1
|
Escribá R, Beksac M, Bennaceur-Griscelli A, Glover JC, Koskela S, Latsoudis H, Querol S, Alvarez-Palomo B. Current Landscape of iPSC Haplobanks. Stem Cell Rev Rep 2024; 20:2155-2164. [PMID: 39276260 DOI: 10.1007/s12015-024-10783-7] [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] [Accepted: 08/20/2024] [Indexed: 09/16/2024]
Abstract
The use of allogeneic induced pluripotent stem cell (iPSC)-derived cell therapies for regenerative medicine offers an affordable and realistic alternative to producing individual iPSC lines for each patient in need. Human Leukocyte Antigens (HLA)-homozygous iPSCs matched in hemi-similarity could provide cell therapies with reduced immune rejection covering a wide range of the population with a few iPSC lines. Several banks of HLA-homozygous iPSCs (haplobanks) have been established worldwide or are underway, to provide clinical grade starting material for cell therapies covering the most frequent HLA haplotypes for certain populations. Harmonizing quality standards among haplobanks and creating a global registry could minimize the collective effort and provide a much wider access to HLA-compatible cell therapies for patients with less frequent haplotypes. In this review we present all the current haplobank initiatives and their potential benefits for the global population.
Collapse
Affiliation(s)
- Rubén Escribá
- Cell Therapy Service, Banc de Sang I Teixits, 106 Passeig de Taulat, 08005, Barcelona, Spain
| | - Meral Beksac
- Ankara Liv Hospital Istinye University and Cord Blood Bank, Ankara University, Ankara, Turkey
| | - Annelise Bennaceur-Griscelli
- CiTHERA, Center of iPS Cell Therapy, Infrastructure INGESTEM University Paris Saclay Inserm U1310, 28 Rue Henri Desbruères, 91100, Corbeil-Essonnes, France
| | - Joel C Glover
- Norwegian Center for Stem Cell Research and Norwegian Core Facility for Human Pluripotent Stem Cells, Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
| | - Satu Koskela
- Finnish Red Cross Blood Service Biobank, Härkälenkki 13, 01730, Vantaa, Finland
| | - Helen Latsoudis
- Information Systems Laboratory, Institute of Computer Sciences, Foundation for Research and Technology Hellas, 70013, Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003, Heraklion, Crete, Greece
| | - Sergi Querol
- Cell Therapy Service, Banc de Sang I Teixits, 106 Passeig de Taulat, 08005, Barcelona, Spain
| | - Belén Alvarez-Palomo
- Cell Therapy Service, Banc de Sang I Teixits, 106 Passeig de Taulat, 08005, Barcelona, Spain.
| |
Collapse
|
2
|
Rehman A, Fatima I, Noor F, Qasim M, Wang P, Jia J, Alshabrmi FM, Liao M. Role of small molecules as drug candidates for reprogramming somatic cells into induced pluripotent stem cells: A comprehensive review. Comput Biol Med 2024; 177:108661. [PMID: 38810477 DOI: 10.1016/j.compbiomed.2024.108661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/08/2024] [Accepted: 05/26/2024] [Indexed: 05/31/2024]
Abstract
With the use of specific genetic factors and recent developments in cellular reprogramming, it is now possible to generate lineage-committed cells or induced pluripotent stem cells (iPSCs) from readily available and common somatic cell types. However, there are still significant doubts regarding the safety and effectiveness of the current genetic methods for reprogramming cells, as well as the conventional culture methods for maintaining stem cells. Small molecules that target specific epigenetic processes, signaling pathways, and other cellular processes can be used as a complementary approach to manipulate cell fate to achieve a desired objective. It has been discovered that a growing number of small molecules can support lineage differentiation, maintain stem cell self-renewal potential, and facilitate reprogramming by either increasing the efficiency of reprogramming or acting as a genetic reprogramming factor substitute. However, ongoing challenges include improving reprogramming efficiency, ensuring the safety of small molecules, and addressing issues with incomplete epigenetic resetting. Small molecule iPSCs have significant clinical applications in regenerative medicine and personalized therapies. This review emphasizes the versatility and potential safety benefits of small molecules in overcoming challenges associated with the iPSCs reprogramming process.
Collapse
Affiliation(s)
- Abdur Rehman
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Israr Fatima
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Fatima Noor
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan; Department of Bioinformatics and Biotechnology, Government College University of Faisalabad, 38000, Pakistan
| | - Muhammad Qasim
- Department of Bioinformatics and Biotechnology, Government College University of Faisalabad, 38000, Pakistan
| | - Peng Wang
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Jinrui Jia
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Fahad M Alshabrmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, 51452, Saudi Arabia
| | - Mingzhi Liao
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China.
| |
Collapse
|
3
|
Liu W, Zhang C, Jiang F, Tan Y, Qin B. From theory to therapy: a bibliometric and visual study of stem cell advancements in age-related macular degeneration. Cytotherapy 2024; 26:616-631. [PMID: 38483361 DOI: 10.1016/j.jcyt.2024.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND AIMS Human pluripotent stem cells, including embryonic stem cells and induced pluripotent stem cells, offer groundbreaking therapeutic potential for degenerative diseases and cellular repair. Despite their significance, a comprehensive bibliometric analysis in this field, particularly in relation to age-related macular degeneration (AMD), is yet to be conducted. This study aims to map the foundational and emerging areas in stem cell and AMD research through bibliometric analysis. METHODS This study analyzed articles and reviews on stem cells and AMD from 2000 to 2022, sourced from the Web of Science Core Collection. We used VOSviewer and CiteSpace for analysis and visualization of data pertaining to countries, institutions, authors, journals, references and key words. Statistical analyses were conducted using R language and Microsoft Excel 365. RESULTS In total, 539 publications were included, indicating an increase in global literature on stem cells and AMD from 2000 to 2022. The USA was the leading contributor, with 239 papers and the highest H-index, also the USA had the highest average citation rate per article (59.82). Notably, 50% of the top 10 institutions were from the USA, with the University of California system being the most productive. Key authors included Masayo Takahashi, Michiko Mandai, Dennis Clegg, Pete J. Coffey, Boris Stanzel, and Budd A. Tucker. Investigative Ophthalmology & Visual Science published the majority of relevant papers (n = 27). Key words like "clinical trial," "stem cell therapy," "retinal organoid," and "retinal progenitor cells" were predominant. CONCLUSIONS Research on stem cells and AMD has grown significantly, highlighting the need for increased global cooperation. Current research prioritizes the relationship between "ipsc," "induced pluripotent stem cell," "cell culture," and "human embryonic stem cell." As stem cell culture and safety have advanced, focus has shifted to prognosis and complications post-transplantation, signifying the movement of stem cell research from labs to clinical settings.
Collapse
Affiliation(s)
| | | | | | - Yao Tan
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha, China; Postdoctoral Station of Clinical Medicine, The Third Xiangya Hospital, Central South University, Changsha City, China.
| | - Bo Qin
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China.
| |
Collapse
|
4
|
Cho YD, Kim KH, Lee YM, Ku Y, Seol YJ. Dental-derived cells for regenerative medicine: stem cells, cell reprogramming, and transdifferentiation. J Periodontal Implant Sci 2022; 52:437-454. [PMID: 36468465 PMCID: PMC9807848 DOI: 10.5051/jpis.2103760188] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/08/2021] [Accepted: 01/24/2022] [Indexed: 01/07/2023] Open
Abstract
Embryonic stem cells have been a popular research topic in regenerative medicine owing to their pluripotency and applicability. However, due to the difficulty in harvesting them and their low yield efficiency, advanced cell reprogramming technology has been introduced as an alternative. Dental stem cells have entered the spotlight due to their regenerative potential and their ability to be obtained from biological waste generated after dental treatment. Cell reprogramming, a process of reverting mature somatic cells into stem cells, and transdifferentiation, a direct conversion between different cell types without induction of a pluripotent state, have helped overcome the shortcomings of stem cells and raised interest in their regenerative potential. Furthermore, the potential of these cells to return to their original cell types due to their epigenetic memory has reinforced the need to control the epigenetic background for successful management of cellular differentiation. Herein, we discuss all available sources of dental stem cells, the procedures used to obtain these cells, and their ability to differentiate into the desired cells. We also introduce the concepts of cell reprogramming and transdifferentiation in terms of genetics and epigenetics, including DNA methylation, histone modification, and non-coding RNA. Finally, we discuss a novel therapeutic avenue for using dental-derived cells as stem cells, and explain cell reprogramming and transdifferentiation, which are used in regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Young-Dan Cho
- Department of Periodontology, School of Dentistry and Dental Research Institute, Seoul National University and Seoul National University Dental Hospital, Seoul, Korea
| | - Kyoung-Hwa Kim
- Department of Periodontology, School of Dentistry and Dental Research Institute, Seoul National University and Seoul National University Dental Hospital, Seoul, Korea
| | - Yong-Moo Lee
- Department of Periodontology, School of Dentistry and Dental Research Institute, Seoul National University and Seoul National University Dental Hospital, Seoul, Korea
| | - Young Ku
- Department of Periodontology, School of Dentistry and Dental Research Institute, Seoul National University and Seoul National University Dental Hospital, Seoul, Korea
| | - Yang-Jo Seol
- Department of Periodontology, School of Dentistry and Dental Research Institute, Seoul National University and Seoul National University Dental Hospital, Seoul, Korea
| |
Collapse
|
5
|
Umbilical Cord Tissue as a Source of Young Cells for the Derivation of Induced Pluripotent Stem Cells Using Non-Integrating Episomal Vectors and Feeder-Free Conditions. Cells 2020; 10:cells10010049. [PMID: 33396312 PMCID: PMC7824218 DOI: 10.3390/cells10010049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 12/27/2020] [Accepted: 12/30/2020] [Indexed: 12/18/2022] Open
Abstract
The clinical application of induced pluripotent stem cells (iPSC) needs to balance the use of an autologous source that would be a perfect match for the patient against any safety or efficacy issues that might arise with using cells from an older patient or donor. Drs. Takahashi and Yamanaka and the Office of Cellular and Tissue-based Products (PMDA), Japan, have had concerns over the existence of accumulated DNA mutations in the cells of older donors and the possibility of long-term negative effects. To mitigate the risk, they have chosen to partner with the Umbilical Cord (UC) banks in Japan to source allogeneic-matched donor cells. Production of iPSCs from UC blood cells (UCB) has been successful; however, reprogramming blood cells requires cell enrichment with columns or flow cytometry and specialized growth media. These requirements add to the cost of production and increase the manipulation of the cells, which complicates the regulatory approval process. Alternatively, umbilical cord tissue mesenchymal stromal cells (CT-MSCs) have the same advantage as UCB cells of being a source of young donor cells. Crucially, CT-MSCs are easier and less expensive to harvest and grow compared to UCB cells. Here, we demonstrate that CT-MSCs can be easily isolated without expensive enzymatic treatment or columns and reprogramed well using episomal vectors, which allow for the removal of the reprogramming factors after a few passages. Together the data indicates that CT-MSCs are a viable source of donor cells for the production of clinical-grade, patient matched iPSCs.
Collapse
|
6
|
Induced Pluripotency: A Powerful Tool for In Vitro Modeling. Int J Mol Sci 2020; 21:ijms21238910. [PMID: 33255453 PMCID: PMC7727808 DOI: 10.3390/ijms21238910] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
One of the greatest breakthroughs of regenerative medicine in this century was the discovery of induced pluripotent stem cell (iPSC) technology in 2006 by Shinya Yamanaka. iPSCs originate from terminally differentiated somatic cells that have newly acquired the developmental capacity of self-renewal and differentiation into any cells of three germ layers. Before iPSCs can be used routinely in clinical practice, their efficacy and safety need to be rigorously tested; however, iPSCs have already become effective and fully-fledged tools for application under in vitro conditions. They are currently routinely used for disease modeling, preparation of difficult-to-access cell lines, monitoring of cellular mechanisms in micro- or macroscopic scales, drug testing and screening, genetic engineering, and many other applications. This review is a brief summary of the reprogramming process and subsequent differentiation and culture of reprogrammed cells into neural precursor cells (NPCs) in two-dimensional (2D) and three-dimensional (3D) conditions. NPCs can be used as biomedical models for neurodegenerative diseases (NDs), which are currently considered to be one of the major health problems in the human population.
Collapse
|
7
|
Sharma S, Bhonde R. Genetic and epigenetic stability of stem cells: Epigenetic modifiers modulate the fate of mesenchymal stem cells. Genomics 2020; 112:3615-3623. [DOI: 10.1016/j.ygeno.2020.04.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/08/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022]
|
8
|
Soleimani F, Babaei E, H Feizi MA, Fathi F. CRISPR-Cas9-mediated knockout of the Prkdc in mouse embryonic stem cells leads to the modulation of the expression of pluripotency genes. J Cell Physiol 2019; 235:3994-4000. [PMID: 31603250 DOI: 10.1002/jcp.29295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/30/2019] [Indexed: 12/15/2022]
Abstract
Prkdc encodes for the catalytic subunit of the DNA-dependent protein kinase (DNA-PKcs) playing a key role in nonhomologous end joining pathway during DNA double-strand break repair and also influencing the homologous recombination (HR) repair system by phosphorylation of proteins involved in HR. In addition, Prkdc has other critical functions in biological processes, such as transcriptional regulation, telomere stability, apoptosis, and metabolism. DNA-PKcs upregulates during in vitro differentiation of mouse embryonic stem cells (mESCs). To address the potential role of Prkdc in mESCs pluripotency and in vitro differentiation into ectoderm, mesoderm, and endoderm germ layers under normal physiological conditions, a bi-allelic Prkdc-knockout cell line was generated in the present study by employing CRISPR/Cas9 system, and subsequently, its potential role in stemness and development was studied. The results of the study showed that the expression of pluripotency-associated genes, including Nanog and Sox-2 were overexpressed in the bi-allelic Prkdc-knockout cell line. Also, bi-allelic Prkdc-knockout cell line was shown to have typical mESCs cell morphology, cell cycle distribution, and alkaline phosphatase activity. Furthermore, the results of the study revealed that the expression of several germ layer markers is modulated in Prkdc-knockout lines. In conclusion, the findings of our study demonstrated the role of Prkdc during differentiation and development of ESCs.
Collapse
Affiliation(s)
- Farzad Soleimani
- Department of Biological Sciences, School of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Esmaeil Babaei
- Department of Biological Sciences, School of Natural Sciences, University of Tabriz, Tabriz, Iran.,Institute of Environment, University of Tabriz, Tabriz, Iran
| | - Mohammad A H Feizi
- Department of Biological Sciences, School of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Fardin Fathi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
9
|
Xia K, Gong Z, Zhu J, Yu W, Wang Y, Wang J, Xu A, Zhou X, Tao H, Li F, Liang C. Differentiation of Pluripotent Stem Cells into Nucleus Pulposus Progenitor Cells for Intervertebral Disc Regeneration. Curr Stem Cell Res Ther 2019; 14:57-64. [PMID: 30227822 DOI: 10.2174/1574888x13666180918095121] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 08/31/2018] [Accepted: 09/11/2018] [Indexed: 02/08/2023]
Abstract
Low back pain (LBP) is one of the world's most common musculoskeletal diseases and is frequently associated with intervertebral disc degeneration (IDD). While the main cause of IDD is commonly attributed to a reduced number of nucleus pulposus (NP) cells, current treatment strategies (both surgical and more conservative) fail to replenish NP cells or reverse the pathology. Cell replacement therapies are an attractive alternative for treating IDD. However, injecting intervertebral disc (IVD) cells, chondrocytes, or mesenchymal stem cells into various animal models of IDD indicate that transplanted cells generally fail to survive and engraft into the avascular IVD niche. Whereas pluripotent stem cells (PSCs), including induced pluripotent stem cells (iPSCs) and embryonic stem cells (ESCs), hold great potential for revolutionizing regenerative medicine, current protocols for differentiating these cells into NP-like cells are inadequate. Nucleus pulposus progenitor cells (NPPCs), which are derived from the embryonic notochord, can not only survive within the harsh hypoxic environment of the IVD, but they also efficiently differentiate into NP-like cells. Here we provide an overview of the latest progress in repairing degenerated IVDs using PSCs and NPPCs. We also discuss the molecular pathways by which PSCs differentiate into NPPCs in vitro and in vivo and propose a new, in vivo IDD therapy.
Collapse
Affiliation(s)
- Kaishun Xia
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jie Fang Road, Hangzhou, 310009, Zhejiang, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Zhe Gong
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jie Fang Road, Hangzhou, 310009, Zhejiang, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Jian Zhu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jie Fang Road, Hangzhou, 310009, Zhejiang, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Wei Yu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jie Fang Road, Hangzhou, 310009, Zhejiang, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Yitian Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jie Fang Road, Hangzhou, 310009, Zhejiang, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Junjie Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jie Fang Road, Hangzhou, 310009, Zhejiang, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Ankai Xu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jie Fang Road, Hangzhou, 310009, Zhejiang, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Xiaopeng Zhou
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jie Fang Road, Hangzhou, 310009, Zhejiang, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Huimin Tao
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jie Fang Road, Hangzhou, 310009, Zhejiang, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Fangcai Li
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jie Fang Road, Hangzhou, 310009, Zhejiang, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Chengzhen Liang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jie Fang Road, Hangzhou, 310009, Zhejiang, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| |
Collapse
|
10
|
Dolatshad H, Tatwavedi D, Ahmed D, Tegethoff JF, Boultwood J, Pellagatti A. Application of induced pluripotent stem cell technology for the investigation of hematological disorders. Adv Biol Regul 2019; 71:19-33. [PMID: 30341008 DOI: 10.1016/j.jbior.2018.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 06/08/2023]
Abstract
Induced pluripotent stem cells (iPSCs) were first described over a decade ago and are currently used in various basic biology and clinical research fields. Recent advances in the field of human iPSCs have opened the way to a better understanding of the biology of human diseases. Disease-specific iPSCs provide an unparalleled opportunity to establish novel human cell-based disease models, with the potential to enhance our understanding of the molecular mechanisms underlying human malignancies, and to accelerate the identification of effective new drugs. When combined with genome editing technologies, iPSCs represent a new approach to study single or multiple disease-causing mutations and model specific diseases in vitro. In addition, genetically corrected patient-specific iPSCs could potentially be used for stem cell based therapy. Furthermore, the reprogrammed cells share patient-specific genetic background, offering a new platform to develop personalized therapy/medicine for patients. In this review we discuss the recent advances in iPSC research technology and their potential applications in hematological diseases. Somatic cell reprogramming has presented new routes for generating patient-derived iPSCs, which can be differentiated to hematopoietic stem cells and the various downstream hematopoietic lineages. iPSC technology shows promise in the modeling of both inherited and acquired hematological disorders. A direct reprogramming and differentiation strategy is able to recapitulate hematological disorder progression and capture the earliest molecular alterations that underlie the initiation of hematological malignancies.
Collapse
Affiliation(s)
- Hamid Dolatshad
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Dharamveer Tatwavedi
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Doaa Ahmed
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK; Clinical Pathology Department, Assiut University Hospitals, Faculty of Medicine, Assiut, Egypt
| | - Jana F Tegethoff
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Jacqueline Boultwood
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Andrea Pellagatti
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK.
| |
Collapse
|
11
|
Leonova K, Safina A, Nesher E, Sandlesh P, Pratt R, Burkhart C, Lipchick B, Gitlin I, Frangou C, Koman I, Wang J, Kirsanov K, Yakubovskaya MG, Gudkov AV, Gurova K. TRAIN (Transcription of Repeats Activates INterferon) in response to chromatin destabilization induced by small molecules in mammalian cells. eLife 2018; 7:e30842. [PMID: 29400649 PMCID: PMC5815852 DOI: 10.7554/elife.30842] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 02/04/2018] [Indexed: 12/13/2022] Open
Abstract
Cellular responses to the loss of genomic stability are well-established, while how mammalian cells respond to chromatin destabilization is largely unknown. We previously found that DNA demethylation on p53-deficient background leads to transcription of repetitive heterochromatin elements, followed by an interferon response, a phenomenon we named TRAIN (Transcription of Repeats Activates INterferon). Here, we report that curaxin, an anticancer small molecule, destabilizing nucleosomes via disruption of histone/DNA interactions, also induces TRAIN. Furthermore, curaxin inhibits oncogene-induced transformation and tumor growth in mice in an interferon-dependent manner, suggesting that anticancer activity of curaxin, previously attributed to p53-activation and NF-kappaB-inhibition, may also involve induction of interferon response to epigenetic derepression of the cellular 'repeatome'. Moreover, we observed that another type of drugs decondensing chromatin, HDAC inhibitor, also induces TRAIN. Thus, we proposed that TRAIN may be one of the mechanisms ensuring epigenetic integrity of mammalian cells via elimination of cells with desilenced chromatin.
Collapse
Affiliation(s)
- Katerina Leonova
- Department of Cell Stress BiologyRoswell Park Cancer InstituteBuffaloUnited States
| | - Alfiya Safina
- Department of Cell Stress BiologyRoswell Park Cancer InstituteBuffaloUnited States
| | - Elimelech Nesher
- Department of Cell Stress BiologyRoswell Park Cancer InstituteBuffaloUnited States
- Department of Molecular BiologyAriel UniversityArielIsrael
| | - Poorva Sandlesh
- Department of Cell Stress BiologyRoswell Park Cancer InstituteBuffaloUnited States
| | - Rachel Pratt
- Department of Cell Stress BiologyRoswell Park Cancer InstituteBuffaloUnited States
| | | | - Brittany Lipchick
- Department of Cell Stress BiologyRoswell Park Cancer InstituteBuffaloUnited States
| | - Ilya Gitlin
- Department of Cell Stress BiologyRoswell Park Cancer InstituteBuffaloUnited States
| | - Costakis Frangou
- Department of Cell Stress BiologyRoswell Park Cancer InstituteBuffaloUnited States
| | - Igor Koman
- Department of Molecular BiologyAriel UniversityArielIsrael
| | - Jianmin Wang
- Department of BioinformaticsRoswell Park Cancer InstituteBuffaloUnited States
| | - Kirill Kirsanov
- Department of Chemical CarcinogenesisInstitute of Carcinogenesis, Blokhin Cancer Research Center RAMSMoscowRussia
| | - Marianna G Yakubovskaya
- Department of Chemical CarcinogenesisInstitute of Carcinogenesis, Blokhin Cancer Research Center RAMSMoscowRussia
| | - Andrei V Gudkov
- Department of Cell Stress BiologyRoswell Park Cancer InstituteBuffaloUnited States
| | - Katerina Gurova
- Department of Cell Stress BiologyRoswell Park Cancer InstituteBuffaloUnited States
| |
Collapse
|
12
|
Rossmann MP, Orkin SH, Chute JP. Hematopoietic Stem Cell Biology. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
13
|
Abstract
Somatic cells can be reprogrammed to pluripotent stem cells or transdifferentiate to another lineage cell type. Much efforts have been made to unravel the epigenetic mechanisms underlying the cell fate conversion. Histone modifications as the major epigenetic regulator are implicated in various aspects of reprogramming and transdifferentiation. Here, we discuss the roles of histone modifications on reprogramming and transdifferentiation and hopefully provide new insights into induction and promotion of the cell fate conversion by modulating histone modifications.
Collapse
|
14
|
Mechanism of human somatic reprogramming to iPS cell. J Transl Med 2017; 97:1152-1157. [PMID: 28530648 DOI: 10.1038/labinvest.2017.56] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/10/2017] [Accepted: 04/17/2017] [Indexed: 01/09/2023] Open
Abstract
Somatic reprogramming to induced pluripotent stem cells (iPSC) was realized in the year 2006 in mice, and in 2007 in humans, by transiently forced expression of a combination of exogenous transcription factors. Human and mouse iPSCs are distinctly reprogrammed into a 'primed' and a 'naïve' state, respectively. In the last decade, puzzle pieces of somatic reprogramming have been collected with difficulty. Collectively, dissecting reprogramming events and identification of the hallmark of sequentially activated/silenced genes have revealed mouse somatic reprogramming in fragments, but there is a long way to go toward understanding the molecular mechanisms of human somatic reprogramming, even with developing technologies. Recently, an established human intermediately reprogrammed stem cell (iRSC) line, which has paused reprogramming at the endogenous OCT4-negative/exogenous transgene-positive pre-MET (mesenchymal-to-epithelial-transition) stage can resume reprogramming into endogenous OCT4-positive iPSCs only by change of culture conditions. Genome-editing-mediated visualization of endogenous OCT4 activity with GFP in living iRSCs demonstrates the timing of OCT4 activation and entry to MET in the reprogramming toward iPSCs. Applications of genome-editing technology to pluripotent stem cells will reshape our approaches for exploring molecular mechanisms.
Collapse
|
15
|
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
|
16
|
Interleukin-6-Mediated Induced Pluripotent Stem Cell (iPSC)-Derived Neural Differentiation. Mol Neurobiol 2017; 55:3513-3522. [PMID: 28509081 DOI: 10.1007/s12035-017-0594-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/02/2017] [Indexed: 12/11/2022]
Abstract
In an aging society with an increasing threat to higher brain cognitive functions due to dementia, it becomes imperative to identify new molecular remedies for supporting adult neurogenesis. Interleukin-6 (IL-6) is a promising cytokine that can support neurogenesis under conditions of neurodegeneration, and neuron replacement is eventually possible due to its agonistic acting soluble receptor sIL-6R. Here, we report that activation of the IL-6-signal transducer and activator of transcription 3 (STAT3) axis is neurogenic and has potential therapeutic applications for the treatment of neurodegenerative diseases such as Parkinson's disease (PD).
Collapse
|
17
|
Spitalieri P, Talarico VR, Murdocca M, Novelli G, Sangiuolo F. Human induced pluripotent stem cells for monogenic disease modelling and therapy. World J Stem Cells 2016; 8:118-35. [PMID: 27114745 PMCID: PMC4835672 DOI: 10.4252/wjsc.v8.i4.118] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 01/21/2016] [Accepted: 02/14/2016] [Indexed: 02/06/2023] Open
Abstract
Recent and advanced protocols are now available to derive human induced pluripotent stem cells (hiPSCs) from patients affected by genetic diseases. No curative treatments are available for many of these diseases; thus, hiPSCs represent a major impact on patient' health. hiPSCs represent a valid model for the in vitro study of monogenic diseases, together with a better comprehension of the pathogenic mechanisms of the pathology, for both cell and gene therapy protocol applications. Moreover, these pluripotent cells represent a good opportunity to test innovative pharmacological treatments focused on evaluating the efficacy and toxicity of novel drugs. Today, innovative gene therapy protocols, especially gene editing-based, are being developed, allowing the use of these cells not only as in vitro disease models but also as an unlimited source of cells useful for tissue regeneration and regenerative medicine, eluding ethical and immune rejection problems. In this review, we will provide an up-to-date of modelling monogenic disease by using hiPSCs and the ultimate applications of these in vitro models for cell therapy. We consider and summarize some peculiar aspects such as the type of parental cells used for reprogramming, the methods currently used to induce the transcription of the reprogramming factors, and the type of iPSC-derived differentiated cells, relating them to the genetic basis of diseases and to their inheritance model.
Collapse
Affiliation(s)
- Paola Spitalieri
- Paola Spitalieri, Valentina Rosa Talarico, Michela Murdocca, Giuseppe Novelli, Federica Sangiuolo, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Valentina Rosa Talarico
- Paola Spitalieri, Valentina Rosa Talarico, Michela Murdocca, Giuseppe Novelli, Federica Sangiuolo, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Michela Murdocca
- Paola Spitalieri, Valentina Rosa Talarico, Michela Murdocca, Giuseppe Novelli, Federica Sangiuolo, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Giuseppe Novelli
- Paola Spitalieri, Valentina Rosa Talarico, Michela Murdocca, Giuseppe Novelli, Federica Sangiuolo, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Federica Sangiuolo
- Paola Spitalieri, Valentina Rosa Talarico, Michela Murdocca, Giuseppe Novelli, Federica Sangiuolo, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| |
Collapse
|
18
|
Jackson SA, Olufs ZPG, Tran KA, Zaidan NZ, Sridharan R. Alternative Routes to Induced Pluripotent Stem Cells Revealed by Reprogramming of the Neural Lineage. Stem Cell Reports 2016; 6:302-11. [PMID: 26905202 PMCID: PMC4788781 DOI: 10.1016/j.stemcr.2016.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 01/11/2016] [Accepted: 01/13/2016] [Indexed: 01/03/2023] Open
Abstract
During the reprogramming of mouse embryonic fibroblasts (MEFs) to induced pluripotent stem cells, the activation of pluripotency genes such as NANOG occurs after the mesenchymal to epithelial transition. Here we report that both adult stem cells (neural stem cells) and differentiated cells (astrocytes) of the neural lineage can activate NANOG in the absence of cadherin expression during reprogramming. Gene expression analysis revealed that only the NANOG+E-cadherin+ populations expressed stabilization markers, had upregulated several cell cycle genes; and were transgene independent. Inhibition of DOT1L activity enhanced both the numbers of NANOG+ and NANOG+E-cadherin+ colonies in neural stem cells. Expressing SOX2 in MEFs prior to reprogramming did not alter the ratio of NANOG colonies that express E-cadherin. Taken together these results provide a unique pathway for reprogramming taken by cells of the neural lineage. NANOG expression can be obtained without E-cadherin in cells of the neural lineage Transgene independence and late markers only occur when colonies are E-cadherin+ NANOG+E-cadherin+ colonies also upregulate cell cycle-related genes DOT1L inhibition increases NANOG+E-cadherin+ colony numbers
Collapse
Affiliation(s)
- Steven A Jackson
- Epigenetics Theme, Wisconsin Institute for Discovery, University of Wisconsin, 330 North Orchard Street, Room 2118, Madison, WI 53715, USA
| | - Zachariah P G Olufs
- Epigenetics Theme, Wisconsin Institute for Discovery, University of Wisconsin, 330 North Orchard Street, Room 2118, Madison, WI 53715, USA
| | - Khoa A Tran
- Epigenetics Theme, Wisconsin Institute for Discovery, University of Wisconsin, 330 North Orchard Street, Room 2118, Madison, WI 53715, USA
| | - Nur Zafirah Zaidan
- Epigenetics Theme, Wisconsin Institute for Discovery, University of Wisconsin, 330 North Orchard Street, Room 2118, Madison, WI 53715, USA
| | - Rupa Sridharan
- Epigenetics Theme, Wisconsin Institute for Discovery, University of Wisconsin, 330 North Orchard Street, Room 2118, Madison, WI 53715, USA; Department of Cell and Regenerative Biology, University of Wisconsin, 1111 Highland Avenue, Madison, WI 53715, USA.
| |
Collapse
|
19
|
Rajasingh S, Thangavel J, Czirok A, Samanta S, Roby KF, Dawn B, Rajasingh J. Generation of Functional Cardiomyocytes from Efficiently Generated Human iPSCs and a Novel Method of Measuring Contractility. PLoS One 2015; 10:e0134093. [PMID: 26237415 PMCID: PMC4523188 DOI: 10.1371/journal.pone.0134093] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 07/02/2015] [Indexed: 12/24/2022] Open
Abstract
Human induced pluripotent stem cells (iPSCs) derived cardiomyocytes (iCMCs) would provide an unlimited cell source for regenerative medicine and drug discoveries. The objective of our study is to generate functional cardiomyocytes from human iPSCs and to develop a novel method of measuring contractility of CMCs. In a series of experiments, adult human skin fibroblasts (HSF) and human umbilical vein endothelial cells (HUVECs) were treated with a combination of pluripotent gene DNA and mRNA under specific conditions. The iPSC colonies were identified and differentiated into various cell lineages, including CMCs. The contractile activity of CMCs was measured by a novel method of frame-by-frame cross correlation (particle image velocimetry-PIV) analysis. Our treatment regimen transformed 4% of HSFs into iPSC colonies at passage 0, a significantly improved efficiency compared with use of either DNA or mRNA alone. The iPSCs were capable of differentiating both in vitro and in vivo into endodermal, ectodermal and mesodermal cells, including CMCs with >88% of cells being positive for troponin T (CTT) and Gata4 by flow cytometry. We report a highly efficient combination of DNA and mRNA to generate iPSCs and functional iCMCs from adult human cells. We also report a novel approach to measure contractility of iCMCs.
Collapse
Affiliation(s)
- Sheeja Rajasingh
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Jayakumar Thangavel
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Andras Czirok
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Saheli Samanta
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Katherine F. Roby
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Buddhadeb Dawn
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Johnson Rajasingh
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * E-mail: (JR)
| |
Collapse
|
20
|
Software-aided automatic laser optoporation and transfection of cells. Sci Rep 2015; 5:11185. [PMID: 26053047 PMCID: PMC4459191 DOI: 10.1038/srep11185] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 05/05/2015] [Indexed: 11/12/2022] Open
Abstract
Optoporation, the permeabilization of a cell membrane by laser pulses, has emerged as a powerful non-invasive and highly efficient technique to induce transfection of cells. However, the usual tedious manual targeting of individual cells significantly limits the addressable cell number. To overcome this limitation, we present an experimental setup with custom-made software control, for computer-automated cell optoporation. The software evaluates the image contrast of cell contours, automatically designates cell locations for laser illumination, centres those locations in the laser focus, and executes the illumination. By software-controlled meandering of the sample stage, in principle all cells in a typical cell culture dish can be targeted without further user interaction. The automation allows for a significant increase in the number of treatable cells compared to a manual approach. For a laser illumination duration of 100 ms, 7-8 positions on different cells can be targeted every second inside the area of the microscope field of view. The experimental capabilities of the setup are illustrated in experiments with Chinese hamster ovary cells. Furthermore, the influence of laser power is discussed, with mention on post-treatment cell survival and optoporation-efficiency rates.
Collapse
|
21
|
Shen Y, Gao F, Wang M, Li A. RPdb: a database of experimentally verified cellular reprogramming records. Bioinformatics 2015; 31:3237-9. [PMID: 26026167 PMCID: PMC4576693 DOI: 10.1093/bioinformatics/btv331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 05/25/2015] [Indexed: 12/27/2022] Open
Abstract
UNLABELLED Many cell lines can be reprogrammed to other cell lines by forced expression of a few transcription factors or by specifically designed culture methods, which have attracted a great interest in the field of regenerative medicine and stem cell research. Plenty of cell lines have been used to generate induced pluripotent stem cells (IPSCs) by expressing a group of genes and microRNAs. These IPSCs can differentiate into somatic cells to promote tissue regeneration. Similarly, many somatic cells can be directly reprogrammed to other cells without a stem cell state. All these findings are helpful in searching for new reprogramming methods and understanding the biological mechanism inside. However, to the best of our knowledge, there is still no database dedicated to integrating the reprogramming records. We built RPdb (cellular reprogramming database) to collect cellular reprogramming information and make it easy to access. All entries in RPdb are manually extracted from more than 2000 published articles, which is helpful for researchers in regenerative medicine and cell biology. AVAILABILITY AND IMPLEMENTATION RPdb is freely available on the web at http://bioinformatics.ustc.edu.cn/rpdb with all major browsers supported. CONTACT aoli@ustc.edu.cn SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Yi Shen
- School of Information Science and Technology, University of Science and Technology of China, Hefei, AH230027, China
| | | | - Minghui Wang
- School of Information Science and Technology, University of Science and Technology of China, Hefei, AH230027, China, Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, AH230037, China
| | - Ao Li
- School of Information Science and Technology, University of Science and Technology of China, Hefei, AH230027, China, Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, AH230037, China
| |
Collapse
|
22
|
Popowski M, Tucker H. Repressors of reprogramming. World J Stem Cells 2015; 7:541-546. [PMID: 25914761 PMCID: PMC4404389 DOI: 10.4252/wjsc.v7.i3.541] [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: 07/31/2014] [Revised: 01/10/2015] [Accepted: 01/20/2015] [Indexed: 02/06/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) have been the focal point of ever increasing interest and scrutiny as they hold the promise of personalized regenerative medicine. However, creation of iPSCs is an inefficient process that requires forced expression of potentially oncogenic proteins. In order to unlock the full potential of iPSCs, both for basic and clinical research, we must broaden our search for more reliable ways of inducing pluripotency in somatic cells. This review surveys an area of reprogramming that does not receive as much focus, barriers to reprogramming, in the hope of stimulating new ideas and approaches towards developing safer and more efficient methods of reprogramming. Better methods of iPSC creation will allow for more reliable disease modeling, better basic research into the pluripotent state and safer iPSCs that can be used in a clinical setting.
Collapse
|
23
|
Rony IK, Baten A, Bloomfield JA, Islam ME, Billah MM, Islam KD. Inducing pluripotency in vitro: recent advances and highlights in induced pluripotent stem cells generation and pluripotency reprogramming. Cell Prolif 2015; 48:140-56. [PMID: 25643745 DOI: 10.1111/cpr.12162] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 10/05/2014] [Indexed: 12/12/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are considered patient-specific counterparts of embryonic stem cells as they originate from somatic cells after forced expression of pluripotency reprogramming factors Oct4, Sox2, Klf4 and c-Myc. iPSCs offer unprecedented opportunity for personalized cell therapies in regenerative medicine. In recent years, iPSC technology has undergone substantial improvement to overcome slow and inefficient reprogramming protocols, and to ensure clinical-grade iPSCs and their functional derivatives. Recent developments in iPSC technology include better reprogramming methods employing novel delivery systems such as non-integrating viral and non-viral vectors, and characterization of alternative reprogramming factors. Concurrently, small chemical molecules (inhibitors of specific signalling or epigenetic regulators) have become crucial to iPSC reprogramming; they have the ability to replace putative reprogramming factors and boost reprogramming processes. Moreover, common dietary supplements, such as vitamin C and antioxidants, when introduced into reprogramming media, have been found to improve genomic and epigenomic profiles of iPSCs. In this article, we review the most recent advances in the iPSC field and potent application of iPSCs, in terms of cell therapy and tissue engineering.
Collapse
Affiliation(s)
- I K Rony
- Biotechnology and Genetic Engineering Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| | | | | | | | | | | |
Collapse
|
24
|
Abstract
Pluripotent cells in embryos are situated near the apex of the hierarchy of developmental potential. They are capable of generating all cell types of the mammalian body proper. Therefore, they are the exemplar of stem cells. In vivo, pluripotent cells exist transiently and become expended within a few days of their establishment. Yet, when explanted into artificial culture conditions, they can be indefinitely propagated in vitro as pluripotent stem cell lines. A host of transcription factors and regulatory genes are now known to underpin the pluripotent state. Nonetheless, how pluripotent cells are equipped with their vast multilineage differentiation potential remains elusive. Consensus holds that pluripotency transcription factors prevent differentiation by inhibiting the expression of differentiation genes. However, this does not explain the developmental potential of pluripotent cells. We have presented another emergent perspective, namely, that pluripotency factors function as lineage specifiers that enable pluripotent cells to differentiate into specific lineages, therefore endowing pluripotent cells with their multilineage potential. Here we provide a comprehensive overview of the developmental biology, transcription factors, and extrinsic signaling associated with pluripotent cells, and their accompanying subtypes, in vitro heterogeneity and chromatin states. Although much has been learned since the appreciation of mammalian pluripotency in the 1950s and the derivation of embryonic stem cell lines in 1981, we will specifically emphasize what currently remains unclear. However, the view that pluripotency factors capacitate differentiation, recently corroborated by experimental evidence, might perhaps address the long-standing question of how pluripotent cells are endowed with their multilineage differentiation potential.
Collapse
Affiliation(s)
- Kyle M. Loh
- Department of Developmental Biology and the Stanford Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, California; Genome Institute of Singapore, Stem Cell & Regenerative Biology Group, Agency for Science, Technology & Research, Singapore; and Department of Medicine and the Beth Israel Deaconess Medical Center, Division of Hematology/Oncology, Harvard Medical School, Boston, Massachusetts
| | - Bing Lim
- Department of Developmental Biology and the Stanford Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, California; Genome Institute of Singapore, Stem Cell & Regenerative Biology Group, Agency for Science, Technology & Research, Singapore; and Department of Medicine and the Beth Israel Deaconess Medical Center, Division of Hematology/Oncology, Harvard Medical School, Boston, Massachusetts
| | - Lay Teng Ang
- Department of Developmental Biology and the Stanford Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, California; Genome Institute of Singapore, Stem Cell & Regenerative Biology Group, Agency for Science, Technology & Research, Singapore; and Department of Medicine and the Beth Israel Deaconess Medical Center, Division of Hematology/Oncology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
25
|
|
26
|
Larcher F, Espada J, Díaz-Ley B, Jaén P, Juarranz A, Quintanilla M. New Experimental Models of Skin Homeostasis and Diseases. ACTAS DERMO-SIFILIOGRAFICAS 2015. [DOI: 10.1016/j.adengl.2014.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
27
|
Zhao P, Luo Z, Tian W, Yang J, Ibáñez DP, Huang Z, Tortorella MD, Esteban MA, Fan W. Solving the puzzle of Parkinson's disease using induced pluripotent stem cells. Exp Biol Med (Maywood) 2014; 239:1421-32. [PMID: 24939824 DOI: 10.1177/1535370214538588] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The prevalence and incidence of Parkinson's disease (PD) is increasing due to a prolonged life expectancy. This highlights the need for a better mechanistic understanding and new therapeutic approaches. However, traditional in vitro and in vivo experimental models to study PD are suboptimal, thus hampering the progress in the field. The epigenetic reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) offers a unique way to overcome this problem, as these cells share many properties of embryonic stem cells (ESCs) including the potential to be transformed into different lineages. PD modeling with iPSCs is nowadays facilitated by the growing availability of high-efficiency neural-specific differentiation protocols and the possibility to correct or induce mutations as well as creating marker cell lines using designer nucleases. These technologies, together with steady advances in human genetics, will likely introduce profound changes in the way we interpret PD and develop new treatments. Here, we summarize the different PD iPSCs reported so far and discuss the challenges for disease modeling using these cell lines.
Collapse
Affiliation(s)
- Ping Zhao
- Laboratory of Chromatin and Human Disease, Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou 510530, China
| | - Zhiwei Luo
- Laboratory of Chromatin and Human Disease, Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou 510530, China
| | - Weihua Tian
- Laboratory of Chromatin and Human Disease, Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou 510530, China
| | - Jiayin Yang
- Laboratory of Chromatin and Human Disease, Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou 510530, China
| | - David P Ibáñez
- Laboratory of Chromatin and Human Disease, Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou 510530, China
| | - Zhijian Huang
- Laboratory of Chromatin and Human Disease, Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou 510530, China
| | - Micky D Tortorella
- Drug Discovery Pipeline Group, Guangzhou Institutes of Biomedicine and Health, Guangzhou 510530, China
| | - Miguel A Esteban
- Laboratory of Chromatin and Human Disease, Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou 510530, China Guangdong Stem Cell and Regenerative Medicine Research Centre, University of Hong Kong, Hong Kong 999077, and Guangzhou Institutes of Biomedicine and Health, Guangzhou 510530, China
| | - Wenxia Fan
- Laboratory of Chromatin and Human Disease, Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou 510530, China
| |
Collapse
|
28
|
Pamies D, Hartung T, Hogberg HT. Biological and medical applications of a brain-on-a-chip. Exp Biol Med (Maywood) 2014; 239:1096-1107. [PMID: 24912505 DOI: 10.1177/1535370214537738] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The desire to develop and evaluate drugs as potential countermeasures for biological and chemical threats requires test systems that can also substitute for the clinical trials normally crucial for drug development. Current animal models have limited predictivity for drug efficacy in humans as the large majority of drugs fails in clinical trials. We have limited understanding of the function of the central nervous system and the complexity of the brain, especially during development and neuronal plasticity. Simple in vitro systems do not represent physiology and function of the brain. Moreover, the difficulty of studying interactions between human genetics and environmental factors leads to lack of knowledge about the events that induce neurological diseases. Microphysiological systems (MPS) promise to generate more complex in vitro human models that better simulate the organ's biology and function. MPS combine different cell types in a specific three-dimensional (3D) configuration to simulate organs with a concrete function. The final aim of these MPS is to combine different "organoids" to generate a human-on-a-chip, an approach that would allow studies of complex physiological organ interactions. The recent discovery of induced pluripotent stem cells (iPSCs) gives a range of possibilities allowing cellular studies of individuals with different genetic backgrounds (e.g., human disease models). Application of iPSCs from different donors in MPS gives the opportunity to better understand mechanisms of the disease and can be a novel tool in drug development, toxicology, and medicine. In order to generate a brain-on-a-chip, we have established a 3D model from human iPSCs based on our experience with a 3D rat primary aggregating brain model. After four weeks of differentiation, human 3D aggregates stain positive for different neuronal markers and show higher gene expression of various neuronal differentiation markers compared to 2D cultures. Here we present the applications and challenges of this emerging technology.
Collapse
Affiliation(s)
- David Pamies
- Centers for Alternatives to Animal Testing (CAAT) at Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, Baltimore, MD 21205, USA; University of Konstanz, POB 600, Konstanz 78457, Germany
| | - Thomas Hartung
- Centers for Alternatives to Animal Testing (CAAT) at Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, Baltimore, MD 21205, USA; University of Konstanz, POB 600, Konstanz 78457, Germany
| | - Helena T Hogberg
- Centers for Alternatives to Animal Testing (CAAT) at Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, Baltimore, MD 21205, USA; University of Konstanz, POB 600, Konstanz 78457, Germany
| |
Collapse
|
29
|
Wright LS, Phillips MJ, Pinilla I, Hei D, Gamm DM. Induced pluripotent stem cells as custom therapeutics for retinal repair: progress and rationale. Exp Eye Res 2014; 123:161-72. [PMID: 24534198 PMCID: PMC4047146 DOI: 10.1016/j.exer.2013.12.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 10/25/2013] [Accepted: 12/02/2013] [Indexed: 12/17/2022]
Abstract
Human pluripotent stem cells have made a remarkable impact on science, technology and medicine by providing a potentially unlimited source of human cells for basic research and clinical applications. In recent years, knowledge gained from the study of human embryonic stem cells and mammalian somatic cell reprogramming has led to the routine production of human induced pluripotent stem cells (hiPSCs) in laboratories worldwide. hiPSCs show promise for use in transplantation, high throughput drug screening, "disease-in-a-dish" modeling, disease gene discovery, and gene therapy testing. This review will focus on the first application, beginning with a discussion of methods for producing retinal lineage cells that are lost in inherited and acquired forms of retinal degenerative disease. The selection of appropriate hiPSC-derived donor cell type(s) for transplantation will be discussed, as will the caveats and prerequisite steps to formulating a clinical Good Manufacturing Practice (cGMP) product for clinical trials.
Collapse
Affiliation(s)
- Lynda S Wright
- Waisman Center, University of Wisconsin, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin, Madison, WI, USA
| | - M Joseph Phillips
- Waisman Center, University of Wisconsin, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin, Madison, WI, USA
| | - Isabel Pinilla
- Department of Ophthalmology, Lozano Blesa Hospital and Aragones Health Sciences Institute, Zaragoza, Spain
| | - Derek Hei
- Waisman Center, University of Wisconsin, Madison, WI, USA
| | - David M Gamm
- Waisman Center, University of Wisconsin, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin, Madison, WI, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
30
|
Larcher F, Espada J, Díaz-Ley B, Jaén P, Juarranz A, Quintanilla M. New experimental models of skin homeostasis and diseases. ACTAS DERMO-SIFILIOGRAFICAS 2014; 106:17-28. [PMID: 24878038 DOI: 10.1016/j.ad.2014.03.008] [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] [Received: 10/08/2013] [Revised: 02/25/2014] [Accepted: 03/03/2014] [Indexed: 12/19/2022] Open
Abstract
Homeostasis, whose regulation at the molecular level is still poorly understood, is intimately related to the functions of epidermal stem cells. Five research groups have been brought together to work on new in vitro and in vivo skin models through the SkinModel-CM program, under the auspices of the Spanish Autonomous Community of Madrid. This project aims to analyze the functions of DNA methyltransferase 1, endoglin, and podoplanin in epidermal stem cell activity, homeostasis, and skin cancer. These new models include 3-dimensional organotypic cultures, immunodeficient skin-humanized mice, and genetically modified mice. Another aim of the program is to use skin-humanized mice to model dermatoses such as Gorlin syndrome and xeroderma pigmentosum in order to optimize new protocols for photodynamic therapy.
Collapse
Affiliation(s)
- F Larcher
- Unidad de Medicina Regenerativa, Departamento de Investigación Básica, División de Biomedicina Epitelial, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) y Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, España
| | - J Espada
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid (UAM), Madrid, España; Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-UAM, Madrid, España
| | - B Díaz-Ley
- Unidad de Dermatología, Hospital Ramón y Cajal, Madrid, España
| | - P Jaén
- Unidad de Dermatología, Hospital Ramón y Cajal, Madrid, España
| | - A Juarranz
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid (UAM), Madrid, España.
| | - M Quintanilla
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-UAM, Madrid, España
| |
Collapse
|
31
|
Feltes BC, Bonatto D. Combining small molecules for cell reprogramming through an interatomic analysis. MOLECULAR BIOSYSTEMS 2014; 9:2741-63. [PMID: 24056910 DOI: 10.1039/c3mb70159j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The knowledge available about the application and generation of induced pluripotent stem cells (iPSC) has grown since their discovery, and new techniques to enhance the reprogramming process have been described. Among the new approaches to induce iPSC that have gained great attention is the use of small molecules for reprogramming. The application of small molecules, unlike genetic manipulation, provides for control of the reprogramming process through the shifting of concentrations and the combination of different molecules. However, different researchers have reported the use of "reprogramming cocktails" with variable results and drug combinations. Thus, the proper combination of small molecules for successful and enhanced reprogramming is a matter for discussion. However, testing all potential drug combinations in different cell lineages is very costly and time-consuming. Therefore, in this article, we discuss the use of already employed molecules for iPSC generation, followed by the application of systems chemo-biology tools to create different data sets of protein-protein (PPI) and chemical-protein (CPI) interaction networks based on the knowledge of already used and new reprogramming cocktail combinations. We further analyzed the biological processes associated with PPI-CPI networks and provided new potential protein targets to be inhibited or expressed for stem cell reprogramming. In addition, we applied a new interference analysis to prospective targets that could negatively affect the classical pluripotency-associated factors (SOX2, NANOG, KLF4 and OCT4) and thus potentially improve reprogramming protocols.
Collapse
Affiliation(s)
- Bruno César Feltes
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves 9500 - Prédio 43421 - Sala 219, Porto Alegre, Caixa Postal 15005, RS - Brazil.
| | | |
Collapse
|
32
|
Harding J, Mirochnitchenko O. Preclinical studies for induced pluripotent stem cell-based therapeutics. J Biol Chem 2013; 289:4585-93. [PMID: 24362021 DOI: 10.1074/jbc.r113.463737] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) and their differentiated derivatives can potentially be applied to cell-based therapy for human diseases. The properties of iPSCs are being studied intensively both to understand the basic biology of pluripotency and cellular differentiation and to solve problems associated with therapeutic applications. Examples of specific preclinical applications summarized briefly in this minireview include the use of iPSCs to treat diseases of the liver, nervous system, eye, and heart and metabolic conditions such as diabetes. Early stage studies illustrate the potential of iPSC-derived cells and have identified several challenges that must be addressed before moving to clinical trials. These include rigorous quality control and efficient production of required cell populations, improvement of cell survival and engraftment, and development of technologies to monitor transplanted cell behavior for extended periods of time. Problems related to immune rejection, genetic instability, and tumorigenicity must be solved. Testing the efficacy of iPSC-based therapies requires further improvement of animal models precisely recapitulating human disease conditions.
Collapse
Affiliation(s)
- John Harding
- From the Division of Comparative Medicine, Office of Research Infrastructure Programs, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, Maryland 20892
| | | |
Collapse
|
33
|
Cooper S, Brockdorff N. Genome-wide shRNA screening to identify factors mediating Gata6 repression in mouse embryonic stem cells. Development 2013; 140:4110-5. [PMID: 24046324 PMCID: PMC3775421 DOI: 10.1242/dev.094615] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The use of whole-genome pooled shRNA libraries in loss-of-function screening in tissue culture models provides an effective means to identify novel factors acting in pathways of interest. Embryonic stem cells (ESCs) offer a unique opportunity to study processes involved in stem cell pluripotency and differentiation. Here, we report a genome-wide shRNA screen in ESCs to identify novel components involved in repression of the Gata6 locus, using a cell viability-based screen, which offers the benefits of stable shRNA integration and a robust and simple protocol for hit identification. Candidate factors identified were enriched for transcription factors and included known Polycomb proteins and other chromatin-modifying factors. We identified the protein Bcor, which is known to associate in complexes with the Polycomb protein Ring1B, and verified its importance in Gata6 repression in ESCs. Potential further applications of such a screening strategy could allow the identification of factors important for regulation of gene expression and pluripotency.
Collapse
Affiliation(s)
- Sarah Cooper
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | | |
Collapse
|
34
|
Human induced pluripotent stem cells from basic research to potential clinical applications in cancer. BIOMED RESEARCH INTERNATIONAL 2013; 2013:430290. [PMID: 24288679 PMCID: PMC3830845 DOI: 10.1155/2013/430290] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 09/15/2013] [Indexed: 12/29/2022]
Abstract
The human induced pluripotent stem cells (hiPSCs) are derived from a direct reprogramming of human somatic cells to a pluripotent stage through ectopic expression of specific transcription factors. These cells have two important properties, which are the self-renewal capacity and the ability to differentiate into any cell type of the human body. So, the discovery of hiPSCs opens new opportunities in biomedical sciences, since these cells may be useful for understanding the mechanisms of diseases in the production of new diseases models, in drug development/drug toxicity tests, gene therapies, and cell replacement therapies. However, the hiPSCs technology has limitations including the potential for the development of genetic and epigenetic abnormalities leading to tumorigenicity. Nowadays, basic research in the hiPSCs field has made progress in the application of new strategies with the aim to enable an efficient production of high-quality of hiPSCs for safety and efficacy, necessary to the future application for clinical practice. In this review, we show the recent advances in hiPSCs' basic research and some potential clinical applications focusing on cancer. We also present the importance of the use of statistical methods to evaluate the possible validation for the hiPSCs for future therapeutic use toward personalized cell therapies.
Collapse
|
35
|
Hematopoietic specification from human pluripotent stem cells: current advances and challenges toward de novo generation of hematopoietic stem cells. Blood 2013; 122:4035-46. [PMID: 24124087 DOI: 10.1182/blood-2013-07-474825] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Significant advances in cellular reprogramming technologies and hematopoietic differentiation from human pluripotent stem cells (hPSCs) have already enabled the routine production of multiple lineages of blood cells in vitro and opened novel opportunities to study hematopoietic development, model genetic blood diseases, and manufacture immunologically matched cells for transfusion and cancer immunotherapy. However, the generation of hematopoietic cells with robust and sustained multilineage engraftment has not been achieved. Here, we highlight the recent advances in understanding the molecular and cellular pathways leading to blood development from hPSCs and discuss potential approaches that can be taken to facilitate the development of technologies for de novo production of hematopoietic stem cells.
Collapse
|
36
|
Kolaja K. Stem cell derived tissues and microphysiological systems: a paradigm shifting moment. Vet J 2013; 198:1-2. [PMID: 24129108 DOI: 10.1016/j.tvjl.2013.09.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 09/06/2013] [Accepted: 09/07/2013] [Indexed: 12/11/2022]
Affiliation(s)
- Kyle Kolaja
- Cellular Dynamics International, 525 Science Drive, Madison, WI 53711, USA.
| |
Collapse
|
37
|
Bernal JA. RNA-based tools for nuclear reprogramming and lineage-conversion: towards clinical applications. J Cardiovasc Transl Res 2013; 6:956-68. [PMID: 23852582 PMCID: PMC3838600 DOI: 10.1007/s12265-013-9494-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 06/21/2013] [Indexed: 02/06/2023]
Abstract
The therapeutic potential of induced pluripotent stem cells (iPSCs) is well established. Safety concerns remain, however, and these have driven considerable efforts aimed at avoiding host genome alteration during the reprogramming process. At present, the tools used to generate human iPSCs include (1) DNA-based integrative and non-integrative methods and (2) DNA-free reprogramming technologies, including RNA-based approaches. Because of their combined efficiency and safety characteristics, RNA-based methods have emerged as the most promising tool for future iPSC-based regenerative medicine applications. Here, I will discuss novel recent advances in reprogramming technology, especially those utilizing the Sendai virus (SeV) and synthetic modified mRNA. In the future, these technologies may find utility in iPSC reprogramming for cellular lineage-conversion, and its subsequent use in cell-based therapies.
Collapse
Affiliation(s)
- Juan A Bernal
- Cardiovascular Development and Repair Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain,
| |
Collapse
|
38
|
Hall V, Hinrichs K, Lazzari G, Betts DH, Hyttel P. Early embryonic development, assisted reproductive technologies, and pluripotent stem cell biology in domestic mammals. Vet J 2013; 197:128-42. [PMID: 23810186 DOI: 10.1016/j.tvjl.2013.05.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 05/03/2013] [Accepted: 05/04/2013] [Indexed: 01/01/2023]
Abstract
Over many decades assisted reproductive technologies, including artificial insemination, embryo transfer, in vitro production (IVP) of embryos, cloning by somatic cell nuclear transfer (SCNT), and stem cell culture, have been developed with the aim of refining breeding strategies for improved production and health in animal husbandry. More recently, biomedical applications of these technologies, in particular, SCNT and stem cell culture, have been pursued in domestic mammals in order to create models for human disease and therapy. The following review focuses on presenting important aspects of pre-implantation development in cattle, pigs, horses, and dogs. Biological aspects and impact of assisted reproductive technologies including IVP, SCNT, and culture of pluripotent stem cells are also addressed.
Collapse
Affiliation(s)
- V Hall
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
39
|
Ghosal S, Das S, Chakrabarti J. Long noncoding RNAs: new players in the molecular mechanism for maintenance and differentiation of pluripotent stem cells. Stem Cells Dev 2013; 22:2240-53. [PMID: 23528033 DOI: 10.1089/scd.2013.0014] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Maintenance of the pluripotent state or differentiation of the pluripotent state into any germ layer depends on the factors that orchestrate expression of thousands of genes through epigenetic, transcriptional, and post-transcriptional regulation. Long noncoding RNAs (lncRNAs) are implicated in the complex molecular circuitry in the developmental processes. The ENCODE project has opened up new avenues for studying these lncRNA transcripts with the availability of new datasets for lncRNA annotation and regulation. Expression studies identified hundreds of long noncoding RNAs differentially expressed in the pluripotent state, and many of these lncRNAs are found to control the pluripotency and stemness in embryonic and induced pluripotent stem cells or, in the reverse way, promote differentiation of pluripotent cells. They are generally transcriptionally activated or repressed by pluripotency-associated transcription factors and function as molecular mediators of gene expression that determine the pluripotent state of the cell. They can act as molecular scaffolds or guides for the chromatin-modifying complexes to direct them to bind into specific genomic loci to impart a repressive or activating effect on gene expression, or they can transcriptionally or post-transcriptionally regulate gene expression by diverse molecular mechanisms. This review focuses on recent findings on the regulatory role of lncRNAs in two main aspects of pluripotency, namely, self renewal and differentiation into any lineage, and elucidates the underlying molecular mechanisms that are being uncovered lately.
Collapse
Affiliation(s)
- Suman Ghosal
- Indian Association for the Cultivation of Science, Kolkata, India
| | | | | |
Collapse
|
40
|
Sartipy P. Advancing pluripotent stem cell culture: it is a matter of setting the standard. Stem Cells Dev 2013; 22:1159-61. [PMID: 23330735 DOI: 10.1089/scd.2012.0668] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Human pluripotent stem cells (hPSCs), defined by their ability to proliferate indefinitely and the capacity to differentiate into all tissue cell types of the adult, represent a platform for the realization of breakthrough technologies for industrial and regenerative medicine applications. We have witnessed tremendous developments over the last decade related to methods for establishment, maintenance, differentiation, and applications of hPSCs and their derivatives. Despite all progress made in the hPSC field, there are still fundamental issues yet to be resolved. For example, our understanding of the pluripotent state remains limited, which in turn may have substantial consequences on how we interpret and communicate scientific data concerning hPSCs. This brief commentary aims to highlight recent important findings that demonstrate additional levels of complexity to the current assessment of pluripotent stem cell cultures. In addition, these data may help to provide some explanations for the challenges in reproducing hPSC differentiation protocols across laboratories.
Collapse
Affiliation(s)
- Peter Sartipy
- Cellectis Stem Cells , Cellartis AB, Göteborg, Sweden.
| |
Collapse
|