1
|
Sugimoto C, Fujita H, Wakao H. Mice Generated with Induced Pluripotent Stem Cells Derived from Mucosal-Associated Invariant T Cells. Biomedicines 2024; 12:137. [PMID: 38255242 PMCID: PMC10813358 DOI: 10.3390/biomedicines12010137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/28/2023] [Accepted: 01/01/2024] [Indexed: 01/24/2024] Open
Abstract
The function of mucosal-associated invariant T (MAIT) cells, a burgeoning member of innate-like T cells abundant in humans and implicated in many diseases, remains obscure. To explore this, mice with a rearranged T cell receptor (TCR) α or β locus, specific for MAIT cells, were generated via induced pluripotent stem cells derived from MAIT cells and were designated Vα19 and Vβ8 mice, respectively. Both groups of mice expressed large numbers of MAIT cells. The MAIT cells from these mice were activated by cytokines and an agonist to produce IFN-γ and IL-17. While Vβ8 mice showed resistance in a cancer metastasis model, Vα19 mice did not. Adoptive transfer of MAIT cells from the latter into the control mice, however, recapitulated the resistance. These mice present an implication for understanding the role of MAIT cells in health and disease and in developing treatments for the plethora of diseases in which MAIT cells are implicated.
Collapse
Affiliation(s)
| | | | - Hiroshi Wakao
- Host Defense Division, Research Centre for Advanced Medical Science, Dokkyo Medical University, Mibu 321-0293, Japan; (C.S.)
| |
Collapse
|
2
|
Watanabe N, Hirose M, Hasegawa A, Mochida K, Ogura A, Inoue K. Derivation of embryonic stem cells from wild-derived mouse strains by nuclear transfer using peripheral blood cells. Sci Rep 2023; 13:11175. [PMID: 37430017 DOI: 10.1038/s41598-023-38341-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/06/2023] [Indexed: 07/12/2023] Open
Abstract
Wild-derived mouse strains have been extensively used in biomedical research because of the high level of inter-strain polymorphisms and phenotypic variations. However, they often show poor reproductive performance and are difficult to maintain by conventional in vitro fertilization and embryo transfer. In this study, we examined the technical feasibility of derivation of nuclear transfer embryonic stem cells (ntESCs) from wild-derived mouse strains for their safe genetic preservation. We used leukocytes collected from peripheral blood as nuclear donors without sacrificing them. We successfully established 24 ntESC lines from two wild-derived strains of CAST/Ei and CASP/1Nga (11 and 13 lines, respectively), both belonging to Mus musculus castaneus, a subspecies of laboratory mouse. Most (23/24) of these lines had normal karyotype, and all lines examined showed teratoma formation ability (4 lines) and pluripotent marker gene expression (8 lines). Two male lines examined (one from each strain) were proven to be competent to produce chimeric mice following injection into host embryos. By natural mating of these chimeric mice, the CAST/Ei male line was confirmed to have germline transmission ability. Our results demonstrate that inter-subspecific ntESCs derived from peripheral leukocytes could provide an alternative strategy for preserving invaluable genetic resources of wild-derived mouse strains.
Collapse
Affiliation(s)
- Naomi Watanabe
- RIKEN BioResource Research Center, Tsukuba, Ibaraki, Japan
- Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Michiko Hirose
- RIKEN BioResource Research Center, Tsukuba, Ibaraki, Japan
| | - Ayumi Hasegawa
- RIKEN BioResource Research Center, Tsukuba, Ibaraki, Japan
| | - Keiji Mochida
- RIKEN BioResource Research Center, Tsukuba, Ibaraki, Japan
| | - Atsuo Ogura
- RIKEN BioResource Research Center, Tsukuba, Ibaraki, Japan.
- Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| | - Kimiko Inoue
- RIKEN BioResource Research Center, Tsukuba, Ibaraki, Japan.
- Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
3
|
Aoki T, Motohashi S, Koseki H. Regeneration of invariant natural killer T (iNKT) cells: application of iPSC technology for iNKT cell-targeted tumor immunotherapy. Inflamm Regen 2023; 43:27. [PMID: 37170375 PMCID: PMC10176773 DOI: 10.1186/s41232-023-00275-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 03/29/2023] [Indexed: 05/13/2023] Open
Abstract
Invariant natural killer T (iNKT) cells are a subset of innate-like T cells restricted by a major histocompatibility complex (MHC) class I-like molecule, CD1d. iNKT cells express an invariant T cell receptor (TCR) encoded by Vα14 Jα18 in mice and Vα24 Jα18 in humans and are activated by recognizing glycolipid antigens, such as α-galactosylceramide (αGalCer), presented by CD1d. iNKT cells exhibit anti-tumor activity via their NK-like cytotoxicity and adjuvant activity. Although iNKT cell-targeted immunotherapy is a conceptually promising approach, we still found a technical hurdle for its clinical implementation which is mainly due to the low frequency of iNKT cells, particularly in humans. To compensate for this, we proposed to generate adequate numbers of clinically competent NKT cells from induced pluripotent stem cells (iPSCs) for cancer immunotherapy. Toward this goal, we first obtained the proof of concept (POC) for this approach in mice. We developed a technology to differentiate iPSCs into iNKT cells (iPSC-iNKT cells) and found iPSC-iNKT cells efficiently rejected a syngeneic experimental thymoma by inducing antigen-specific CD8 T cells. After achieving the POC in mice, we developed human iPSC-iNKT cells, which had a high correlation in their gene expression profiles with parental iNKT cells. Human iPSC-iNKT cells also exhibited anti-tumor activity and adjuvant activity for human NK cells in vivo. Based on this supporting evidence for the anti-tumor activity of human iPSC-iNKT cells, we began to generate good manufacturing practice (GMP)-grade iPSC-iNKT cells. As of now, the first-in-human clinical trial of iPSC-iNKT cell therapy is ongoing as a single-agent, dose-escalation study for patients with advanced head and neck cancer. Demonstration of the safety of iPSC-iNKT cell therapy may allow us to improve the strategy by further reinforcing the therapeutic activity of iPSC-iNKT, cells either by gene-editing or combinatorial use with other immune cell products such as dendritic cells. Sixteen years after the establishment of the iPSC technology, we are reaching the first checkpoint to evaluate the clinical efficacy of iPSC-derived immune cells.
Collapse
Affiliation(s)
- Takahiro Aoki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8670, Japan.
| | - Shinichiro Motohashi
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8670, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
4
|
Moura MT. Cloning by SCNT: Integrating Technical and Biology-Driven Advances. Methods Mol Biol 2023; 2647:1-35. [PMID: 37041327 DOI: 10.1007/978-1-0716-3064-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Somatic cell nuclear transfer (SCNT) into enucleated oocytes initiates nuclear reprogramming of lineage-committed cells to totipotency. Pioneer SCNT work culminated with cloned amphibians from tadpoles, while technical and biology-driven advances led to cloned mammals from adult animals. Cloning technology has been addressing fundamental questions in biology, propagating desired genomes, and contributing to the generation of transgenic animals or patient-specific stem cells. Nonetheless, SCNT remains technically complex and cloning efficiency relatively low. Genome-wide technologies revealed barriers to nuclear reprogramming, such as persistent epigenetic marks of somatic origin and reprogramming resistant regions of the genome. To decipher the rare reprogramming events that are compatible with full-term cloned development, it will likely require technical advances for large-scale production of SCNT embryos alongside extensive profiling by single-cell multi-omics. Altogether, cloning by SCNT remains a versatile technology, while further advances should continuously refresh the excitement of its applications.
Collapse
Affiliation(s)
- Marcelo Tigre Moura
- Chemical Biology Graduate Program, Federal University of São Paulo - UNIFESP, Campus Diadema, Diadema - SP, Brazil
| |
Collapse
|
5
|
da Silva CG, Martins CF. Stem Cells as Nuclear Donors for Mammalian Cloning. Methods Mol Biol 2023; 2647:105-119. [PMID: 37041331 DOI: 10.1007/978-1-0716-3064-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Mammals are routinely cloned by introducing somatic nuclei into enucleated oocytes. Cloning contributes to propagating desired animals, to germplasm conservation efforts, among other applications. A challenge to more broader use of this technology is the relatively low cloning efficiency, which inversely correlates with donor cell differentiation status. Emerging evidence suggests that adult multipotent stem cells improve cloning efficiency, while the greater potential of embryonic stem cells for cloning remains restricted to the mouse. The derivation of pluripotent or totipotent stem cells from livestock and wild species and their association with modulators of epigenetic marks in donor cells should increase cloning efficiency.
Collapse
Affiliation(s)
- Carolina Gonzales da Silva
- Federal Institute of Education, Science and Technology of Bahia, Campus Xique-Xique, Xique-Xique, Bahia, Brazil
| | - Carlos Frederico Martins
- Brazilian Agricultural Research Corporation (Embrapa Cerrados), Brasília, Federal District, Brazil.
| |
Collapse
|
6
|
Bolton RL, Mooney A, Pettit MT, Bolton AE, Morgan L, Drake GJ, Appeltant R, Walker SL, Gillis JD, Hvilsom C. Resurrecting biodiversity: advanced assisted reproductive technologies and biobanking. REPRODUCTION AND FERTILITY 2022; 3:R121-R146. [PMID: 35928671 PMCID: PMC9346332 DOI: 10.1530/raf-22-0005] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 06/30/2022] [Indexed: 11/21/2022] Open
Abstract
Biodiversity is defined as the presence of a variety of living organisms on the Earth that is essential for human survival. However, anthropogenic activities are causing the sixth mass extinction, threatening even our own species. For many animals, dwindling numbers are becoming fragmented populations with low genetic diversity, threatening long-term species viability. With extinction rates 1000-10,000 times greater than natural, ex situ and in situ conservation programmes need additional support to save species. The indefinite storage of cryopreserved (-196°C) viable cells and tissues (cryobanking), followed by assisted or advanced assisted reproductive technology (ART: utilisation of oocytes and spermatozoa to generate offspring; aART: utilisation of somatic cell genetic material to generate offspring), may be the only hope for species' long-term survival. As such, cryobanking should be considered a necessity for all future conservation strategies. Following cryopreservation, ART/aART can be used to reinstate lost genetics back into a population, resurrecting biodiversity. However, for this to be successful, species-specific protocol optimisation and increased knowledge of basic biology for many taxa are required. Current ART/aART is primarily focused on mammalian taxa; however, this needs to be extended to all, including to some of the most endangered species: amphibians. Gamete, reproductive tissue and somatic cell cryobanking can fill the gap between losing genetic diversity today and future technological developments. This review explores species prioritisation for cryobanking and the successes and challenges of cryopreservation and multiple ARTs/aARTs. We here discuss the value of cryobanking before more species are lost and the potential of advanced reproductive technologies not only to halt but also to reverse biodiversity loss. Lay summary The world is undergoing its sixth mass extinction; however, unlike previous events, the latest is caused by human activities and is resulting in the largest loss of biodiversity (all living things on Earth) for 65 million years. With an extinction rate 1000-10,000-fold greater than natural, this catastrophic decline in biodiversity is threatening our own survival. As the number of individuals within a species declines, genetic diversity reduces, threatening their long-term existence. In this review, the authors summarise approaches to indefinitely preserve living cells and tissues at low temperatures (cryobanking) and the technologies required to resurrect biodiversity. In the future when appropriate techniques become available, these living samples can be thawed and used to reinstate genetic diversity and produce live young ones of endangered species, enabling their long-term survival. The successes and challenges of genome resource cryopreservation are discussed to enable a move towards a future of stable biodiversity.
Collapse
Affiliation(s)
- Rhiannon L Bolton
- Nature’s SAFE, Chapel Field Stud, Ash Lane, Whitchurch, Shropshire, UK
| | | | - Matt T Pettit
- Nature’s SAFE, Chapel Field Stud, Ash Lane, Whitchurch, Shropshire, UK
- IMT International Limited, Tattenhall, Chester, UK
| | - Anthony E Bolton
- Nature’s SAFE, Chapel Field Stud, Ash Lane, Whitchurch, Shropshire, UK
| | - Lucy Morgan
- Gemini Genetics, Chapel Field Stud, Ash Lane, Whitchurch, UK
| | | | - Ruth Appeltant
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Women’s Centre, Level 3, John Radcliffe Hospital, Oxford, UK
| | - Susan L Walker
- Nature’s SAFE, Chapel Field Stud, Ash Lane, Whitchurch, Shropshire, UK
- Chester Zoo, Upton-by-Chester, UK
| | - James D Gillis
- South-East Zoo Alliance for Reproduction & Conservation, Yulee, Florida, USA
| | | |
Collapse
|
7
|
Kawamoto H, Masuda K, Nagano S. Regeneration of antigen-specific T cells by using induced pluripotent stem cell (iPSC) technology. Int Immunol 2021; 33:827-833. [PMID: 34661676 DOI: 10.1093/intimm/dxab091] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/15/2021] [Indexed: 01/20/2023] Open
Abstract
In currently ongoing adoptive T-cell therapies, T cells collected from the patient are given back to the patient after ex vivo cell activation and expansion. In some cases, T cells are transduced with chimeric antigen receptor (CAR) or T-cell receptor (TCR) genes during the ex vivo culture period. Although such strategies have been shown to be effective in some types of cancer, there remain issues to be solved; these methods (i) are time-consuming, (ii) are costly and (iii) it is difficult to guarantee the quality because the products depend on patient-derived T cells. To address these issues, several groups including ours have developed methods in which cytotoxic cells are mass-produced by using induced pluripotent stem cell (iPSC) technology. For the regeneration of T cells, the basic idea is as follows: iPSCs produced from T cells inherit rearranged TCR genes, and thus all regenerated T cells should express the same TCR. Based on this idea, various types of T cells have been regenerated, including conventional cytotoxic T lymphocytes (CTLs), γδT cells, NKT cells and mucosal-associated invariant T (MAIT) cells. On the other hand, any cytotoxic cells can be used as the base cells into which CAR is introduced, and thus iPSC-derived NK cells have been developed. To apply the iPSC-based cell therapy in an allogeneic setting, the authors' group developed a method in which non-T-cell-derived iPSCs are transduced with exogenous TCR genes (TCR-iPSC method). This approach is being prepared for a clinical trial to be realized in Kyoto University Hospital, in which acute myeloid leukemia patients will be treated by the regenerated WT1 antigen-specific CTLs.
Collapse
Affiliation(s)
- Hiroshi Kawamoto
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Regenerative Immunology, International Center for Cell and Gene Therapy, Fujita Health University, Toyoake, Japan
| | - Kyoko Masuda
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Seiji Nagano
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
8
|
Role of CD4 + T Cells in Allergic Airway Diseases: Learning from Murine Models. Int J Mol Sci 2020; 21:ijms21207480. [PMID: 33050549 PMCID: PMC7589900 DOI: 10.3390/ijms21207480] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 01/18/2023] Open
Abstract
The essential contribution of CD4+ T cells in allergic airway diseases has been demonstrated, especially by using various murine models of antigen-induced airway inflammation. In addition to antigen-immunized mouse models employing mast cell-deficient mice and CD4+ T cell-depleting procedure, antigen-specific CD4+ T cell transfer models have revealed the possible development of allergic inflammation solely dependent on CD4+ T cells. Regardless of the classical Th1/Th2 theory, various helper T cell subsets have the potential to induce different types of allergic inflammation. T cell receptor (TCR)-transgenic (Tg) mice have been used for investigating T cell-mediated immune responses. Besides, we have recently generated cloned mice from antigen-specific CD4+ T cells through somatic cell nuclear transfer. In contrast to TCR-Tg mice that express artificially introduced TCR, the cloned mice express endogenously regulated antigen-specific TCR. Upon antigen exposure, the mite antigen-reactive T cell-cloned mice displayed strong airway inflammation accompanied by bronchial hyperresponsiveness in a short time period. Antigen-specific CD4+ T cell-cloned mice are expected to be useful for investigating the detailed role of CD4+ T cells in various allergic diseases and for evaluating novel anti-allergic drugs.
Collapse
|
9
|
Abstract
The mouse is the most extensively used mammalian laboratory species in biology and medicine because of the ready availability of a wide variety of defined genetic and gene-modified strains and abundant genetic information. Its small size and rapid generation turnover are also advantages compared with other experimental animals. Using these advantages, somatic cell nuclear transfer (SCNT) in mice has provided invaluable information on epigenetics related to SCNT technology and cloning, playing a leading role in relevant technical improvements. These improvements include treatment with histone deacetylase inhibitors, correction of Xist gene expression (controlling X chromosome inactivation), and removal of methylated histones from SCNT-generated embryos, which have proven to be effective for SCNT cloning of other species. However, even with the best combination of these treatments, the birth rate in cloned offspring is still lower than intracytoplasmic sperm injection (ICSI) or in vitro fertilization (IVF). One remaining issue associated with SCNT is placental enlargement (hyperplasia) found in late pregnancy, but this abnormality might not be a major cause for the low efficiency of SCNT because many SCNT-derived embryos die before their placentas start to enlarge at midgestation (early postimplantation stage). It is known that, at this stage, undifferentiated trophoblast cells in the extraembryonic tissue of SCNT-derived embryos fail to proliferate. Understanding the molecular mechanisms is essential for further technical improvements of mouse SCNT, which might also provide clues for technical breakthroughs in mammalian SCNT and cloning in general.
Collapse
Affiliation(s)
- Atsuo Ogura
- RIKEN BioResource Research Center, Ibaraki, 305-0074, Japan; Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, 305-8572, Japan; RIKEN Cluster for Pioneering Research, Saitama, 351-0198, Japan.
| |
Collapse
|
10
|
Dashtsoodol N, Bortoluzzi S, Schmidt-Supprian M. T Cell Receptor Expression Timing and Signal Strength in the Functional Differentiation of Invariant Natural Killer T Cells. Front Immunol 2019; 10:841. [PMID: 31080448 PMCID: PMC6497757 DOI: 10.3389/fimmu.2019.00841] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/01/2019] [Indexed: 12/19/2022] Open
Abstract
The CD1d-restricted Vα14 invariant NKT (iNKT) cell lineage in mice (Vα24 in humans) represents an evolutionary conserved innate-like immune cell type that recognizes glycolipid antigens. Because of their unique ability to promptly secrete copious amounts of both pro-inflammatory and anti-inflammatory cytokines, typically produced by different T helper cell types, iNKT cells are implicated in the regulation of various pathologic conditions such as infection, allergy, autoimmune disease, maintenance of transplantation tolerance, and cancer. This striking multifaceted role in immune regulation is correlated with the presence of multiple functionally distinct iNKT cell subsets that can be distinguished based on the expression of characteristic surface markers and transcription factors. However, to date it, remains largely unresolved how this puzzling diversity of iNKT cell functional subsets emerges and what factors dictate the type of effector cell differentiation during the thymic differentiation considering the mono-specific nature of their T cell receptor (TCR) and their selecting molecule CD1d. Here, we summarize recent findings focusing on the role of TCR-mediated signaling and discuss possible mechanisms that may influence the sub-lineage choice of iNKT cells.
Collapse
Affiliation(s)
- Nyambayar Dashtsoodol
- Department of Hematology and Medical Oncology, Klinikum rechts der Isar and TranslaTUM Cancer Center, Technische Universität München, München, Germany.,Department of Microbiology and Immunology, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Sabrina Bortoluzzi
- Department of Hematology and Medical Oncology, Klinikum rechts der Isar and TranslaTUM Cancer Center, Technische Universität München, München, Germany
| | - Marc Schmidt-Supprian
- Department of Hematology and Medical Oncology, Klinikum rechts der Isar and TranslaTUM Cancer Center, Technische Universität München, München, Germany
| |
Collapse
|
11
|
Fabrèges D, Daniel N, Duranthon V, Peyriéras N. Control of the proportion of inner cells by asymmetric divisions and the ensuing resilience of cloned rabbit embryos. Development 2018; 145:dev.152041. [PMID: 29567671 PMCID: PMC5964649 DOI: 10.1242/dev.152041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 03/13/2018] [Indexed: 01/14/2023]
Abstract
Mammalian embryo cloning by nuclear transfer has a low success rate. This is hypothesized to correlate with a high variability of early developmental steps that segregate outer cells, which are fated to extra-embryonic tissues, from inner cells, which give rise to the embryo proper. Exploring the cell lineage of wild-type embryos and clones, imaged in toto until hatching, highlights the respective contributions of cell proliferation, death and asymmetric divisions to phenotypic variability. Preferential cell death of inner cells in clones, probably pertaining to the epigenetic plasticity of the transferred nucleus, is identified as a major difference with effects on the proportion of inner cell. In wild type and clones, similar patterns of outer cell asymmetric divisions are shown to be essential to the robust proportion of inner cells observed in wild type. Asymmetric inner cell division, which is not described in mice, is identified as a regulator of the proportion of inner cells and likely gives rise to resilient clones. Summary: A unique quantitative approach based on complete reconstruction of the cell lineage that unveils an unknown mechanism of size control in cell populations of rabbit blastocysts, wild types or clones.
Collapse
Affiliation(s)
- Dimitri Fabrèges
- BioEmergences Laboratory, CNRS USR 3695, 91190 Gif-sur-Yvette, France
| | - Nathalie Daniel
- UMR BDR, INRA, ENVA, Université Paris Saclay, 78350, Jouy en Josas, France
| | | | - Nadine Peyriéras
- BioEmergences Laboratory, CNRS USR 3695, 91190 Gif-sur-Yvette, France
| |
Collapse
|
12
|
Kawamoto H, Masuda K, Nagano S, Maeda T. Cloning and expansion of antigen-specific T cells using iPS cell technology: development of "off-the-shelf" T cells for the use in allogeneic transfusion settings. Int J Hematol 2018; 107:271-277. [PMID: 29388165 DOI: 10.1007/s12185-018-2399-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 01/12/2018] [Indexed: 10/18/2022]
Abstract
Recent advances in adoptive immunotherapy using cytotoxic T lymphocytes (CTLs) have led to moderate therapeutic anti-cancer effects in clinical trials. However, a critical issue, namely that CTLs collected from patients are easily exhausted during expansion culture, has yet to be solved. To address this issue, we have been developing a strategy which utilizes induced pluripotent stem cell (iPSC) technology. This strategy is based on the idea that when iPSCs are produced from antigen-specific CTLs, CTLs regenerated from such iPSCs should show the same antigen specificity as the original CTLs. Pursuing this idea, we previously succeeded in regenerating melanoma antigen MART1-specific CTLs, and more recently in producing potent CTLs expressing CD8αβ heterodimer. We are now developing a novel method by which non-T derived iPSCs are transduced with exogenous T cell receptor genes. If this method is applied to Human Leukocyte Antigen (HLA) haplotype-homozygous iPSC stock, it will be possible to prepare "off-the-shelf" T cells. As a first-in-human trial, we are planning to apply our strategy to relapsed acute myeloid leukemia patients by targeting the WT1 antigen.
Collapse
Affiliation(s)
- Hiroshi Kawamoto
- Laboratory of Immunology, Institute for Frontier Life and Medical Science, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Kyoko Masuda
- Laboratory of Immunology, Institute for Frontier Life and Medical Science, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Seiji Nagano
- Laboratory of Immunology, Institute for Frontier Life and Medical Science, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.,Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takuya Maeda
- Laboratory of Immunology, Institute for Frontier Life and Medical Science, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.,Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
13
|
Clancy-Thompson E, Chen GZ, Tyler PM, Servos MM, Barisa M, Brennan PJ, Ploegh HL, Dougan SK. Monoclonal Invariant NKT (iNKT) Cell Mice Reveal a Role for Both Tissue of Origin and the TCR in Development of iNKT Functional Subsets. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:159-171. [PMID: 28576977 PMCID: PMC5518629 DOI: 10.4049/jimmunol.1700214] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 05/02/2017] [Indexed: 01/10/2023]
Abstract
Invariant NKT (iNKT) cell functional subsets are defined by key transcription factors and output of cytokines, such as IL-4, IFN-γ, IL-17, and IL-10. To examine how TCR specificity determines iNKT function, we used somatic cell nuclear transfer to generate three lines of mice cloned from iNKT nuclei. Each line uses the invariant Vα14Jα18 TCRα paired with unique Vβ7 or Vβ8.2 subunits. We examined tissue homing, expression of PLZF, T-bet, and RORγt, and cytokine profiles and found that, although monoclonal iNKT cells differentiated into all functional subsets, the NKT17 lineage was reduced or expanded depending on the TCR expressed. We examined iNKT thymic development in limited-dilution bone marrow chimeras and show that higher TCR avidity correlates with higher PLZF and reduced T-bet expression. iNKT functional subsets showed distinct tissue distribution patterns. Although each individual monoclonal TCR showed an inherent subset distribution preference that was evident across all tissues examined, the iNKT cytokine profile differed more by tissue of origin than by TCR specificity.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/immunology
- B-Lymphocytes/physiology
- Cell Differentiation
- Cytokines/genetics
- Cytokines/immunology
- Cytotoxicity, Immunologic/immunology
- Interleukin-10/immunology
- Interleukin-10/metabolism
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Kruppel-Like Transcription Factors/genetics
- Mice
- Mice, Inbred C57BL
- Natural Killer T-Cells/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
- Nuclear Transfer Techniques
- Organ Specificity
- Promyelocytic Leukemia Zinc Finger Protein
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- T-Box Domain Proteins/genetics
- T-Box Domain Proteins/metabolism
- T-Lymphocyte Subsets/immunology
Collapse
Affiliation(s)
- Eleanor Clancy-Thompson
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215
| | - Gui Zhen Chen
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215
| | - Paul M Tyler
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215
| | - Mariah M Servos
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215
| | - Marta Barisa
- Whitehead Institute for Biomedical Research, Cambridge, MA 02242; and
| | - Patrick J Brennan
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA 02215
| | - Hidde L Ploegh
- Whitehead Institute for Biomedical Research, Cambridge, MA 02242; and
| | - Stephanie K Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215;
- Whitehead Institute for Biomedical Research, Cambridge, MA 02242; and
| |
Collapse
|
14
|
Kaminuma O, Katayama K, Inoue K, Saeki M, Nishimura T, Kitamura N, Shimo Y, Tofukuji S, Ishida S, Ogonuki N, Kamimura S, Oikawa M, Katoh S, Mori A, Shichijo M, Hiroi T, Ogura A. Hyper-reactive cloned mice generated by direct nuclear transfer of antigen-specific CD4 + T cells. EMBO Rep 2017; 18:885-893. [PMID: 28468955 DOI: 10.15252/embr.201643321] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 11/09/2022] Open
Abstract
T-cell receptor (TCR)-transgenic mice have been employed for evaluating antigen-response mechanisms, but their non-endogenous TCR might induce immune response differently than the physiologically expressed TCR Nuclear transfer cloning produces animals that retain the donor genotype in all tissues including germline and immune systems. Taking advantage of this feature, we generated cloned mice that carry endogenously rearranged TCR genes from antigen-specific CD4+ T cells. We show that T cells of the cloned mice display distinct developmental pattern and antigen reactivity because of their endogenously pre-rearranged TCRα (rTα) and TCRβ (rTβ) alleles. These alleles were transmitted to the offspring, allowing us to establish a set of mouse lines that show chronic-type allergic phenotypes, that is, bronchial and nasal inflammation, upon local administrations of the corresponding antigens. Intriguingly, the existence of either rTα or rTβ is sufficient to induce in vivo hypersensitivity. These cloned mice expressing intrinsic promoter-regulated antigen-specific TCR are a unique animal model with allergic predisposition for investigating CD4+ T-cell-mediated pathogenesis and cellular commitment in immune diseases.
Collapse
Affiliation(s)
- Osamu Kaminuma
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan .,Bioresource Center RIKEN, Tsukuba, Japan.,Clinical Research Center for Allergy and Rheumatology, National Hospital Organization, Sagamihara National Hospital, Sagamihara, Japan.,Center for Life Science Research, University of Yamanashi, Chuo, Japan
| | - Kazufumi Katayama
- Drug Discovery & Disease Research Laboratory, SHIONOGI & Co., Ltd., Osaka, Japan
| | | | - Mayumi Saeki
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Tomoe Nishimura
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Noriko Kitamura
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yusuke Shimo
- Drug Discovery & Disease Research Laboratory, SHIONOGI & Co., Ltd., Osaka, Japan
| | - Soichi Tofukuji
- Drug Discovery & Disease Research Laboratory, SHIONOGI & Co., Ltd., Osaka, Japan
| | - Satoru Ishida
- Drug Discovery & Disease Research Laboratory, SHIONOGI & Co., Ltd., Osaka, Japan
| | | | | | | | - Shigeki Katoh
- Department of Respiratory Medicine, Kawasaki Medical School, Kurashiki, Japan
| | - Akio Mori
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Clinical Research Center for Allergy and Rheumatology, National Hospital Organization, Sagamihara National Hospital, Sagamihara, Japan
| | - Michitaka Shichijo
- Drug Discovery & Disease Research Laboratory, SHIONOGI & Co., Ltd., Osaka, Japan
| | - Takachika Hiroi
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | |
Collapse
|
15
|
Abstract
Reproductive engineering techniques are essential for assisted reproduction of animals
and generation of genetically modified animals. They may also provide invaluable research
models for understanding the mechanisms involved in the developmental and reproductive
processes. At the RIKEN BioResource Center (BRC), I have sought to develop new
reproductive engineering techniques, especially those related to cryopreservation,
microinsemination (sperm injection), nuclear transfer, and generation of new stem cell
lines and animals, hoping that they will support the present and future projects at BRC. I
also want to combine our techniques with genetic and biochemical analyses to solve
important biological questions. We expect that this strategy makes our research more
unique and refined by providing deeper insights into the mechanisms that govern the
reproductive and developmental systems in mammals. To make this strategy more effective,
it is critical to work with experts in different scientific fields. I have enjoyed
collaborations with about 100 world-recognized laboratories, and all our collaborations
have been successful and fruitful. This review summarizes development of reproductive
engineering techniques at BRC during these 15 years.
Collapse
Affiliation(s)
- Atsuo Ogura
- RIKEN BioResource Center, Tsukuba, Ibaraki 305-0074, Japan
| |
Collapse
|
16
|
Yamada D, Iyoda T, Vizcardo R, Shimizu K, Sato Y, Endo TA, Kitahara G, Okoshi M, Kobayashi M, Sakurai M, Ohara O, Taniguchi M, Koseki H, Fujii SI. Efficient Regeneration of Human Vα24 + Invariant Natural Killer T Cells and Their Anti-Tumor Activity In Vivo. Stem Cells 2016; 34:2852-2860. [PMID: 27422351 DOI: 10.1002/stem.2465] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/14/2016] [Accepted: 06/30/2016] [Indexed: 01/31/2023]
Abstract
Reprogramming of antigen-specific T lymphocytes into induced pluripotent stem cells (iPSCs) and their subsequent re-differentiation has enabled expansion of functional T lymphocytes in vitro, thus opening up new approaches for immunotherapy of cancer and other diseases. In this study, we have established a robust protocol to reprogram human invariant NKT (Vα24+ iNKT) cells, which have been shown to act as cellular adjuvants and thus exert anti-tumor activity in mice and humans, and to re-differentiate the iNKT cell-derived iPSCs into functional iNKT cells. These iPSC-derived iNKT cells (iPS-Vα24+ iNKT cells) can be activated by ligand-pulsed dendritic cells (DCs) and produce a large amount of interferon-γ upon activation, as much as parental Vα24+ iNKT cells, but exhibit even better cytotoxic activity against various tumor cell lines. The iPS-Vα24+ iNKT cells possess significant anti-tumor activity in tumor-bearing mice and can activate autologous NK cells upon activation by ligand-pulsed DCs in the NOG mouse model in vivo, further extending their therapeutic potential. This study thus provides a first proof of concept for the clinical application of human iPS-Vα24+ iNKT cells for cancer immunotherapy. Stem Cells 2016;34:2852-2860.
Collapse
Affiliation(s)
- Daisuke Yamada
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Science (IMS), Yokohama, Japan
| | - Tomonori Iyoda
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Science (IMS)
| | - Raul Vizcardo
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Science (IMS), Yokohama, Japan
| | - Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Science (IMS)
| | - Yusuke Sato
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Science (IMS)
| | - Takaho A Endo
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Science (IMS), Yokohama, Japan
| | - Genta Kitahara
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Science (IMS), Yokohama, Japan
| | - Momoko Okoshi
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Science (IMS), Yokohama, Japan
| | - Midori Kobayashi
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Science (IMS), Yokohama, Japan
| | - Maki Sakurai
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Science (IMS)
| | - Osamu Ohara
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Science (IMS), Yokohama, Japan
| | - Masaru Taniguchi
- Laboratory for Immunoregulation, RIKEN Center for Integrative Medical Science (IMS), Yokohama, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Science (IMS), Yokohama, Japan
| | - Shin-Ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Science (IMS)
| |
Collapse
|
17
|
Jyotsana B, Sahare AA, Raja AK, Singh KP, Nala N, Singla SK, Chauhan MS, Manik RS, Palta P. Use of peripheral blood for production of buffalo (Bubalus bubalis) embryos by handmade cloning. Theriogenology 2016; 86:1318-1324.e1. [PMID: 27242179 DOI: 10.1016/j.theriogenology.2016.04.073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 04/17/2016] [Accepted: 04/22/2016] [Indexed: 12/27/2022]
Abstract
Buffalo embryos were produced by handmade cloning using peripheral blood-derived lymphocytes as donor cells. Although the blastocyst rate was lower (P < 0.01) for lymphocyte- than control skin fibroblast-derived embryos (6.6 ± 0.84% vs. 31.15 ± 2.97%), the total cell number (152.6 ± 23.06 vs. 160.1 ± 13.25) and apoptotic index (6.54 ± 0.95 vs. 8.45 ± 1.32) were similar. The global level of H3K9ac was higher (P < 0.05) in lymphocyte- than that in skin-derived blastocysts; whereas in IVF blastocysts, the level was not significantly different from the two cloned groups. The level of H3K27me3 was similar among the three groups. The expression level of DNMT1, DNMT3a, HDAC1, and IGF-1R was higher (P < 0.01) in lymphocytes than that in skin fibroblasts. The expression level of CDX2 was higher (P < 0.05) than that of DNMT3a, IGF-1R, OCT4, and NANOG was lower (P < 0.05) in lymphocyte-derived than in IVF blastocysts; that of DNMT1 and HDAC1 was similar in the two groups. The expression level of all these genes, except that of NANOG, was lower (P < 0.05) in lymphocyte- than in skin fibroblast-derived blastocysts. It is concluded that, peripheral blood-derived lymphocytes can be used for producing handmade cloning embryos in bubaline buffaloes.
Collapse
Affiliation(s)
- Basanti Jyotsana
- Animal Biotechnology Section, Central Sheep and Wool Research Institute, Avikanagar, Rajasthan, India
| | - Amol A Sahare
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana, India
| | - Anuj K Raja
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana, India
| | - Karn P Singh
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana, India
| | - Narendra Nala
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana, India
| | - S K Singla
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana, India
| | - M S Chauhan
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana, India
| | - R S Manik
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana, India
| | - P Palta
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana, India.
| |
Collapse
|
18
|
Differential developmental competence and gene expression patterns in buffalo (Bubalus bubalis) nuclear transfer embryos reconstructed with fetal fibroblasts and amnion mesenchymal stem cells. Cytotechnology 2015; 68:1827-48. [PMID: 26660476 DOI: 10.1007/s10616-015-9936-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 12/01/2015] [Indexed: 01/01/2023] Open
Abstract
The developmental ability and gene expression pattern at 8- to 16-cell and blastocyst stages of buffalo (Bubalus bubalis) nuclear transfer (NT) embryos from fetal fibroblasts (FFs), amnion mesenchymal stem cells (AMSCs) and in vitro fertilized (IVF) embryos were compared in the present studies. The in vitro expanded buffalo FFs showed a typical "S" shape growth curve with a doubling time of 41.4 h and stained positive for vimentin. The in vitro cultured undifferentiated AMSCs showed a doubling time of 39.5 h and stained positive for alkaline phosphatase, and these cells also showed expression of pluripotency markers (OCT 4, SOX 2, NANOG), and mesenchymal stem cell markers (CD29, CD44) and were negative for haematopoietic marker (CD34) genes at different passages. Further, when AMSCs were exposed to corresponding induction conditions, these cells differentiated into adipogenic, chondrogenic and osteogenic lineages which were confirmed through oil red O, alcian blue and alizarin staining, respectively. Donor cells at 3-4 passage were employed for NT. The cleavage rate was significantly (P < 0.05) higher in IVF than in FF-NT and AMSC-NT embryos (82.6 ± 8.2 vs. 64.6 ± 1.3 and 72.3 ± 2.2 %, respectively). However, blastocyst rates in IVF and AMSC-NT embryos (30.6 ± 2.7 and 28.9 ± 3.1 %) did not differ and were significantly (P < 0.05) higher than FF-NT (19.5 ± 1.8 %). Total cell number did not show significant (P > 0.05) differences between IVF and AMSC-NT embryos (186.7 ± 4.2, 171.2 ± 3.8, respectively) but were significantly (P < 0.05) higher than that from FF-NT (151.3 ± 4.1). Alterations in the expression pattern of genes implicated in transcription and pluripotency (OCT4, STAT3, NANOG), DNA methylation (DNMT1, DNMT3A), histone deacetylation (HDAC2), growth factor signaling and imprinting (IGF2, IGF2R), apoptosis (BAX, BCL2), metabolism (GLUT1) and oxidative stress (MnSOD) regulation were observed in cloned embryos. The transcripts or expression patterns in AMSC-NT embryos more closely followed that of the in vitro derived embryos compared with FF-NT embryos. The results demonstrate that multipotent amnion MSCs have a greater potential as donor cells than FFs in achieving enhanced production of cloned buffalo embryos.
Collapse
|
19
|
Abstract
Induced pluripotency defines the process by which somatic cells are converted into induced pluripotent stem cells (iPSCs) upon overexpression of a small set of transcription factors. In this article, we put transcription factor-induced pluripotency into a historical context, review current methods to generate iPSCs, and discuss mechanistic insights that have been gained into the process of reprogramming. In addition, we focus on potential therapeutic applications of induced pluripotency and emerging technologies to efficiently engineer the genomes of human pluripotent cells for scientific and therapeutic purposes.
Collapse
Affiliation(s)
- Konrad Hochedlinger
- Howard Hughes Medical Institute at Massachusetts General Hospital, Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Medical School, Boston, Massachusetts 02114
| | - Rudolf Jaenisch
- Whitehead Institute and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142
| |
Collapse
|
20
|
Handmade cloning: recent advances, potential and pitfalls. J Anim Sci Biotechnol 2015; 6:43. [PMID: 26473031 PMCID: PMC4606838 DOI: 10.1186/s40104-015-0043-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 09/21/2015] [Indexed: 12/02/2022] Open
Abstract
Handmade cloning (HMC) is the most awaited, simple and micromanipulator-free version of somatic cell nuclear transfer (SCNT). The requirement of expensive micromanipulators and skilled expertise is eliminated in this technique, proving it as a major revolution in the field of embryology. During the past years, many modifications have been incorporated in this technique to boost its efficiency. This alternative approach to micromanipulator based traditional cloning (TC) works wonder in generating comparable or even higher birth rates in addition to declining costs drastically and enabling cryopreservation. This technique is not only applicable to intraspecies nuclear transfer but also to interspecies nuclear transfer (iSCNT) thus permitting conservation of endangered species. It also offers unique possibilities for automation of SCNT which aims at production of transgenic animals that can cure certain human diseases by producing therapeutics hence, providing a healthier future for the wellbeing of humans. The present review aims at highlighting certain aspects of HMC including recent advancements in procedure and factors involved in elevating its efficiency besides covering the potentials and pitfalls of this technique.
Collapse
|
21
|
Abstract
It should be emphasized that "129" is not simply a number but is also the designation of a mouse strain that has made a great contribution to modern biological science and technology. Embryonic stem cells derived from 129 mice were essential components of gene-targeting strategies in early research. More recently, 129 mice have provided superior donor genomes for cloning by nuclear transfer. Some factor or factors conferring genomic plasticity must exist in the 129 genome, but these remain unidentified.
Collapse
Affiliation(s)
- Kimiko Inoue
- RIKEN BioResource Center, Tsukuba, Ibaraki 305-0074, Japan
| | | |
Collapse
|
22
|
Mizutani E, Oikawa M, Kassai H, Inoue K, Shiura H, Hirasawa R, Kamimura S, Matoba S, Ogonuki N, Nagatomo H, Abe K, Wakayama T, Aiba A, Ogura A. Generation of Cloned Mice from Adult Neurons by Direct Nuclear Transfer1. Biol Reprod 2015; 92:81. [DOI: 10.1095/biolreprod.114.123455] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
23
|
Chestkov IV, Khomyakova EA, Vasilieva EA, Lagarkova MA, Kiselev SL. Molecular barriers to processes of genetic reprogramming and cell transformation. BIOCHEMISTRY. BIOKHIMIIA 2014; 79:1297-1307. [PMID: 25716723 DOI: 10.1134/s0006297914120037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Genetic reprogramming by ectopic expression of transcription factor genes induces the pluripotent state in somatic cells. This technology provides an opportunity to establish pluripotent stem cells for each person, as well as to get better understanding of epigenetic mechanisms controlling cell state. Interestingly, some of the molecular processes that accompany somatic cell reprogramming in vitro are also characteristic for tumor manifestation. Thus, similar "molecular barriers" that control the stability of epigenetic state exist for both processes of pluripotency induction and malignant transformation. The reprogramming of tumor cells is interesting in two aspects: first, it will determine the contribution of epigenetic changes in carcinogenesis; second, it gives an approach to evaluate tumor stem cells that are supposed to form the entire cell mass of the tumor. This review discusses the key stages of genetic reprogramming, the similarity and difference between the reprogramming process and malignant transformation.
Collapse
Affiliation(s)
- I V Chestkov
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, 119991, Russia.
| | | | | | | | | |
Collapse
|
24
|
Ren Y, Dashtsoodol N, Watarai H, Koseki H, Quan C, Taniguchi M. Generation of induced pluripotent stem cell-derived mice by reprogramming of a mature NKT cell. Int Immunol 2014; 26:551-61. [PMID: 24854340 PMCID: PMC4169672 DOI: 10.1093/intimm/dxu057] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The successful generation of iPSC-derived mouse strains to study NKT cells NKT cells are characterized by their expression of an NKT-cell-specific invariant antigen-receptor α chain encoded by Vα14Jα18 gene segments. These NKT cells bridge the innate and acquired immune systems to mediate effective and augmented responses; however, the limited number of NKT cells in vivo hampers their analysis. Here, two lines of induced pluripotent stem cell-derived mice (NKT-iPSC-derived mice) were generated by reprogramming of mature NKT cells, where one harbors both rearranged Vα14Jα18 and Vβ7 genes and the other carries rearranged Vα14Jα18 on both alleles but germline Vβ loci. The analysis of NKT-iPSC-derived mice showed a significant increase in NKT cell numbers with relatively normal frequencies of functional subsets, but significantly enhanced in some cases, and acquired functional NKT cell maturation in peripheral lymphoid organs. NKT-iPSC-derived mice also showed normal development of other immune cells except for the absence of γδT cells and disturbed development of conventional CD4 αβT cells. These results suggest that the NKT-iPSC-derived mice are a better model for NKT cell development and function study rather than transgenic mouse models reported previously and also that the presence of a pre-rearranged Vα14Jα18 in the natural chromosomal context favors the developmental fate of NKT cells.
Collapse
Affiliation(s)
- Yue Ren
- Laboratory for Immune Regulation, RCAI, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), 230-0045 Kanagawa, Japan The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021 Jilin, People's Republic of China
| | - Nyambayar Dashtsoodol
- Laboratory for Immune Regulation, RCAI, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), 230-0045 Kanagawa, Japan
| | - Hiroshi Watarai
- Laboratory for Immune Regulation, RCAI, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), 230-0045 Kanagawa, Japan Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), 102-0076 Tokyo, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RCAI, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), 230-0045 Kanagawa, Japan
| | - Chengshi Quan
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021 Jilin, People's Republic of China
| | - Masaru Taniguchi
- Laboratory for Immune Regulation, RCAI, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), 230-0045 Kanagawa, Japan
| |
Collapse
|
25
|
TANABE K, TAKAHASHI K, YAMANAKA S. Induction of pluripotency by defined factors. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2014; 90:83-96. [PMID: 24621955 PMCID: PMC3997808 DOI: 10.2183/pjab.90.83] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The "reversion of cell fate from differentiated states back into totipotent or pluripotent states" has been an interest of many scientists for a long time. With the help of knowledge accumulated by those scientists, we succeeded in converting somatic cells to a pluripotent cell lineage by the forced expression of defined factors. These established induced pluripotent stem (iPS) cells have similar features to embryonic stem (ES) cells, including pluripotency and immortality. The iPS cell technology provides unprecedented opportunities for regenerative medicine and drug discovery.
Collapse
Affiliation(s)
- Koji TANABE
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Kazutoshi TAKAHASHI
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Shinya YAMANAKA
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, U.S.A.
- Correspondence should be addressed: S. Yamanaka, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin Yoshida, Sakyo-ku, Kyoto 606-8507, Japan (e-mail: )
| |
Collapse
|
26
|
Electrical activation of rabbit oocytes increases fertilization and embryo development by intracytoplasmic sperm injection using sperm from deceased male. J Assist Reprod Genet 2013; 30:1605-10. [PMID: 24114632 DOI: 10.1007/s10815-013-0113-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/30/2013] [Indexed: 10/26/2022] Open
Abstract
PURPOSE We investigated the effect of electrical stimulation on rabbit oocyte activation using intracytoplasmic sperm injection (ICSI) to determine whether viable offspring can be produced from deceased rabbit sperm using ICSI. METHODS Sperm were collected from a heterozygote GFP male rabbit 5 h after sacrifice and cryopreserved in liquid nitrogen. Mature oocytes were fertilized using ICSI. A series of electrical pulse procedures were used to activate oocytes before and/or after ICSI. Following ICSI, zygotes were cultured in B2 medium for 4 days or transferred into the oviducts of recipient rabbits at the 2- or 4-cell stage. RESULTS The blastocyst formation rate was significantly greater in oocytes that received one or two pulses prior to ICSI compared to controls and other electrically stimulated groups. In the single pulse before ICSI group, 23 % of the blastocysts expressed GFP, which was significantly greater than all other groups. However, those that received treatment before and after, or just following ICSI, showed a significant decrease in embryo survival. Finally, embryos from the single pulse before ICSI group were transferred into recipient female rabbits and a full-term kit was successfully delivered. CONCLUSIONS One pulse of electrical stimulation prior to sperm injection was an effective method to activate rabbit oocytes for fertilization. Sperm collected from a deceased rabbit is able to produce viable embryos through ISCI that are capable of normal fetal and kit development.
Collapse
|
27
|
Abstract
Abstract Professor Keith Campbell's critical contribution to the discovery that a somatic cell from an adult animal can be fully reprogrammed by oocyte factors to form a cloned individual following nuclear transfer (NT)(Wilmut et al., 1997 ) overturned a dogma concerning the reversibility of cell fate that many scientists had considered to be biologically impossible. This seminal experiment proved the totipotency of adult somatic nuclei and finally confirmed that adult cells could differentiate without irreversible changes to the genetic material.
Collapse
|
28
|
Kamimura S, Inoue K, Ogonuki N, Hirose M, Oikawa M, Yo M, Ohara O, Miyoshi H, Ogura A. Mouse Cloning Using a Drop of Peripheral Blood1. Biol Reprod 2013; 89:24. [DOI: 10.1095/biolreprod.113.110098] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
29
|
Vahl JC, Heger K, Knies N, Hein MY, Boon L, Yagita H, Polic B, Schmidt-Supprian M. NKT cell-TCR expression activates conventional T cells in vivo, but is largely dispensable for mature NKT cell biology. PLoS Biol 2013; 11:e1001589. [PMID: 23853545 PMCID: PMC3708704 DOI: 10.1371/journal.pbio.1001589] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 05/07/2013] [Indexed: 12/24/2022] Open
Abstract
Natural killer T (NKT) cell development depends on recognition of self-glycolipids via their semi-invariant Vα14i-TCR. However, to what extent TCR-mediated signals determine identity and function of mature NKT cells remains incompletely understood. To address this issue, we developed a mouse strain allowing conditional Vα14i-TCR expression from within the endogenous Tcrα locus. We demonstrate that naïve T cells are activated upon replacement of their endogenous TCR repertoire with Vα14i-restricted TCRs, but they do not differentiate into NKT cells. On the other hand, induced TCR ablation on mature NKT cells did not affect their lineage identity, homeostasis, or innate rapid cytokine secretion abilities. We therefore propose that peripheral NKT cells become unresponsive to and thus are independent of their autoreactive TCR. Immune system natural killer T (NKT) cells help to protect against certain strains of bacteria and viruses, and suppress the development of autoimmune diseases and cancer. However, NKT cells are also central mediators of allergic responses. The recognition of one's own glycolipid antigens (self-glycolipids) in the thymus via the unique Vα14i T cell receptor, Vα14i-TCR, triggers the NKT cell developmental program, which differs considerably from that of conventional T cells. We generated a mouse model to investigate whether the Vα14i-TCR on mature NKT cells constantly recognizes self-glycolipids and to assess whether this TCR is required for survival and continued NKT cell identity. Switching the peptide-recognizing TCR of a mature conventional T cell to a glycolipid-recognizing Vα14i-TCR led to activation of the T cells, indicating that this TCR is also autoreactive on peripheral T cells or can signal autonomously. But TCR ablation did not affect the half-life, characteristic gene expression or innate functions of mature NKT cells. Therefore, the inherently autoreactive Vα14i-TCR is dispensable for the functions of mature peripheral NKT cells after instructing thymic NKT cell development. Thus the Vα14i-TCR serves a similar function to pattern-recognition receptors, in mediating immune recognition of foreign invasion or diseased cells.
Collapse
Affiliation(s)
- J. Christoph Vahl
- Molecular Immunology and Signaltransduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Klaus Heger
- Molecular Immunology and Signaltransduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Nathalie Knies
- Molecular Immunology and Signaltransduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Marco Y. Hein
- Molecular Immunology and Signaltransduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Louis Boon
- Bioceros, Yalelaan 46, Utrecht, The Netherlands
| | - Hideo Yagita
- Juntendo University School of Medicine, Tokyo, Japan
| | - Bojan Polic
- University of Rijeka School of Medicine, Rijeka, Croatia
| | - Marc Schmidt-Supprian
- Molecular Immunology and Signaltransduction, Max Planck Institute of Biochemistry, Martinsried, Germany
- * E-mail:
| |
Collapse
|
30
|
Ogura A, Inoue K, Wakayama T. Recent advancements in cloning by somatic cell nuclear transfer. Philos Trans R Soc Lond B Biol Sci 2013; 368:20110329. [PMID: 23166393 DOI: 10.1098/rstb.2011.0329] [Citation(s) in RCA: 148] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Somatic cell nuclear transfer (SCNT) cloning is the sole reproductive engineering technology that endows the somatic cell genome with totipotency. Since the first report on the birth of a cloned sheep from adult somatic cells in 1997, many technical improvements in SCNT have been made by using different epigenetic approaches, including enhancement of the levels of histone acetylation in the chromatin of the reconstructed embryos. Although it will take a considerable time before we fully understand the nature of genomic programming and totipotency, we may expect that somatic cell cloning technology will soon become broadly applicable to practical purposes, including medicine, pharmaceutical manufacturing and agriculture. Here we review recent progress in somatic cell cloning, with a special emphasis on epigenetic studies using the laboratory mouse as a model.
Collapse
Affiliation(s)
- Atsuo Ogura
- RIKEN BioResource Center, Tsukuba, Ibaraki, Japan.
| | | | | |
Collapse
|
31
|
Abstract
The crucial facts underlying the low efficiency of cellular reprogramming are poorly understood. Cellular reprogramming occurs in nuclear transfer, induced pluripotent stem cell (iPSC) formation, cell fusion, and lineage-switching experiments. Despite these advances, there are three fundamental problems to be addressed: (1) the majority of cells cannot be reprogrammed, (2) the efficiency of reprogramming cells is usually low, and (3) the reprogrammed cells developed from a patient's own cells activate immune responses. These shortcomings present major obstacles for using reprogramming approaches in customised cell therapy. In this Perspective, the author synthesises past and present observations in the field of cellular reprogramming to propose a theoretical picture of the cellular memory disc. The current hypothesis is that all cells undergo an endogenous and exogenous holographic memorisation such that parts of the cellular memory dramatically decrease the efficiency of reprogramming cells, act like a barrier against reprogramming in the majority of cells, and activate immune responses. Accordingly, the focus of this review is mainly to describe the cellular memory disc (CMD). Based on the present theory, cellular memory includes three parts: a reprogramming-resistance memory (RRM), a switch-promoting memory (SPM) and a culture-induced memory (CIM). The cellular memory arises genetically, epigenetically and non-genetically and affects cellular behaviours. [corrected].
Collapse
Affiliation(s)
- Seyed Hadi Anjamrooz
- Cellular and Molecular Research Center, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
32
|
Abstract
The nuclear transfer (NT) technique in the mouse has enabled us to generate cloned mice and to establish NT embryonic stem (ntES) cells. Direct nuclear injection into mouse oocytes with a piezo impact drive unit can aid in the bypass of several steps of the original cell fusion procedure. It is important to note that only the NT approach can reveal dynamic and global modifications in the epigenome without using genetic modification as well as generating live animals from single cells. Thus, these techniques could also be applied to the preservation of genetic material from any mouse strain instead of preserving embryos or gametes. Moreover, with this technique, we can use not only living cells but also the nuclei of dead cells from frozen mouse carcasses for NT. This chapter describes our most recent protocols of NT into the mouse oocyte for cloning mice and for the establishment of ntES cells from cloned embryos.
Collapse
Affiliation(s)
- Eiji Mizutani
- Center for Developmental Biology, RIKEN Kobe institute, Kobe, Japan,
| | | | | |
Collapse
|
33
|
|
34
|
Rezanejad H, Matin MM. Induced Pluripotent Stem Cells: Progress and Future Perspectives in the Stem Cell World. Cell Reprogram 2012; 14:459-70. [DOI: 10.1089/cell.2012.0039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Habib Rezanejad
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Maryam M. Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Cell and Molecular Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
35
|
Pereira CF, Lemischka IR, Moore K. Reprogramming cell fates: insights from combinatorial approaches. Ann N Y Acad Sci 2012; 1266:7-17. [PMID: 22901251 DOI: 10.1111/j.1749-6632.2012.06508.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Epigenetic reprogramming can be achieved in different ways, including nuclear transfer, cell fusion, or the expression of transcription factors (TFs). Combinatorial overexpression provides an opportunity to define the minimal core network of TFs that instructs specific cell fates. This approach has been employed to induce mouse and human pluripotency and differentiated cell types from cells that can be also as distant as cells from different germ layers. This suggests the possibility that any specific cell type may be directly converted into another if the appropriate reprogramming TF core is determined. Herein, we review the factors used for reprogramming multiple cell identities and raise the question of whether there is a common underlying blueprint for reprogramming factors. In addition to the generation of human cell types of interest for cell-replacement therapies, we propose that the TF-mediated conversion of differentiated cell types, especially somatic stem cells, will have an impact on our understanding of their biological development.
Collapse
Affiliation(s)
- Carlos-Filipe Pereira
- Department of Developmental and Regenerative Biology, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, New York, USA.
| | | | | |
Collapse
|
36
|
Rouaux C, Bhai S, Arlotta P. Programming and reprogramming neuronal subtypes in the central nervous system. Dev Neurobiol 2012; 72:1085-98. [PMID: 22378700 DOI: 10.1002/dneu.22018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent discoveries in nuclear reprogramming have challenged the dogma that the identity of terminally differentiated cells cannot be changed. The identification of molecular mechanisms that reprogram differentiated cells to a new identity carries profound implications for regenerative medicine across organ systems. The central nervous system (CNS) has historically been considered to be largely immutable. However, recent studies indicate that even the adult CNS is imparted with the potential to change under the appropriate stimuli. Here, we review current knowledge regarding the capability of distinct cells within the CNS to reprogram their identity and consider the role of developmental signals in directing these cell fate decisions. Finally, we discuss the progress and current challenges of using developmental signals to precisely direct the generation of individual neuronal subtypes in the postnatal CNS and in the dish.
Collapse
Affiliation(s)
- Caroline Rouaux
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | | | | |
Collapse
|
37
|
Rodriguez-Osorio N, Urrego R, Cibelli JB, Eilertsen K, Memili E. Reprogramming mammalian somatic cells. Theriogenology 2012; 78:1869-86. [PMID: 22979962 DOI: 10.1016/j.theriogenology.2012.05.030] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 05/20/2012] [Accepted: 05/31/2012] [Indexed: 01/23/2023]
Abstract
Somatic cell nuclear transfer (SCNT), the technique commonly known as cloning, permits transformation of a somatic cell into an undifferentiated zygote with the potential to develop into a newborn animal (i.e., a clone). In somatic cells, chromatin is programmed to repress most genes and express some, depending on the tissue. It is evident that the enucleated oocyte provides the environment in which embryonic genes in a somatic cell can be expressed. This process is controlled by a series of epigenetic modifications, generally referred to as "nuclear reprogramming," which are thought to involve the removal of reversible epigenetic changes acquired during cell differentiation. A similar process is thought to occur by overexpression of key transcription factors to generate induced pluripotent stem cells (iPSCs), bypassing the need for SCNT. Despite its obvious scientific and medical importance, and the great number of studies addressing the subject, the molecular basis of reprogramming in both reprogramming strategies is largely unknown. The present review focuses on the cellular and molecular events that occur during nuclear reprogramming in the context of SCNT and the various approaches currently being used to improve nuclear reprogramming. A better understanding of the reprogramming mechanism will have a direct impact on the efficiency of current SCNT procedures, as well as iPSC derivation.
Collapse
|
38
|
Progress and bottleneck in induced pluripotency. CELL REGENERATION 2012; 1:5. [PMID: 25408868 PMCID: PMC4230504 DOI: 10.1186/2045-9769-1-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 05/03/2012] [Indexed: 01/05/2023]
Abstract
With their capability to undergo unlimited self-renewal and to differentiate into all cell types in the body, induced pluripotent stem cells (iPSCs), reprogrammed from somatic cells of individual patients with defined factors, have unlimited potential in cell therapy and in modeling complex human diseases. Significant progress has been achieved to improve the safety of iPSCs and the reprogramming efficiency. To avoid the cancer risk and spontaneous reactivation of the reprogramming factors associated with the random integration of viral vectors into the genome, several approaches have been established to deliver the reprogramming factors into the somatic cells without inducing genetic modification. In addition, a panel of small molecule compounds, many of which targeting the epigenetic machinery, have been identified to increase the reprogramming efficiency. Despite these progresses, recent studies have identified genetic and epigenetic abnormalities of iPSCs as well as the immunogenicity of some cells derived from iPSCs. In addition, due to the oncogenic potential of the reprogramming factors and the reprogramming-induced DNA damage, the critical tumor suppressor pathways such as p53 and ARF are activated to act as the checkpoints that suppress induced pluripotency. The inactivation of these tumor suppression pathways even transiently during reprogramming processes could have significant adverse impact on the genome integrity. These safety concerns must be resolved to improve the feasibility of the clinic development of iPSCs into human cell therapy.
Collapse
|
39
|
Kim J, Ambasudhan R, Ding S. Direct lineage reprogramming to neural cells. Curr Opin Neurobiol 2012; 22:778-84. [PMID: 22652035 DOI: 10.1016/j.conb.2012.05.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 05/03/2012] [Indexed: 01/11/2023]
Abstract
Recently we have witnessed an array of studies on direct reprogramming that describe induced inter conversion of mature cell types from higher organisms including human. While these studies reveal an unexpected level of plasticity of differentiated somatic cells, they also provide unprecedented opportunities to develop regenerative therapies for many debilitating disorders and model these 'diseases-in-a-dish' for studying their pathophysiology. Here we review the current state of the art in direct lineage reprogramming to neural cells, and discuss the challenges that need to be addressed toward achieving the full potential of this exciting new technology.
Collapse
Affiliation(s)
- Janghwan Kim
- Regenerative Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 305-806, Republic of Korea
| | | | | |
Collapse
|
40
|
Watarai H, Yamada D, Fujii SI, Taniguchi M, Koseki H. Induced pluripotency as a potential path towards iNKT cell-mediated cancer immunotherapy. Int J Hematol 2012; 95:624-31. [DOI: 10.1007/s12185-012-1091-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 04/19/2012] [Accepted: 04/20/2012] [Indexed: 10/28/2022]
|
41
|
LIU YX, ZHANG ZW, FU XP, JIN P, LI AM, ZHANG Y. Cell Reprogramming by Cell Extract Treatment*. PROG BIOCHEM BIOPHYS 2011. [DOI: 10.3724/sp.j.1206.2011.00346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
42
|
Dutta R, Malakar D, Khate K, Sahu S, Akshey Y, Mukesh M. A comparative study on efficiency of adult fibroblast, putative embryonic stem cell and lymphocyte as donor cells for production of handmade cloned embryos in goat and characterization of putative ntES cells obtained from these embryos. Theriogenology 2011; 76:851-63. [DOI: 10.1016/j.theriogenology.2011.03.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 02/22/2011] [Accepted: 03/04/2011] [Indexed: 10/18/2022]
|
43
|
Nowak-Imialek M, Kues W, Carnwath JW, Niemann H. Pluripotent stem cells and reprogrammed cells in farm animals. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2011; 17:474-497. [PMID: 21682936 DOI: 10.1017/s1431927611000080] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Pluripotent cells are unique because of their ability to differentiate into the cell lineages forming the entire organism. True pluripotent stem cells with germ line contribution have been reported for mice and rats. Human pluripotent cells share numerous features of pluripotentiality, but confirmation of their in vivo capacity for germ line contribution is impossible due to ethical and legal restrictions. Progress toward derivation of embryonic stem cells from domestic species has been made, but the derived cells were not able to produce germ line chimeras and thus are termed embryonic stem-like cells. However, domestic animals, in particular the domestic pig (Sus scrofa), are excellent large animals models, in which the clinical potential of stem cell therapies can be studied. Reprogramming technologies for somatic cells, including somatic cell nuclear transfer, cell fusion, in vitro culture in the presence of cell extracts, in vitro conversion of adult unipotent spermatogonial stem cells into germ line derived pluripotent stem cells, and transduction with reprogramming factors have been developed with the goal of obtaining pluripotent, germ line competent stem cells from domestic animals. This review summarizes the present state of the art in the derivation and maintenance of pluripotent stem cells in domestic animals.
Collapse
Affiliation(s)
- Monika Nowak-Imialek
- Institute of Farm Animal Genetics (FLI), Biotechnology, Mariensee, 31535 Neustadt, Germany
| | | | | | | |
Collapse
|
44
|
Abstract
We review experiments in which somatic cell nuclei are transplanted singly to enucleated eggs (metaphase II) in amphibia and mammals and as multiple nuclei to the germinal vesicle of amphibian oocytes (prophase I). These experiments have shown the totipotency of some somatic cell nuclei, as well as switches in cell type and changes in gene expression. Abnormalities of nuclear transplant embryo development increase greatly as nuclei are taken from progressively more differentiated donor cells. The molecular changes that accompany the reprogramming of transplanted nuclei help to indicate the mechanisms used by eggs and oocytes to reprogram gene expression. We discuss the importance of chromosomal protein exchange, of transcription factor supply, and of chromatin access in reprogramming.
Collapse
Affiliation(s)
- J B Gurdon
- Wellcome Trust Cancer Research UK Gurdon Institute, Cambridge, United Kingdom.
| | | |
Collapse
|
45
|
Zipori D. À la recherche d’une définition moléculaire plus que descriptive pour les cellules souches. Med Sci (Paris) 2011; 27:303-7. [DOI: 10.1051/medsci/2011273303] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
46
|
Wada H, Seino KI. Immune cells and iPS cells. Inflamm Regen 2011. [DOI: 10.2492/inflammregen.31.361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
47
|
Stadtfeld M, Hochedlinger K. Induced pluripotency: history, mechanisms, and applications. Genes Dev 2010; 24:2239-63. [PMID: 20952534 DOI: 10.1101/gad.1963910] [Citation(s) in RCA: 557] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The generation of induced pluripotent stem cells (iPSCs) from somatic cells demonstrated that adult mammalian cells can be reprogrammed to a pluripotent state by the enforced expression of a few embryonic transcription factors. This discovery has raised fundamental questions about the mechanisms by which transcription factors influence the epigenetic conformation and differentiation potential of cells during reprogramming and normal development. In addition, iPSC technology has provided researchers with a unique tool to derive disease-specific stem cells for the study and possible treatment of degenerative disorders with autologous cells. In this review, we summarize the progress that has been made in the iPSC field over the last 4 years, with an emphasis on understanding the mechanisms of cellular reprogramming and its potential applications in cell therapy.
Collapse
Affiliation(s)
- Matthias Stadtfeld
- Howard Hughes Medical Institute, Harvard University and Harvard Medical School, Cambridge, Massachusetts 02138, USA
| | | |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Recent advances in molecular biology research have culminated in development of technologies to generate pluripotent stem cells from somatic cells. In addition to skin fibroblasts, hematopoietic cells also have been shown to be amenable to reprogramming to pluripotency. The present review discusses the relevance of these findings to basic researches and regenerative medicine, and how researchers can take advantage of hematopoietic cell reprogramming technologies. RECENT FINDINGS In 2006, Yamanaka and his colleagues published their amazing observation that murine somatic cells can be reprogrammed to the embryonic stem cell-like state simply by retroviral-mediated introduction of three or four defined factors. Soon after, human cells also were shown to be amenable to similar reprogramming. Generation of induced pluripotent cells from several types of hematopoietic cells of both murine and human origins now has been reported. SUMMARY Reprogramming adult hematopoietic cells will provide opportunities to obtain valuable materials with minimum risk and burden to patients. Reprogrammed cells can be used in research to elucidate disease mechanisms and in drug or toxicity screening. In clinical settings, patient-derived induced pluripotent stem cells may be used to generate mature functional cells for various therapies.
Collapse
|
49
|
Fukuda A, Cao F, Morita S, Yamada K, Jincho Y, Tane S, Sotomaru Y, Kono T. Identification of inappropriately reprogrammed genes by large-scale transcriptome analysis of individual cloned mouse blastocysts. PLoS One 2010; 5:e11274. [PMID: 20614022 PMCID: PMC2894852 DOI: 10.1371/journal.pone.0011274] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Accepted: 06/02/2010] [Indexed: 01/11/2023] Open
Abstract
Although cloned embryos generated by somatic/embryonic stem cell nuclear transfer (SECNT) certainly give rise to viable individuals, they can often undergo embryonic arrest at any stage of embryogenesis, leading to diverse morphological abnormalities. In an effort to gain further insights into reprogramming and the properties of SECNT embryos, we performed a large-scale gene expression profiling of 87 single blastocysts using GeneChip microarrays. Sertoli cells, cumulus cells, and embryonic stem cells were used as donor cells. The gene expression profiles of 87 blastocysts were subjected to microarray analysis. Using principal component analysis and hierarchical clustering, the gene expression profiles were clearly classified into 3 clusters corresponding to the type of donor cell. The results revealed that each type of SECNT embryo had a unique gene expression profile that was strictly dependent upon the type of donor cells, although there was considerable variation among the individual profiles within each group. This suggests that the reprogramming process is distinct for embryos cloned from different types of donor cells. Furthermore, on the basis of the results of comparison analysis, we identified 35 genes that were inappropriately reprogrammed in most of the SECNT embryos; our findings demonstrated that some of these genes, such as Asz1, Xlr3a and App, were appropriately reprogrammed only in the embryos with a transcriptional profile that was the closest to that of the controls. Our findings provide a framework to further understand the reprogramming in SECNT embryos.
Collapse
Affiliation(s)
- Atsushi Fukuda
- Department of Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Feng Cao
- Department of Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Shinnosuke Morita
- Department of Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Kaori Yamada
- Department of Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Yuko Jincho
- Department of Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Shouji Tane
- Department of Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Yusuke Sotomaru
- Natural Science Centre for Basic Research and Development, Hiroshima University, Hiroshima, Japan
| | - Tomohiro Kono
- Department of Bioscience, Tokyo University of Agriculture, Tokyo, Japan
- * E-mail:
| |
Collapse
|
50
|
Chang G, Miao YL, Zhang Y, Liu S, Kou Z, Ding J, Chen DY, Sun QY, Gao S. Linking incomplete reprogramming to the improved pluripotency of murine embryonal carcinoma cell-derived pluripotent stem cells. PLoS One 2010; 5:e10320. [PMID: 20436676 PMCID: PMC2859941 DOI: 10.1371/journal.pone.0010320] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Accepted: 03/30/2010] [Indexed: 12/18/2022] Open
Abstract
Somatic cell nuclear transfer (SCNT) has been proved capable of reprogramming various differentiated somatic cells into pluripotent stem cells. Recently, induced pluripotent stem cells (iPS) have been successfully derived from mouse and human somatic cells by the over-expression of a combination of transcription factors. However, the molecular mechanisms underlying the reprogramming mediated by either the SCNT or iPS approach are poorly understood. Increasing evidence indicates that many tumor pathways play roles in the derivation of iPS cells. Embryonal carcinoma (EC) cells have the characteristics of both stem cells and cancer cells and thus they might be the better candidates for elucidating the details of the reprogramming process. Although previous studies indicate that EC cells cannot be reprogrammed into real pluripotent stem cells, the reasons for this remain unclear. Here, nuclei from mouse EC cells (P19) were transplanted into enucleated oocytes and pluripotent stem cells (P19 NTES cells) were subsequently established. Interestingly, P19 NTES cells prolonged the development of tetraploid aggregated embryos compared to EC cells alone. More importantly, we found that the expression recovery of the imprinted H19 gene was dependent on the methylation state in the differential methylation region (DMR). The induction of Nanog expression, however, was independent of the promoter region DNA methylation state in P19 NTES cells. A whole-genome transcriptome analysis further demonstrated that P19 NTES cells were indeed the intermediates between P19 cells and ES cells and many interesting genes were uncovered that may be responsible for the failed reprogramming of P19 cells. To our knowledge, for the first time, we linked incomplete reprogramming to the improved pluripotency of EC cell-derived pluripotent stem cells. The candidate genes we discovered may be useful not only for understanding the mechanisms of reprogramming, but also for deciphering the transition between tumorigenesis and pluripotency.
Collapse
Affiliation(s)
- Gang Chang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- National Institute of Biological Sciences, Beijing, China
- Department of Histology and Embryology, Capital Medical University, Beijing, China
| | - Yi-Liang Miao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- National Institute of Biological Sciences, Beijing, China
| | - Yu Zhang
- National Institute of Biological Sciences, Beijing, China
| | - Sheng Liu
- National Institute of Biological Sciences, Beijing, China
| | - Zhaohui Kou
- National Institute of Biological Sciences, Beijing, China
| | - Junjun Ding
- National Institute of Biological Sciences, Beijing, China
| | - Da-Yuan Chen
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- * E-mail: (SG); (QYS)
| | - Shaorong Gao
- National Institute of Biological Sciences, Beijing, China
- * E-mail: (SG); (QYS)
| |
Collapse
|