from parotid gland (hPMSCs) as a suitable candidate for iPSC production. In the present study, hPMSCs were isolated from parotid gland specimens in patients with squamous cell carcinoma of the oral cavity. The mesenchymal stem cell properties of cultured hPMSCs were confirmed by expression of surface markers and induced differentiation into osteogenic, chondrogenic and adipogenic cell lineages. hPMSCs were then reprogrammed to pluripotent cells by episomal vector-mediated transduction of reprogramming factors (OCT3/4, SOX2, KLF4, c-MYC, LIN28 and TP53 shRNA). The resulting hPMSC-iPSCs showed similar characteristics as human embryonic stem cells (ESCs) with regard to morphology, pluripotent markers, global gene expression, and methylation status of pluripotent cell-specific genes OCT4 and NANOG. These hPMSC-iPSCs were able to differentiate into cells of all three germ layers both in vitro and in vivo. Our results indicate that hPMSCs could be an alternative cell source for generation of iPSCs and have the potential to be used in cell-based regenerative medicine.
Collapse
Affiliation(s)
- Xing Yan
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical UniversityBeijing, PR China
| | - Nuo Xu
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical UniversityBeijing, PR China
| | - Cen Meng
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical UniversityBeijing, PR China
| | - Bianhong Wang
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical UniversityBeijing, PR China
| | - Jinghong Yuan
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical UniversityBeijing, PR China
| | - Caiyun Wang
- Beijing Cellapy Biotechnology Co., LTDBeijing, PR China
| | - Yang Li
- Stem Cell Research Center and Department of Cell Biology, School of Basic Medical Sciences, Peking UniversityBeijing, PR China
| |
Collapse
|
35
|
Abstract
Induced pluripotent stem (iPS) cells have great potential in regenerative medicine, including cell replacement therapies and disease modelling in vitro. However, with this potential comes several challenges, including clinical safety, reprogramming and differentiation efficiency, and compromised functionality of differentiated cell types after transplantation. Many of these issues arise from imprecise control of cell fate. With large-scale sequencing and genome-editing technologies we can now precisely manipulate the genome, which has expanded our knowledge of functional cell types and cell identity. These technologies may improve our efforts in generating iPS-derived therapeutic cells and in development of therapies for human diseases.
Collapse
|
36
|
An Overview of Direct Somatic Reprogramming: The Ins and Outs of iPSCs. Int J Mol Sci 2016; 17:ijms17010141. [PMID: 26805822 PMCID: PMC4730380 DOI: 10.3390/ijms17010141] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/12/2016] [Accepted: 01/13/2016] [Indexed: 02/07/2023] Open
Abstract
Stem cells are classified into embryonic stem cells and adult stem cells. An evolving alternative to conventional stem cell therapies is induced pluripotent stem cells (iPSCs), which have a multi-lineage potential comparable to conventionally acquired embryonic stem cells with the additional benefits of being less immunoreactive and avoiding many of the ethical concerns raised with the use of embryonic material. The ability to generate iPSCs from somatic cells provides tremendous promise for regenerative medicine. The breakthrough of iPSCs has raised the possibility that patient-specific iPSCs can provide autologous cells for cell therapy without the concern for immune rejection. iPSCs are also relevant tools for modeling human diseases and drugs screening. However, there are still several hurdles to overcome before iPSCs can be used for translational purposes. Here, we review the recent advances in somatic reprogramming and the challenges that must be overcome to move this strategy closer to clinical application.
Collapse
|
37
|
The Application of Human iPSCs in Neurological Diseases: From Bench to Bedside. Stem Cells Int 2016; 2016:6484713. [PMID: 26880979 PMCID: PMC4736583 DOI: 10.1155/2016/6484713] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 11/23/2015] [Accepted: 11/26/2015] [Indexed: 12/20/2022] Open
Abstract
In principle, induced pluripotent stem cells (iPSCs) are generated from somatic cells by reprogramming and gaining the capacity to self-renew indefinitely as well as the ability to differentiate into cells of different lineages. Human iPSCs have absolute advantages over human embryonic stem cells (ESCs) and animal models in disease modeling, drug screening, and cell replacement therapy. Since Takahashi and Yamanaka first described in 2007 that iPSCs can be generated from human adult somatic cells by retroviral transduction of the four transcription factors, Oct3/4, Sox2, Klf4, and c-Myc, disease specific iPSC lines have sprung up worldwide like bamboo shoots after a spring rain, making iPSC one of the hottest and fastest moving topics in modern science. The craze for iPSCs has spread throughout main branches of clinical medicine, covering neurology, hematology, cardiology, endocrinology, hepatology, ophthalmology, and so on. Here in this paper, we will focus on the clinical application of human iPSCs in disease modeling, drug screening, and cell replacement therapy for neurological diseases.
Collapse
|
38
|
iPSCs: A Minireview from Bench to Bed, including Organoids and the CRISPR System. Stem Cells Int 2016; 2016:5934782. [PMID: 26880972 PMCID: PMC4736429 DOI: 10.1155/2016/5934782] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 10/19/2015] [Accepted: 10/25/2015] [Indexed: 12/22/2022] Open
Abstract
When Dolly the sheep was born, the first probe into an adult mammalian genome traveling back in time and generating a whole new animal appeared. Ten years later, the reprogramming process became a defined method of producing induced pluripotent stem cells (iPSCs) through the overexpression of four transcription factors. iPSCs are capable of originating virtually all types of cells and tissues, including a whole new animal. The reprogramming strategies based on patient-derived cells should make the development of clinical applications of cell based therapy much more straightforward. Here, we analyze the current state, opportunities, and challenges of iPSCs from bench to bed, including organoids and the CRISPR system.
Collapse
|
39
|
Iizuka H, Kagoya Y, Kataoka K, Yoshimi A, Miyauchi M, Taoka K, Kumano K, Yamamoto T, Hotta A, Arai S, Kurokawa M. Targeted gene correction of RUNX1 in induced pluripotent stem cells derived from familial platelet disorder with propensity to myeloid malignancy restores normal megakaryopoiesis. Exp Hematol 2015; 43:849-57. [DOI: 10.1016/j.exphem.2015.05.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 05/09/2015] [Accepted: 05/10/2015] [Indexed: 12/26/2022]
|
40
|
Qin Y, Gao WQ. Concise Review: Patient-Derived Stem Cell Research for Monogenic Disorders. Stem Cells 2015; 34:44-54. [DOI: 10.1002/stem.2112] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 06/05/2015] [Accepted: 06/20/2015] [Indexed: 12/24/2022]
Affiliation(s)
- Yiren Qin
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine; hanghai Jiao Tong University; Shanghai People's Republic of China
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine; hanghai Jiao Tong University; Shanghai People's Republic of China
- School of Biomedical Engineering & Med-X Research Institute; Shanghai Jiao Tong University; Shanghai People's Republic of China
- Collaborative Innovation Center of Systems Biomedicine; Shanghai Jiao Tong University; Shanghai People's Republic of China
| |
Collapse
|
41
|
Gupta MK, Teo AKK, Rao TN, Bhatt S, Kleinridders A, Shirakawa J, Takatani T, Hu J, De Jesus DF, Windmueller R, Wagers AJ, Kulkarni RN. Excessive Cellular Proliferation Negatively Impacts Reprogramming Efficiency of Human Fibroblasts. Stem Cells Transl Med 2015; 4:1101-8. [PMID: 26253715 DOI: 10.5966/sctm.2014-0217] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 06/22/2015] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED The impact of somatic cell proliferation rate on induction of pluripotent stem cells remains controversial. Herein, we report that rapid proliferation of human somatic fibroblasts is detrimental to reprogramming efficiency when reprogrammed using a lentiviral vector expressing OCT4, SOX2, KLF4, and cMYC in insulin-rich defined medium. Human fibroblasts grown in this medium showed higher proliferation, enhanced expression of insulin signaling and cell cycle genes, and a switch from glycolytic to oxidative phosphorylation metabolism, but they displayed poor reprogramming efficiency compared with cells grown in normal medium. Thus, in contrast to previous studies, our work reveals an inverse correlation between the proliferation rate of somatic cells and reprogramming efficiency, and also suggests that upregulation of proteins in the growth factor signaling pathway limits the ability to induce pluripotency in human somatic fibroblasts. SIGNIFICANCE The efficiency with which human cells can be reprogrammed is of interest to stem cell biology. In this study, human fibroblasts cultured in media containing different concentrations of growth factors such as insulin and insulin-like growth factor-1 exhibited variable abilities to proliferate, with consequences on pluripotency. This occurred in part because of changes in the expression of proteins involved in the growth factor signaling pathway, glycolysis, and oxidative phosphorylation. These findings have implications for efficient reprogramming of human cells.
Collapse
Affiliation(s)
- Manoj K Gupta
- Sections of Islet Cell and Regenerative Biology and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Adrian Kee Keong Teo
- Sections of Islet Cell and Regenerative Biology and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tata Nageswara Rao
- Sections of Islet Cell and Regenerative Biology and Howard Hughes Medical Institute, Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Shweta Bhatt
- Sections of Islet Cell and Regenerative Biology and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andre Kleinridders
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Integrative Physiology and Metabolism, Joslin Diabetes Center, and
| | - Jun Shirakawa
- Sections of Islet Cell and Regenerative Biology and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tomozumi Takatani
- Sections of Islet Cell and Regenerative Biology and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jiang Hu
- Sections of Islet Cell and Regenerative Biology and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dario F De Jesus
- Sections of Islet Cell and Regenerative Biology and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca Windmueller
- Sections of Islet Cell and Regenerative Biology and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Amy J Wagers
- Sections of Islet Cell and Regenerative Biology and Howard Hughes Medical Institute, Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Rohit N Kulkarni
- Sections of Islet Cell and Regenerative Biology and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
42
|
Velho RV, Sperb-Ludwig F, Schwartz IVD. New approaches to the treatment of orphan genetic disorders: Mitigating molecular pathologies using chemicals. AN ACAD BRAS CIENC 2015; 87:1375-88. [PMID: 26247150 DOI: 10.1590/0001-3765201520140711] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
With the advance and popularization of molecular techniques, the identification of genetic mutations that cause diseases has increased dramatically. Thus, the number of laboratories available to investigate a given disorder and the number of subsequent diagnosis have increased over time. Although it is necessary to identify mutations and provide diagnosis, it is also critical to develop specific therapeutic approaches based on this information. This review aims to highlight recent advances in mutation-targeted therapies with chemicals that mitigate mutational pathology at the molecular level, for disorders that, for the most part, have no effective treatment. Currently, there are several strategies being used to correct different types of mutations, including the following: the identification and characterization of translational readthrough compounds; antisense oligonucleotide-mediated splicing redirection; mismatch repair; and exon skipping. These therapies and other approaches are reviewed in this paper.
Collapse
Affiliation(s)
- Renata V Velho
- Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, BR
| | | | | |
Collapse
|
43
|
Arai S, Miyauchi M, Kurokawa M. Modeling of hematologic malignancies by iPS technology. Exp Hematol 2015; 43:654-60. [PMID: 26135030 DOI: 10.1016/j.exphem.2015.06.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 06/16/2015] [Indexed: 01/11/2023]
Abstract
Induced pluripotent stem cells (iPSCs) can be generated from various types of cells with transduction of defined transcription factors. Patient-derived iPSCs are becoming commonly utilized for understanding the molecular pathways involved in disease and for the development of novel targeted therapies. With the use of patient-derived iPSCs differentiated to specific-lineage cells, the potency and toxicity of drug candidates can be evaluated. In the past, patient-derived iPSCs were mainly established from patients of inherited hematologic diseases, followed by the expansion of target to acquired diseases like myeloproliferative neoplasms. Thanks to the rapid development of novel genome editing technologies, we can now utilize genetically modified and unprocessed iPSCs more readily than before. These technologies, which enable us to modulate genetic status or even chromosome structure at the right time, could help the elucidation of pathogenesis of hematologic diseases. If iPSC-derived hematopoietic cells are to be robustly reconstituted in vivo as a consequence of the development of reprogramming and conversion technology, research on leukemic stem cells must be widely promoted. Therefore, iPSC technology has great potential on oncology research using patient samples.
Collapse
Affiliation(s)
- Shunya Arai
- Department of Hematology & Oncology, Graduate School of Medicine, The University of Tokyo, Hongo, Bunkyo-ku; and CREST, Japan Science and Technology Agency, Chiyoda-ku, Tokyo, Japan.
| | - Masashi Miyauchi
- Department of Hematology & Oncology, Graduate School of Medicine, The University of Tokyo, Hongo, Bunkyo-ku; and CREST, Japan Science and Technology Agency, Chiyoda-ku, Tokyo, Japan
| | - Mineo Kurokawa
- Department of Hematology & Oncology, Graduate School of Medicine, The University of Tokyo, Hongo, Bunkyo-ku; and CREST, Japan Science and Technology Agency, Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
44
|
Long-term and efficient expression of human β-globin gene in a hematopoietic cell line using a new site-specific integrating non-viral system. Gene Ther 2015; 22:663-74. [DOI: 10.1038/gt.2015.30] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 03/07/2015] [Accepted: 03/16/2015] [Indexed: 11/08/2022]
|
45
|
Song B, Fan Y, He W, Zhu D, Niu X, Wang D, Ou Z, Luo M, Sun X. Improved hematopoietic differentiation efficiency of gene-corrected beta-thalassemia induced pluripotent stem cells by CRISPR/Cas9 system. Stem Cells Dev 2015; 24:1053-65. [PMID: 25517294 DOI: 10.1089/scd.2014.0347] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The generation of beta-thalassemia (β-Thal) patient-specific induced pluripotent stem cells (iPSCs), subsequent homologous recombination-based gene correction of disease-causing mutations/deletions in the β-globin gene (HBB), and their derived hematopoietic stem cell (HSC) transplantation offers an ideal therapeutic solution for treating this disease. However, the hematopoietic differentiation efficiency of gene-corrected β-Thal iPSCs has not been well evaluated in the previous studies. In this study, we used the latest gene-editing tool, clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated 9 (Cas9), to correct β-Thal iPSCs; gene-corrected cells exhibit normal karyotypes and full pluripotency as human embryonic stem cells (hESCs) showed no off-targeting effects. Then, we evaluated the differentiation efficiency of the gene-corrected β-Thal iPSCs. We found that during hematopoietic differentiation, gene-corrected β-Thal iPSCs showed an increased embryoid body ratio and various hematopoietic progenitor cell percentages. More importantly, the gene-corrected β-Thal iPSC lines restored HBB expression and reduced reactive oxygen species production compared with the uncorrected group. Our study suggested that hematopoietic differentiation efficiency of β-Thal iPSCs was greatly improved once corrected by the CRISPR/Cas9 system, and the information gained from our study would greatly promote the clinical application of β-Thal iPSC-derived HSCs in transplantation.
Collapse
Affiliation(s)
- Bing Song
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University , Guangzhou City, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Orban M, Goedel A, Haas J, Sandrock-Lang K, Gärtner F, Jung CB, Zieger B, Parrotta E, Kurnik K, Sinnecker D, Wanner G, Laugwitz KL, Massberg S, Moretti A. Functional comparison of induced pluripotent stem cell- and blood-derived GPIIbIIIa deficient platelets. PLoS One 2015; 10:e0115978. [PMID: 25607928 PMCID: PMC4301811 DOI: 10.1371/journal.pone.0115978] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 11/28/2014] [Indexed: 12/16/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) represent a versatile tool to model genetic diseases and are a potential source for cell transfusion therapies. However, it remains elusive to which extent patient-specific hiPSC-derived cells functionally resemble their native counterparts. Here, we generated a hiPSC model of the primary platelet disease Glanzmann thrombasthenia (GT), characterized by dysfunction of the integrin receptor GPIIbIIIa, and compared side-by-side healthy and diseased hiPSC-derived platelets with peripheral blood platelets. Both GT-hiPSC-derived platelets and their peripheral blood equivalents showed absence of membrane expression of GPIIbIIIa, a reduction of PAC-1 binding, surface spreading and adherence to fibrinogen. We demonstrated that GT-hiPSC-derived platelets recapitulate molecular and functional aspects of the disease and show comparable behavior to their native counterparts encouraging the further use of hiPSC-based disease models as well as the transition towards a clinical application.
Collapse
Affiliation(s)
- Mathias Orban
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximillians-Universität, Munich, Germany
| | - Alexander Goedel
- I. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Jessica Haas
- I. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Kirstin Sandrock-Lang
- Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Florian Gärtner
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximillians-Universität, Munich, Germany
| | - Christian Billy Jung
- I. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Barbara Zieger
- Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Elvira Parrotta
- I. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany; Department of Experimental and Clinical Medicine, University of Magna Graecia, Medical School, Catanzaro, Italy
| | - Karin Kurnik
- Paediatric Haemophilia Centre, Dr. von Hauner Children's Hospital, Ludwig-Maximillians-Universität, Munich, Germany
| | - Daniel Sinnecker
- I. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Gerhard Wanner
- Ultrastructural Research, Department Biology I, Biozentrum, Ludwig-Maximillians-Universität, Planegg-Martinsried, Germany
| | - Karl-Ludwig Laugwitz
- I. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany; DZHK (German Centre for Cardiovascular Research)-partner site Munich Heart Alliance, Munich, Germany
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximillians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research)-partner site Munich Heart Alliance, Munich, Germany
| | - Alessandra Moretti
- I. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany; DZHK (German Centre for Cardiovascular Research)-partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
47
|
Chintawar S, Graf M, Cader Z. Utility of Human Stem Cells for Drug Discovery. HUMAN-BASED SYSTEMS FOR TRANSLATIONAL RESEARCH 2014. [DOI: 10.1039/9781782620136-00162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The pharmaceutical industry continues to struggle to deliver novel and innovative medicines to the market. One of the major challenges in deriving new therapeutics is to more accurately predict the safety and efficacy of the candidate molecule. The current paradigm of drug discovery has several limitations but perhaps the most conspicuous deficiency is the lack of human-based experimental models. The advent of human embryonic stem cells followed by the discovery of induced pluripotent stem (iPS) cells offers unprecedented opportunities for integrating human cellular assays in drug discovery and development. Human iPS cell lines of many diseases have been obtained and iPSC-derived disease affected cells have been utilised for proof-of-concept drug screens to assess efficacy or potential toxicology. The incorporation of iPSC technology thus provides an invaluable opportunity to reduce drug attrition during the process of drug development.
Collapse
Affiliation(s)
- Satyan Chintawar
- Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford Oxford OX3 9DU UK
| | - Martin Graf
- Roche Pharmaceutical Research and Early Development, Discovery Technologies, Roche Innovation Center Basel 124 Grenzacherstrasse CH 4070 Basel Switzerland
| | - Zameel Cader
- Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford Oxford OX3 9DU UK
| |
Collapse
|
48
|
Generation of Human β-Thalassemia Induced Pluripotent Cell Lines by Reprogramming of Bone Marrow–Derived Mesenchymal Stromal Cells Using Modified mRNA. Cell Reprogram 2014; 16:447-55. [DOI: 10.1089/cell.2014.0050] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
49
|
Wang J, Huang V, Ye L, Bárcena A, Lin G, Lue TF, Li LC. Identification of small activating RNAs that enhance endogenous OCT4 expression in human mesenchymal stem cells. Stem Cells Dev 2014; 24:345-53. [PMID: 25232932 DOI: 10.1089/scd.2014.0290] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Ectopic overexpression of transcription factors has been used to reprogram cell fate. For example, virus-mediated overexpression of four transcription factors OCT4, SOX2, MYC, and KLF4, known as Yamanaka factors, can convert somatic cells to induced pluripotent stem (iPS) cells. However, gene-specific switch-on of endogenous gene production without the use of foreign DNA remains a challenge. The small RNA machinery that comprised small RNAs and Argonaute proteins is known to silence gene expression, but can be repurposed to activate gene expression when directed to gene promoters, a phenomenon known as RNA activation or RNAa. By screening of dsRNAs targeting OCT4 promoter, we identified a small activating RNA (saRNA) that activated OCT4 expression in several types of human mesenchymal stem cells (MSCs). We found that saRNA-induced OCT4 activation can be further enhanced by a histone deacetylase inhibitor, valproic acid. Furthermore, introducing OCT4 saRNA in combination with viruses encoding the remaining three Yamanaka factors (SOX2, MYC, and KLF4) into MSCs led to the derivation of partially reprogrammed iPS cells. Findings from this study suggest that, with further optimization, RNAa can be a powerful tool to reprogram cell fate by inducing the expression of endogenous genes.
Collapse
Affiliation(s)
- Ji Wang
- 1 Department of Urology, University of California , San Francisco, San Francisco, California
| | | | | | | | | | | | | |
Collapse
|
50
|
Jiang G, Di Bernardo J, DeLong CJ, Monteiro da Rocha A, O'Shea KS, Kunisaki SM. Induced Pluripotent Stem Cells from Human Placental Chorion for Perinatal Tissue Engineering Applications. Tissue Eng Part C Methods 2014; 20:731-40. [DOI: 10.1089/ten.tec.2013.0480] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Affiliation(s)
- Guihua Jiang
- From the Consortium for Stem Cell Therapies, C.S. Mott Children's Hospital, Von Voigtlander Women's Hospital, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Surgery, C.S. Mott Children's Hospital, Von Voigtlander Women's Hospital, University of Michigan Medical School, Ann Arbor, Michigan
| | - Julie Di Bernardo
- Department of Surgery, C.S. Mott Children's Hospital, Von Voigtlander Women's Hospital, University of Michigan Medical School, Ann Arbor, Michigan
| | - Cynthia J. DeLong
- From the Consortium for Stem Cell Therapies, C.S. Mott Children's Hospital, Von Voigtlander Women's Hospital, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, C.S. Mott Children's Hospital, Von Voigtlander Women's Hospital, University of Michigan Medical School, Ann Arbor, Michigan
| | - André Monteiro da Rocha
- From the Consortium for Stem Cell Therapies, C.S. Mott Children's Hospital, Von Voigtlander Women's Hospital, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Obstetrics and Gynecology, C.S. Mott Children's Hospital, Von Voigtlander Women's Hospital, University of Michigan Medical School, Ann Arbor, Michigan
| | - K. Sue O'Shea
- From the Consortium for Stem Cell Therapies, C.S. Mott Children's Hospital, Von Voigtlander Women's Hospital, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, C.S. Mott Children's Hospital, Von Voigtlander Women's Hospital, University of Michigan Medical School, Ann Arbor, Michigan
| | - Shaun M. Kunisaki
- From the Consortium for Stem Cell Therapies, C.S. Mott Children's Hospital, Von Voigtlander Women's Hospital, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Surgery, C.S. Mott Children's Hospital, Von Voigtlander Women's Hospital, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Obstetrics and Gynecology, C.S. Mott Children's Hospital, Von Voigtlander Women's Hospital, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|