1
|
Da-Veiga MA, Rogister B, Lombard A, Neirinckx V, Piette C. Glioma Stem Cells in Pediatric High-Grade Gliomas: From Current Knowledge to Future Perspectives. Cancers (Basel) 2022; 14:cancers14092296. [PMID: 35565425 PMCID: PMC9099564 DOI: 10.3390/cancers14092296] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Pediatric high-grade glioma (pHGG) has a dismal prognosis in which the younger the patient, the more restricted the treatments are, in regard to the incurred risks. Current therapies destroy many tumor cells but fail to target the highly malignant glioma stem cells (GSCs) that adapt quickly to give rise to recurring, treatment-resistant cancers. Despite a lack of consensus around an efficient detection, GSCs are well described in adult brain tumors but remain poorly investigated in pediatric cases, mostly due to their rarity. An improved knowledge about GSC roles in pediatric tumors would provide a key leverage towards the elimination of this sub-population, based on targeted treatments. The aim of this review is to sum up the state of art about GSCs in pHGG. Abstract In children, high-grade gliomas (HGG) and diffuse midline gliomas (DMG) account for a high proportion of death due to cancer. Glioma stem cells (GSCs) are tumor cells in a specific state defined by a tumor-initiating capacity following serial transplantation, self-renewal, and an ability to recapitulate tumor heterogeneity. Their presence was demonstrated several decades ago in adult glioblastoma (GBM), and more recently in pediatric HGG and DMG. In adults, we and others have previously suggested that GSCs nest into the subventricular zone (SVZ), a neurogenic niche, where, among others, they find shelter from therapy. Both bench and bedside evidence strongly indicate a role for the GSCs and the SVZ in GBM progression, fostering the development of innovative targeting treatments. Such new therapeutic approaches are of particular interest in infants, in whom standard therapies are often limited due to the risk of late effects. The aim of this review is to describe current knowledge about GSCs in pediatric HGG and DMG, i.e., their characterization, the models that apply to their development and maintenance, the specific signaling pathways that may underlie their activity, and their specific interactions with neurogenic niches. Finally, we will discuss the clinical relevance of these observations and the therapeutic advantages of targeting the SVZ and/or the GSCs in infants.
Collapse
Affiliation(s)
- Marc-Antoine Da-Veiga
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000 Liège, Belgium; (M.-A.D.-V.); (B.R.); (A.L.); (V.N.)
| | - Bernard Rogister
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000 Liège, Belgium; (M.-A.D.-V.); (B.R.); (A.L.); (V.N.)
- Department of Neurology, CHU of Liège, 4000 Liège, Belgium
| | - Arnaud Lombard
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000 Liège, Belgium; (M.-A.D.-V.); (B.R.); (A.L.); (V.N.)
- Department of Neurosurgery, CHU of Liège, 4000 Liège, Belgium
| | - Virginie Neirinckx
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000 Liège, Belgium; (M.-A.D.-V.); (B.R.); (A.L.); (V.N.)
| | - Caroline Piette
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000 Liège, Belgium; (M.-A.D.-V.); (B.R.); (A.L.); (V.N.)
- Department of Pediatrics, Division of Hematology-Oncology, CHU Liège, 4000 Liège, Belgium
- Correspondence:
| |
Collapse
|
2
|
Fisher CL, Marks H, Cho LTY, Andrews R, Wormald S, Carroll T, Iyer V, Tate P, Rosen B, Stunnenberg HG, Fisher AG, Skarnes WC. An efficient method for generation of bi-allelic null mutant mouse embryonic stem cells and its application for investigating epigenetic modifiers. Nucleic Acids Res 2017; 45:e174. [PMID: 28981838 PMCID: PMC5716182 DOI: 10.1093/nar/gkx811] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 08/25/2017] [Accepted: 09/04/2017] [Indexed: 11/18/2022] Open
Abstract
Mouse embryonic stem (ES) cells are a popular model system to study biological processes, though uncovering recessive phenotypes requires inactivating both alleles. Building upon resources from the International Knockout Mouse Consortium (IKMC), we developed a targeting vector for second allele inactivation in conditional-ready IKMC 'knockout-first' ES cell lines. We applied our technology to several epigenetic regulators, recovering bi-allelic targeted clones with a high efficiency of 60% and used Flp recombinase to restore expression in two null cell lines to demonstrate how our system confirms causality through mutant phenotype reversion. We designed our strategy to select against re-targeting the 'knockout-first' allele and identify essential genes in ES cells, including the histone methyltransferase Setdb1. For confirmation, we exploited the flexibility of our system, enabling tamoxifen inducible conditional gene ablation while controlling for genetic background and tamoxifen effects. Setdb1 ablated ES cells exhibit severe growth inhibition, which is not rescued by exogenous Nanog expression or culturing in naive pluripotency '2i' media, suggesting that the self-renewal defect is mediated through pluripotency network independent pathways. Our strategy to generate null mutant mouse ES cells is applicable to thousands of genes and repurposes existing IKMC Intermediate Vectors.
Collapse
Affiliation(s)
- Cynthia L. Fisher
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- MRC London Institute of Medical Sciences and Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Hendrik Marks
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, 6525 GA, Nijmegen, The Netherlands
| | - Lily Ting-yin Cho
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Robert Andrews
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - Sam Wormald
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Thomas Carroll
- MRC London Institute of Medical Sciences and Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Vivek Iyer
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Peri Tate
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Barry Rosen
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Hendrik G. Stunnenberg
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, 6525 GA, Nijmegen, The Netherlands
| | - Amanda G. Fisher
- MRC London Institute of Medical Sciences and Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - William C. Skarnes
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| |
Collapse
|
3
|
Svobodova M, Raudenska M, Gumulec J, Balvan J, Fojtu M, Kratochvilova M, Polanska H, Horakova Z, Kostrica R, Babula P, Heger Z, Masarik M. Establishment of oral squamous cell carcinoma cell line and magnetic bead-based isolation and characterization of its CD90/CD44 subpopulations. Oncotarget 2017; 8:66254-66269. [PMID: 29029509 PMCID: PMC5630409 DOI: 10.18632/oncotarget.19914] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 06/28/2017] [Indexed: 12/26/2022] Open
Abstract
In this study, we describe the establishment of the human papillomavirus 18-positive, stage II, grade 1, T2N0M0 head and neck tumor primary cell line derived from oral squamous cell carcinoma of a non-smoking patient by using two different protocols. Furthermore, a preparation of subpopulations derived from this primary cell line according to the cluster of differentiation molecules CD44/CD90 status using magnetic bead-based separation and their characterization was performed. Impedance-based real-time cell analysis, enzyme-linked immunsorbant assay (ELISA), wound-healing assay, flow-cytometry, gene expression analysis, and MTT assay were used to characterize these four subpopulations (CD44+/CD90-, CD44-/CD90-, CD44+/CD90+, CD44-/CD90-). We optimised methodics for establishement of primary cell lines derived from oral squamous cell carcinoma tissue samples and subsequent separation of mesenchymal (CD90+) and epithelial (CD90-) types of tumorous cells. Primary cell line prepared by using trypsin proteolysis was more viable than the one prepared by using collagenase. According to our results, CD90 separation is a necessary step in preparation of permanent tumor-tissue derived cell lines. Based on the wound-healing assay, CD44+ cells exhibited stronger migratory capacity than CD44- subpopulations. CD44+ subpopulations had also significantly higher expression of BIRC5 and SOX2, lower expression of FLT1 and IL6, and higher levels of basal autophagy compared to CD44- subpopulations. Furthermore, co-cultivation experiments revealed that CD44-/CD90+ cells supported growth of epithelial tumor cells (CD44+/CD90-). On the contrary, factors released by CD44+/CD90+ type of cells seem to have rather inhibiting effect. The most cisplatin-resistant subpopulation with the shortest doubling time was CD44-/CD90+, but this subpopulation had a low migratory capacity.
Collapse
Affiliation(s)
- Marketa Svobodova
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, CZ-61600 Brno, Czech Republic
| | - Martina Raudenska
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, CZ-61600 Brno, Czech Republic
| | - Jaromir Gumulec
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, CZ-61600 Brno, Czech Republic
| | - Jan Balvan
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Michaela Fojtu
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Monika Kratochvilova
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, CZ-61600 Brno, Czech Republic
| | - Hana Polanska
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, CZ-61600 Brno, Czech Republic
| | - Zuzana Horakova
- Department of Otorhinolaryngology and Head and Neck Surgery, St. Anne’s Faculty Hospital, CZ-65691 Brno, Czech Republic
| | - Rom Kostrica
- Department of Otorhinolaryngology and Head and Neck Surgery, St. Anne’s Faculty Hospital, CZ-65691 Brno, Czech Republic
| | - Petr Babula
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Zbynek Heger
- Central European Institute of Technology, Brno University of Technology, CZ-61600 Brno, Czech Republic
- Department of Chemistry and Biochemistry, Mendel University in Brno, CZ-61300 Brno, Czech Republic
| | - Michal Masarik
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
| |
Collapse
|
4
|
McKee C, Chaudhry GR. Advances and challenges in stem cell culture. Colloids Surf B Biointerfaces 2017; 159:62-77. [PMID: 28780462 DOI: 10.1016/j.colsurfb.2017.07.051] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/04/2017] [Accepted: 07/22/2017] [Indexed: 12/12/2022]
Abstract
Stem cells (SCs) hold great promise for cell therapy, tissue engineering, and regenerative medicine as well as pharmaceutical and biotechnological applications. They have the capacity to self-renew and the ability to differentiate into specialized cell types depending upon their source of isolation. However, use of SCs for clinical applications requires a high quality and quantity of cells. This necessitates large-scale expansion of SCs followed by efficient and homogeneous differentiation into functional derivatives. Traditional methods for maintenance and expansion of cells rely on two-dimensional (2-D) culturing techniques using plastic culture plates and xenogenic media. These methods provide limited expansion and cells tend to lose clonal and differentiation capacity upon long-term passaging. Recently, new approaches for the expansion of SCs have emphasized three-dimensional (3-D) cell growth to mimic the in vivo environment. This review provides a comprehensive compendium of recent advancements in culturing SCs using 2-D and 3-D techniques involving spheroids, biomaterials, and bioreactors. In addition, potential challenges to achieve billion-fold expansion of cells are discussed.
Collapse
Affiliation(s)
- Christina McKee
- Department of Biological Sciences , Oakland University, Rochester, MI, 48309, USA; OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA
| | - G Rasul Chaudhry
- Department of Biological Sciences , Oakland University, Rochester, MI, 48309, USA; OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA.
| |
Collapse
|
5
|
Gawlik-Rzemieniewska N, Bednarek I. The role of NANOG transcriptional factor in the development of malignant phenotype of cancer cells. Cancer Biol Ther 2016; 17:1-10. [PMID: 26618281 DOI: 10.1080/15384047.2015.1121348] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
NANOG is a transcription factor that is involved in the self-renewal of embryonic stem cells (ES) and is a critical factor for the maintenance of the undifferentiated state of pluripotent cells. Extensive data in the literature show that the NANOG gene is aberrantly expressed during the development of malignancy in cancer cells. ES and cancer stem cells (CSCs), a subpopulation of cancer cells within the tumor, are thought to share common phenotypic properties. This review describes the role of NANOG in cancer cell proliferation, epithelial-mesenchymal transition (EMT), apoptosis and metastasis. In addition, this paper illustrates a correlation between NANOG and signal transducer and activator of transcription 3 (STAT3) in the maintenance of cancer stem cell properties and multidrug resistance. Together, the available data demonstrate that NANOG is strictly involved in the process of carcinogenesis and is a potential prognostic marker of malignant tumors.
Collapse
Affiliation(s)
- Natalia Gawlik-Rzemieniewska
- a School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia , Katowice , Poland
| | - Ilona Bednarek
- a School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia , Katowice , Poland
| |
Collapse
|
6
|
Gawlik-Rzemieniewska N, Galilejczyk A, Krawczyk M, Bednarek I. Silencing expression of the NANOG gene and changes in migration and metastasis of urinary bladder cancer cells. Arch Med Sci 2016; 12:889-97. [PMID: 27478472 PMCID: PMC4947613 DOI: 10.5114/aoms.2015.55368] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 01/01/2015] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION It has been proved that expression of the NANOG gene is observed not only in embryonic-derived malignancies, but also in breast cancer, ovarian cancer, cervix cancer and bladder cancer. NANOG overexpression is correlated with high activity of MMP-2 and MMP-9. The aim of the study was to evaluate the changes in the malignant phenotype of T24 bladder cancer cells with modulated expression of the NANOG gene. MATERIAL AND METHODS Human urinary bladder cancer cells T24 (HTB-4) were cultivated under standard conditions. Transfection of the cells with silencing constructions was performed with the application of Lipofectamine 2000 (Invitrogen) reagent. Evaluation of changes in the expression level of individual genes was performed using qRTPCR. Changes in the protein level were evaluated using the Human ELISA Kit (Abcam). The invasion capability of transfected cells was tested using Matrigel Invasion Chambers (BD Biosciences). The changes in cell migration were assessed with a wound-healing assay. RESULTS The qRTPCR evaluation showed that silencing the NANOG gene in T24 cells led to the decrease of mRNA for the MMP-2 gene to the level of 62.4% and the MMP-9 gene to the level of 76%. The cells with modulated expression of the NANOG gene migrated slower in the Matrigel invasion assay and in the wound-healing assay. The immunoenzymatic test showed a decrease in the protein level of MMP-9. CONCLUSIONS The transcriptional activity of the NANOG gene might be connected with some aspects of bladder cancer cell metastasis in vitro and has an influence on MMP-2 and MMP-9 expression levels.
Collapse
Affiliation(s)
- Natalia Gawlik-Rzemieniewska
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia, Katowice, Poland
| | - Anna Galilejczyk
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia, Katowice, Poland
| | - Michał Krawczyk
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia, Katowice, Poland
| | - Ilona Bednarek
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
7
|
Wang X, Wang Y, Zuo Q, Li D, Zhang W, Lian C, Tang B, Xiao T, Wang M, Wang K, Li B, Zhang Y. The synergistic effect of 5Azadc and TSA on maintenance of pluripotency of chicken ESCs by overexpression of NANOG gene. In Vitro Cell Dev Biol Anim 2016; 52:488-96. [PMID: 26822431 DOI: 10.1007/s11626-015-9993-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 12/20/2015] [Indexed: 11/30/2022]
Abstract
NANOG is a transcription factor that functions in embryonic stem cells (ESCs) and a key factor in maintaining pluripotency. Here, we cloned the NANOG gene promoter from the Rugao yellow chicken and constructed a dual luciferase reporter vector to detect its transcriptional activity and analyze the effects of 5-aza-2'-deoxycytidine (5-Azadc) and trichostatin A (TSA) on NANOG promoter activity and ESC pluripotency maintenance in vitro. NANOG transcriptional activity was enhanced when 5-Azadc and TSA were used alone or together, suggesting the possibility of elevated methylation of the CpG island in the NANOG regulatory region. When ESCs were cultured in basic medium with 5-Azadc and TSA in vitro, significantly more cell colonies were maintained in the 5-Azadc + TSA group than in the control group, which had many differentiated cells and few cell colonies after 6 d of induction. On the tenth day of induction, the cells in the control group fully differentiated and no cell colonies remained, but many cell colonies were present in the 5-Azadc + TSA group. The expression of NANOG in the cell colonies was confirmed by indirect immunofluorescence. Furthermore, ESCs could be passaged to the 12th generation under 5-Azadc and TSA treatment and maintained their pluripotency. Thus, we showed that 5-Azadc and TSA can effectively maintain chicken ESC pluripotency in vitro by increasing NANOG gene expression.
Collapse
Affiliation(s)
- Xiaoyan Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, Jiangsu Province, People's Republic of China.,Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Yingjie Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, Jiangsu Province, People's Republic of China.,Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Qisheng Zuo
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, Jiangsu Province, People's Republic of China.,Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Dong Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, Jiangsu Province, People's Republic of China.,Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Wenhui Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, Jiangsu Province, People's Republic of China.,Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Chao Lian
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, Jiangsu Province, People's Republic of China.,Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Beibei Tang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, Jiangsu Province, People's Republic of China.,Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Tianrong Xiao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, Jiangsu Province, People's Republic of China.,Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Man Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, Jiangsu Province, People's Republic of China.,Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Kehua Wang
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, 225125, Jiangsu Province, People's Republic of China
| | - Bichun Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, Jiangsu Province, People's Republic of China. .,Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, 225009, Jiangsu Province, People's Republic of China.
| | - Yani Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, Jiangsu Province, People's Republic of China. .,Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, 225009, Jiangsu Province, People's Republic of China.
| |
Collapse
|
8
|
Krause MN, Sancho-Martinez I, Izpisua Belmonte JC. Understanding the molecular mechanisms of reprogramming. Biochem Biophys Res Commun 2015; 473:693-7. [PMID: 26655812 DOI: 10.1016/j.bbrc.2015.11.120] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/25/2015] [Indexed: 12/28/2022]
Abstract
Despite the profound and rapid advancements in reprogramming technologies since the generation of the first induced pluripotent stem cells (iPSCs) in 2006[1], the molecular basics of the process and its implications are still not fully understood. Recent work has suggested that a subset of TFs, so called "Pioneer TFs", play an important role during the stochastic phase of iPSC reprogramming [2-6]. Pioneer TFs activities differ from conventional transcription factors in their mechanism of action. They bind directly to condensed chromatin and elicit a series of chromatin remodeling events that lead to opening of the chromatin. Chromatin decondensation by pioneer factors progressively occurs during cell division and in turn exposes specific gene promoters in the DNA to which TFs can now directly bind to promoters that are readily accessible[2, 6]. Here, we will summarize recent advancements on our understanding of the molecular mechanisms underlying reprogramming to iPSC as well as the implications that pioneer Transcription Factor activities might play during different lineage conversion processes.
Collapse
Affiliation(s)
- Marie N Krause
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla 92037, CA, USA; University Hospital of Würzburg, Department of Pediatrics, 2 Josef-Schneiderstrasse, 97080 Würzburg, Germany
| | - Ignacio Sancho-Martinez
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla 92037, CA, USA; Centre for Stem Cells and Regenerative Medicine, King's College London, 28th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London, UK
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla 92037, CA, USA.
| |
Collapse
|
9
|
Allouba MH, ElGuindy AM, Krishnamoorthy N, Yacoub MH, Aguib YE. NaNog: A pluripotency homeobox (master) molecule. Glob Cardiol Sci Pract 2015; 2015:36. [PMID: 26566529 PMCID: PMC4625207 DOI: 10.5339/gcsp.2015.36] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 06/30/2015] [Indexed: 01/06/2023] Open
Abstract
One of the most intriguing aspects of cell biology is the state of pluripotency, where the cell is capable of self-renewal for as many times as deemed “necessary”, then at a specified time can differentiate into any type of cell. This fundamental process is required during organogenesis in foetal life and importantly during tissue repair in health and disease. Pluripotency is very tightly regulated, as any dysregulation can result in congenital defects, inability to repair damage, or cancer. Fuelled by the relatively recent interest in stem cell biology and tissue regeneration, the molecules implicated in regulating pluripotency have been the subject of extensive research. One of the important molecules involved in pluripotency, is NaNog, the subject of this article.
Collapse
Affiliation(s)
| | | | - Navaneethakrishnan Krishnamoorthy
- Qatar Cardiovascular Research Centre, Doha, Qatar ; Harefield Heart Science Centre, National Heart and Lung Institute, Imperial College London, UK
| | | | | |
Collapse
|
10
|
Voutsadakis IA. The network of pluripotency, epithelial-mesenchymal transition, and prognosis of breast cancer. BREAST CANCER-TARGETS AND THERAPY 2015; 7:303-19. [PMID: 26379447 PMCID: PMC4567227 DOI: 10.2147/bctt.s71163] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Breast cancer is the leading female cancer in terms of prevalence. Progress in molecular biology has brought forward a better understanding of its pathogenesis that has led to better prognostication and treatment. Subtypes of breast cancer have been identified at the genomic level and guide therapeutic decisions based on their biology and the expected benefit from various interventions. Despite this progress, a significant percentage of patients die from their disease and further improvements are needed. The cancer stem cell theory and the epithelial-mesenchymal transition are two comparatively novel concepts that have been introduced in the area of cancer research and are actively investigated. Both processes have their physiologic roots in normal development and common mediators have begun to surface. This review discusses the associations of these networks as a prognostic framework in breast cancer.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Division of Medical Oncology, Department of Internal Medicine, Sault Area Hospital, Sault Ste Marie, ON, Canada ; Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada
| |
Collapse
|
11
|
Satheesha S, Manzella G, Bovay A, Casanova EA, Bode PK, Belle R, Feuchtgruber S, Jaaks P, Dogan N, Koscielniak E, Schäfer BW. Targeting hedgehog signaling reduces self-renewal in embryonal rhabdomyosarcoma. Oncogene 2015; 35:2020-30. [PMID: 26189795 PMCID: PMC5399168 DOI: 10.1038/onc.2015.267] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 04/14/2014] [Accepted: 06/19/2015] [Indexed: 12/11/2022]
Abstract
Current treatment regimens for rhabdomyosarcoma (RMS), the most common pediatric soft tissue cancer, rely on conventional chemotherapy, and although they show clinical benefit, there is a significant risk of adverse side effects and secondary tumors later in life. Therefore, identifying and targeting sub-populations with higher tumorigenic potential and self-renewing capacity would offer improved patient management strategies. Hedgehog signaling has been linked to the development of embryonal RMS (ERMS) through mouse genetics and rare human syndromes. However, activating mutations in this pathway in sporadic RMS are rare and therefore the contribution of hedgehog signaling to oncogenesis remains unclear. Here, we show by genetic loss- and gain-of-function experiments and the use of clinically relevant small molecule modulators that hedgehog signaling is important for controlling self-renewal of a subpopulation of RMS cells in vitro and tumor initiation in vivo. In addition, hedgehog activity altered chemoresistance, motility and differentiation status. The core stem cell gene NANOG was determined to be important for ERMS self-renewal, possibly acting downstream of hedgehog signaling. Crucially, evaluating the presence of a subpopulation of tumor-propagating cells in patient biopsies identified by GLI1 and NANOG expression had prognostic significance. Hence, this work identifies novel functional aspects of hedgehog signaling in ERMS, redefines the rationale for its targeting as means to control ERMS self-renewal and underscores the importance of studying functional tumor heterogeneity in pediatric cancers.
Collapse
Affiliation(s)
- S Satheesha
- Department of Oncology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - G Manzella
- Department of Oncology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - A Bovay
- Department of Oncology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - E A Casanova
- Department of Oncology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - P K Bode
- Department of Oncology and Children's Research Center, University Children's Hospital, Zurich, Switzerland.,Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - R Belle
- Department of Oncology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - S Feuchtgruber
- Department of Oncology/Hematology/Immunology, Olgahospital, Klinikum Stuttgart, Stuttgart, Germany
| | - P Jaaks
- Department of Oncology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - N Dogan
- Department of Oncology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - E Koscielniak
- Department of Oncology/Hematology/Immunology, Olgahospital, Klinikum Stuttgart, Stuttgart, Germany
| | - B W Schäfer
- Department of Oncology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| |
Collapse
|
12
|
Kim J, Liu Y, Qiu M, Xu Y. Pluripotency factor Nanog is tumorigenic by deregulating DNA damage response in somatic cells. Oncogene 2015; 35:1334-40. [DOI: 10.1038/onc.2015.205] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/27/2015] [Accepted: 04/14/2015] [Indexed: 12/19/2022]
|
13
|
Squillaro T, Severino V, Alessio N, Farina A, Di Bernardo G, Cipollaro M, Peluso G, Chambery A, Galderisi U. De-regulated expression of the BRG1 chromatin remodeling factor in bone marrow mesenchymal stromal cells induces senescence associated with the silencing of NANOG and changes in the levels of chromatin proteins. Cell Cycle 2015; 14:1315-1326. [PMID: 25724006 PMCID: PMC4614278 DOI: 10.4161/15384101.2014.995053] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 12/01/2014] [Accepted: 12/02/2014] [Indexed: 12/17/2022] Open
Abstract
Stem cells have a peculiar chromatin architecture that contributes to their unique properties, including uncommitted status, multi/pluripotency and self-renewal. We analyzed the effect of the de-regulation of the SWI/SNF chromatin remodeling complex in mesenchymal stromal cells (MSC) through the silencing and up-regulation of BRG1, which is the ATPase subunit of the complex. The altered expression of BRG1 promoted the senescence of MSC with suppression of the NANOG transcription, which is part of the transcriptional circuitry governing stem cell functions. To gain insight on the way NANOG was silenced, we evaluated how the de-regulated BRG1 expression affect the binding of activators and repressors on the NANOG promoter. We found 4 E2F binding motifs on NANOG promoter, which can be occupied by RB1 and RB2/P130. These are members of the retinoblastoma gene family. In MSC with a silenced BRG1, the relative binding of the 2 retinoblastoma proteins increased, and this was associated with the recruitment of DNMT1. This induced the methylation of CpG on the NANOG promoter. Opposingly, when a high level of BRG1 was present, the same E2F binding motifs were docking sites for BRG1, which induced chromatin compaction without CpG methylation but with increased histone deacetylation, associated with the presence of HDAC1 on E2F binding sites. Besides the sharp regulation of the NANOG expression, we evidenced, through proteomic analysis, that the de-regulation of the SWI/SNF function affected the expression of histones and other nuclear proteins involved in "nuclear architecture," suggesting that BRG1 may act as global regulator of gene expression.
Collapse
Affiliation(s)
- Tiziana Squillaro
- Department of Experimental Medicine; Biotechnology and Molecular Biology Section; Second University of Naples; Naples, Italy
| | - Valeria Severino
- Department of Environmental; Biological and Pharmaceutical Sciences and Technologies; Second University of Naples; Naples, Italy
| | - Nicola Alessio
- Biomedical Proteomics Research Group; Department of Human Protein Science; Geneva University; Geneva, Switzerland
| | - Annarita Farina
- Biomedical Proteomics Research Group; Department of Human Protein Science; Geneva University; Geneva, Switzerland
| | - Giovanni Di Bernardo
- Department of Experimental Medicine; Biotechnology and Molecular Biology Section; Second University of Naples; Naples, Italy
| | - Marilena Cipollaro
- Department of Experimental Medicine; Biotechnology and Molecular Biology Section; Second University of Naples; Naples, Italy
| | | | - Angela Chambery
- Department of Environmental; Biological and Pharmaceutical Sciences and Technologies; Second University of Naples; Naples, Italy
- IRCCS; Multimedica, Milano, Italy
| | - Umberto Galderisi
- Department of Experimental Medicine; Biotechnology and Molecular Biology Section; Second University of Naples; Naples, Italy
- Institute Bioscience and BioResources; CNR; Naples, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine; Center for Biotechnology; Temple University; Philadelphia, PA USA
| |
Collapse
|
14
|
Peter Y, Weingarten M, Akhavan N, Hanau J. A Place to Call Home: Bioengineering Pluripotential Stem Cell Cultures. AIMS BIOENGINEERING 2015. [DOI: 10.3934/bioeng.2015.2.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
15
|
Sun LT, Yamaguchi S, Hirano K, Ichisaka T, Kuroda T, Tada T. Nanog co-regulated by Nodal/Smad2 and Oct4 is required for pluripotency in developing mouse epiblast. Dev Biol 2014; 392:182-92. [DOI: 10.1016/j.ydbio.2014.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 05/30/2014] [Accepted: 06/03/2014] [Indexed: 10/25/2022]
|
16
|
Guasti L, Vagaska B, Bulstrode NW, Seifalian AM, Ferretti P. Chondrogenic differentiation of adipose tissue-derived stem cells within nanocaged POSS-PCU scaffolds: A new tool for nanomedicine. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:279-89. [DOI: 10.1016/j.nano.2013.08.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 08/19/2013] [Accepted: 08/22/2013] [Indexed: 02/06/2023]
|
17
|
Golubovskaya VM. FAK and Nanog cross talk with p53 in cancer stem cells. Anticancer Agents Med Chem 2014; 13:576-80. [PMID: 22934707 DOI: 10.2174/1871520611313040006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 05/21/2012] [Accepted: 08/01/2012] [Indexed: 12/31/2022]
Abstract
This review is focused on the role of Focal Adhesion Kinase (FAK) signaling in cancer stem cells. The recent data demonstrate the important role of FAK in cancer stem cell proliferation, differentiation, motility, and invasion. We showed recently that the transcription factor Nanog binds the FAK promoter and up-regulates FAK expression, and that FAK binds Nanog and phosphorylates it. This review discusses the interaction of FAK, Nanog, Oct-3/4, and Sox-2 signaling pathways that are critical for the regulation of cancer stem cells. The cross-linked signaling of FAK with p53 and Nanog signaling in cancer stem cell and function and targeted therapeutics approaches are discussed.
Collapse
Affiliation(s)
- Vita M Golubovskaya
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.
| |
Collapse
|
18
|
Hunt MM, Meng G, Rancourt DE, Gates ID, Kallos MS. Factorial Experimental Design for the Culture of Human Embryonic Stem Cells as Aggregates in Stirred Suspension Bioreactors Reveals the Potential for Interaction Effects Between Bioprocess Parameters. Tissue Eng Part C Methods 2014; 20:76-89. [DOI: 10.1089/ten.tec.2013.0040] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Megan M. Hunt
- Pharmaceutical Production Research Facility (PPRF), Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - Guoliang Meng
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Derrick E. Rancourt
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ian D. Gates
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - Michael S. Kallos
- Pharmaceutical Production Research Facility (PPRF), Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
19
|
Verma V, Huang B, Kallingappa PK, Oback B. Dual Kinase Inhibition Promotes Pluripotency in Finite Bovine Embryonic Cell Lines. Stem Cells Dev 2013; 22:1728-42. [DOI: 10.1089/scd.2012.0481] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Vinod Verma
- AgResearch, Ruakura Research Centre, Hamilton, New Zealand
| | - Ben Huang
- AgResearch, Ruakura Research Centre, Hamilton, New Zealand
| | | | - Björn Oback
- AgResearch, Ruakura Research Centre, Hamilton, New Zealand
| |
Collapse
|
20
|
Wu J, Tzanakakis ES. Contribution of stochastic partitioning at human embryonic stem cell division to NANOG heterogeneity. PLoS One 2012; 7:e50715. [PMID: 23226362 PMCID: PMC3511357 DOI: 10.1371/journal.pone.0050715] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 10/23/2012] [Indexed: 12/16/2022] Open
Abstract
Heterogeneity is an often unappreciated characteristic of stem cell populations yet its importance in fate determination is becoming increasingly evident. Although gene expression noise has received greater attention as a source of non-genetic heterogeneity, the effects of stochastic partitioning of cellular material during mitosis on population variability have not been researched to date. We examined self-renewing human embryonic stem cells (hESCs), which typically exhibit a dispersed distribution of the pluripotency marker NANOG. In conjunction with our experiments, a multiscale cell population balance equation (PBE) model was constructed accounting for transcriptional noise and stochastic partitioning at division as sources of population heterogeneity. Cultured hESCs maintained time-invariant profiles of size and NANOG expression and the data were utilized for parameter estimation. Contributions from both sources considered in this study were significant on the NANOG profile, although elimination of the gene expression noise resulted in greater changes in the dispersion of the NANOG distribution. Moreover, blocking of division by treating hESCs with nocodazole or colcemid led to a 39% increase in the average NANOG content and over 68% of the cells had higher NANOG level than the mean NANOG expression of untreated cells. Model predictions, which were in excellent agreement with these findings, revealed that stochastic partitioning accounted for 17% of the total noise in the NANOG profile of self-renewing hESCs. The computational framework developed in this study will aid in gaining a deeper understanding of how pluripotent stem/progenitor cells orchestrate processes such as gene expression and proliferation for maintaining their pluripotency or differentiating along particular lineages. Such models will be essential in designing and optimizing efficient differentiation strategies and bioprocesses for the production of therapeutically suitable stem cell progeny.
Collapse
Affiliation(s)
- Jincheng Wu
- Department of Chemical and Biological Engineering, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Emmanuel S. Tzanakakis
- Department of Chemical and Biological Engineering, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, New York, United States of America
- New York State Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
21
|
Chalut KJ, Höpfler M, Lautenschläger F, Boyde L, Chan CJ, Ekpenyong A, Martinez-Arias A, Guck J. Chromatin decondensation and nuclear softening accompany Nanog downregulation in embryonic stem cells. Biophys J 2012. [PMID: 23200040 DOI: 10.1016/j.bpj.2012.10.015] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The interplay between epigenetic modification and chromatin compaction is implicated in the regulation of gene expression, and it comprises one of the most fascinating frontiers in cell biology. Although a complete picture is still lacking, it is generally accepted that the differentiation of embryonic stem (ES) cells is accompanied by a selective condensation into heterochromatin with concomitant gene silencing, leaving access only to lineage-specific genes in the euchromatin. ES cells have been reported to have less condensed chromatin, as they are capable of differentiating into any cell type. However, pluripotency itself-even prior to differentiation-is a split state comprising a naïve state and a state in which ES cells prime for differentiation. Here, we show that naïve ES cells decondense their chromatin in the course of downregulating the pluripotency marker Nanog before they initiate lineage commitment. We used fluorescence recovery after photobleaching, and histone modification analysis paired with a novel, to our knowledge, optical stretching method, to show that ES cells in the naïve state have a significantly stiffer nucleus that is coupled to a globally more condensed chromatin state. We link this biophysical phenotype to coinciding epigenetic differences, including histone methylation, and show a strong correlation of chromatin condensation and nuclear stiffness with the expression of Nanog. Besides having implications for transcriptional regulation and embryonic cell sorting and suggesting a putative mechanosensing mechanism, the physical differences point to a system-level regulatory role of chromatin in maintaining pluripotency in embryonic development.
Collapse
Affiliation(s)
- Kevin J Chalut
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Manoli DS, Subramanyam D, Carey C, Sudin E, Van Westerhuyzen JA, Bales KL, Blelloch R, Shah NM. Generation of induced pluripotent stem cells from the prairie vole. PLoS One 2012; 7:e38119. [PMID: 22675440 PMCID: PMC3365000 DOI: 10.1371/journal.pone.0038119] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 05/01/2012] [Indexed: 01/01/2023] Open
Abstract
The vast majority of animals mate more or less promiscuously. A few mammals, including humans, utilize more restrained mating strategies that entail a longer term affiliation with a single mating partner. Such pair bonding mating strategies have been resistant to genetic analysis because of a lack of suitable model organisms. Prairie voles are small mouse-like rodents that form enduring pair bonds in the wild as well as in the laboratory, and consequently they have been used widely to study social bonding behavior. The lack of targeted genetic approaches in this species however has restricted the study of the molecular and neural circuit basis of pair bonds. As a first step in rendering the prairie vole amenable to reverse genetics, we have generated induced pluripotent stem cell (IPSC) lines from prairie vole fibroblasts using retroviral transduction of reprogramming factors. These IPSC lines display the cellular and molecular hallmarks of IPSC cells from other organisms, including mice and humans. Moreover, the prairie vole IPSC lines have pluripotent differentiation potential since they can give rise to all three germ layers in tissue culture and in vivo. These IPSC lines can now be used to develop conditions that facilitate homologous recombination and eventually the generation of prairie voles bearing targeted genetic modifications to study the molecular and neural basis of pair bond formation.
Collapse
Affiliation(s)
- Devanand S. Manoli
- Department of Anatomy, University of California San Francisco, San Francisco, California, United States of America
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
| | - Deepa Subramanyam
- Department of Urology, University of California San Francisco, San Francisco, California, United States of America
| | - Catriona Carey
- Department of Anatomy, University of California San Francisco, San Francisco, California, United States of America
| | - Erik Sudin
- Department of Anatomy, University of California San Francisco, San Francisco, California, United States of America
| | - Julie A. Van Westerhuyzen
- Department of Psychology, University of California Davis, Davis, California, United States of America
| | - Karen L. Bales
- Department of Psychology, University of California Davis, Davis, California, United States of America
| | - Robert Blelloch
- Department of Urology, University of California San Francisco, San Francisco, California, United States of America
- Center for Reproductive Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Nirao M. Shah
- Department of Anatomy, University of California San Francisco, San Francisco, California, United States of America
- Center for Reproductive Sciences, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
23
|
Zheng Z, Jian J, Zhang X, Zara JN, Yin W, Chiang M, Liu Y, Wang J, Pang S, Ting K, Soo C. Reprogramming of human fibroblasts into multipotent cells with a single ECM proteoglycan, fibromodulin. Biomaterials 2012; 33:5821-31. [PMID: 22622142 DOI: 10.1016/j.biomaterials.2012.04.049] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 04/21/2012] [Indexed: 01/01/2023]
Abstract
Pluripotent and/or multipotent stem cell-based therapeutics are a vital component of tissue engineering and regenerative medicine. The generation or isolation of safer and readily available stem cell sources will significantly aid clinical applications. We report here a technique using a single molecule, recombinant human fibromodulin protein (FMOD), to reprogram human fibroblasts into multipotent cells. Like virally-induced pluripotent stem (iPS) cells, FMOD reprogrammed (FReP) cells express pluripotency markers, form embryoid bodies (EBs), and differentiate into ectoderm, mesoderm, and endoderm derivatives in vitro. Notably, FReP cells regenerate muscle and bone tissues but do not generate teratomas in vivo. Unlike iPS cells, undifferentiated FReP cells proliferate slowly and express low proto-oncogene c-MYC and unexpectedly high levels of cyclin-dependent kinase inhibitors p15(Ink4B) and p21(WAF1/Cip1). Remarkably, in a fashion reminiscent of quiescent stem cells, the slow replicative phenotype of undifferentiated FReP cells reverses after differentiation induction, with differentiating FReP cells proliferating faster and expressing less p15(Ink4B) and p21(WAF1/Cip1) than differentiating iPS cells. Overall, single protein, FMOD-based, cell reprograming bypasses the risks of mutation, gene instability, and malignancy associated with genetically-modified iPS cells, and provides an alternative strategy for engineering patient-specific multipotent cells for basic research and therapeutic application.
Collapse
Affiliation(s)
- Zhong Zheng
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA 90095-1759, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Park SW, Do HJ, Huh SH, Sung B, Uhm SJ, Song H, Kim NH, Kim JH. Identification of a putative nuclear export signal motif in human NANOG homeobox domain. Biochem Biophys Res Commun 2012; 421:484-9. [PMID: 22516749 DOI: 10.1016/j.bbrc.2012.04.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 04/03/2012] [Indexed: 12/29/2022]
Abstract
NANOG is a homeobox-containing transcription factor that plays an important role in pluripotent stem cells and tumorigenic cells. To understand how nuclear localization of human NANOG is regulated, the NANOG sequence was examined and a leucine-rich nuclear export signal (NES) motif ((125)MQELSNILNL(134)) was found in the homeodomain (HD). To functionally validate the putative NES motif, deletion and site-directed mutants were fused to an EGFP expression vector and transfected into COS-7 cells, and the localization of the proteins was examined. While hNANOG HD exclusively localized to the nucleus, a mutant with both NLSs deleted and only the putative NES motif contained (hNANOG HD-ΔNLSs) was predominantly cytoplasmic, as observed by nucleo/cytoplasmic fractionation and Western blot analysis as well as confocal microscopy. Furthermore, site-directed mutagenesis of the putative NES motif in a partial hNANOG HD only containing either one of the two NLS motifs led to localization in the nucleus, suggesting that the NES motif may play a functional role in nuclear export. Furthermore, CRM1-specific nuclear export inhibitor LMB blocked the hNANOG potent NES-mediated export, suggesting that the leucine-rich motif may function in CRM1-mediated nuclear export of hNANOG. Collectively, a NES motif is present in the hNANOG HD and may be functionally involved in CRM1-mediated nuclear export pathway.
Collapse
Affiliation(s)
- Sung-Won Park
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-si, Gyeonggi-Do, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Lin Y, Cheng Z, Yang Z, Zheng J, Lin T. DNp73 improves generation efficiency of human induced pluripotent stem cells. BMC Cell Biol 2012; 13:9. [PMID: 22449255 PMCID: PMC3348002 DOI: 10.1186/1471-2121-13-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 03/26/2012] [Indexed: 11/10/2022] Open
Abstract
Background Recent studies have found that p53 and its' associated cell cycle pathways are major inhibitors of human induced pluripotent stem (iPS) cell generation. In the same family as p53 is p73, which shares sequence similarities with p53. However, p73 also has distinct properties of its own, such as two alternative promoters to express transactivation of p73 (TAp73) and N terminal deleted p73 (DNp73). Functionally, TAp73 acts similarly to p53 in tumor suppression. However, DNp73, on the other hand acts as an oncogene to suppress p53 and p73 induced apoptosis. Therefore, how can p73 have opposing roles in human iPS cell generation? Results Transcription factors, Oct4, Sox2, Klf4 and cMyc (4TF, Yamanaka factors) are used as basal conditions to generate iPS cells. In addition, the factor of DNp73(actually alpha splicing DNp73, DNp73α) is used to generate iPS cells. The experiment found that the addition of DNp73 gene increases human iPS cell generation efficiency by 12.6 folds in comparison to human fibroblast cells transduced with only the basal conditions. Also, iPS cells generated with DNp73 expression are more resistant to in vitro and in vivo differentiation. Conclusions This study found DNp73, a family member of p53, is also involved in the human iPS cell generation. Specifically, that the involvement of DNp73 generates iPS cells that are more resistant to in vitro and in vivo differentiation. Therefore, this data may prove to be useful in future developmental studies and cancer researches.
Collapse
Affiliation(s)
- Yi Lin
- Stem Cell Research Center, Fujian Agriculture and Forestry University, Cangshan District, Fuzhou, Fujian, PR China
| | | | | | | | | |
Collapse
|
26
|
Cheng F, Ke Q, Chen F, Cai B, Gao Y, Ye C, Wang D, Zhang L, Lahn BT, Li W, Xiang AP. Protecting against wayward human induced pluripotent stem cells with a suicide gene. Biomaterials 2012; 33:3195-204. [PMID: 22269649 DOI: 10.1016/j.biomaterials.2012.01.023] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2011] [Accepted: 01/09/2012] [Indexed: 11/30/2022]
Abstract
The generation of human induced pluripotent stem cells (hiPSCs) opens a prospect for regenerative medicine. However, transplantation of somatic cells derived from hiPSCs still harbor many risks such as cells' incorrect differentiation or over-proliferation, and the worst, tumor formation. Therefore, it's essential to ravel out these obstacles before their clinical application. Herein, we genetically modified hiPSCs and human embryonic stem cells (hESCs) with a truncated herpes simplex virus delta thymidine kinase (deltaTK) gene driven by EF1α or Nanog promoter to selectively ablate wayward pluripotent stem cells. The results showed that insertion of deltaTK gene did not alter their pluripotency and self-renewal capacity but rendered them sensitive to ganciclovir, which induced elimination of deltaTK(+) cells in vitro in a dose and time-dependent manner, most importantly, facilitated both prevention and ablation of tumors in vivo. Furthermore, comparative analysis between transduced hiPSCs and hESCs showed that there was no difference in ganciclovir sensitivity between them. This approach may help to develop safety strategies for clinical application of hiPSCs in regenerative medicine in the future.
Collapse
Affiliation(s)
- Fuyi Cheng
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Theunissen TW, Costa Y, Radzisheuskaya A, van Oosten AL, Lavial F, Pain B, Castro LFC, Silva JCR. Reprogramming capacity of Nanog is functionally conserved in vertebrates and resides in a unique homeodomain. Development 2011; 138:4853-65. [PMID: 22028025 PMCID: PMC3201656 DOI: 10.1242/dev.068775] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Pluripotency is a developmental ground state that can be recreated by direct reprogramming. Establishment of pluripotency is crucially dependent on the homeodomain-containing transcription factor Nanog. Compared with other pluripotency-associated genes, however, Nanog shows relatively low sequence conservation. Here, we investigated whether Nanog orthologs have the capacity to orchestrate establishment of pluripotency in Nanog(-/-) somatic cells. Mammalian, avian and teleost orthologs of Nanog enabled efficient reprogramming to full pluripotency, despite sharing as little as 13% sequence identity with mouse Nanog. Nanog orthologs supported self-renewal of pluripotent cells in the absence of leukemia inhibitory factor, and directly regulated mouse Nanog target genes. Related homeodomain transcription factors showed no reprogramming activity. Nanog is distinguished by the presence of two unique residues in the DNA recognition helix of its homeodomain, and mutations in these positions impaired reprogramming. On the basis of genome analysis and homeodomain identity, we propose that Nanog is a vertebrate innovation, which shared an ancestor with the Bsx gene family prior to the vertebrate radiation. However, cephalochordate Bsx did not have the capacity to replace mouse Nanog in reprogramming. Surprisingly, the Nanog homeodomain, a short sequence that contains the only recognizable conservation between Nanog orthologs, was sufficient to induce naive pluripotency in Nanog(-/-) somatic cells. This shows that control of the pluripotent state resides within a unique DNA-binding domain, which appeared at least 450 million years ago in a common ancestor of vertebrates. Our results support the hypothesis that naive pluripotency is a generic feature of vertebrate development.
Collapse
Affiliation(s)
- Thorold W Theunissen
- Wellcome Trust Centre for Stem Cell Research and Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Wilmut I, Sullivan G, Chambers I. The evolving biology of cell reprogramming. Philos Trans R Soc Lond B Biol Sci 2011; 366:2183-97. [PMID: 21727124 DOI: 10.1098/rstb.2011.0051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Modern stem cell biology has achieved a transformation that was thought by many to be every bit as unattainable as the ancient alchemists' dream of transforming base metals into gold. Exciting opportunities arise from the process known as 'cellular reprogramming' in which cells can be reliably changed from one tissue type to another. This is enabling novel approaches to more deeply investigate the fundamental basis of cell identity. In addition, new opportunities have also been created to study (perhaps even to treat) human genetic and degenerative diseases. Specific cell types that are affected in inherited disease can now be generated from easily accessible cells from the patient and compared with equivalent cells from healthy donors. The differences in cellular phenotype between the two may then be identified, and assays developed to establish therapies that prevent the development or progression of disease symptoms. Cellular reprogramming also has the potential to create new cells to replace those whose death or dysfunction causes disease symptoms. For patients suffering from inherited cases of degenerative diseases like Parkinson's disease or amyotrophic lateral sclerosis (also known as motor neuron disease), the future realization of such cell-based therapies would truly be worth its weight in gold. However, before this enormous potential can become a reality, several significant biological and technical challenges must be overcome. Furthermore, to maintain the credibility of the scientific community with the general public, it is important that hope-inspiring advances are not over-hyped. The papers in this issue of the Philosophical Transactions of the Royal Society B: Biological Sciences cover many areas relevant to this topic. In this Introduction, we provide an overall context in which to consider these individual papers.
Collapse
Affiliation(s)
- Ian Wilmut
- MRC Centre for Regenerative Medicine, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, Scotland, UK.
| | | | | |
Collapse
|