1
|
Chernyi N, Gavrilova D, Saruhanyan M, Oloruntimehin ES, Karabelsky A, Bezsonov E, Malogolovkin A. Recent Advances in Gene Therapy for Hemophilia: Projecting the Perspectives. Biomolecules 2024; 14:854. [PMID: 39062568 PMCID: PMC11274510 DOI: 10.3390/biom14070854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
One of the well-known X-linked genetic disorders is hemophilia, which could be hemophilia A as a result of a mutation in the F8 (factor VIII) gene or hemophilia B as a result of a mutation in the F9 (factor IX) gene, leading to insufficient levels of the proteins essential for blood coagulation cascade. In patients with severe hemophilia, factor VIII or factor IX activities in the blood plasma are considerably low, estimated to be less than 1%. This is responsible for spontaneous or post-traumatic bleeding episodes, or both, leading to disease complications and death. Current treatment of hemophilia relies on the prevention of bleeding, which consists of expensive lifelong replacement infusion therapy of blood plasma clotting factors, their recombinant versions, or therapy with recombinant monoclonal antibodies. Recently emerged gene therapy approaches may be a potential game changer that could reshape the therapeutic outcomes of hemophilia A or B using a one-off vector in vivo delivery and aim to achieve long-term endogenous expression of factor VIII or IX. This review examines both traditional approaches to the treatment of hemophilia and modern methods, primarily focusing on gene therapy, to update knowledge in this area. Recent technological advances and gene therapeutics in the pipeline are critically reviewed and summarized. We consider gene therapy to be the most promising method as it may overcome the problems associated with more traditional treatments, such as the need for constant and expensive infusions and the presence of an immune response to the antibody drugs used to treat hemophilia.
Collapse
Affiliation(s)
- Nikita Chernyi
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
| | - Darina Gavrilova
- Department of Biology and General Genetics, First Moscow State Medical University (Sechenov University), Moscow 105043, Russia
| | - Mane Saruhanyan
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
| | - Ezekiel S. Oloruntimehin
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
| | - Alexander Karabelsky
- Center for Translational Medicine, Sirius University of Science and Technology, Sochi 354530, Russia
| | - Evgeny Bezsonov
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
- Department of Biology and General Genetics, First Moscow State Medical University (Sechenov University), Moscow 105043, Russia
| | - Alexander Malogolovkin
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
- Center for Translational Medicine, Sirius University of Science and Technology, Sochi 354530, Russia
| |
Collapse
|
2
|
Behl T, Kaur I, Sehgal A, Singh S, Sharma N, Chigurupati S, Felemban SG, Alsubayiel AM, Iqbal MS, Bhatia S, Al-Harrasi A, Bungau S, Mostafavi E. "Cutting the Mustard" with Induced Pluripotent Stem Cells: An Overview and Applications in Healthcare Paradigm. Stem Cell Rev Rep 2022; 18:2757-2780. [PMID: 35793037 DOI: 10.1007/s12015-022-10390-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2022] [Indexed: 12/09/2022]
Abstract
Treatment of numerous ailments has been made accessible by the advent of genetic engineering, where the self-renewal property has unfolded the mysteries of regeneration, i.e., stem cells. This is narrowed down to pluripotency, the cell property of differentiating into other adult cells. The generation of induced pluripotent stem cells (iPSCs) was a major breakthrough in 2006, which was generated by a cocktail of 4 Yamanaka Factors, following which significant advancements have been reported in medical science and therapeutics. The iPSCs are reprogrammed from somatic cells, and the fascinating results focused on developing authentic techniques for their generation via molecular reprogramming mechanisms, with a plethora of molecules, like NANOG, miRNAs, and DNA modifying agents, etc. The iPSCs have exhibited reliable results in assessing the etiology and molecular mechanisms of diseases, followed by the development of possible treatments and the elimination of risks of immune rejection. The authors formulate a comprehensive review to develop a clear understanding of iPSC generation, their advantages and limitations, with potential challenges associated with their medical utility. In addition, a wide compendium of applications of iPSCs in regenerative medicine and disease modeling has been discussed, alongside bioengineering technologies for iPSC reprogramming, expansion, isolation, and differentiation. The manuscript aims to provide a holistic picture of the booming advancement of iPSC therapy, to attract the attention of global researchers, to investigate this versatile approach in treatment of multiple disorders, subsequently overcoming the challenges, in order to effectively expand its therapeutic window.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Shatha Ghazi Felemban
- Department of Medical Laboratory Science, Fakeeh College for Medical Sciences, Jeddah, Kingdom of Saudi Arabia
| | - Amal M Alsubayiel
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Muhammad Shahid Iqbal
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Oman.,School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Ebrahim Mostafavi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA. .,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
3
|
Barbon S, Stocco E, Rajendran S, Zardo L, Macchi V, Grandi C, Tagariello G, Porzionato A, Radossi P, De Caro R, Parnigotto PP. In Vitro Conditioning of Adipose-Derived Mesenchymal Stem Cells by the Endothelial Microenvironment: Modeling Cell Responsiveness towards Non-Genetic Correction of Haemophilia A. Int J Mol Sci 2022; 23:ijms23137282. [PMID: 35806285 PMCID: PMC9266329 DOI: 10.3390/ijms23137282] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 11/24/2022] Open
Abstract
In recent decades, the use of adult multipotent stem cells has paved the way for the identification of new therapeutic approaches for the treatment of monogenic diseases such as Haemophilia A. Being already studied for regenerative purposes, adipose-derived mesenchymal stem cells (Ad-MSCs) are still poorly considered for Haemophilia A cell therapy and their capacity to produce coagulation factor VIII (FVIII) after proper stimulation and without resorting to gene transfection. In this work, Ad-MSCs were in vitro conditioned towards the endothelial lineage, considered to be responsible for coagulation factor production. The cells were cultured in an inductive medium enriched with endothelial growth factors for up to 21 days. In addition to significantly responding to the chemotactic endothelial stimuli, the cell populations started to form capillary-like structures and up-regulated the expression of specific endothelial markers (CD34, PDGFRα, VEGFR2, VE-cadherin, CD31, and vWF). A dot blot protein study detected the presence of FVIII in culture media collected from both unstimulated and stimulated Ad-MSCs. Remarkably, the activated partial thromboplastin time test demonstrated that the clot formation was accelerated, and FVIII activity was enhanced when FVIII deficient plasma was mixed with culture media from the untreated/stimulated Ad-MSCs. Overall, the collected evidence supported a possible Ad-MSC contribution to HA correction via specific stimulation by the endothelial microenvironment and without any need for gene transfection.
Collapse
Affiliation(s)
- Silvia Barbon
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy; (S.B.); (E.S.); (V.M.); (A.P.); (R.D.C.)
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling—TES, Onlus, 35030 Padova, Italy; (C.G.); (P.P.P.)
| | - Elena Stocco
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy; (S.B.); (E.S.); (V.M.); (A.P.); (R.D.C.)
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling—TES, Onlus, 35030 Padova, Italy; (C.G.); (P.P.P.)
| | - Senthilkumar Rajendran
- Department of Surgery Oncology and Gastroenterology, University of Padova, 35124 Padova, Italy;
| | - Lorena Zardo
- Haematology and Haemophilia Centre, Castelfranco Veneto Hospital, 31033 Castelfranco Veneto, Italy; (L.Z.); (G.T.)
| | - Veronica Macchi
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy; (S.B.); (E.S.); (V.M.); (A.P.); (R.D.C.)
| | - Claudio Grandi
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling—TES, Onlus, 35030 Padova, Italy; (C.G.); (P.P.P.)
| | - Giuseppe Tagariello
- Haematology and Haemophilia Centre, Castelfranco Veneto Hospital, 31033 Castelfranco Veneto, Italy; (L.Z.); (G.T.)
| | - Andrea Porzionato
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy; (S.B.); (E.S.); (V.M.); (A.P.); (R.D.C.)
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling—TES, Onlus, 35030 Padova, Italy; (C.G.); (P.P.P.)
| | - Paolo Radossi
- Haematology and Haemophilia Centre, Castelfranco Veneto Hospital, 31033 Castelfranco Veneto, Italy; (L.Z.); (G.T.)
- Correspondence:
| | - Raffaele De Caro
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy; (S.B.); (E.S.); (V.M.); (A.P.); (R.D.C.)
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling—TES, Onlus, 35030 Padova, Italy; (C.G.); (P.P.P.)
| | - Pier Paolo Parnigotto
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling—TES, Onlus, 35030 Padova, Italy; (C.G.); (P.P.P.)
| |
Collapse
|
4
|
Liu M, Yang F, Xu Y. Global Trends of Stem Cell Precision Medicine Research (2018–2022): A Bibliometric Analysis. Front Surg 2022; 9:888956. [PMID: 35813047 PMCID: PMC9260221 DOI: 10.3389/fsurg.2022.888956] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundStem cells are a group of cells that can self-renew and have multiple differentiation capabilities. Shinya Yamanaka first discovered a method to convert somatic cells into pluripotent stem cells in 2006. Stem cell therapy can be summarized into three aspects (regenerative treatment, therapy targeted at stem cells, and establishment of disease models). Disease models are mainly established by induced pluripotent stem cells, and the research of stem cell precision medicine has been promising in recent years. Based on the construction of 3D, patient-specific disease models from pluripotent induced stem cells, proper research on disease development and treatment prognosis can be realized. Bibliometric analysis is an efficient way to quickly understand global trends and hotspots in this field.MethodsA literature search of stem cell precision medicine research from 2018 to 2022 was carried out using the Web of Science Core Collection.VOSviewer, R-bibliometrix, and CiteSpace software programs were employed to perform the bibliometric analysis.ResultsA total of 552 publications were retrieved from 2018 to 2022. Annual publication outputs trended upward and reached a peak of 172 in 2021. The United States contributed the most publications (160, 29.0%) to the field, followed by China (63, 11.4%) and Italy (60, 10.9%). International academic collaborations were active. CANCERS was considered the most productive journal with 18 documents. NATURE was the most co-cited journal with 1860 times citations. The most cited document was entitled “Induced Pluripotent Stem Cells for Cardiovascular Disease Modeling and Precision Medicine: A Scientific Statement From the American Heart Association” with 9 times local citations. “ precision medicine” (n = 89, 12.64%), “personalized medicine” (n = 72, 10.23%), “stem cells” (n = 43, 4.40%), and “induced pluripotent stem cells” (n = 41, 5.82%), “cancer stem cells” (n = 31, 4%), “organoids” (n = 26, 3.69%) were the top 6 frequent keywords.ConclusionThe present study performs a comprehensive investigation concerning stem cell precision medicine (2018–2022) for the first time. This research field is developing, and a deeper exploration of 3D patient-specific organoid disease models is worth more research in the future.
Collapse
|
5
|
Son JS, Park CY, Lee G, Park JY, Kim HJ, Kim G, Chi KY, Woo DH, Han C, Kim SK, Park HJ, Kim DW, Kim JH. Therapeutic correction of hemophilia A using 2D endothelial cells and multicellular 3D organoids derived from CRISPR/Cas9-engineered patient iPSCs. Biomaterials 2022; 283:121429. [DOI: 10.1016/j.biomaterials.2022.121429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 01/26/2022] [Accepted: 02/17/2022] [Indexed: 01/19/2023]
|
6
|
Serrano LJ, de la Torre P, Liras A, Flores AI. Cell therapy for factor V deficiency: An approach based on human decidua mesenchymal stem cells. Biomed Pharmacother 2021; 142:112059. [PMID: 34467894 DOI: 10.1016/j.biopha.2021.112059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 02/08/2023] Open
Abstract
Deficiency of factor V is a congenital autosomal recessive coagulopathy associated with mutations in the F5 gene that results in mild-to-severe bleeding episodes. Factor V is a component of the prothrombinase complex responsible for accelerating conversion of prothrombin to thrombin. At the present time there are no therapeutic factor V concentrates available. This study was designed to lay the preliminary foundations for future cell-based therapy for patients with severe factor V deficiency. The study showed that hepatospheres, which produce coagulation factors VIII, IX, and V, synthetize and store intracellular glycogen and express albumin levels up to 8 times higher than those of undifferentiated cells. Factor IX and factor V gene expression increased significantly in hepatospheres as compared to undifferentiated cells, whereas factor VIII gene expression remained constant. The factor V protein was detected in the hepatospheres´ secretome. Considering the enormous potential of mesenchymal stem cells as therapeutic agents, this study proposes a highly reproducible method to induce differentiation of mesenchymal stem cells from human placenta to factor V-producing hepatospheres. This strategy constitutes a preliminary step towards a curative treatment of factor V deficiency through advanced therapies such as cell therapy.
Collapse
Affiliation(s)
- Luis J Serrano
- Department of Genetic, Physiology and Microbiology, Biology School, Complutense University of Madrid, Spain
| | - Paz de la Torre
- Regenerative Medicine Group, 12 de Octubre Hospital Research Institute, Madrid, Spain
| | - Antonio Liras
- Regenerative Medicine Group, 12 de Octubre Hospital Research Institute, Madrid, Spain; Department of Genetic, Physiology and Microbiology, Biology School, Complutense University of Madrid, Spain.
| | - Ana I Flores
- Regenerative Medicine Group, 12 de Octubre Hospital Research Institute, Madrid, Spain.
| |
Collapse
|
7
|
Ou M, Zhao M, Li C, Tang D, Xu Y, Dai W, Sui W, Zhang Y, Xiang Z, Mo C, Lin H, Dai Y. Single-cell sequencing reveals the potential oncogenic expression atlas of human iPSC-derived cardiomyocytes. Biol Open 2021; 10:10/2/bio053348. [PMID: 33589441 PMCID: PMC7903994 DOI: 10.1242/bio.053348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Human induced pluripotent stem cells (iPSCs) are important source for regenerative medicine. However, the links between pluripotency and oncogenic transformation raise safety issues. To understand the characteristics of iPSC-derived cells at single-cell resolution, we directly reprogrammed two human iPSC lines into cardiomyocytes and collected cells from four time points during cardiac differentiation for single-cell sequencing. We captured 32,365 cells and identified five molecularly distinct clusters that aligned well with our reconstructed differentiation trajectory. We discovered a set of dynamic expression events related to the upregulation of oncogenes and the decreasing expression of tumor suppressor genes during cardiac differentiation, which were similar to the gain-of-function and loss-of-function patterns during oncogenesis. In practice, we characterized the dynamic expression of the TP53 and Yamanaka factor genes (OCT4, SOX2, KLF4 and MYC), which were widely used for human iPSCs lines generation; and revealed the co-occurrence of MYC overexpression and TP53 silencing in some of human iPSC-derived TNNT2+ cardiomyocytes. In summary, our oncogenic expression atlas is valuable for human iPSCs application and the single-cell resolution highlights the clues potentially associated with the carcinogenic risk of human iPSC-derived cells. Summary: The single-cell expression atlas in the cardiomyocytes generated from human iPSCs provide potential carcinogenic information for the clinical application of human iPSC-derived cells.
Collapse
Affiliation(s)
- Minglin Ou
- Central Laboratory, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin 541000, China.,Clinical Medical Research Center, The Second Clinical Medical College, Jinan University, Shenzhen People's Hospital, Shenzhen 518020, China
| | - Min Zhao
- GeneCology Research Centre/Seaweed Research Group, School of Science and Engineering, University of the Sunshine Coast, Queensland 4556, Australia
| | - Chunhong Li
- Guangxi Key laboratory of Metabolic Diseases Research, Central Laboratory of Guilin No. 181 Hospital, Guilin 541002, China.,College of Life Science, Guangxi Normal University, Guilin 541006, China
| | - Donge Tang
- Clinical Medical Research Center, The Second Clinical Medical College, Jinan University, Shenzhen People's Hospital, Shenzhen 518020, China
| | - Yong Xu
- Clinical Medical Research Center, The Second Clinical Medical College, Jinan University, Shenzhen People's Hospital, Shenzhen 518020, China
| | - Weier Dai
- College of Natural Science, University of Texas at Austin, Austin 78712, Texas, USA
| | - Weiguo Sui
- Guangxi Key laboratory of Metabolic Diseases Research, Central Laboratory of Guilin No. 181 Hospital, Guilin 541002, China
| | - Yue Zhang
- Guangxi Key laboratory of Metabolic Diseases Research, Central Laboratory of Guilin No. 181 Hospital, Guilin 541002, China
| | - Zhen Xiang
- Guangxi Key laboratory of Metabolic Diseases Research, Central Laboratory of Guilin No. 181 Hospital, Guilin 541002, China
| | - Chune Mo
- Guangxi Key laboratory of Metabolic Diseases Research, Central Laboratory of Guilin No. 181 Hospital, Guilin 541002, China
| | - Hua Lin
- Guangxi Key laboratory of Metabolic Diseases Research, Central Laboratory of Guilin No. 181 Hospital, Guilin 541002, China
| | - Yong Dai
- Clinical Medical Research Center, The Second Clinical Medical College, Jinan University, Shenzhen People's Hospital, Shenzhen 518020, China .,Guangxi Key laboratory of Metabolic Diseases Research, Central Laboratory of Guilin No. 181 Hospital, Guilin 541002, China
| |
Collapse
|
8
|
Bioengineering hemophilia A-specific microvascular grafts for delivery of full-length factor VIII into the bloodstream. Blood Adv 2020; 3:4166-4176. [PMID: 31851760 DOI: 10.1182/bloodadvances.2019000848] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/15/2019] [Indexed: 01/19/2023] Open
Abstract
Hemophilia A (HA) is a bleeding disorder caused by mutations in the F8 gene encoding coagulation factor VIII (FVIII). Current treatments are based on regular infusions of FVIII concentrates throughout a patient's life. Alternatively, viral gene therapies that directly deliver F8 in vivo have shown preliminary successes. However, hurdles remain, including lack of infection specificity and the inability to deliver the full-length version of F8 due to restricted viral cargo sizes. Here, we developed an alternative nonviral ex vivo gene-therapy approach that enables the overexpression of full-length F8 in patients' endothelial cells (ECs). We first generated HA patient-specific induced pluripotent stem cells (HA-iPSCs) from urine epithelial cells and genetically modified them using a piggyBac DNA transposon system to insert multiple copies of full-length F8. We subsequently differentiated the modified HA-iPSCs into competent ECs with high efficiency, and demonstrated that the cells (termed HA-FLF8-iECs) were capable of producing high levels of FVIII. Importantly, following subcutaneous implantation into immunodeficient hemophilic (SCID-f8ko) mice, we demonstrated that HA-FLF8-iECs were able to self-assemble into vascular networks, and that the newly formed microvessels had the capacity to deliver functional FVIII directly into the bloodstream of the mice, effectively correcting the clotting deficiency. Moreover, our implant maintains cellular confinement, which reduces potential safety concerns and allows effective monitoring and reversibility. We envision that this proof-of-concept study could become the basis for a novel autologous ex vivo gene-therapy approach to treat HA.
Collapse
|
9
|
Rose M, Gao K, Cortez-Toledo E, Agu E, Hyllen AA, Conroy K, Pan G, Nolta JA, Wang A, Zhou P. Endothelial cells derived from patients' induced pluripotent stem cells for sustained factor VIII delivery and the treatment of hemophilia A. Stem Cells Transl Med 2020; 9:686-696. [PMID: 32162786 PMCID: PMC7214661 DOI: 10.1002/sctm.19-0261] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/29/2020] [Indexed: 12/12/2022] Open
Abstract
Hemophilia A (HA) is a bleeding disorder characterized by spontaneous and prolonged hemorrhage. The disease is caused by mutations in the coagulation factor 8 gene (F8) leading to factor VIII (FVIII) deficiency. Since FVIII is primarily produced in endothelial cells (ECs) in a non‐diseased human being, ECs hold great potential for development as a cell therapy for HA. We showed that HA patient‐specific induced pluripotent stem cells (HA‐iPSCs) could provide a renewable supply of ECs. The HA‐iPSC‐derived ECs were transduced with lentiviral vectors to stably express the functional B domain deleted F8 gene, the luciferase gene, and the enhanced green fluorescent protein gene (GFP). When transplanted intramuscularly into neonatal and adult immune deficient mice, the HA‐iPSC‐derived ECs were retained in the animals for at least 10‐16 weeks and maintained their expression of FVIII, GFP, and the endothelial marker CD31, as demonstrated by bioluminescence imaging and immunostaining, respectively. When transplanted into HA mice, these transduced HA‐iPSC‐derived ECs significantly reduced blood loss in a tail‐clip bleeding test and produced therapeutic plasma levels (11.2%‐369.2%) of FVIII. Thus, our studies provide proof‐of‐concept that HA‐iPSC‐derived ECs can serve as a factory to deliver FVIII for the treatment of HA not only in adults but also in newborns.
Collapse
Affiliation(s)
- Melanie Rose
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California
| | - Kewa Gao
- Department of Surgery, University of California Davis Medical Center, Sacramento, California.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California
| | - Elizabeth Cortez-Toledo
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California
| | - Emmanuel Agu
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California
| | - Alicia A Hyllen
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California
| | - Kelsey Conroy
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California
| | - Guangjin Pan
- Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jan A Nolta
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California.,University of California Davis Gene Therapy Center, Sacramento, California
| | - Aijun Wang
- Department of Surgery, University of California Davis Medical Center, Sacramento, California.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California.,Department of Biomedical Engineering, University of California Davis, Davis, California
| | - Ping Zhou
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California.,University of California Davis Gene Therapy Center, Sacramento, California
| |
Collapse
|
10
|
Yasen A, Tuxun T, Apaer S, Li W, Maimaitinijiati Y, Wang H, Aisan M, Aji T, Shao Y, Hao W. Fetal liver stem cell transplantation for liver diseases. Regen Med 2019; 14:703-714. [PMID: 31393226 DOI: 10.2217/rme-2018-0160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Stem cell transplantation exhibited a promising lifesaving therapy for various end-stage liver diseases and could serve as a salvaging bridge until curative methods can be performed. In past decades, mature hepatocytes, liver progenitor cells, mesenchymal stem cells and induced pluripotent stem cells have been practiced in above settings. However, long-term survival rates and continuous proliferation ability of these cells in vivo are unsatisfactory, whereas, fetal liver stem cells (FLSCs), given their unique superiority, may be the best candidate for stem cell transplantation technique. Recent studies have revealed that FLSCs could be used as an attractive genetic therapy or regenerative treatments for inherited metabolic or other hepatic disorders. In this study, we reviewed current status and advancements of FLSCs-based treatment.
Collapse
Affiliation(s)
- Aimaiti Yasen
- Xinjiang Medical University, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China.,State Key Laboratory on Pathogenesis, Prevention & Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, 393 Xin Yi Road, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China.,Department of Liver & Laparoscopic Surgery, Digestive & Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China.,Department of Hepatobiliary and Hydatid Disease, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China
| | - Tuerhongjiang Tuxun
- Department of Liver & Laparoscopic Surgery, Digestive & Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China
| | - Shadike Apaer
- State Key Laboratory on Pathogenesis, Prevention & Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, 393 Xin Yi Road, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China.,Department of Liver & Laparoscopic Surgery, Digestive & Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China
| | - Wending Li
- Xinjiang Medical University, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China.,State Key Laboratory on Pathogenesis, Prevention & Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, 393 Xin Yi Road, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China.,Department of Liver & Laparoscopic Surgery, Digestive & Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China
| | - Yusufukadier Maimaitinijiati
- Xinjiang Medical University, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China.,State Key Laboratory on Pathogenesis, Prevention & Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, 393 Xin Yi Road, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China.,Department of Hepatobiliary and Hydatid Disease, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China
| | - Hui Wang
- State Key Laboratory on Pathogenesis, Prevention & Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, 393 Xin Yi Road, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China
| | - Meiheriayi Aisan
- Xinjiang Medical University, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China
| | - Tuerganaili Aji
- Department of Hepatobiliary and Hydatid Disease, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China
| | - Yingmei Shao
- Department of Hepatobiliary and Hydatid Disease, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China
| | - Wen Hao
- State Key Laboratory on Pathogenesis, Prevention & Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, 393 Xin Yi Road, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China.,Department of Hepatobiliary and Hydatid Disease, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang Uyghur Autonomous Region, PR China
| |
Collapse
|
11
|
ssODN-Mediated In-Frame Deletion with CRISPR/Cas9 Restores FVIII Function in Hemophilia A-Patient-Derived iPSCs and ECs. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:198-209. [PMID: 31261034 PMCID: PMC6610636 DOI: 10.1016/j.omtn.2019.05.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 05/25/2019] [Accepted: 05/27/2019] [Indexed: 12/31/2022]
Abstract
Given that the cDNA of F8 is too large to be packaged into adeno-associated virus (AAV) capsids, gene transfer of some versions of B-domain-deleted F8 (BDD-F8) for hemophilia A (HA) treatment has been attempted with promising results. Here, we describe an efficient gene correction via single-stranded-oligodeoxynucleotide (ssODN)-mediated in-frame deletion within the B domain of F8 with CRISPR/Cas9 in HA-patient-derived induced pluripotent stem cells (HA-iPSCs). The expression and activity of FVIII was restored in corrected HA-iPSC-derived induced endothelial progenitor cells (C-iEPCs) in vitro and in vivo. The bleeding phenotype was rescued in HA mice after C-iEPC infusion. Our results demonstrate an efficient approach for in situ gene correction via introduction of a tiny deletion using ssODN and CRISPR/Cas9 to reframe the F8 transcript and restore FVIII function in HA-iPSC-derived EPCs with potential clinical impact in HA gene therapy. For the first time, we demonstrated in vitro and in vivo the FVIII function that is encoded by the endogenous F8 gene with a partially deleted B domain. This work also suggests an applicable strategy for genetic correction of other gene frameshift mutations.
Collapse
|
12
|
Kumar P, Gao K, Wang C, Pivetti C, Lankford L, Farmer D, Wang A. In Utero Transplantation of Placenta-Derived Mesenchymal Stromal Cells for Potential Fetal Treatment of Hemophilia A. Cell Transplant 2019; 27:130-139. [PMID: 29562772 PMCID: PMC6434487 DOI: 10.1177/0963689717728937] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Hemophilia A (HA) is an X-linked recessive disorder caused by mutations in the factor VIII (FVIII) gene leading to deficient blood coagulation. The current standard of care is frequent infusions of plasma-derived FVIII or recombinant B-domain-deleted FVIII (BDD-FVIII). While this treatment is effective, many patients eventually develop FVIII inhibitors that limit the effectiveness of the infused FVIII. As a monogenic disorder, HA is an ideal target for gene or cell-based therapy. Several studies have investigated allogeneic stem cell therapy targeting in utero or postnatal treatment of HA but have not been successful in completely correcting HA. Autologous in utero transplantation of mesenchymal stem cells is promising for treatment of HA due to the naive immune status of the fetal environment as well as its potential to prevent transplant rejection and long-term FVIII inhibitor formation. HA can be diagnosed by chorionic villus sampling performed during the first trimester (10 to 13 wk) of gestation. In this study, we used an established protocol and isolated placenta-derived mesenchymal stromal cells (PMSCs) from first trimester chorionic villus tissue and transduced them with lentiviral vector encoding the BDD-FVIII gene. We show that gene-modified PMSCs maintain their immunophenotype and multipotency, express, and secrete high levels of active FVIII. PMSCs were then transplanted at embryonic day 14.5 (E14.5) into wild-type fetuses from time-mated pregnant mice. Four days after birth, pups were checked for engraftment, and varying levels of expression of human green fluorescent protein were found in the organs tested. This study shows feasibility of the approach to obtain PMSCs from first trimester chorionic villus tissue, genetically modify them with the FVIII gene, and transplant them in utero for cell-mediated gene therapy of HA. Future studies will involve evaluation of long-term engraftment, phenotypic correction in HA mice, and prevention of FVIII inhibitor development by this approach.
Collapse
Affiliation(s)
- Priyadarsini Kumar
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA
| | - Kewa Gao
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA.,2 Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Chuwang Wang
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA.,2 Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Christopher Pivetti
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA
| | - Lee Lankford
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA
| | - Diana Farmer
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA
| | - Aijun Wang
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
13
|
Olgasi C, Talmon M, Merlin S, Cucci A, Richaud-Patin Y, Ranaldo G, Colangelo D, Di Scipio F, Berta GN, Borsotti C, Valeri F, Faraldi F, Prat M, Messina M, Schinco P, Lombardo A, Raya A, Follenzi A. Patient-Specific iPSC-Derived Endothelial Cells Provide Long-Term Phenotypic Correction of Hemophilia A. Stem Cell Reports 2018; 11:1391-1406. [PMID: 30416049 PMCID: PMC6294075 DOI: 10.1016/j.stemcr.2018.10.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 11/15/2022] Open
Abstract
We generated patient-specific disease-free induced pluripotent stem cells (iPSCs) from peripheral blood CD34+ cells and differentiated them into functional endothelial cells (ECs) secreting factor VIII (FVIII) for gene and cell therapy approaches to cure hemophilia A (HA), an X-linked bleeding disorder caused by F8 mutations. iPSCs were transduced with a lentiviral vector carrying FVIII transgene driven by an endothelial-specific promoter (VEC) and differentiated into bona fide ECs using an optimized protocol. FVIII-expressing ECs were intraportally transplanted in monocrotaline-conditioned non-obese diabetic (NOD) severe combined immune-deficient (scid)-IL2rγ null HA mice generating a chimeric liver with functional human ECs. Transplanted cells engrafted and proliferated in the liver along sinusoids, in the long term showed stable therapeutic FVIII activity (6%). These results demonstrate that the hemophilic phenotype can be rescued by transplantation of ECs derived from HA FVIII-corrected iPSCs, confirming the feasibility of cell-reprogramming strategy in patient-derived cells as an approach for HA gene and cell therapy.
Collapse
Affiliation(s)
- Cristina Olgasi
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy
| | - Maria Talmon
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy
| | - Simone Merlin
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy
| | - Alessia Cucci
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy
| | - Yvonne Richaud-Patin
- Center of Regenerative Medicine in Barcelona (CMRB), Hospital Durans Reynals, Hospitalet de Llobregat, 08908 Barcelona, Spain; Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Gabriella Ranaldo
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy
| | - Donato Colangelo
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy
| | | | | | - Chiara Borsotti
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy
| | - Federica Valeri
- A.O.U. Città della Salute e della Scienza, 10126 Torino, Italy
| | | | - Maria Prat
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy
| | - Maria Messina
- A.O.U. Città della Salute e della Scienza, 10126 Torino, Italy
| | | | - Angelo Lombardo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Milan, Italy; San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Angel Raya
- Center of Regenerative Medicine in Barcelona (CMRB), Hospital Durans Reynals, Hospitalet de Llobregat, 08908 Barcelona, Spain; Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Antonia Follenzi
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy.
| |
Collapse
|
14
|
Hu Z, Wu Y, Zhou M, Wang X, Pang J, Li Z, Feng M, Wang Y, Hu Q, Zhao J, Liu X, Wu L, Liang D. Generation of reporter hESCs by targeting EGFP at the CD144 locus to facilitate the endothelial differentiation. Dev Growth Differ 2018; 60:205-215. [PMID: 29696633 DOI: 10.1111/dgd.12433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/14/2018] [Accepted: 03/19/2018] [Indexed: 12/31/2022]
Abstract
Reporter embryonic stem cell (ESC) lines with tissue-specific reporter genes may contribute to optimizing the differentiation conditions in vitro as well as trafficking transplanted cells in vivo. To optimize and monitor endothelial cell (EC) differentiation specifically, here we targeted the enhanced green fluorescent protein (EGFP) reporter gene at the junction of 5'UTR and exon2 of the endothelial specific marker gene CD144 using TALENs in human ESCs (H9) to generate a EGFP-CD144-reporter ESC line. The reporter cells expressed EGFP and CD144 increasingly and specifically without unexpected effects during the EC differentiation. The EC differentiation protocol was optimized and applied to EC differentiation from hiPSCs, resulting in an efficient and simplified endothelial differentiation approach. Here we created our own optimized and robust protocol for EC differentiation of hESCs and hiPSCs by generating the lineage-specific site-specific integration reporter cell lines, showing great potential to be applied in the fields such as trafficking gene and cell fate in vivo in preclinical animal models.
Collapse
Affiliation(s)
- Zhiqing Hu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Yong Wu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Miaojin Zhou
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Xiaolin Wang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Jialun Pang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Zhuo Li
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Mai Feng
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Yanchi Wang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Qian Hu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Junya Zhao
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Xionghao Liu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Lingqian Wu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Desheng Liang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| |
Collapse
|
15
|
Evaluation of ex vivo produced endothelial progenitor cells for autologous transplantation in primates. Stem Cell Res Ther 2018; 9:14. [PMID: 29357928 PMCID: PMC5778763 DOI: 10.1186/s13287-018-0769-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 01/03/2018] [Accepted: 01/04/2018] [Indexed: 02/05/2023] Open
Abstract
Background Autologous transplantation of endothelial progenitor cells (EPCs) is a promising therapeutic approach in the treatment of various vascular diseases. We previously reported a two-step culture system for scalable generation of human EPCs derived from cord blood CD34+ cells ex vivo. Here, we now apply this culture system to expand and differentiate human and nonhuman primate EPCs from mobilized peripheral blood (PB) CD34+ cells for the therapeutic potential of autologous transplantation. Methods The human and nonhuman primate EPCs from mobilized PB CD34+ cells were cultured according to our previously reported system. The generated adherent cells were then characterized by the morphology, surface markers, nitric oxide (NO)/endothelial NO synthase (eNOS) levels and Dil-acetylated low-density lipoprotein (Dil-Ac-LDL) uptake/fluorescein isothiocyanate (FITC)-lectin binding actives. Furthermore, the efficacy and safety studies were performed by autologous transplantation via hepatic portal vein injection in a nonhuman primate model with acute liver sinusoidal endothelial cell injury. Results The mobilized PB CD34+ cells from both human and nonhuman primate were efficiently expanded and differentiated. Over 2 × 108 adherent cells were generated from 20 mL mobilized primate PB (1.51 × 106 ± 3.39 × 105 CD34+ cells) by 36-day culture and more than 80% of the produced cells were identified as EPCs/endothelial cells (ECs). In the autologous transplant model, the injected EPC/ECs from nonhuman primate PB were scattered in the intercellular spaces of hepatocytes at the hepatic tissues 14 days post-transplantation, indicating successful migration and reconstitution in the liver structure as the functional EPCs/ECs. Conclusions We successfully applied our previous two-step culture system for the generation of primate EPCs from mobilized PB CD34+ cells, evaluated the phenotypes ex vivo, and transplanted autologous EPCs/ECs in a nonhuman primate model. Our study indicates that it may be possible for these ex-vivo high-efficient expanded EPCs to be used in clinical cell therapy.
Collapse
|
16
|
Tatsumi K, Okano T. Hepatocyte Transplantation: Cell Sheet Technology for Liver Cell Transplantation. CURRENT TRANSPLANTATION REPORTS 2017; 4:184-192. [PMID: 28932649 PMCID: PMC5577064 DOI: 10.1007/s40472-017-0156-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Purpose of Review We will review the recent developments of cell sheet technology as a feasible tissue engineering approach. Specifically, we will focus on the technological advancement for engineering functional liver tissue using cell sheet technology, and the associated therapeutic effect of cell sheets for liver diseases, highlighting hemophilia. Recent Findings Cell-based therapies using hepatocytes have recently been explored as a new therapeutic modality for patients with many forms of liver disease. We have developed a cell sheet technology, which allows cells to be harvested in a monolithic layer format. We have succeeded in fabricating functional liver tissues in mice by stacking the cell sheets composed of primary hepatocytes. As a curative measure for hemophilia, we have also succeeded in treating hemophilia mice by transplanting of cells sheets composed of genetically modified autologous cells. Summary Tissue engineering using cell sheet technology provides the opportunity to create new therapeutic options for patients with various types of liver diseases.
Collapse
Affiliation(s)
- Kohei Tatsumi
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka 589-8511 Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan.,Cell Sheet Tissue Engineering Center and Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112 USA
| |
Collapse
|
17
|
Junqueira Reis LC, Picanço-Castro V, Paes BCMF, Pereira OA, Gerdes Gyuricza I, de Araújo FT, Morato-Marques M, Moreira LF, Costa EDBO, dos Santos TPM, Covas DT, Pereira Carramaschi LDV, Russo EMDS. Induced Pluripotent Stem Cell for the Study and Treatment of Sickle Cell Anemia. Stem Cells Int 2017; 2017:7492914. [PMID: 28814957 PMCID: PMC5549510 DOI: 10.1155/2017/7492914] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/05/2017] [Indexed: 12/14/2022] Open
Abstract
Sickle cell anemia (SCA) is a monogenic disease of high mortality, affecting millions of people worldwide. There is no broad, effective, and safe definitive treatment for SCA, so the palliative treatments are the most used. The establishment of an in vitro model allows better understanding of how the disease occurs, besides allowing the development of more effective tests and treatments. In this context, iPSC technology is a powerful tool for basic research and disease modeling, and a promise for finding and screening more effective and safe drugs, besides the possibility of use in regenerative medicine. This work obtained a model for study and treatment of SCA using iPSC. Then, episomal vectors were used for reprogramming peripheral blood mononuclear cells to obtain integration-free iPSC. Cells were collected from patients treated with hydroxyurea and without treatment. The iPSCP Bscd lines were characterized for pluripotent and differentiation potential. The iPSC lines were differentiated into HSC, so that we obtained a dynamic and efficient protocol of CD34+CD45+ cells production. We offer a valuable tool for a better understanding of how SCA occurs, in addition to making possible the development of more effective drugs and treatments and providing better understanding of widely used treatments, such as hydroxyurea.
Collapse
Affiliation(s)
- Luiza Cunha Junqueira Reis
- Pharmaceutical Sciences School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
- Blood Center Foundation of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Virgínia Picanço-Castro
- Blood Center Foundation of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Bárbara Cristina Martins Fernandes Paes
- Blood Center Foundation of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
- Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Olívia Ambrozini Pereira
- Philosophy, Sciences and Languages School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | | | | | | | | | - Dimas Tadeu Covas
- Blood Center Foundation of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
- Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | - Elisa Maria de Sousa Russo
- Pharmaceutical Sciences School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
- Blood Center Foundation of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
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
|
Qin M, Guan X, Wang H, Zhang Y, Shen B, Zhang Q, Dai W, Ma Y, Jiang Y. An effective ex-vivo approach for inducing endothelial progenitor cells from umbilical cord blood CD34 + cells. Stem Cell Res Ther 2017; 8:25. [PMID: 28173870 PMCID: PMC5297174 DOI: 10.1186/s13287-017-0482-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 01/10/2017] [Accepted: 01/14/2017] [Indexed: 11/11/2022] Open
Abstract
Background Transplantation of endothelial progenitor cells (EPCs)/endothelial cells (ECs) has been used for the treatment of ischemic diseases and hemophilia A, due to their great capacity for producing factor VIII and for repairing vascular damage. We established an effective approach to stimulate the expansion and differentiation of EPCs for potential therapeutic applications. Methods CD34+ cells isolated from human cord blood were cultured in a two-step system for 21 days. The generated adherent cells were characterized via flow cytometry and immunofluorescent staining. Moreover, single-cell clonogenic and tube-forming assays were carried out to evaluate their potential to proliferate and form vessel networks. Furthermore, these cells were transplanted into a mouse model of hepatic sinusoidal endothelium injury by hepatic portal vein injection to investigate their in-vivo behavior. Results The two-step culture protocol promoted the expansion and differentiation of human cord blood CD34+ cells efficiently, resulting in a large number of adherent cells within 3 weeks. The generated adherent cells were identified as EPCs/ECs based on the expression of CD31, CD144, vWF, and FVIII, and cell numbers showed a 1400-fold increase compared with the initial number. Moreover, these EPCs/ECs were capable of proliferating and establishing colonies as individual cells, and forming tube-like structures. More significantly, tissue examination of mice after transplantation revealed that the injected EPCs/ECs migrated and integrated into the liver, reconstituting the sinusoidal endothelial compartment. Conclusions We developed an approach for the generation of cord blood-derived EPCs/ECs on a large scale, characterized them phenotypically, and demonstrated their in-vivo functional capacity. Our approach provides an excellent source of healthy EPCs/ECs for use in cell therapy in a clinical setting.
Collapse
Affiliation(s)
- Meng Qin
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,Biopharmagen Corp., Suzhou, China
| | - Xin Guan
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Huihui Wang
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,Biopharmagen Corp., Suzhou, China
| | - Yu Zhang
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Bin Shen
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Qingyu Zhang
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,School of Public Health, University at Albany, Albany, NY, USA
| | - Wei Dai
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, NY, USA
| | - Yupo Ma
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,Department of Pathology, BST-9C, The State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Yongping Jiang
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China. .,Biopharmagen Corp., Suzhou, China.
| |
Collapse
|
20
|
Abstract
Despite decades of basic research, biliary diseases remain prevalent, highly morbid, and notoriously difficult to treat. We have, however, dramatically increased our understanding of biliary developmental biology, cholangiocyte pathophysiology, and the endogenous mechanisms of biliary regeneration and repair. All of this complex and rapidly evolving knowledge coincides with an explosion of new technological advances in the area of regenerative medicine. New breakthroughs such as induced pluripotent stem cells and organoid culture are increasingly being applied to the biliary system; it is only a matter of time until new regenerative therapeutics for the cholangiopathies are unveiled. In this review, the authors integrate what is known about biliary development, regeneration, and repair, and link these conceptual advances to the technological breakthroughs that are collectively driving the emergence of a new global field in biliary regenerative medicine.
Collapse
Affiliation(s)
- Thiago M. De Assuncao
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN,Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN
| | - Nidhi Jalan-Sakrikar
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN,Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN
| | - Robert C. Huebert
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN,Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN,Center for Cell Signaling in Gastroenterology; Mayo Clinic and Foundation, Rochester, MN
| |
Collapse
|
21
|
Aznar J, Tudela J. Diez años desde el descubrimiento de las células iPS: estado actual de su aplicación clínica. Rev Clin Esp 2017; 217:30-34. [DOI: 10.1016/j.rce.2016.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 06/29/2016] [Accepted: 08/17/2016] [Indexed: 02/02/2023]
|
22
|
Aznar J, Tudela J. Ten years since the discovery of iPS cells: The current state of their clinical application. Rev Clin Esp 2017. [DOI: 10.1016/j.rceng.2016.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
23
|
Hedgehog Signaling Overcomes an EZH2-Dependent Epigenetic Barrier to Promote Cholangiocyte Expansion. PLoS One 2016; 11:e0168266. [PMID: 27936185 PMCID: PMC5148157 DOI: 10.1371/journal.pone.0168266] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/29/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND & AIMS Developmental morphogens play an important role in coordinating the ductular reaction and portal fibrosis occurring in the setting of cholangiopathies. However, little is known about how membrane signaling events in ductular reactive cells (DRCs) are transduced into nuclear transcriptional changes to drive cholangiocyte maturation and matrix deposition. Therefore, the aim of this study was to investigate potential mechanistic links between cell signaling events and epigenetic regulators in DRCs. METHODS Using directed differentiation of induced pluripotent stem cells (iPSC), isolated DRCs, and in vivo models, we examine the mechanisms whereby sonic hedgehog (Shh) overcomes an epigenetic barrier in biliary precursors and promotes both cholangiocyte maturation and deposition of fibronectin (FN). RESULTS We demonstrate, for the first time, that Gli1 influences the differentiation state and fibrogenic capacity of iPSC-derived hepatic progenitors and isolated DRCs. We outline a novel pathway wherein Shh-mediated Gli1 binding in key cholangiocyte gene promoters overcomes an epigenetic barrier conferred by the polycomb protein, enhancer of zeste homolog 2 (EZH2) and initiates the transcriptional program of cholangiocyte maturation. We also define previously unknown functional Gli1 binding sites in the promoters of cytokeratin (CK)7, CK19, and FN. Our in vivo results show that EZH2 KO mice fed the choline-deficient, ethanolamine supplemented (CDE) diet have an exaggerated cholangiocyte expansion associated with more robust ductular reaction and increased peri-portal fibrosis. CONCLUSION We conclude that Shh/Gli1 signaling plays an integral role in cholangiocyte maturation in vitro by overcoming an EZH2-dependent epigenetic barrier and this mechanism also promotes biliary expansion in vivo.
Collapse
|
24
|
TGF-β1 along with other platelet contents augments Treg cells to suppress anti-FVIII immune responses in hemophilia A mice. Blood Adv 2016; 1:139-151. [PMID: 28164173 DOI: 10.1182/bloodadvances.2016001453] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Platelets are a rich source of many cytokines and chemokines including transforming growth factor β 1 (TGF-β1). TGF-β1 is required to convert conventional CD4+ T (Tconv) cells into induced regulatory T (iTreg) cells that express the transcription factor Foxp3. Whether platelet contents will affect Treg cell properties has not been explored. In this study, we show that unfractionated platelet lysates (pltLys) containing TGF-β1 efficiently induced Foxp3 expression in Tconv cells. The common Treg cell surface phenotype and in vitro suppressive activity of unfractionated pltLys-iTreg cells were similar to those of iTreg cells generated using purified TGF-β1 (purTGFβ-iTreg) cells. However, there were substantial differences in gene expression between pltLys-iTreg and purTGFβ-iTreg cells, especially in granzyme B, interferon γ, and interleukin-2 (a 30.99-, 29.18-, and 17.94-fold difference, respectively) as determined by gene microarray analysis. In line with these gene signatures, we found that pltLys-iTreg cells improved cell recovery after transfer and immune suppressive function compared with purTGFβ-iTreg cells in factor VIII (FVIII)-deficient (F8null, hemophilia A model) mice after recombinant human FVIII (rhF8) infusion. Acute antibody-mediated platelet destruction in F8null mice followed by rhF8 infusion increased the number of Treg cells and suppressed the antibody response to rhF8. Consistent with these data, ex vivo proliferation of F8-specific Treg cells from platelet-depleted animals increased when restimulated with rhF8. Together, our data suggest that pltLys-iTreg cells may have advantages in emerging clinical applications and that platelet contents impact the properties of iTreg cells induced by TGF-β1.
Collapse
|
25
|
Kasuda S, Tatsumi K, Sakurai Y, Shima M, Hatake K. Therapeutic approaches for treating hemophilia A using embryonic stem cells. Hematol Oncol Stem Cell Ther 2016; 9:64-70. [PMID: 27131224 DOI: 10.1016/j.hemonc.2016.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 04/09/2016] [Indexed: 01/14/2023] Open
Abstract
Hemophilia A is an X-linked rescessive bleeding disorder that results from F8 gene aberrations. Previously, we established embryonic stem (ES) cells (tet-226aa/N6-Ainv18) that secrete human factor VIII (hFVIII) by introducing the human F8 gene in mouse Ainv18 ES cells. Here, we explored the potential of cell transplantation therapy for hemophilia A using the ES cells. Transplant tet-226aa/N6-Ainv18 ES cells were injected into the spleens of severe combined immunodeficiency (SCID) mice, carbon tetrachloride (CCl4)-pretreated wild-type mice, and CCl4-pretreated hemophilia A mice. F8 expression was induced by doxycycline in drinking water, and hFVIII-antigen production was assessed in all cell transplantation experiments. Injecting the ES cells into SCID mice resulted in an enhanced expression of the hFVIII antigen; however, teratoma generation was confirmed in the spleen. Transplantation of ES cells into wild-type mice after CCl4-induced liver injury facilitated survival and engraftment of transplanted cells without teratoma formation, resulting in hFVIII production in the plasma. Although CCl4 was lethal to most hemophilia A mice, therapeutic levels of FVIII activity, as well as the hFVIII antigen, were detected in surviving hemophilia A mice after cell transplantation. Immunolocalization results for hFVIII suggested that transplanted ES cells might be engrafted at the periportal area in the liver. Although the development of a safer induction method for liver regeneration is required, our results suggested the potential for developing an effective ES-cell transplantation therapeutic model for treating hemophilia A in the future.
Collapse
Affiliation(s)
- Shogo Kasuda
- Department of Legal Medicine, Nara Medical University School of Medicine, Kashihara, Japan
| | - Kohei Tatsumi
- Department of Pediatrics, Nara Medical University School of Medicine, Kashihara, Japan
| | - Yoshihiko Sakurai
- Department of Pediatrics, Nara Medical University School of Medicine, Kashihara, Japan; Department of Pediatrics, Matsubara Tokushukai Hospital, Matsubara, Japan.
| | - Midori Shima
- Department of Pediatrics, Nara Medical University School of Medicine, Kashihara, Japan
| | - Katsuhiko Hatake
- Department of Legal Medicine, Nara Medical University School of Medicine, Kashihara, Japan
| |
Collapse
|
26
|
Martin F, Gutierrez-Guerrero A, Sánchez S, Galvani G, Benabdellah K. Genome editing: An alternative to retroviral vectors for Wiskott-Aldrich Syndrome (WAS) Gene Therapy? Expert Opin Orphan Drugs 2016. [DOI: 10.1517/21678707.2016.1142870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
27
|
Wu Y, Hu Z, Li Z, Pang J, Feng M, Hu X, Wang X, Lin-Peng S, Liu B, Chen F, Wu L, Liang D. In situ genetic correction of F8 intron 22 inversion in hemophilia A patient-specific iPSCs. Sci Rep 2016; 6:18865. [PMID: 26743572 PMCID: PMC4705535 DOI: 10.1038/srep18865] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 11/27/2015] [Indexed: 11/09/2022] Open
Abstract
Nearly half of severe Hemophilia A (HA) cases are caused by F8 intron 22 inversion (Inv22). This 0.6-Mb inversion splits the 186-kb F8 into two parts with opposite transcription directions. The inverted 5' part (141 kb) preserves the first 22 exons that are driven by the intrinsic F8 promoter, leading to a truncated F8 transcript due to the lack of the last 627 bp coding sequence of exons 23-26. Here we describe an in situ genetic correction of Inv22 in patient-specific induced pluripotent stem cells (iPSCs). By using TALENs, the 627 bp sequence plus a polyA signal was precisely targeted at the junction of exon 22 and intron 22 via homologous recombination (HR) with high targeting efficiencies of 62.5% and 52.9%. The gene-corrected iPSCs retained a normal karyotype following removal of drug selection cassette using a Cre-LoxP system. Importantly, both F8 transcription and FVIII secretion were rescued in the candidate cell types for HA gene therapy including endothelial cells (ECs) and mesenchymal stem cells (MSCs) derived from the gene-corrected iPSCs. This is the first report of an efficient in situ genetic correction of the large inversion mutation using a strategy of targeted gene addition.
Collapse
Affiliation(s)
- Yong Wu
- State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.,Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhiqing Hu
- State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Zhuo Li
- State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Jialun Pang
- State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Mai Feng
- State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Xuyun Hu
- State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Xiaolin Wang
- State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | | | - Bo Liu
- State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Fangping Chen
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lingqian Wu
- State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.,Hunan Jiahui Genetics Hospital, Changsha, Hunan, China
| | - Desheng Liang
- State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| |
Collapse
|
28
|
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: 15] [Impact Index Per Article: 1.9] [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
|
29
|
Reprogramming of Human Fibroblasts to Induced Pluripotent Stem Cells with Sleeping Beauty Transposon-Based Stable Gene Delivery. Methods Mol Biol 2016; 1400:419-27. [PMID: 26895068 DOI: 10.1007/978-1-4939-3372-3_26] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human induced pluripotent stem (iPS) cells are a source of patient-specific pluripotent stem cells and resemble human embryonic stem (ES) cells in gene expression profiles, morphology, pluripotency, and in vitro differentiation potential. iPS cells are applied in disease modeling, drug screenings, toxicology screenings, and autologous cell therapy. In this protocol, we describe how to derive human iPS cells from fibroblasts by Sleeping Beauty (SB) transposon-mediated gene transfer of reprogramming factors. First, the components of the non-viral Sleeping Beauty transposon system, namely a transposon vector encoding reprogramming transcription factors and a helper plasmid expressing the SB transposase, are electroporated into human fibroblasts. The reprogramming cassette undergoes transposition from the transfected plasmids into the fibroblast genome, thereby resulting in stable delivery of the reprogramming factors. Reprogramming by using this protocol takes ~4 weeks, after which the iPS cells are isolated and clonally propagated.
Collapse
|
30
|
Oceguera-Yanez F, Kim SI, Matsumoto T, Tan GW, Xiang L, Hatani T, Kondo T, Ikeya M, Yoshida Y, Inoue H, Woltjen K. Engineering the AAVS1 locus for consistent and scalable transgene expression in human iPSCs and their differentiated derivatives. Methods 2015; 101:43-55. [PMID: 26707206 DOI: 10.1016/j.ymeth.2015.12.012] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 12/14/2015] [Accepted: 12/16/2015] [Indexed: 11/30/2022] Open
Abstract
The potential use of induced pluripotent stem cells (iPSCs) in personalized regenerative medicine applications may be augmented by transgenics, including the expression of constitutive cell labels, differentiation reporters, or modulators of disease phenotypes. Thus, there is precedence for reproducible transgene expression amongst iPSC sub-clones with isogenic or diverse genetic backgrounds. Using virus or transposon vectors, transgene integration sites and copy numbers are difficult to control, and nearly impossible to reproduce across multiple cell lines. Moreover, randomly integrated transgenes are often subject to pleiotropic position effects as a consequence of epigenetic changes inherent in differentiation, undermining applications in iPSCs. To address this, we have adapted popular TALEN and CRISPR/Cas9 nuclease technologies in order to introduce transgenes into pre-defined loci and overcome random position effects. AAVS1 is an exemplary locus within the PPP1R12C gene that permits robust expression of CAG promoter-driven transgenes. Gene targeting controls transgene copy number such that reporter expression patterns are reproducible and scalable by ∼2-fold. Furthermore, gene expression is maintained during long-term human iPSC culture and in vitro differentiation along multiple lineages. Here, we outline our AAVS1 targeting protocol using standardized donor vectors and construction methods, as well as provide practical considerations for iPSC culture, drug selection, and genotyping.
Collapse
Affiliation(s)
- Fabian Oceguera-Yanez
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Shin-Il Kim
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Tomoko Matsumoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Ghee Wan Tan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Long Xiang
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan; iPS Portal Inc., Kyoto 602-0841, Japan
| | - Takeshi Hatani
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Takayuki Kondo
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Makoto Ikeya
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Yoshinori Yoshida
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Knut Woltjen
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan; Hakubi Center for Advanced Research, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
31
|
Wu YM, Huang YJ, Chen P, Hsu YC, Lin SW, Lai HS, Lee HS. Hepatocyte-Like Cells Derived From Mouse Induced Pluripotent Stem Cells Produce Functional Coagulation Factor IX in a Hemophilia B Mouse Model. Cell Transplant 2015; 25:1237-46. [PMID: 26395869 DOI: 10.3727/096368915x689541] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hemophilia B (HB) is an inherited deficiency in coagulation factor IX (FIX) that leads to prolonged bleeding after injury. Although hepatocyte transplantation has been demonstrated to be an effective therapeutic strategy for HB, the quality and sources of hepatocytes still limit their application. Recently, stem cells were proposed as an alternative source of donor cells for cell-based therapy. Much research has been devoted to the properties of stem cells that can be differentiated into functional hepatocytes, thereby providing a new cell source for cell-based therapy. Induced pluripotent stem cells (iPSCs) represent a renewable source of hepatocytes for cell-based therapy; these cells exhibit pluripotency and differentiation ability and can be derived from somatic cells. These iPSCs are highly similar to embryonic stem cells (ESCs). We hypothesized that hepatocyte-like cells derived from iPSCs would have therapeutic efficiency in a mouse model of HB. To test this hypothesis, we differentiated iPSCs toward hepatocytes by stepwise protocol and then transplanted these cells into HB mice. We found that these cells shared many characteristics with hepatocytes, such as albumin synthesis, metabolic capacity, glycogen storage, and ureagenesis. Moreover, iPSC-derived hepatocyte transplantation led to increased coagulation factor IX activity, improved thrombus generation, and better hemostasis parameters, and the transferred cells were localized in the liver in recipient HB mice. In conclusion, our results clearly demonstrate that hepatocyte-like cells derived from iPSCs represent a potential cell source for cell-based therapy in the treatment of HB.
Collapse
Affiliation(s)
- Yao-Ming Wu
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
32
|
Li X, Zhang J, Zhang L, Cheng T, Zhang X. [Research advances on gene therapy for hemophilia A]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2015; 36:620-5. [PMID: 26304093 PMCID: PMC7342641 DOI: 10.3760/cma.j.issn.0253-2727.2015.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Indexed: 11/22/2022]
Affiliation(s)
- Xiaolan Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, CAMS & PUMC, Tianjin 300020, China
| | - Jianping Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, CAMS & PUMC, Tianjin 300020, China
| | - Lei Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, CAMS & PUMC, Tianjin 300020, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, CAMS & PUMC, Tianjin 300020, China
| | - Xiaobing Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, CAMS & PUMC, Tianjin 300020, China
| |
Collapse
|
33
|
Ohmori T, Mizukami H, Ozawa K, Sakata Y, Nishimura S. New approaches to gene and cell therapy for hemophilia. J Thromb Haemost 2015; 13 Suppl 1:S133-42. [PMID: 26149014 DOI: 10.1111/jth.12926] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Hemophilia is considered suitable for gene therapy because it is caused by a single gene abnormality, and therapeutic coagulation factor levels may vary across a broad range. Recent success of hemophilia B gene therapy with an adeno-associated virus (AAV) vector in a clinical trial showed the real prospect that, through gene therapy, a cure for hemophilia may become a reality. However, AAV-mediated gene therapy is not applicable to patients with hemophilia A at present, and neutralizing antibodies against AAV reduce the efficacy of AAV-mediated strategies. Because patients that benefit from AAV treatment (hemophilia B without neutralizing antibodies) are estimated to represent only 15% of total patients with hemophilia, the development of basic technologies for hemophilia A and those that result in higher therapeutic effects are critical. In this review, we present an outline of gene therapy methods for hemophilia, including the transition of technical developments thus far and our novel techniques.
Collapse
Affiliation(s)
- T Ohmori
- Research Division of Cell and Molecular Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - H Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - K Ozawa
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Y Sakata
- Research Division of Cell and Molecular Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - S Nishimura
- Research Division of Cell and Molecular Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
- Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan
- Translational Systems Biology and Medicine Initiative, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
34
|
De Assuncao TM, Sun Y, Jalan-Sakrikar N, Drinane M, Huang BQ, Li Y, Davila JI, Wang R, O’Hara SP, Lomberk GA, Urrutia RA, Ikeda Y, Huebert RC. Development and characterization of human-induced pluripotent stem cell-derived cholangiocytes. J Transl Med 2015; 95:684-96. [PMID: 25867762 PMCID: PMC4447567 DOI: 10.1038/labinvest.2015.51] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/26/2015] [Accepted: 02/04/2015] [Indexed: 12/25/2022] Open
Abstract
Cholangiocytes are the target of a heterogeneous group of liver diseases known as the cholangiopathies. An evolving understanding of the mechanisms driving biliary development provides the theoretical underpinnings for rational development of induced pluripotent stem cell (iPSC)-derived cholangiocytes (iDCs). Therefore, the aims of this study were to develop an approach to generate iDCs and to fully characterize the cells in vitro and in vivo. Human iPSC lines were generated by forced expression of the Yamanaka pluripotency factors. We then pursued a stepwise differentiation strategy toward iDCs, using precise temporal exposure to key biliary morphogens, and we characterized the cells, using a variety of morphologic, molecular, cell biologic, functional, and in vivo approaches. Morphology shows a stepwise phenotypic change toward an epithelial monolayer. Molecular analysis during differentiation shows appropriate enrichment in markers of iPSC, definitive endoderm, hepatic specification, hepatic progenitors, and ultimately cholangiocytes. Immunostaining, western blotting, and flow cytometry demonstrate enrichment of multiple functionally relevant biliary proteins. RNA sequencing reveals that the transcriptome moves progressively toward that of human cholangiocytes. iDCs generate intracellular calcium signaling in response to ATP, form intact primary cilia, and self-assemble into duct-like structures in three-dimensional culture. In vivo, the cells engraft within mouse liver, following retrograde intrabiliary infusion. In summary, we have developed a novel approach to generate mature cholangiocytes from iPSCs. In addition to providing a model of biliary differentiation, iDCs represent a platform for in vitro disease modeling, pharmacologic testing, and individualized, cell-based, regenerative therapies for the cholangiopathies.
Collapse
Affiliation(s)
- Thiago M. De Assuncao
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN,Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN
| | - Yan Sun
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN,Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN
| | - Nidhi Jalan-Sakrikar
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN,Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN
| | - Mary Drinane
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN,Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN
| | - Bing Q. Huang
- Center for Basic Research in Digestive Diseases, Mayo Clinic and Foundation, Rochester, MN
| | - Ying Li
- Division of Biomedical Statistics and Informatics, Mayo Clinic and Foundation, Rochester, MN
| | - Jaime I. Davila
- Division of Biomedical Statistics and Informatics, Mayo Clinic and Foundation, Rochester, MN
| | - Ruisi Wang
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN,Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN
| | - Steven P. O’Hara
- Center for Basic Research in Digestive Diseases, Mayo Clinic and Foundation, Rochester, MN
| | - Gwen A. Lomberk
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN,Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN,Center for Cell Signaling in Gastroenterology, Mayo Clinic and Foundation, Rochester, MN
| | - Raul A. Urrutia
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN,Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN,Center for Cell Signaling in Gastroenterology, Mayo Clinic and Foundation, Rochester, MN
| | - Yasuhiro Ikeda
- Department of Molecular Medicine; Mayo Clinic and Foundation, Rochester, MN
| | - Robert C. Huebert
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN,Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN,Center for Cell Signaling in Gastroenterology, Mayo Clinic and Foundation, Rochester, MN
| |
Collapse
|
35
|
Liras A. Biological therapies for inherited diseases: social and bioethical considerations. Hemophilia as an example. Expert Opin Biol Ther 2015; 15:713-22. [PMID: 25826280 DOI: 10.1517/14712598.2015.1029451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION In hemophilia, advanced therapies are warranted from a conceptual and methodological standpoint. Current advanced therapy strategies are centered on the use of adeno-associated viral vectors, although problems related to immunogenicity and hepatotoxicity still remain. AREAS COVERED Future clinical trials will have to scrupulously observe international bioethical standards in terms of patient selection, particularly children. Patient recruitment rates are likely to remain low due to the stringent exclusion criteria usually imposed on the trial population regarding their hepatic and immunological markers and the presence of viral coinfection; and to the existence of an optimal palliative treatment. EXPERT OPINION Accordingly, the results obtained are likely to be of low statistical significance, which could hinder their application to clinical practice. Another important issue is the degree to which society embraces these new emerging therapies. The unfamiliarity of society with these new methods, together with the many unresolved questions about them that remain, may threaten their acceptance not only by society at large but also by health-care professionals, which would limit their translational application to clinical practice.
Collapse
Affiliation(s)
- Antonio Liras
- Complutense University of Madrid, Research Institute 12 October Hospital of Madrid, Department of Physiology , Madrid , Spain +34649907879 ;
| |
Collapse
|
36
|
Noh MJ, Copeland O, O’Mara M, Lee KH. Cell mediated gene therapy: A guide for doctors in the clinic. World J Med Genet 2015; 5:1-13. [DOI: 10.5496/wjmg.v5.i1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/16/2014] [Accepted: 01/19/2015] [Indexed: 02/06/2023] Open
Abstract
The recent approval of gene therapy products in Europe and Asia and the upsurge of gene therapy products in clinical trials signal the rebound of this technology not only for many orphan diseases but also for non-life threatening diseases. Following the success of induced pluripotent stem (iPS) cells in research, other modified ex vivo gene therapies are also knocking on the door of the clinic. Historically, gene therapy has experienced many ups and downs and still faces many challenges. During the past 10 years, many new ideas have been tried, and the goal of making this technology a more effective treatment modality through greater safety and control is coming within reach. The first clinical trial of iPS cells has begun, and cell mediated gene therapy products have reached phase III in some countries. The potential for tumorigenicity and immunogenicity are still concerns with these products, so physicians should understand the biological aspects of engineered cells in the clinic. In this review article, we attempted to provide a summary update of the current state of knowledge regarding this technology: that is, we reviewed products that have finished clinical trials, are still in clinical trials and/or are at the research stage. We also focused on the challenges, future directions, and strategies for making this technology available in the clinic. In addition, the available measures for making gene therapy products safer are within the scope of this article. It is also important to understand the manufacturing process for gene therapy products, because cell characteristics can change during the cell expansion process. When physicians use gene therapy products in the clinic, they should be aware of the viability, temperature sensitivity and stability of these cells because biologic products are different from chemical products. Although we may not be able to answer all possible questions and concerns, we believe that this is the right time for physicians to increase their interest in and understanding of this evolving technology.
Collapse
|
37
|
Singh VK, Kalsan M, Kumar N, Saini A, Chandra R. Induced pluripotent stem cells: applications in regenerative medicine, disease modeling, and drug discovery. Front Cell Dev Biol 2015; 3:2. [PMID: 25699255 PMCID: PMC4313779 DOI: 10.3389/fcell.2015.00002] [Citation(s) in RCA: 250] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/06/2015] [Indexed: 12/12/2022] Open
Abstract
Recent progresses in the field of Induced Pluripotent Stem Cells (iPSCs) have opened up many gateways for the research in therapeutics. iPSCs are the cells which are reprogrammed from somatic cells using different transcription factors. iPSCs possess unique properties of self renewal and differentiation to many types of cell lineage. Hence could replace the use of embryonic stem cells (ESC), and may overcome the various ethical issues regarding the use of embryos in research and clinics. Overwhelming responses prompted worldwide by a large number of researchers about the use of iPSCs evoked a large number of peple to establish more authentic methods for iPSC generation. This would require understanding the underlying mechanism in a detailed manner. There have been a large number of reports showing potential role of different molecules as putative regulators of iPSC generating methods. The molecular mechanisms that play role in reprogramming to generate iPSCs from different types of somatic cell sources involves a plethora of molecules including miRNAs, DNA modifying agents (viz. DNA methyl transferases), NANOG, etc. While promising a number of important roles in various clinical/research studies, iPSCs could also be of great use in studying molecular mechanism of many diseases. There are various diseases that have been modeled by uing iPSCs for better understanding of their etiology which maybe further utilized for developing putative treatments for these diseases. In addition, iPSCs are used for the production of patient-specific cells which can be transplanted to the site of injury or the site of tissue degeneration due to various disease conditions. The use of iPSCs may eliminate the chances of immune rejection as patient specific cells may be used for transplantation in various engraftment processes. Moreover, iPSC technology has been employed in various diseases for disease modeling and gene therapy. The technique offers benefits over other similar techniques such as animal models. Many toxic compounds (different chemical compounds, pharmaceutical drugs, other hazardous chemicals, or environmental conditions) which are encountered by humans and newly designed drugs may be evaluated for toxicity and effects by using iPSCs. Thus, the applications of iPSCs in regenerative medicine, disease modeling, and drug discovery are enormous and should be explored in a more comprehensive manner.
Collapse
Affiliation(s)
- Vimal K Singh
- INSPIRE Faculty, Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Manisha Kalsan
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Neeraj Kumar
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Abhishek Saini
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Ramesh Chandra
- B. R. Ambedkar Centre for Biomedical Research, University of Delhi Delhi, India
| |
Collapse
|
38
|
Hodgetts SI, Edel M, Harvey AR. The State of Play with iPSCs and Spinal Cord Injury Models. J Clin Med 2015; 4:193-203. [PMID: 26237027 PMCID: PMC4470248 DOI: 10.3390/jcm4010193] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/08/2014] [Indexed: 01/10/2023] Open
Abstract
The application of induced pluripotent stem cell (iPSC) technologies in cell based strategies, for the repair of the central nervous system (with particular focus on the spinal cord), is moving towards the potential use of clinical grade donor cells. The ability of iPSCs to generate donor neuronal, glial and astrocytic phenotypes for transplantation is highlighted here, and we review recent research using iPSCs in attempts to treat spinal cord injury in various animal models. Also discussed are issues relating to the production of clinical grade iPSCs, recent advances in transdifferentiation protocols for iPSC-derived donor cell populations, concerns about tumourogenicity, and whether iPSC technologies offer any advantages over previous donor cell candidates or tissues already in use as therapeutic tools in experimental spinal cord injury studies.
Collapse
Affiliation(s)
- Stuart I Hodgetts
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia 6009, Australia.
| | - Michael Edel
- Control of Pluripotency Laboratory, Department of Physiological Sciences I, Faculty of Medicine, University of Barcelona, Hospital Clinic, Casanova 143, Barcelona 08036, Spain.
- Faculty of Medicine, The University of Sydney Medical School, Division of Pediatrics and Child Health, Westmead Children's Hospital, Sydney 2010, Australia.
- School of Anatomy, Physiology and Human Biology, and the Harry Perkins Institute for Medical Research (CCTRM), The University of Western Australia, Western Australia 6009, Australia.
| | - Alan R Harvey
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia 6009, Australia.
| |
Collapse
|
39
|
Abstract
Hemophilia is an X-linked inherited bleeding disorder consisting of two classifications, hemophilia A and hemophilia B, depending on the underlying mutation. Although the disease is currently treatable with intravenous delivery of replacement recombinant clotting factor, this approach represents a significant cost both monetarily and in terms of quality of life. Gene therapy is an attractive alternative approach to the treatment of hemophilia that would ideally provide life-long correction of clotting activity with a single injection. In this review, we will discuss the multitude of approaches that have been explored for the treatment of both hemophilia A and B, including both in vivo and ex vivo approaches with viral and nonviral delivery vectors.
Collapse
Affiliation(s)
- Geoffrey L Rogers
- University of Florida, Department of Pediatrics, Division of Cellular and Molecular Therapy, Gainesville, FL 32610
| | - Roland W Herzog
- University of Florida, Department of Pediatrics, Division of Cellular and Molecular Therapy, Gainesville, FL 32610
| |
Collapse
|
40
|
Doering CB, Spencer HT. Replacing bad (F)actors: hemophilia. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2014; 2014:461-467. [PMID: 25696895 DOI: 10.1182/asheducation-2014.1.461] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Hemophilia A and B are bleeding disorders that result from functional deficiencies in specific circulating blood clotting factors termed factor VIII (FVIII) and factor IX (FIX), respectively, and collectively display an incidence of 1 in 4000 male births. Stem cell transplantation therapies hold the promise of providing a cure for hemophilia, but currently available transplantable stem cell products do not confer endogenous FIX or FVIII biosynthesis. For this reason, stem cell-based approaches for hemophilia have focused primarily on genetic engineering of pluripotent or multipotent stem cells. While pluripotent stem cells have been branded with high expectation and promise, they remain poorly characterized in terms of clinical utility and safety. In contrast, adult-lineage-restricted stem cells are established agents in the clinical armamentarium. Of the clinically established stem cell types, hematopoietic stem cells (HSCs) are the most utilized and represent the standard of care for several genetic and acquired diseases. Furthermore, HSCs are ideal cellular vehicles for gene therapy applications because they self-renew, repopulate the entire blood lineage while concurrently amplifying the transgene copy number >10(6) fold, and also have direct access to the bloodstream. Current research on HSC transplantation gene therapy approaches for hemophilia A and B is focused on the following: (1) identification of safe and efficient methods of nucleic acid transfer, (2) optimization of transgene product expression, (3) minimization of conditioning-regimen-related toxicity while maintaining HSC engraftment, and (4) overcoming preexisting immunity. Based on the existing data and current rate of progress, clinical trials of HSC transplantation gene therapy for hemophilia are predicted to begin in the coming years.
Collapse
Affiliation(s)
- Christopher B Doering
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - H Trent Spencer
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
41
|
Xu D, Nishimura T, Zheng M, Wu M, Su H, Sato N, Lee G, Michie S, Glenn J, Peltz G. Enabling Autologous Human Liver Regeneration with Differentiated Adipocyte Stem Cells. Cell Transplant 2014; 23:1573-84. [DOI: 10.3727/096368913x673432] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We developed a novel method for differentiating adipocyte-derived stem cells (ASCs) into hepatocyte-like cells (iHeps). ASCs are cultured as spherical cellular aggregates and are then induced by culture in chemically defined media for a short time period to differentiate into spherical culture iHeps (SCi-Heps). SCi-Heps have many of the in vitro functional properties of mature hepatocytes, and they can stably reconstitute functioning human liver in vivo in a murine model system. Implantation studies demonstrate that SCi-Heps have a very low malignant potential. All human liver regenerative procedures, including ultrasound-guided direct liver implantation, are scalable and appropriate for human clinical use. These methods can be used to achieve the major promise of regenerative medicine. It may now be possible to regenerate human liver using autologous stem cells obtained from a readily accessible tissue.
Collapse
Affiliation(s)
- Dan Xu
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA, USA
| | - Toshihiko Nishimura
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA, USA
| | - Ming Zheng
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA, USA
| | - Manhong Wu
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA, USA
| | - Hua Su
- Department of Biochemistry, University of California-Riverside, Riverside, CA, USA
| | - Noboru Sato
- Department of Biochemistry, University of California-Riverside, Riverside, CA, USA
| | - Gordon Lee
- Department of Plastic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Sara Michie
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeffrey Glenn
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Gary Peltz
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
42
|
Fomin ME, Togarrati PP, Muench MO. Progress and challenges in the development of a cell-based therapy for hemophilia A. J Thromb Haemost 2014; 12:1954-65. [PMID: 25297648 PMCID: PMC4388483 DOI: 10.1111/jth.12750] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Indexed: 12/11/2022]
Abstract
Hemophilia A results from an insufficiency of factor VIII (FVIII). Although replacement therapy with plasma-derived or recombinant FVIII is a life-saving therapy for hemophilia A patients, such therapy is a life-long treatment rather than a cure for the disease. In this review, we discuss the possibilities, progress, and challenges that remain in the development of a cell-based cure for hemophilia A. The success of cell therapy depends on the type and availability of donor cells, the age of the host and method of transplantation, and the levels of engraftment and production of FVIII by the graft. Early therapy, possibly even prenatal transplantation, may yield the highest levels of engraftment by avoiding immunological rejection of the graft. Potential cell sources of FVIII include a specialized subset of endothelial cells known as liver sinusoidal endothelial cells (LSECs) present in the adult and fetal liver, or patient-specific endothelial cells derived from induced pluripotent stem cells that have undergone gene editing to produce FVIII. Achieving sufficient engraftment of transplanted LSECs is one of the obstacles to successful cell therapy for hemophilia A. We discuss recent results from transplants performed in animals that show production of functional and clinically relevant levels of FVIII obtained from donor LSECs. Hence, the possibility of treating hemophilia A can be envisioned through persistent production of FVIII from transplanted donor cells derived from a number of potential cell sources or through creation of donor endothelial cells from patient-specific induced pluripotent stem cells.
Collapse
Affiliation(s)
- Marina E. Fomin
- Cell Therapy Core, Blood Systems Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California, San Francisco, CA
| | - Padma Priya Togarrati
- Cell Therapy Core, Blood Systems Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California, San Francisco, CA
| | - Marcus O. Muench
- Cell Therapy Core, Blood Systems Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California, San Francisco, CA
- Liver Center, University of California, San Francisco, CA
| |
Collapse
|
43
|
Kashiwakura Y, Ohmori T, Mimuro J, Madoiwa S, Inoue M, Hasegawa M, Ozawa K, Sakata Y. Production of functional coagulation factor VIII from iPSCs using a lentiviral vector. Haemophilia 2014; 20:e40-4. [PMID: 24354485 DOI: 10.1111/hae.12311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2013] [Indexed: 12/17/2022]
Abstract
The use of induced pluripotent stem cells (iPSCs) as an autologous cell source has shed new light on cell replacement therapy with respect to the treatment of numerous hereditary disorders. We focused on the use of iPSCs for cell-based therapy of haemophilia. We generated iPSCs from mesenchymal stem cells that had been isolated from C57BL/6 mice. The mouse iPSCs were generated through the induction of four transcription factor genes Oct3/4, Klf-4, Sox-2 and c-Myc. The derived iPSCs released functional coagulation factor VIII (FVIII) following transduction with a simian immunodeficiency virus vector. The subcutaneous transplantation of iPSCs expressing FVIII into nude mice resulted in teratoma formation, and significantly increased plasma levels of FVIII. The plasma concentration of FVIII was at levels appropriate for human therapy at 2-4 weeks post transplantation. Our data suggest that iPSCs could be an attractive and prospective autologous cell source for the production of coagulation factor, and that engineered iPSCs expressing coagulation factor might provide a cell-based therapeutic strategy appropriate for haemophilia.
Collapse
Affiliation(s)
- Y Kashiwakura
- Research Division of Cell and Molecular Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan; Department of Immunology, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Mo X, Li N, Wu S. Generation and characterization of bat-induced pluripotent stem cells. Theriogenology 2014; 82:283-93. [PMID: 24853281 PMCID: PMC7103130 DOI: 10.1016/j.theriogenology.2014.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 04/04/2014] [Accepted: 04/08/2014] [Indexed: 12/17/2022]
Abstract
Induced pluripotent stem cells (iPSCs) were first generated from mouse embryonic fibroblasts in the year 2006. These cells resemble the typical morphology of embryonic stem cells, express pluripotency markers, and are able to transmit through germlines. To date, iPSCs of many species have been generated, whereas generation of bat iPSCs (biPSCs) has not been reported. To facilitate in-depth study of bats at the molecular and cellular levels, we describe the successful derivation of biPSCs with a piggyBac (PB) vector that contains eight reprogramming factors Oct4, Sox2, Klf4, Nanog, cMyc, Lin28, Nr5a2, and miR302/367. These biPSCs were cultured in media containing leukemia inhibitory factor and three small molecule inhibitors (CHIR99021, PD0325901, and A8301). They retained normal karyotype, displayed alkaline phosphatase activity, and expressed pluripotency markers Oct4, Sox2, Nanog, TBX3, and TRA-1-60. They could differentiate in vitro to form embryoid bodies and in vivo to form teratomas that contained tissue cells of all three germ layers. Generation of biPSCs will facilitate future studies on the mechanisms of antiviral immunity and longevity of bats at the cellular level.
Collapse
Affiliation(s)
- Xiaohui Mo
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Ning Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Sen Wu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
45
|
Jia B, Chen S, Zhao Z, Liu P, Cai J, Qin D, Du J, Wu C, Chen Q, Cai X, Zhang H, Yu Y, Pei D, Zhong M, Pan G. Modeling of hemophilia A using patient-specific induced pluripotent stem cells derived from urine cells. Life Sci 2014; 108:22-9. [PMID: 24834837 DOI: 10.1016/j.lfs.2014.05.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 04/26/2014] [Accepted: 05/01/2014] [Indexed: 12/13/2022]
Abstract
AIMS Hemophilia A (HA) is a severe, congenital bleeding disorder caused by the deficiency of clotting factor VIII (FVIII). For years, traditional laboratory animals have been used to study HA and its therapies, although animal models may not entirely mirror the human pathophysiology. Human induced pluripotent stem cells (iPSCs) can undergo unlimited self-renewal and differentiate into all cell types. This study aims to generate hemophilia A (HA) patient-specific iPSCs that differentiate into disease-affected hepatocyte cells. These hepatocytes are potentially useful for in vitro disease modeling and provide an applicable cell source for autologous cell therapy after genetic correction. MAIN METHODS In this study, we mainly generated iPSCs from urine collected from HA patients with integration-free episomal vectors PEP4-EO2S-ET2K containing human genes OCT4, SOX2, SV40LT and KLF4, and differentiated these iPSCs into hepatocyte-like cells. We further identified the genetic phenotype of the FVIII genes and the FVIII activity in the patient-specific iPSC derived hepatic cells. KEY FINDINGS HA patient-specific iPSCs (HA-iPSCs) exhibited typical pluripotent properties evident by immunostaining, in vitro assays and in vivo assays. Importantly, we showed that HA-iPSCs could differentiate into functional hepatocyte-like cells and the HA-iPSC-derived hepatocytes failed to produce FVIII, but otherwise functioned normally, recapitulating the phenotype of HA disease in vitro. SIGNIFICANCE HA-iPSCs, particular those generated from the urine using a non-viral approach, provide an efficient way for modeling HA in vitro. Furthermore, HA-iPSCs and their derivatives serve as an invaluable cell source that can be used for gene and cell therapy in regenerative medicine.
Collapse
Affiliation(s)
- Bei Jia
- The Center for Prenatal and Hereditary Disease Diagnosis, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Shen Chen
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P. R. China
| | - Zhiju Zhao
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P. R. China
| | - Pengfei Liu
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P. R. China; Department of Regeneration Medicine, School of Pharmaceutical Science, Jilin University, Changchun, P. R. China
| | - Jinglei Cai
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P. R. China
| | - Dajiang Qin
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P. R. China
| | - Juan Du
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P. R. China
| | - Changwei Wu
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P. R. China
| | - Qianyu Chen
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P. R. China
| | - Xiujuan Cai
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P. R. China
| | - Hui Zhang
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P. R. China
| | - Yanhong Yu
- The Center for Prenatal and Hereditary Disease Diagnosis, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Duanqing Pei
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P. R. China
| | - Mei Zhong
- The Center for Prenatal and Hereditary Disease Diagnosis, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China.
| | - Guangjin Pan
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P. R. China.
| |
Collapse
|
46
|
A conditional knockout mouse model reveals endothelial cells as the principal and possibly exclusive source of plasma factor VIII. Blood 2014; 123:3706-13. [PMID: 24705491 DOI: 10.1182/blood-2014-02-555151] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The cellular source of coagulation factor VIII (FVIII) remains controversial. Like many coagulation proteins, FVIII is produced in the liver, and FVIII synthesis has long been associated with hepatocytes. But extrahepatic synthesis also occurs, and mounting evidence suggests that hepatocytes are not responsible for FVIII production. To determine the tissue that synthesizes FVIII, we developed a Cre/lox-dependent conditional knockout (KO) model in which exons 17 and 18 of the murine factor VIII gene (F8) are flanked by loxP sites, or floxed (F8(F)). In cells expressing Cre-recombinase, the floxed sequence is deleted, resulting in F8(F→KO) gene inactivation. When F8(F) mice were crossed with various tissue-specific Cre strains, we found that hepatocyte-specific F8-KO mice are indistinguishable from controls, whereas efficient endothelial-KO models display a severe hemophilic phenotype with no detectable plasma FVIII activity. A hematopoietic Cre model was more equivocal, so experimental bone marrow transplantation was used to examine hematopoietic FVIII synthesis. FVIII(null) mice that received bone marrow transplants from wild-type donors were still devoid of plasma FVIII activity after hematopoietic donor cell engraftment. Our results indicate that endothelial cells are the predominant, and possibly exclusive, source of plasma FVIII.
Collapse
|
47
|
Jalali M, Kirkpatrick WNA, Cameron MG, Pauklin S, Vallier L. Human stem cells for craniomaxillofacial reconstruction. Stem Cells Dev 2014; 23:1437-51. [PMID: 24564584 DOI: 10.1089/scd.2013.0576] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Human stem cell research represents an exceptional opportunity for regenerative medicine and the surgical reconstruction of the craniomaxillofacial complex. The correct architecture and function of the vastly diverse tissues of this important anatomical region are critical for life supportive processes, the delivery of senses, social interaction, and aesthetics. Craniomaxillofacial tissue loss is commonly associated with inflammatory responses of the surrounding tissue, significant scarring, disfigurement, and psychological sequelae as an inevitable consequence. The in vitro production of fully functional cells for skin, muscle, cartilage, bone, and neurovascular tissue formation from human stem cells, may one day provide novel materials for the reconstructive surgeon operating on patients with both hard and soft tissue deficit due to cancer, congenital disease, or trauma. However, the clinical translation of human stem cell technology, including the application of human pluripotent stem cells (hPSCs) in novel regenerative therapies, faces several hurdles that must be solved to permit safe and effective use in patients. The basic biology of hPSCs remains to be fully elucidated and concerns of tumorigenicity need to be addressed, prior to the development of cell transplantation treatments. Furthermore, functional comparison of in vitro generated tissue to their in vivo counterparts will be necessary for confirmation of maturity and suitability for application in reconstructive surgery. Here, we provide an overview of human stem cells in disease modeling, drug screening, and therapeutics, while also discussing the application of regenerative medicine for craniomaxillofacial tissue deficit and surgical reconstruction.
Collapse
Affiliation(s)
- Morteza Jalali
- 1 Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge , Cambridge, United Kingdom
| | | | | | | | | |
Collapse
|
48
|
Zhang DM, Li JJ, Yan P, Hu JT. Establishment and identification of induced pluripotent stem cells in liver cancer patients. ASIAN PAC J TROP MED 2014; 7:253-6. [PMID: 24507670 DOI: 10.1016/s1995-7645(14)60032-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 02/15/2014] [Accepted: 03/15/2014] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE To induce pluripotent stem (IPS) cells from fibrocytes that are separated from liver cancer patients. METHODS The fibrocytes were reprogrammed to IPS cells by lentiviral vector, stained and identified by immunohistochemistry. RESULTS The IPS cells were successfully established from fibrocytes after infection, and IPS cell clones formed in round shape under a microscopy. The induction rate was 0.013%±0.007%. No tumor formed at the back of nude mice within 8 weeks after the inoculation of cell clones. However, tetatoma appeared in nude mice within 1 week after IPS inoculation. A few tumors formed in nude mice within 4 weeks after the inoculation of cell clones. However, subcutaneous tumors formed within 1 week after IPS inoculation. The induced IPS cells showed three germ layers in tetatoma. Nanog and OCT4 in the induced IPS cells showed hypomethylation. SSEA-A, TRA-1-6-, TRA-1-81 and Nanog were highly expressed in the induced IPS cells, indicating the IPS cells possessed the similar ability as the stem cells. CONCLUSIONS The IPS cells of liver cancer patients can be established effectively from fibrocytes and can be cultured stably in vitro, which provides an approach for the treatment of intermediate or advanced stage liver cancer.
Collapse
Affiliation(s)
- Da-Ming Zhang
- Department of Laparoscopic Surgery, People's Hospital of Zhengzhou, Zhengzhou 450003, China
| | - Jian-Jun Li
- Department of Laparoscopic Surgery, People's Hospital of Zhengzhou, Zhengzhou 450003, China
| | - Peng Yan
- Department of Laparoscopic Surgery, People's Hospital of Zhengzhou, Zhengzhou 450003, China
| | - Jian-Ting Hu
- Department of Laparoscopic Surgery, People's Hospital of Zhengzhou, Zhengzhou 450003, China.
| |
Collapse
|
49
|
Comparison of different protocols for neural differentiation of human induced pluripotent stem cells. Mol Biol Rep 2014; 41:1713-21. [DOI: 10.1007/s11033-014-3020-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 01/02/2014] [Indexed: 12/27/2022]
|
50
|
Li G, Cheng G, Wu J, Ma S, Sun C. New iPSC for old long QT syndrome modeling: putting the evidence into perspective. Exp Biol Med (Maywood) 2013; 239:131-40. [PMID: 24363251 DOI: 10.1177/1535370213514000] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Induced pluripotent stem cells (iPS cells or iPSCs) are typically derived by transfection of certain stem cell-associated genes into non-pluripotent cells, such as adult fibroblasts (typically adult somatic cells). Various diseases can be modeled through iPSC technology. The important implication of iPSCs to offer an unprecedented opportunity to recapitulate pathologic human tissue formation in vitro has generated great excitement and interest in the whole biomedical research community. Long QT syndrome (LQTS), an inherited heart disease, is characterized by prolonged QT interval on a surface electrocardiogram. LQTS presents with life-threatening cardiac arrhythmias, which can lead to fainting, syncope, and sudden death. The iPSC-derived cardiomyocytes from LQTS patients offer a potentially unlimited source of materials for biomedical study. They can be used to recapitulate complex physiological phenotypes, probe toxicological testing and drug screening, clarify the novel mechanistic insights and may also rectify gene defects at the cellular and molecular level. Despite the emerging challenges, iPSC technology has been increasingly recognized as a valuable and growing toolkit for modeling LQTS over other various models of human diseases.
Collapse
Affiliation(s)
- Guoliang Li
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | | | | | | | | |
Collapse
|