1
|
Barata T, Duarte I, Futschik ME. Integration of Stemness Gene Signatures Reveals Core Functional Modules of Stem Cells and Potential Novel Stemness Genes. Genes (Basel) 2023; 14:genes14030745. [PMID: 36981016 PMCID: PMC10048104 DOI: 10.3390/genes14030745] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Stem cells encompass a variety of different cell types which converge on the dual capacity to self-renew and differentiate into one or more lineages. These characteristic features are key for the involvement of stem cells in crucial biological processes such as development and ageing. To decipher their underlying genetic substrate, it is important to identify so-called stemness genes that are common to different stem cell types and are consistently identified across different studies. In this meta-analysis, 21 individual stemness signatures for humans and another 21 for mice, obtained from a variety of stem cell types and experimental techniques, were compared. Although we observed biological and experimental variability, a highly significant overlap between gene signatures was identified. This enabled us to define integrated stemness signatures (ISSs) comprised of genes frequently occurring among individual stemness signatures. Such integrated signatures help to exclude false positives that can compromise individual studies and can provide a more robust basis for investigation. To gain further insights into the relevance of ISSs, their genes were functionally annotated and connected within a molecular interaction network. Most importantly, the present analysis points to the potential roles of several less well-studied genes in stemness and thus provides promising candidates for further experimental validation.
Collapse
Affiliation(s)
- Tânia Barata
- SysBioLab, Centre for Biomedical Research (CBMR), Universidade do Algarve, 8005-139 Faro, Portugal
| | - Isabel Duarte
- Center for Research in Health Technologies and Information Systems (CINTESIS), Universidade do Algarve, 8005-139 Faro, Portugal
| | - Matthias E Futschik
- SysBioLab, Centre for Biomedical Research (CBMR), Universidade do Algarve, 8005-139 Faro, Portugal
- School of Biomedical Sciences, Faculty of Health, Derriford Research Facility, University of Plymouth, Plymouth PL6 8BU, UK
- MRC London Institute of Medical Sciences (LMS), Imperial College London, London W12 0NN, UK
- NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
| |
Collapse
|
2
|
TRANSPLANTATION OF SUBRETINAL STEM CELL-DERIVED RETINAL PIGMENT EPITHELIUM FOR STARGARDT DISEASE: A Phase I Clinical Trial. Retina 2023; 43:263-274. [PMID: 36223778 DOI: 10.1097/iae.0000000000003655] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
PURPOSE To assess the safety of injecting human embryonic stem cell retinal pigment epithelial cell dose to treat Stargardt disease. METHODS In this prospective, Phase I clinical trial, human embryonic stem cell retinal pigment epithelial cells in suspension were injected into the subretinal space in eyes with the worse best-corrected visual acuity (BCVA). After vitrectomy/posterior hyaloid removal, a partial retinal detachment was created and the human embryonic stem cell retinal pigment epithelial cells were administered. Phacoemulsification with intraocular lens implantation was performed in eyes with lens opacity. All procedures were optical coherence tomography-guided. The 12-month follow-up included retinal imaging, optical coherence tomography, visual field/electrophysiologic testing, and systemic evaluation. The main outcome was the absence of ocular/systemic inflammation or rejection, tumor formation, or toxicity during follow-up. RESULTS The mean baseline BCVAs in the phacoemulsification and no phacoemulsification groups were similar (1.950 ± 0.446 and 1.575 ± 0.303, respectively). One year postoperatively, treated eyes showed a nonsignificant increase in BCVA. No adverse effects occurred during follow-up. Intraoperative optical coherence tomography was important for guiding all procedures. CONCLUSION This surgical procedure was feasible and safe without cellular migration, rejection, inflammation, or development of ocular or systemic tumors during follow-up.
Collapse
|
3
|
Yadav S, Garrido A, Hernández MC, Oliveros JC, Pérez-García V, Fraga MF, Carrera AC. PI3Kβ-regulated β-catenin mediates EZH2 removal from promoters controlling primed human ESC stemness and primitive streak gene expression. Stem Cell Reports 2022; 17:2239-2255. [PMID: 36179694 PMCID: PMC9561645 DOI: 10.1016/j.stemcr.2022.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/21/2022] Open
Abstract
The mechanism governing the transition of human embryonic stem cells (hESCs) toward differentiated cells is only partially understood. To explore this transition, the activity and expression of the ubiquitous phosphatidylinositol 3-kinase (PI3Kα and PI3Kβ) were modulated in primed hESCs. The study reports a pathway that dismantles the restraint imposed by the EZH2 polycomb repressor on an essential stemness gene, NODAL, and on transcription factors required to trigger primitive streak formation. The primitive streak is the site where gastrulation begins to give rise to the three embryonic cell layers from which all human tissues derive. The pathway involves a PI3Kβ non-catalytic action that controls nuclear/active RAC1 levels, activation of JNK (Jun N-terminal kinase) and nuclear β-catenin accumulation. β-Catenin deposition at promoters triggers release of the EZH2 repressor, permitting stemness maintenance (through control of NODAL) and correct differentiation by allowing primitive streak master gene expression. PI3Kβ epigenetic control of EZH2/β-catenin might be modulated to direct stem cell differentiation. PI3Kβ directs epigenetic control of stemness and primitive streak (PS) essential genes PI3Kβ directs RAC1/JNK/β-catenin activation and induces EZH2 promoter displacement β-Catenin/EZH2 control NODAL, a gene essential for stemness and the master PS genes PI3Kβ/PI3K activities cooperate at stemness; PI3Kβ directs PS gene expression
Collapse
Affiliation(s)
- Sudhanshu Yadav
- Department of Immunology and Oncology, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | - Antonio Garrido
- Department of Immunology and Oncology, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | - M Carmen Hernández
- Department of Immunology and Oncology, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | - Juan C Oliveros
- Department of Systems Biology, Bioinformatics, Centro Nacional de Biotecnología/CSIC, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | - Vicente Pérez-García
- Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera, 46013 Valencia, Spain
| | - Mario F Fraga
- Nanomaterials and Nanotechnology Research Center/CSIC, Health Research Institute of Asturias (ISPA), Institute of Oncology of Asturias (IUOPA), Research Center for Rare Diseases (CIBERER), 33011 Oviedo, Asturias, Spain
| | - Ana C Carrera
- Department of Immunology and Oncology, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain.
| |
Collapse
|
4
|
Kusena JWT, Shariatzadeh M, Thomas RJ, Wilson SL. Understanding cell culture dynamics: a tool for defining protocol parameters for improved processes and efficient manufacturing using human embryonic stem cells. Bioengineered 2021; 12:979-996. [PMID: 33757391 PMCID: PMC8806349 DOI: 10.1080/21655979.2021.1902696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/09/2021] [Accepted: 03/09/2021] [Indexed: 12/16/2022] Open
Abstract
Standardization is crucial when culturing cells including human embryonic stem cells (hESCs) which are valuable for therapy development and disease modeling. Inherent issues regarding reproducibility of protocols are problematic as they hinder translation to good manufacturing practice (GMP), thus reducing clinical efficacy and uptake. Pluripotent cultures require standardization to ensure that input material is consistent prior to differentiation, as inconsistency of input cells creates end-product variation. To improve protocols, developers first must understand the cells they are working with and their related culture dynamics. This innovative work highlights key conditions required for optimized and cost-effective bioprocesses compared to generic protocols typically implemented. This entailed investigating conditions affecting growth, metabolism, and phenotype dynamics to ensure cell quality is appropriate for use. Results revealed critical process parameters (CPPs) including feeding regime and seeding density impact critical quality attributes (CQAs) including specific metabolic rate (SMR) and specific growth rate (SGR). This implied that process understanding, and control is essential to maintain key cell characteristics, reduce process variation and retain CQAs. Examination of cell dynamics and CPPs permitted the formation of a defined protocol for culturing H9 hESCs. The authors recommend that H9 seeding densities of 20,000 cells/cm2, four-day cultures or three-day cultures following a recovery passage from cryopreservation and 100% medium exchange after 48 hours are optimal. These parameters gave ~SGR of 0.018 hour-1 ± 1.5x10-3 over three days and cell viabilities ≥95%±0.4, while producing cells which highly expressed pluripotent and proliferation markers, Oct3/4 (>99% positive) and Ki-67 (>99% positive).
Collapse
Affiliation(s)
- J W T Kusena
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough University, Loughborough, Leicestershire, UK
| | - M Shariatzadeh
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough University, Loughborough, Leicestershire, UK
| | - R J Thomas
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough University, Loughborough, Leicestershire, UK
| | - S L Wilson
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough University, Loughborough, Leicestershire, UK
| |
Collapse
|
5
|
Reed CJ, Hutinet G, de Crécy-Lagard V. Comparative Genomic Analysis of the DUF34 Protein Family Suggests Role as a Metal Ion Chaperone or Insertase. Biomolecules 2021; 11:1282. [PMID: 34572495 PMCID: PMC8469502 DOI: 10.3390/biom11091282] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 12/12/2022] Open
Abstract
Members of the DUF34 (domain of unknown function 34) family, also known as the NIF3 protein superfamily, are ubiquitous across superkingdoms. Proteins of this family have been widely annotated as "GTP cyclohydrolase I type 2" through electronic propagation based on one study. Here, the annotation status of this protein family was examined through a comprehensive literature review and integrative bioinformatic analyses that revealed varied pleiotropic associations and phenotypes. This analysis combined with functional complementation studies strongly challenges the current annotation and suggests that DUF34 family members may serve as metal ion insertases, chaperones, or metallocofactor maturases. This general molecular function could explain how DUF34 subgroups participate in highly diversified pathways such as cell differentiation, metal ion homeostasis, pathogen virulence, redox, and universal stress responses.
Collapse
Affiliation(s)
- Colbie J. Reed
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA; (C.J.R.); (G.H.)
| | - Geoffrey Hutinet
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA; (C.J.R.); (G.H.)
| | - Valérie de Crécy-Lagard
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA; (C.J.R.); (G.H.)
- Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
6
|
Li H, Gao L, Du J, Ma T, Ye Z, Li Z. To Better Generate Organoids, What Can We Learn From Teratomas? Front Cell Dev Biol 2021; 9:700482. [PMID: 34336851 PMCID: PMC8324104 DOI: 10.3389/fcell.2021.700482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
The genomic profile of animal models is not completely matched with the genomic profile of humans, and 2D cultures do not represent the cellular heterogeneity and tissue architecture found in tissues of their origin. Derived from 3D culture systems, organoids establish a crucial bridge between 2D cell cultures and in vivo animal models. Organoids have wide and promising applications in developmental research, disease modeling, drug screening, precision therapy, and regenerative medicine. However, current organoids represent only single or partial components of a tissue, which lack blood vessels, native microenvironment, communication with near tissues, and a continuous dorsal-ventral axis within 3D culture systems. Although efforts have been made to solve these problems, unfortunately, there is no ideal method. Teratoma, which has been frequently studied in pathological conditions, was recently discovered as a new in vivo model for developmental studies. In contrast to organoids, teratomas have vascularized 3D structures and regions of complex tissue-like organization. Studies have demonstrated that teratomas can be used to mimic multilineage human development, enrich specific somatic progenitor/stem cells, and even generate brain organoids. These results provide unique opportunities to promote our understanding of the vascularization and maturation of organoids. In this review, we first summarize the basic characteristics, applications, and limitations of both organoids and teratomas and further discuss the possibility that in vivo teratoma systems can be used to promote the vascularization and maturation of organoids within an in vitro 3D culture system.
Collapse
Affiliation(s)
- Hongyu Li
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lixiong Gao
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jinlin Du
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Tianju Ma
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zi Ye
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhaohui Li
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
7
|
Dziedzicka D, Tewary M, Keller A, Tilleman L, Prochazka L, Östblom J, Couvreu De Deckersberg E, Markouli C, Franck S, Van Nieuwerburgh F, Spits C, Zandstra PW, Sermon K, Geens M. Endogenous suppression of WNT signalling in human embryonic stem cells leads to low differentiation propensity towards definitive endoderm. Sci Rep 2021; 11:6137. [PMID: 33731744 PMCID: PMC7969605 DOI: 10.1038/s41598-021-85447-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/23/2021] [Indexed: 02/06/2023] Open
Abstract
Low differentiation propensity towards a targeted lineage can significantly hamper the utility of individual human pluripotent stem cell (hPSC) lines in biomedical applications. Here, we use monolayer and micropatterned cell cultures, as well as transcriptomic profiling, to investigate how variability in signalling pathway activity between human embryonic stem cell lines affects their differentiation efficiency towards definitive endoderm (DE). We show that endogenous suppression of WNT signalling in hPSCs at the onset of differentiation prevents the switch from self-renewal to DE specification. Gene expression profiling reveals that this inefficient switch is reflected in NANOG expression dynamics. Importantly, we demonstrate that higher WNT stimulation or inhibition of the PI3K/AKT signalling can overcome the DE commitment blockage. Our findings highlight that redirection of the activity of Activin/NODAL pathway by WNT signalling towards mediating DE fate specification is a vulnerable spot, as disruption of this process can result in poor hPSC specification towards DE.
Collapse
Affiliation(s)
- Dominika Dziedzicka
- grid.8767.e0000 0001 2290 8069Research Group Reproduction and Genetics, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Mukul Tewary
- grid.17063.330000 0001 2157 2938Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3E1 Canada ,grid.13097.3c0000 0001 2322 6764Centre for Stem Cells and Regenerative Medicine, King’s College London, Guy’s Hospital, London, SE1 9RT UK
| | - Alexander Keller
- grid.8767.e0000 0001 2290 8069Research Group Reproduction and Genetics, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Laurentijn Tilleman
- grid.5342.00000 0001 2069 7798Laboratory of Pharmaceutical Biotechnology, Ghent University, 9000 Ghent, Belgium
| | - Laura Prochazka
- grid.17063.330000 0001 2157 2938Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - Joel Östblom
- grid.17063.330000 0001 2157 2938Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - Edouard Couvreu De Deckersberg
- grid.8767.e0000 0001 2290 8069Research Group Reproduction and Genetics, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Christina Markouli
- grid.8767.e0000 0001 2290 8069Research Group Reproduction and Genetics, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Silvie Franck
- grid.8767.e0000 0001 2290 8069Research Group Reproduction and Genetics, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Filip Van Nieuwerburgh
- grid.5342.00000 0001 2069 7798Laboratory of Pharmaceutical Biotechnology, Ghent University, 9000 Ghent, Belgium
| | - Claudia Spits
- grid.8767.e0000 0001 2290 8069Research Group Reproduction and Genetics, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Peter W. Zandstra
- grid.17063.330000 0001 2157 2938Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3E1 Canada ,grid.17091.3e0000 0001 2288 9830Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4 Canada ,grid.17091.3e0000 0001 2288 9830School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Karen Sermon
- grid.8767.e0000 0001 2290 8069Research Group Reproduction and Genetics, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Mieke Geens
- grid.8767.e0000 0001 2290 8069Research Group Reproduction and Genetics, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| |
Collapse
|
8
|
The Impact of DNA Methylation Dynamics on the Mutation Rate During Human Germline Development. G3-GENES GENOMES GENETICS 2020; 10:3337-3346. [PMID: 32727923 PMCID: PMC7466984 DOI: 10.1534/g3.120.401511] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
DNA methylation is a dynamic epigenetic modification found in most eukaryotic genomes. It is known to lead to a high CpG to TpG mutation rate. However, the relationship between the methylation dynamics in germline development and the germline mutation rate remains unexplored. In this study, we used whole genome bisulfite sequencing (WGBS) data of cells at 13 stages of human germline development and rare variants from the 1000 Genome Project as proxies for germline mutations to investigate the correlation between dynamic methylation levels and germline mutation rates at different scales. At the single-site level, we found a significant correlation between methylation and the germline point mutation rate at CpG sites during germline developmental stages. Then we explored the mutability of methylation dynamics in all stages. Our results also showed a broad correlation between the regional methylation level and the rate of C > T mutation at CpG sites in all genomic regions, especially in intronic regions; a similar link was also seen at all chromosomal levels. Our findings indicate that the dynamic DNA methylome during human germline development has a broader mutational impact than is commonly assumed.
Collapse
|
9
|
Abbas MN, Liang H, Kausar S, Dong Z, Cui H. Zinc finger protein RP-8, the Bombyx mori ortholog of programmed cell death 2, regulates cell proliferation. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 104:103542. [PMID: 31730828 DOI: 10.1016/j.dci.2019.103542] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/11/2019] [Accepted: 11/11/2019] [Indexed: 06/10/2023]
Abstract
Programmed cell death 2 (PDCD2) is a highly conserved eukaryotic protein indispensable for various physiological processes such as cell proliferation, development, and apoptosis. In the present study, we identified a Zinc finger protein RP-8 from the silkworm, Bombyx mori (BmZfrp8), the ortholog of PDCD2 protein. The quantitative real-time PCR analysis revealed the ubiquitous distribution of BmZfrp8 in the different tissues; however, the gene's transcription level was highest in those of the silk gland, testis, and ovary. Additionally, the expression levels of BmZfrp8 were unequal on different days of embryonic development, and it reached the highest level on the 5th day of early development. The challenge with pathogens influenced the expression level of BmZfrp8 in both hemocyte and fat body when compared with the control. Administration of 20-hydroxyecdysone significantly enhanced the BmZfrp8 expression in hemocyte. The knock-down of BmZfrp8 by double-stranded RNA suppressed the expression of developmental pathway associated genes as well as cell cycle-associated genes. Furthermore, the RNAi treated cells also showed cell cycle arrest compared to the control group. Taken together, BmZfrp8 may have a critical biological role in of B. mori, since it regulates the expression of the developmental pathway and cell cycle-associated genes.
Collapse
Affiliation(s)
- Muhammad Nadeem Abbas
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Beibei, Chongqing, 400715, China; Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing, 400715, China.
| | - Hanghua Liang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Beibei, Chongqing, 400715, China; Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing, 400715, China.
| | - Saima Kausar
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Beibei, Chongqing, 400715, China; Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing, 400715, China.
| | - Zhen Dong
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Beibei, Chongqing, 400715, China; Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing, 400715, China.
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Beibei, Chongqing, 400715, China; Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing, 400715, China.
| |
Collapse
|
10
|
Phelps HM, Pierce JM, Murphy AJ, Correa H, Qian J, Massion PP, Lovvorn HN. FXR1 expression domain in Wilms tumor. J Pediatr Surg 2019; 54:1198-1205. [PMID: 30894247 PMCID: PMC6545243 DOI: 10.1016/j.jpedsurg.2019.02.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 02/21/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND/PURPOSE Wilms tumor (WT) is the most common childhood kidney cancer globally. Our prior unbiased proteomic screen of WT disparities revealed increased expression of Fragile X-Related 1 (FXR1) in Kenyan specimens where survival is dismal. FXR1 is an RNA-binding protein that associates with poor outcomes in multiple adult cancers. The aim of this study therefore was to validate and characterize the FXR1 expression domain in WT. METHODS Quantitative FXR1 gene expression was compared between WT, adjacent, adult, and fetal kidney specimens. The cellular and subcellular expression domain of FXR1 was characterized across these tissues using immunoperoxidase staining. RNA-sequencing of FXR1 was performed from WT and other pediatric malignancies to examine its broader target potential. RESULTS FXR1 was detected in all clinical WT specimens evaluated (n = 82), and as a result appeared independent of demographic, histology, or adverse event. Specific cytosolic staining was strongest in blastema, intermediate and variable in epithelia, and weakest in stroma. When present, areas of skeletal muscle differentiation stained strongly for FXR1. qPCR revealed increased FXR1 expression in WT compared to adult and adjacent kidney (p < 0.0002) but was similar to fetal kidney (p = 0.648). RNA-sequencing revealed expression of FXR1 in multiple pediatric tumors, greatest in rhabdomyosarcoma and WT. CONCLUSIONS FXR1 was expressed consistently across this broad sampling of WT and most robustly in the primitive blastema. Notably, FXR1 labeled a specific self-renewing progenitor population of the fetal kidney.
Collapse
Affiliation(s)
| | - Janene M. Pierce
- Vanderbilt University Medical Center, Department of Pediatric Surgery, Nashville, TN
| | - Andrew J. Murphy
- St. Jude Children’s Research Hospital, Department of Surgery, Memphis, TN
| | - Hernan Correa
- Vanderbilt University Medical Center, Department of Pathology, Microbiology, and Immunology, Nashville, TN
| | - Jun Qian
- Vanderbilt University Medical Center, Department of Medicine and Vanderbilt Ingram Cancer Center, Nashville, TN
| | - Pierre P. Massion
- Vanderbilt University Medical Center, Department of Medicine and Vanderbilt Ingram Cancer Center, Nashville, TN
| | - Harold N. Lovvorn
- Vanderbilt University Medical Center, Department of Pediatric Surgery, Nashville, TN
| |
Collapse
|
11
|
Harkness L, Chen X, Gillard M, Gray PP, Davies AM. Media composition modulates human embryonic stem cell morphology and may influence preferential lineage differentiation potential. PLoS One 2019; 14:e0213678. [PMID: 30889226 PMCID: PMC6424453 DOI: 10.1371/journal.pone.0213678] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 02/26/2019] [Indexed: 12/22/2022] Open
Abstract
Undifferentiated human embryonic stem cells have a distinct morphology (hESC). Changes in cell morphology during culture can be indicative of differentiation. hESC, maintained in diverse medias, demonstrated alterations in morphological parameters and subsequent alterations in underlying transcript expression and lineage differentiation. Analysis of morphological parameters showed distinct and significant differences between the undefined, less defined and Xeno-free medias while still maintaining pluripotency markers. This suggested that the less defined media may be creating dynamic instability in the cytoskeleton, with the cytoskeleton becoming more stabilised in the Xeno-free media as demonstrated by smaller and rounder cells. Examination of early lineage markers during undirected differentiation using d5 embryoid bodies demonstrated increased mesodermal lineage preference as compared to endodermal or ectoderm in cells originally cultured in Xeno-free media. Undefined media showed preference for mesoderm and ectoderm lineages, while less defined media (BSA present) demonstrated no preference. These data reveal that culture media may produce fundamental changes in cell morphology which are reflected in early lineage differentiation choice.
Collapse
Affiliation(s)
- Linda Harkness
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Australia
- * E-mail:
| | - Xiaoli Chen
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Australia
| | - Marianne Gillard
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Australia
| | - Peter Paul Gray
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Australia
| | - Anthony Mitchell Davies
- Translational Cell Imaging Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
12
|
Abstract
The very small embryonic-like stem cells (VSELs) are known as a subset of adult pluripotent stem cells able to differentiate to all three germ layers. However, their small number and quiescence restrict the possibility of their use in cell therapy. In the present study, we first delineate different subpopulation of VSELs from human cord blood CD34+ cells to define their purity. We next determine genes expression levels in the whole transcriptome of VSELs expressing the pluripotent marker NANOG and control cells under the steady state condition. We found that more than a thousand of genes are downregulated in VSELs, as well as many membrane receptors, cells signaling molecules and CDKs mRNAs. In addition, we observed discordance in some pluripotent genes expression levels with embryonic stem cells (ESCs), which could explain VSELs quiescence. We then evaluate VSELs capacity to expand and differentiate in vitro in specific and appropriate media. After 12 days culture in specific medium containing a pyrimidoindole derivative (UM171), VSELs were significantly expanded for the first time without feeder cells and importantly preserve their capacities to differentiate into hematopoietic and endothelial cells. Interestingly, this stimulation of VSELs self-renewal restores the expression of some downregulated genes known as key regulators of cell proliferation and differentiation. The properties of such pluripotent expanded cells make them a potential candidate in regenerative medicine.
Collapse
|
13
|
Farhadian M, Rafat SA, Hasanpur K, Ebrahimi M, Ebrahimie E. Cross-Species Meta-Analysis of Transcriptomic Data in Combination With Supervised Machine Learning Models Identifies the Common Gene Signature of Lactation Process. Front Genet 2018; 9:235. [PMID: 30050559 PMCID: PMC6052129 DOI: 10.3389/fgene.2018.00235] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 06/13/2018] [Indexed: 01/13/2023] Open
Abstract
Lactation, a physiologically complex process, takes place in mammary gland after parturition. The expression profile of the effective genes in lactation has not comprehensively been elucidated. Herein, meta-analysis, using publicly available microarray data, was conducted identify the differentially expressed genes (DEGs) between pre- and post-peak milk production. Three microarray datasets of Rat, Bos Taurus, and Tammar wallaby were used. Samples related to pre-peak (n = 85) and post-peak (n = 24) milk production were selected. Meta-analysis revealed 31 DEGs across the studied species. Interestingly, 10 genes, including MRPS18B, SF1, UQCRC1, NUCB1, RNF126, ADSL, TNNC1, FIS1, HES5 and THTPA, were not detected in original studies that highlights meta-analysis power in biosignature discovery. Common target and regulator analysis highlighted the high connectivity of CTNNB1, CDD4 and LPL as gene network hubs. As data originally came from three different species, to check the effects of heterogeneous data sources on DEGs, 10 attribute weighting (machine learning) algorithms were applied. Attribute weighting results showed that the type of organism had no or little effect on the selected gene list. Systems biology analysis suggested that these DEGs affect the milk production by improving the immune system performance and mammary cell growth. This is the first study employing both meta-analysis and machine learning approaches for comparative analysis of gene expression pattern of mammary glands in two important time points of lactation process. The finding may pave the way to use of publically available to elucidate the underlying molecular mechanisms of physiologically complex traits such as lactation in mammals.
Collapse
Affiliation(s)
- Mohammad Farhadian
- Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Seyed A Rafat
- Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Karim Hasanpur
- Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | | | - Esmaeil Ebrahimie
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia.,Institute of Biotechnology, Shiraz University, Shiraz, Iran.,Division of Information Technology, Engineering and the Environment, School of Information Technology & Mathematical Sciences, University of South Australia, Adelaide, SA, Australia.,School of Biological Sciences, Faculty of Science and Engineering, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
14
|
Transcriptome variations among human embryonic stem cell lines are associated with their differentiation propensity. PLoS One 2018; 13:e0192625. [PMID: 29444173 PMCID: PMC5812638 DOI: 10.1371/journal.pone.0192625] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/26/2018] [Indexed: 12/20/2022] Open
Abstract
Human embryonic stem cells (hESCs) have the potential to form any cell type in the body, making them attractive cell sources in drug screening, regenerative medicine, disease and developmental processes modeling. However, not all hESC lines have the equal potency to generate desired cell types in vitro. Significant variations have been observed for the differentiation efficiency of various human ESC lines. The precise underpinning molecular mechanisms are still unclear. In this work, we compared transcriptome variations of four hESC lines H7, HUES1, HUES8 and HUES9. We found that hESC lines have different gene expression profiles, and these differentially expressed genes (DEGs) are significantly enriched in developmental processes, such as ectodermal, mesodermal and endodermal development. The enrichment difference between hESC lines was consistent with its lineage bias. Among these DEGs, some pluripotency factors and genes involved in signaling transduction showed great variations as well. The pleiotropic functions of these genes in controlling hESC identity and early lineage specification, implicated that different hESC lines may utilize distinct balance mechanisms to maintain pluripotent state. When the balance is broken in a certain environment, gene expression variation between them could impact on their different lineage specification behavior.
Collapse
|
15
|
(Re-)programming of subtype specific cardiomyocytes. Adv Drug Deliv Rev 2017; 120:142-167. [PMID: 28916499 DOI: 10.1016/j.addr.2017.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/29/2017] [Accepted: 09/07/2017] [Indexed: 01/10/2023]
Abstract
Adult cardiomyocytes (CMs) possess a highly restricted intrinsic regenerative potential - a major barrier to the effective treatment of a range of chronic degenerative cardiac disorders characterized by cellular loss and/or irreversible dysfunction and which underlies the majority of deaths in developed countries. Both stem cell programming and direct cell reprogramming hold promise as novel, potentially curative approaches to address this therapeutic challenge. The advent of induced pluripotent stem cells (iPSCs) has introduced a second pluripotent stem cell source besides embryonic stem cells (ESCs), enabling even autologous cardiomyocyte production. In addition, the recent achievement of directly reprogramming somatic cells into cardiomyocytes is likely to become of great importance. In either case, different clinical scenarios will require the generation of highly pure, specific cardiac cellular-subtypes. In this review, we discuss these themes as related to the cardiovascular stem cell and programming field, including a focus on the emergent topic of pacemaker cell generation for the development of biological pacemakers and in vitro drug testing.
Collapse
|
16
|
Skelton RJP, Kamp TJ, Elliott DA, Ardehali R. Biomarkers of Human Pluripotent Stem Cell-Derived Cardiac Lineages. Trends Mol Med 2017; 23:651-668. [PMID: 28576602 DOI: 10.1016/j.molmed.2017.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 04/24/2017] [Accepted: 05/04/2017] [Indexed: 02/07/2023]
Abstract
Human pluripotent stem cells (hPSCs) offer a practical source for the de novo generation of cardiac tissues and a unique opportunity to investigate cardiovascular lineage commitment. Numerous strategies have focused on the in vitro production of cardiomyocytes, smooth muscle, and endothelium from hPSCs. However, these differentiation protocols often yield undesired cell types. Thus, establishing a set of stage-specific markers for pure cardiac subpopulations will assist in defining the hierarchy of cardiac differentiation, aid in the development of cellular therapy, and facilitate drug screening and disease modeling. The recent characterization of many such markers is enabling the isolation of major cardiac lineages and subpopulations from differentiating hPSCs. We provide here a comprehensive review detailing the suite of biomarkers used to differentiate cardiac lineages from mixed hPSC-derived populations.
Collapse
Affiliation(s)
- Rhys J P Skelton
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA
| | - Timothy J Kamp
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - David A Elliott
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
17
|
Zhao Z, Ma Y, Chen Z, Liu Q, Li Q, Kong D, Yuan K, Hu L, Wang T, Chen X, Peng Y, Jiang W, Yu Y, Liu X. Effects of Feeder Cells on Dopaminergic Differentiation of Human Embryonic Stem Cells. Front Cell Neurosci 2016; 10:291. [PMID: 28066186 PMCID: PMC5168467 DOI: 10.3389/fncel.2016.00291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 12/05/2016] [Indexed: 01/30/2023] Open
Abstract
Mouse embryonic fibroblasts (MEFs) and human foreskin fibroblasts (HFFs) are used for the culture of human embryonic stem cells (hESCs). MEFs and HFFs differed in their capacity to support the proliferation and pluripotency of hESCs and could affect cardiac differentiation potential of hESCs. The aim of this study was to evaluate the effect of MEFs and HFFs feeders on dopaminergic differentiation of hESCs lines. To minimize the impact of culture condition variation, two hESCs lines were cultured on mixed feeder cells (MFCs, MEFs: HFFs = 1:1) and HFFs feeder, respectively, and then were differentiated into dopaminergic (DA) neurons under the identical protocol. Dopaminergic differentiation was evaluated by immunocytochemistry, quantitative fluorescent real-time PCR, transmission and scanning electron microscopy, and patch clamp. Our results demonstrated that these hESCs-derived neurons were genuine and functional DA neurons. However, compared to hESCs line on MFCs feeder, hESCs line on HFFs feeder had a higher proportion of tyrosine hydroxylase (TH) positive cells and expressed higher levels of FOXA2, PITX3, NURR1, and TH genes. In addition, the values of threshold intensity and threshold membrane potential of DA neurons from hESCs line on HFFs feeder were lower than those of DA neurons from hESCs line on the MFCs feeder. In conclusion, HFFs feeder not only facilitated the differentiation of hESCs cells into dopaminergic neurons, but also induced hESCs-derived DA neurons to express higher electrophysiological excitability. Therefore, feeder cells could affect not only dopaminergic differentiation potential of different hESCs lines, but also electrophysiological properties of hESCs-derived DA neurons.
Collapse
Affiliation(s)
- Zhenqiang Zhao
- Department of Neurology, Jinling Hospital, Southern Medical UniversityNanjing, China; Department of Neurology, First Affiliated Hospital, Hainan Medical UniversityHaikou, China
| | - Yanlin Ma
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical UniversityGuangzhou, China; Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Reproductive Medical Center, First Affiliated Hospital, Hainan Medical UniversityHaikou, China
| | - Zhibin Chen
- Department of Neurology, First Affiliated Hospital, Hainan Medical University Haikou, China
| | - Qian Liu
- Department of Neurology, Jinling Hospital, Southern Medical University Nanjing, China
| | - Qi Li
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Reproductive Medical Center, First Affiliated Hospital, Hainan Medical University Haikou, China
| | - Deyan Kong
- Department of Neurology, Jinling Hospital, Southern Medical UniversityNanjing, China; Department of Neurology, Affiliated Ruikang Hospital, Guangxi Traditional Chinese Medical UniversityNanning, China
| | - Kunxiong Yuan
- Department of Neurology, Jinling Hospital, Southern Medical UniversityNanjing, China; Department of Neurology, Central HospitalShenzhen, China
| | - Lan Hu
- Department of Laboratory Medicines, First Affiliated Hospital, Hainan Medical University Haikou, China
| | - Tan Wang
- Department of Neurology, First Affiliated Hospital, Hainan Medical University Haikou, China
| | - Xiaowu Chen
- Department of Neurology, First Affiliated Hospital, Hainan Medical University Haikou, China
| | - Yanan Peng
- Department of Neurology, First Affiliated Hospital, Hainan Medical University Haikou, China
| | - Weimin Jiang
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Reproductive Medical Center, First Affiliated Hospital, Hainan Medical University Haikou, China
| | - Yanhong Yu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University Guangzhou, China
| | - Xinfeng Liu
- Department of Neurology, Jinling Hospital, Southern Medical University Nanjing, China
| |
Collapse
|
18
|
Ohyashiki K, Umezu T, Katagiri S, Kobayashi C, Azuma K, Tauchi T, Okabe S, Fukuoka Y, Ohyashiki JH. Downregulation of Plasma miR-215 in Chronic Myeloid Leukemia Patients with Successful Discontinuation of Imatinib. Int J Mol Sci 2016; 17:570. [PMID: 27092489 PMCID: PMC4849026 DOI: 10.3390/ijms17040570] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/14/2016] [Accepted: 04/08/2016] [Indexed: 12/11/2022] Open
Abstract
Approximately 40% of chronic myeloid leukemia (CML) patients who discontinue imatinib (IM) therapy maintain undetectable minimal residual disease (UMRD) for more than one year (stopping IM (STOP-IM)). To determine a possible biomarker for STOP-IM CML, we examined plasma miRNA expression in CML patients who were able to discontinue IM. We first screened candidate miRNAs in unselected STOP-IM patients, who had sustained UMRD after discontinuing IM for more than six months, in comparison with healthy volunteers, by using a TaqMan low-density array for plasma or exosomes. Exosomal miR-215 and plasma miR-215 were downregulated in the STOP-IM group compared to the control, indicating that the biological relevance of the plasma miR-215 level is equivalent to that of the exosomal level. Next, we performed real-time quantitative RT-PCR in 20 STOP-IM patients, 32 patients with UMRD on continued IM therapy (IM group) and 28 healthy volunteers. The plasma miRNA-215 level was significantly downregulated in the STOP-IM group (p < 0.0001); we determined the cut-off level and divided the IM group patients into two groups according to whether the plasma miR-215 was downregulated or not. The IM group patients with a low plasma miR-215 level had a significantly higher total IM intake, compared to the patients with elevated miR-215 levels (p = 0.0229). Functional annotation of miR-215 target genes estimated by the Database for Annotation, Visualization and Integrated Discovery (DAVID) bioinformatic tools involved cell cycle, mitosis, DNA repair and cell cycle checkpoint. Our study suggests a possible role of miR-215 in successful IM discontinuation.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Down-Regulation/drug effects
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Imatinib Mesylate/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- MicroRNAs/blood
- MicroRNAs/genetics
Collapse
Affiliation(s)
- Kazuma Ohyashiki
- Department of Hematology, Tokyo Medical University, Tokyo 160-0023, Japan.
- Department of Molecular Science, Tokyo Medical University, Tokyo 160-0023, Japan.
| | - Tomohiro Umezu
- Department of Molecular Science, Tokyo Medical University, Tokyo 160-0023, Japan.
| | - Seiichiro Katagiri
- Department of Hematology, Tokyo Medical University, Tokyo 160-0023, Japan.
| | - Chiaki Kobayashi
- Department of Molecular Science, Tokyo Medical University, Tokyo 160-0023, Japan.
| | - Kenko Azuma
- Department of Molecular Oncology, Institute of Medical Science, Tokyo Medical University, Tokyo 160-0023, Japan.
| | - Tetsuzo Tauchi
- Department of Hematology, Tokyo Medical University, Tokyo 160-0023, Japan.
| | - Seiichi Okabe
- Department of Hematology, Tokyo Medical University, Tokyo 160-0023, Japan.
| | - Yutaka Fukuoka
- Department of Electrical Engineering, Kogakuin University, Tokyo 163-8677, Japan.
| | - Junko H Ohyashiki
- Department of Molecular Oncology, Institute of Medical Science, Tokyo Medical University, Tokyo 160-0023, Japan.
| |
Collapse
|
19
|
Ovando-Roche P, Yu JSL, Testori S, Ho C, Cui W. TRF2-mediated stabilization of hREST4 is critical for the differentiation and maintenance of neural progenitors. Stem Cells 2015; 32:2111-22. [PMID: 24740933 DOI: 10.1002/stem.1725] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 03/08/2014] [Accepted: 03/16/2014] [Indexed: 12/25/2022]
Abstract
Telomere repeat binding factor 2 (TRF2) is a component of the shelterin complex that is known to bind and protect telomeric DNA, yet the detection of TRF2 in extra-telomeric regions of chromosomes suggests other roles for TRF2 besides telomere protection. Here, we demonstrate that TRF2 plays a critical role in antagonizing the repressive function of neuron-restrictive silencer factor, also known as repressor element-1 silencing transcription factor (REST), during the neural differentiation of human embryonic stem cells (hESCs) by enhancing the expression of a truncated REST splice isoform we term human REST4 (hREST4) due to its similarity to rodent REST4. We show that TRF2 is specifically upregulated during hESC neural differentiation concordantly with an increase in the expression of hREST4 and that both proteins are highly expressed in NPCs. Overexpression of TRF2 in hESCs increases hREST4 levels and induces their neural differentiation, whereas TRF2 knockdown in hESCs and NPCs reduces hREST4 expression, hindering their ability to differentiate to the neural lineage. Concurrently, we show that TRF2 directly interacts with the C-terminal of hREST4 through its TRF2 core binding motif [F/Y]xL, protecting hREST4 from ubiquitin-mediated proteasomal degradation and consequently furthering neural induction. Thus, the TRF2-mediated counterbalance between hREST4 and REST is vital for both the generation and maintenance of NPCs, suggesting an important role for TRF2 in both neurogenesis and function of the central nervous system.
Collapse
Affiliation(s)
- Patrick Ovando-Roche
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Campus, London, United Kingdom
| | | | | | | | | |
Collapse
|
20
|
Girlovanu M, Susman S, Soritau O, Rus-Ciuca D, Melincovici C, Constantin AM, Mihu CM. Stem cells - biological update and cell therapy progress. ACTA ACUST UNITED AC 2015; 88:265-71. [PMID: 26609255 PMCID: PMC4632881 DOI: 10.15386/cjmed-483] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/12/2015] [Indexed: 12/17/2022]
Abstract
In recent years, the advances in stem cell research have suggested that the human body may have a higher plasticity than it was originally expected. Until now, four categories of stem cells were isolated and cultured in vivo: embryonic stem cells, fetal stem cells, adult stem cells and induced pluripotent stem cells (hiPSCs). Although multiple studies were published, several issues concerning the stem cells are still debated, such as: the molecular mechanisms of differentiation, the methods to prevent teratoma formation or the ethical and religious issues regarding especially the embryonic stem cell research. The direct differentiation of stem cells into specialized cells: cardiac myocytes, neural cells, pancreatic islets cells, may represent an option in treating incurable diseases such as: neurodegenerative diseases, type I diabetes, hematologic or cardiac diseases. Nevertheless, stem cell-based therapies, based on stem cell transplantation, remain mainly at the experimental stages and their major limitation is the development of teratoma and cancer after transplantation. The induced pluripotent stem cells (hiPSCs) represent a prime candidate for future cell therapy research because of their significant self-renewal and differentiation potential and the lack of ethical issues. This article presents an overview of the biological advances in the study of stem cells and the current progress made in the field of regenerative medicine.
Collapse
Affiliation(s)
- Mihai Girlovanu
- Morphological Sciences Department 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Sergiu Susman
- Morphological Sciences Department 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Olga Soritau
- Research Department, Prof. Dr. I. Chiricuta Oncology Institute, Cluj-Napoca, Romania
| | - Dan Rus-Ciuca
- Department of Pathology, Karlstad Central Hospital, Sweden
| | - Carmen Melincovici
- Morphological Sciences Department 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Anne-Marie Constantin
- Morphological Sciences Department 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Carmen Mihaela Mihu
- Morphological Sciences Department 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
21
|
Alajem A, Biran A, Harikumar A, Sailaja BS, Aaronson Y, Livyatan I, Nissim-Rafinia M, Sommer AG, Mostoslavsky G, Gerbasi VR, Golden DE, Datta A, Sze SK, Meshorer E. Differential association of chromatin proteins identifies BAF60a/SMARCD1 as a regulator of embryonic stem cell differentiation. Cell Rep 2015; 10:2019-31. [PMID: 25818293 DOI: 10.1016/j.celrep.2015.02.064] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 01/09/2015] [Accepted: 02/25/2015] [Indexed: 11/19/2022] Open
Abstract
Embryonic stem cells (ESCs) possess a distinct chromatin conformation maintained by specialized chromatin proteins. To identify chromatin regulators in ESCs, we developed a simple biochemical assay named D-CAP (differential chromatin-associated proteins), using brief micrococcal nuclease digestion of chromatin, followed by liquid chromatography tandem mass spectrometry (LC-MS/MS). Using D-CAP, we identified several differentially chromatin-associated proteins between undifferentiated and differentiated ESCs, including the chromatin remodeling protein SMARCD1. SMARCD1 depletion in ESCs led to altered chromatin and enhanced endodermal differentiation. Gene expression and chromatin immunoprecipitation sequencing (ChIP-seq) analyses suggested that SMARCD1 is both an activator and a repressor and is enriched at developmental regulators and that its chromatin binding coincides with H3K27me3. SMARCD1 knockdown caused H3K27me3 redistribution and increased H3K4me3 around the transcription start site (TSS). One of the identified SMARCD1 targets was Klf4. In SMARCD1-knockdown clones, KLF4, as well as H3K4me3 at the Klf4 locus, remained high and H3K27me3 was abolished. These results propose a role for SMARCD1 in restricting pluripotency and activating lineage pathways by regulating H3K27 methylation.
Collapse
Affiliation(s)
- Adi Alajem
- Department of Genetics, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Alva Biran
- Department of Genetics, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Arigela Harikumar
- Department of Genetics, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Badi Sri Sailaja
- Department of Genetics, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Yair Aaronson
- Department of Genetics, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Ilana Livyatan
- Department of Genetics, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Malka Nissim-Rafinia
- Department of Genetics, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Andreia Gianotti Sommer
- Section of Gastroenterology, Department of Medicine, Center for Regenerative Medicine (CReM), Boston University School of Medicine, 670 Albany Street, Suite 209, Boston, MA 02118, USA
| | - Gustavo Mostoslavsky
- Section of Gastroenterology, Department of Medicine, Center for Regenerative Medicine (CReM), Boston University School of Medicine, 670 Albany Street, Suite 209, Boston, MA 02118, USA
| | - Vincent R Gerbasi
- Department of Molecular Biosciences, Northwestern University, 2205 Tech Drive, Evanston, IL 60208, USA; Naval Medical Research Center, Silver Spring, MD 20910, USA
| | | | - Arnab Datta
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Eran Meshorer
- Department of Genetics, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel; The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel.
| |
Collapse
|
22
|
The L1TD1 protein interactome reveals the importance of post-transcriptional regulation in human pluripotency. Stem Cell Reports 2015; 4:519-28. [PMID: 25702638 PMCID: PMC4376047 DOI: 10.1016/j.stemcr.2015.01.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 01/16/2015] [Accepted: 01/16/2015] [Indexed: 11/24/2022] Open
Abstract
The RNA-binding protein L1TD1 is one of the most specific and abundant proteins in pluripotent stem cells and is essential for the maintenance of pluripotency in human cells. Here, we identify the protein interaction network of L1TD1 in human embryonic stem cells (hESCs) and provide insights into the interactome network constructed in human pluripotent cells. Our data reveal that L1TD1 has an important role in RNA splicing, translation, protein traffic, and degradation. L1TD1 interacts with multiple stem-cell-specific proteins, many of which are still uncharacterized in the context of development. Further, we show that L1TD1 is a part of the pluripotency interactome network of OCT4, SOX2, and NANOG, bridging nuclear and cytoplasmic regulation and highlighting the importance of RNA biology in pluripotency. A protein interactome is constructed in human embryonic stem cells L1TD1 interacts with U2af1 and Srsf3, which are vital for somatic cell reprogramming L1TD1 is involved in the high proteasome activity of pluripotent cells L1TD1 connects uncharacterized factors to the OCT4, SOX2, and NANOG network
Collapse
|
23
|
Zhang J, Wei W, Jin HC, Ying RC, Zhu AK, Zhang FJ. Programmed cell death 2 protein induces gastric cancer cell growth arrest at the early S phase of the cell cycle and apoptosis in a p53-dependent manner. Oncol Rep 2014; 33:103-10. [PMID: 25334010 DOI: 10.3892/or.2014.3551] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 08/02/2014] [Indexed: 11/06/2022] Open
Abstract
Programmed cell death 2 (PDCD2) is a highly conserved nuclear protein, and aberrant PDCD2 expression alters cell apoptosis. The present study aimed to investigate PDCD2 expression in gastric cancer. Tissue specimens from 34 gastric cancer patients were collected for analysis of PDCD2 expression using immunohistochemistry, western blotting and qRT-PCR. Gastric cancer cell lines (a p53-mutated MKN28 line and a wild-type p53 MKN45 line) were used to assess the effects of PDCD2 overexpression. p53-/- nude mice were used to investigate the effect of PDCD2 on ultraviolet B (UVB)-induced skin carcinogenesis. The data showed that PDCD2 expression was reduced in gastric cancer tissue specimens, and loss of PDCD2 expression was associated with the poor survival of patients. PDCD2 expression induced gastric cancer cell growth arrest at the early S phase of the cell cycle and apoptosis. The antitumor effects of PDCD2 expression were dependent on p53 expression in gastric cancer cells. Moreover, PDCD2 expression inhibited activity of the ATM/Chk1/2/p53 signaling pathway. In addition, PDCD2 expression suppressed UVB-induced skin carcinogenesis in p53+/+ nude mice, but not in p53-/- mice. The data from the present study demonstrated that loss of PDCD2 expression could contribute to gastric cancer development and progression and that PDCD2-induced gastric cancer cell growth arrest at the early S phase of the cell cycle and apoptosis are p53-dependent.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| | - Wei Wei
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| | - Hui-Cheng Jin
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| | - Rong-Chao Ying
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| | - A-Kao Zhu
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| | - Fang-Jie Zhang
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| |
Collapse
|
24
|
Sterthaus O, Feutz AC, Zhang H, Pletscher F, Bruder E, Miny P, Lezzi G, De Geyter M, De Geyter C. Gene expression profiles of similarly derived human embryonic stem cell lines correlate with their distinct propensity to exit stemness and their different differentiation behavior in culture. Cell Reprogram 2014; 16:185-95. [PMID: 24811852 DOI: 10.1089/cell.2013.0089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Four normal-karyotype human embryonic stem cell (hESC) lines were generated using the same protocol and maintained under identical conditions. Despite these precautions, gene expression patterns were found to be dissimilar among the four lines. The observed differences were typical of each cell line, correlated with their distinct propensity to exit stemness, created heterogeneity among the cells during cell line maintenance, and correlated with their altered capacity as a source of differentiated cells. The capacity of some cell lines to give rise to more, and more mature, neurons within comparable time frames of directed differentiation reflected the distinct proportions of cells already predifferentiated at the onset. These findings demonstrate that the subsequent stages of neural differentiation were altered both in a quantitative and timely fashion. As a consequence, cell lines with apparent better and quicker ability to produce neurons were actually the less capable of reproducing proper differentiation. Previous data suggested that cell lines able to generate more neurons faster would be more suitable to clinical application. Our analysis of the differentiation process strongly suggests the opposite. The spontaneous tendency to predifferentiate of any particular hESC line should be known because it clearly impacts further experimental results.
Collapse
Affiliation(s)
- Oliver Sterthaus
- 1 Clinic of Gynecological Endocrinology and Reproductive Medicine, University of Basel , CH-4031, Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Jones GN, Moschidou D, Abdulrazzak H, Kalirai BS, Vanleene M, Osatis S, Shefelbine SJ, Horwood NJ, Marenzana M, De Coppi P, Bassett JD, Williams GR, Fisk NM, Guillot PV. Potential of human fetal chorionic stem cells for the treatment of osteogenesis imperfecta. Stem Cells Dev 2014; 23:262-76. [PMID: 24028330 PMCID: PMC3904514 DOI: 10.1089/scd.2013.0132] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 09/12/2013] [Indexed: 12/13/2022] Open
Abstract
Osteogenesis imperfecta (OI) is a genetic bone pathology with prenatal onset, characterized by brittle bones in response to abnormal collagen composition. There is presently no cure for OI. We previously showed that human first trimester fetal blood mesenchymal stem cells (MSCs) transplanted into a murine OI model (oim mice) improved the phenotype. However, the clinical use of fetal MSC is constrained by their limited number and low availability. In contrast, human fetal early chorionic stem cells (e-CSC) can be used without ethical restrictions and isolated in high numbers from the placenta during ongoing pregnancy. Here, we show that intraperitoneal injection of e-CSC in oim neonates reduced fractures, increased bone ductility and bone volume (BV), increased the numbers of hypertrophic chondrocytes, and upregulated endogenous genes involved in endochondral and intramembranous ossification. Exogenous cells preferentially homed to long bone epiphyses, expressed osteoblast genes, and produced collagen COL1A2. Together, our data suggest that exogenous cells decrease bone brittleness and BV by directly differentiating to osteoblasts and indirectly stimulating host chondrogenesis and osteogenesis. In conclusion, the placenta is a practical source of stem cells for the treatment of OI.
Collapse
Affiliation(s)
- Gemma N. Jones
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Dafni Moschidou
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Hassan Abdulrazzak
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Bhalraj Singh Kalirai
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Maximilien Vanleene
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Suchaya Osatis
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | | | - Nicole J. Horwood
- Kennedy Institute of Rheumatology, Imperial College London, London, United Kingdom
| | - Massimo Marenzana
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Paolo De Coppi
- Surgery Unit, UCL Institute of Child Health, London, United Kingdom
| | - J.H. Duncan Bassett
- Molecular Endocrinology Group, Department of Medicine, Imperial College London, London, United Kingdom
| | - Graham R. Williams
- Molecular Endocrinology Group, Department of Medicine, Imperial College London, London, United Kingdom
| | - Nicholas M. Fisk
- UQ Centre for Clinical Research, University of Queensland, Brisbane, Australia
| | - Pascale V. Guillot
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| |
Collapse
|
26
|
Hosseinpour B, Bakhtiarizadeh MR, Khosravi P, Ebrahimie E. Predicting distinct organization of transcription factor binding sites on the promoter regions: a new genome-based approach to expand human embryonic stem cell regulatory network. Gene 2013; 531:212-9. [DOI: 10.1016/j.gene.2013.09.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 09/01/2013] [Accepted: 09/04/2013] [Indexed: 12/23/2022]
|
27
|
Ting S, Lecina M, Chan YC, Tse HF, Reuveny S, Oh SKW. Nutrient supplemented serum-free medium increases cardiomyogenesis efficiency of human pluripotent stem cells. World J Stem Cells 2013; 5:86-97. [PMID: 23904910 PMCID: PMC3723792 DOI: 10.4252/wjsc.v5.i3.86] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 05/07/2013] [Accepted: 06/10/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To develop an improved p38 MAPK inhibitor-based serum-free medium for embryoid body cardiomyocyte differentiation of human pluripotent stem cells.
METHODS: Human embryonic stem cells (hESC) differentiated to cardiomyocytes (CM) using a p38 MAPK inhibitor (SB203580) based serum-free medium (SB media). Nutrient supplements known to increase cell viability were added to SB medium. The ability of these supplements to improve cardiomyogenesis was evaluated by measurements of cell viability, total cell count, and the expression of cardiac markers via flow cytometry. An improved medium containing Soy hydrolysate (HySoy) and bovine serum albumin (BSA) (SupSB media) was developed and tested on 2 additional cell lines (H1 and Siu-hiPSC). Characterization of the cardiomyocytes was done by immunohistochemistry, electrophysiology and quantitative real-time reverse transcription-polymerase chain reaction.
RESULTS: hESC cell line, HES-3, differentiating in SB medium for 16 d resulted in a cardiomyocyte yield of 0.07 ± 0.03 CM/hESC. A new medium (SupSB media) was developed with the addition of HySoy and BSA to SB medium. This medium resulted in 2.6 fold increase in cardiomyocyte yield (0.21 ± 0.08 CM/hESC). The robustness of SupSB medium was further demonstrated using two additional pluripotent cell lines (H1, hESC and Siu1, hiPSC), showing a 15 and 9 fold increase in cardiomyocyte yield respectively. The age (passage number) of the pluripotent cells did not affect the cardiomyocyte yields. Embryoid body (EB) cardiomyocytes formed in SupSB medium expressed canonical cardiac markers (sarcomeric α-actinin, myosin heavy chain and troponin-T) and demonstrated all three major phenotypes: nodal-, atrial- and ventricular-like. Electrophysiological characteristics (maximum diastolic potentials and action potential durations) of cardiomyocytes derived from SB and SupSB media were similar.
CONCLUSION: The nutrient supplementation (HySoy and BSA) leads to increase in cell viability, cell yield and cardiac marker expression during cardiomyocyte differentiation, translating to an overall increase in cardiomyocyte yield.
Collapse
|
28
|
Calloni R, Cordero EAA, Henriques JAP, Bonatto D. Reviewing and updating the major molecular markers for stem cells. Stem Cells Dev 2013; 22:1455-76. [PMID: 23336433 PMCID: PMC3629778 DOI: 10.1089/scd.2012.0637] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Stem cells (SC) are able to self-renew and to differentiate into many types of committed cells, making SCs interesting for cellular therapy. However, the pool of SCs in vivo and in vitro consists of a mix of cells at several stages of differentiation, making it difficult to obtain a homogeneous population of SCs for research. Therefore, it is important to isolate and characterize unambiguous molecular markers that can be applied to SCs. Here, we review classical and new candidate molecular markers that have been established to show a molecular profile for human embryonic stem cells (hESCs), mesenchymal stem cells (MSCs), and hematopoietic stem cells (HSCs). The commonly cited markers for embryonic ESCs are Nanog, Oct-4, Sox-2, Rex-1, Dnmt3b, Lin-28, Tdgf1, FoxD3, Tert, Utf-1, Gal, Cx43, Gdf3, Gtcm1, Terf1, Terf2, Lefty A, and Lefty B. MSCs are primarily identified by the expression of CD13, CD29, CD44, CD49e, CD54, CD71, CD73, CD90, CD105, CD106, CD166, and HLA-ABC and lack CD14, CD31, CD34, CD45, CD62E, CD62L, CD62P, and HLA-DR expression. HSCs are mainly isolated based on the expression of CD34, but the combination of this marker with CD133 and CD90, together with a lack of CD38 and other lineage markers, provides the most homogeneous pool of SCs. Here, we present new and alternative markers for SCs, along with microRNA profiles, for these cells.
Collapse
Affiliation(s)
- Raquel Calloni
- Departamento de Biologia Molecular e Biotecnologia, Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | | | | | | |
Collapse
|
29
|
Human neural stem/progenitor cells derived from embryonic stem cells and fetal nervous system present differences in immunogenicity and immunomodulatory potentials in vitro. Stem Cell Res 2013; 10:325-37. [PMID: 23416350 DOI: 10.1016/j.scr.2013.01.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Revised: 12/18/2012] [Accepted: 01/02/2013] [Indexed: 12/29/2022] Open
Abstract
To develop cell therapies for damaged nervous tissue with human neural stem/progenitor cells (hNPCs), the risk of an immune response and graft rejection must be considered. There are conflicting results and lack of knowledge concerning the immunocompetence of hNPCs of different origin. Here, we studied the immunogenicity and immunomodulatory potentials of hNPCs cultured under equivalent conditions after derivation from human embryonic stem cells (hESC-NPCs) or human fetal spinal cord tissue (hfNPCs). The expression patterns of human leukocyte antigen, co-stimulatory and adhesion molecules in hESC-NPCs and hfNPCs were relatively similar and mostly not affected by inflammatory cytokines. Unstimulated hfNPCs secreted more transforming growth factor-β1 (TGF-β1) and β2 but similar level of interleukin (IL)-10 compared to hESC-NPCs. In contrast to hfNPCs, hESC-NPCs displayed 4-6 fold increases in TGF-β1, TGF-β2 and IL-10 under inflammatory conditions. Both hNPCs reduced the alloreaction between allogeneic peripheral blood mononuclear cells (PBMCs) and up-regulated CD4(+)CD25(+)forkhead box P3 (FOXP3)(+) T cells. However, hESC-NPCs but not hfNPCs dose-dependently triggered PBMC proliferation, which at least partly may be due to TGF-β signaling. To conclude, hESC-NPCs and hfNPCs displayed similarities but also significant differences in their immunocompetence and interaction with allogeneic PBMCs, differences may be crucial for the outcome of cell therapy.
Collapse
|
30
|
Intrinsic properties and external factors determine the differentiation bias of human embryonic stem cell lines. Cell Biol Int 2013; 34:1021-31. [DOI: 10.1042/cbi20100283] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
31
|
Mandal A, Mathew S, Saha D, Viswanathan C. Establishment, characterization, and differentiation of a karyotypically normal human embryonic stem cell line from a trisomy-affected embryo. In Vitro Cell Dev Biol Anim 2012; 49:15-26. [PMID: 23242925 DOI: 10.1007/s11626-012-9567-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 10/22/2012] [Indexed: 01/25/2023]
Abstract
Derivation of human embryonic stem cell (hESC) lines from chromosomally or genetically abnormal embryos obtained following preimplantation genetic diagnosis (PGD) is of immense interest to study various kinds of genetic disorders. In this study, we have established a new hESC line Relicell(®)hES4, isolated from an aneuploid embryo. Derivation of this cell line was achieved by isolation of the inner cell mass (ICM) by mechanical method. Karyotype analysis showed that the hESC line is euploid having 46 chromosomes, contrary to our expectations. The undifferentiated cells exhibited long-term proliferation capacity and expressed markers typical for hESC, such as OCT4, NANOG, and SSEA4. A comparative microarray study was carried out to analyze the transcription profile of Relicell(®)hES4 along with three other normal hESC line generated earlier in our lab. Relicell(®)hES4 manifested pluripotent differentiation potential both in vivo and in vitro. The cells were also induced to form neurons, cardiomyocytes, and pancreatic β islets. The generation of a normal hESC line from an abnormal embryo points to the fact that even such embryos can be considered for deriving new hESC lines instead of discarding them. The data represented here are the first detailed report on characterization and differentiation of an Indian hESC line generated from a PGD analyzed embryo.
Collapse
Affiliation(s)
- Arundhati Mandal
- Regenerative Medicine, Reliance Life Sciences Pvt Ltd, Dhirubhai Ambani Life Sciences Centre, R-282, TTC Industrial Area of MIDC, Thane Belapur Road, Rabale, Navi Mumbai, 400 701, India.
| | | | | | | |
Collapse
|
32
|
Kokorina NA, Granier CJ, Zakharkin SO, Davis S, Rabson AB, Sabaawy HE. PDCD2 knockdown inhibits erythroid but not megakaryocytic lineage differentiation of human hematopoietic stem/progenitor cells. Exp Hematol 2012; 40:1028-1042.e3. [PMID: 22922207 PMCID: PMC5218995 DOI: 10.1016/j.exphem.2012.08.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 07/19/2012] [Accepted: 08/01/2012] [Indexed: 11/27/2022]
Abstract
Programmed cell death-2 (PDCD2) protein is enriched in embryonic, hematopoietic, and neural stem cells, however, its function in stem/progenitor cell differentiation is unclear. We investigated the effects of PDCD2 knockdown on the development and differentiation of hematopoietic progenitor cells (HPC). CD34(+) cells derived from normal human bone marrow and K562 leukemic cells were effectively transduced with short-hairpin RNA to knockdown PDCD2. Colony-forming assays were used to investigate the effects of PDCD2 loss on HPC clonogenic potential and on 12-O-tetradecanoyl-phorbol-13-acetate-and arabinofuranosylcytosine-induced terminal differentiation. In CD34(+) clonogenic progenitors, PDCD2 knockdown decreased the total number of colony-forming units, increased the number of colony-forming units-granulocyte-erythroid-macrophage-megakaryocyte and burst-forming unit-erythroid primitive colonies, and decreased the number of burst-forming unit-erythroid mature colonies. Similar results were observed in K562 cells, suggesting that PDCD2 is important for HPC differentiation and/or survival, and for erythroid lineage commitment. Furthermore, 12-O-tetradecanoyl-phorbol-13-acetate-induced megakaryocytic differentiation and proliferation of K562 cells was not affected by PDCD2 knockdown. In contrast, arabinofuranosylcytosine-induced erythroid differentiation of K562 cells was significantly reduced with PDCD2 knockdown, with no effect on cell proliferation. The effects of PDCD2 knockdown were attributed to a cell cycle arrest at G(0)/G(1), along with increased messenger RNA expression of early progenitor factors c-MYB and GATA-2, and decreased expression of erythroid factors GATA-1, EpoR, and γ-globin. We conclude that PDCD2 loss of function(s) impedes erythroid differentiation by inducing cell cycle arrest and increasing expression of early hematopoietic progenitor factors. These findings suggest that PDCD2 has a novel regulatory role in human hematopoiesis and is essential for erythroid development.
Collapse
Affiliation(s)
| | | | | | - Stephani Davis
- The Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Arnold B. Rabson
- Child Health Institute of New Jersey, New Brunswick, NJ, USA
- Department of Pediatrics, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ, USA
- Department of Molecular Genetics, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ, USA
- Department of Microbiology and Immunology, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ, USA
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ, USA
| | - Hatem E. Sabaawy
- The Cancer Institute of New Jersey, New Brunswick, NJ, USA
- Department of Pharmacology, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ, USA
- Department of Medicine, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
33
|
Datta I, Ganapathy K, Tattikota SM, Bhonde R. Directed differentiation of human embryonic stem cell-line HUES9 to dopaminergic neurons in a serum-free defined culture niche. Cell Biol Int 2012; 37:54-64. [DOI: 10.1002/cbin.10012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 09/17/2012] [Indexed: 01/24/2023]
Affiliation(s)
- Indrani Datta
- Manipal Institute of Regenerative Medicine; Constituent Institute of Manipal University; GKVK Post, Bellary Road, Allalasandra, Yelahanka, Bangalore, Karnataka India
| | - Kavina Ganapathy
- Manipal Institute of Regenerative Medicine; Constituent Institute of Manipal University; GKVK Post, Bellary Road, Allalasandra, Yelahanka, Bangalore, Karnataka India
| | - Surendra Mohan Tattikota
- Manipal Institute of Regenerative Medicine; Constituent Institute of Manipal University; GKVK Post, Bellary Road, Allalasandra, Yelahanka, Bangalore, Karnataka India
| | - Ramesh Bhonde
- Manipal Institute of Regenerative Medicine; Constituent Institute of Manipal University; GKVK Post, Bellary Road, Allalasandra, Yelahanka, Bangalore, Karnataka India
| |
Collapse
|
34
|
Ojala M, Rajala K, Pekkanen-Mattila M, Miettinen M, Huhtala H, Aalto-Setälä K. Culture conditions affect cardiac differentiation potential of human pluripotent stem cells. PLoS One 2012; 7:e48659. [PMID: 23119085 PMCID: PMC3485380 DOI: 10.1371/journal.pone.0048659] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 09/28/2012] [Indexed: 12/21/2022] Open
Abstract
Human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), are capable of differentiating into any cell type in the human body and thus can be used in studies of early human development, as cell models for different diseases and eventually also in regenerative medicine applications. Since the first derivation of hESCs in 1998, a variety of culture conditions have been described for the undifferentiated growth of hPSCs. In this study, we cultured both hESCs and hiPSCs in three different culture conditions: on mouse embryonic fibroblast (MEF) and SNL feeder cell layers together with conventional stem cell culture medium containing knockout serum replacement and basic fibroblast growth factor (bFGF), as well as on a Matrigel matrix in mTeSR1 medium. hPSC lines were subjected to cardiac differentiation in mouse visceral endodermal-like (END-2) co-cultures and the cardiac differentiation efficiency was determined by counting both the beating areas and Troponin T positive cells, as well as studying the expression of OCT-3/4, mesodermal Brachyury T and NKX2.5 and endodermal SOX-17 at various time points during END-2 differentiation by q-RT-PCR analysis. The most efficient cardiac differentiation was observed with hPSCs cultured on MEF or SNL feeder cell layers in stem cell culture medium and the least efficient cardiac differentiation was observed on a Matrigel matrix in mTeSR1 medium. Further, hPSCs cultured on a Matrigel matrix in mTeSR1 medium were found to be more committed to neural lineage than hPSCs cultured on MEF or SNL feeder cell layers. In conclusion, culture conditions have a major impact on the propensity of the hPSCs to differentiate into a cardiac lineage.
Collapse
Affiliation(s)
- Marisa Ojala
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
| | - Kristiina Rajala
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
| | - Mari Pekkanen-Mattila
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
| | - Marinka Miettinen
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
| | - Heini Huhtala
- School of Health Sciences, University of Tampere, Tampere, Finland
| | - Katriina Aalto-Setälä
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
- Heart Center, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
35
|
Mallon BS, Chenoweth JG, Johnson KR, Hamilton RS, Tesar PJ, Yavatkar AS, Tyson LJ, Park K, Chen KG, Fann YC, McKay RDG. StemCellDB: the human pluripotent stem cell database at the National Institutes of Health. Stem Cell Res 2012; 10:57-66. [PMID: 23117585 DOI: 10.1016/j.scr.2012.09.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 09/12/2012] [Accepted: 09/18/2012] [Indexed: 01/06/2023] Open
Abstract
Much of the excitement generated by induced pluripotent stem cell technology is concerned with the possibility of disease modeling as well as the potential for personalized cell therapy. However, to pursue this it is important to understand the 'normal' pluripotent state including its inherent variability. We have performed various molecular profiling assays for 21 hESC lines and 8 hiPSC lines to generate a comprehensive snapshot of the undifferentiated state of pluripotent stem cells. Analysis of the gene expression data revealed no iPSC-specific gene expression pattern in accordance with previous reports. We further compared cells, differentiated as embryoid bodies in 2 media proposed to initiate differentiation towards separate cell fates, as well as 20 adult tissues. From this analysis we have generated a gene list which defines pluripotency and establishes a baseline for the pluripotent state. Finally, we provide lists of genes enriched under both differentiation conditions which show the proposed bias toward independent cell fates.
Collapse
Affiliation(s)
- Barbara S Mallon
- NIH Stem Cell Unit, Division of Intramural Research, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Classification of human genomic regions based on experimentally determined binding sites of more than 100 transcription-related factors. Genome Biol 2012; 13:R48. [PMID: 22950945 PMCID: PMC3491392 DOI: 10.1186/gb-2012-13-9-r48] [Citation(s) in RCA: 187] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 05/06/2012] [Accepted: 06/08/2012] [Indexed: 01/22/2023] Open
Abstract
Background Transcription factors function by binding different classes of regulatory elements. The Encyclopedia of DNA Elements (ENCODE) project has recently produced binding data for more than 100 transcription factors from about 500 ChIP-seq experiments in multiple cell types. While this large amount of data creates a valuable resource, it is nonetheless overwhelmingly complex and simultaneously incomplete since it covers only a small fraction of all human transcription factors. Results As part of the consortium effort in providing a concise abstraction of the data for facilitating various types of downstream analyses, we constructed statistical models that capture the genomic features of three paired types of regions by machine-learning methods: firstly, regions with active or inactive binding; secondly, those with extremely high or low degrees of co-binding, termed HOT and LOT regions; and finally, regulatory modules proximal or distal to genes. From the distal regulatory modules, we developed computational pipelines to identify potential enhancers, many of which were validated experimentally. We further associated the predicted enhancers with potential target transcripts and the transcription factors involved. For HOT regions, we found a significant fraction of transcription factor binding without clear sequence motifs and showed that this observation could be related to strong DNA accessibility of these regions. Conclusions Overall, the three pairs of regions exhibit intricate differences in chromosomal locations, chromatin features, factors that bind them, and cell-type specificity. Our machine learning approach enables us to identify features potentially general to all transcription factors, including those not included in the data.
Collapse
|
37
|
Harvey AJ, Mao S, Lalancette C, Krawetz SA, Brenner CA. Transcriptional differences between rhesus embryonic stem cells generated from in vitro and in vivo derived embryos. PLoS One 2012; 7:e43239. [PMID: 23028448 PMCID: PMC3445581 DOI: 10.1371/journal.pone.0043239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 07/18/2012] [Indexed: 01/16/2023] Open
Abstract
Numerous studies have focused on the transcriptional signatures that underlie the maintenance of embryonic stem cell (ESC) pluripotency. However, it remains unclear whether ESC retain transcriptional aberrations seen in in vitro cultured embryos. Here we report the first global transcriptional profile comparison between ESC generated from either in vitro cultured or in vivo derived primate embryos by microarray analysis. Genes involved in pluripotency, oxygen regulation and the cell cycle were downregulated in rhesus ESC generated from in vitro cultured embryos (in vitro ESC). Significantly, several gene differences are similarly downregulated in preimplantation embryos cultured in vitro, which have been associated with long term developmental consequences and disease predisposition. This data indicates that prior to derivation, embryo quality may influence the molecular signature of ESC lines, and may differentially impact the physiology of cells prior to or following differentiation.
Collapse
Affiliation(s)
- Alexandra J Harvey
- Department of Physiology, Wayne State University, Detroit, Michigan, United States of America.
| | | | | | | | | |
Collapse
|
38
|
Jones GN, Moschidou D, Puga-Iglesias TI, Kuleszewicz K, Vanleene M, Shefelbine SJ, Bou-Gharios G, Fisk NM, David AL, De Coppi P, Guillot PV. Ontological differences in first compared to third trimester human fetal placental chorionic stem cells. PLoS One 2012; 7:e43395. [PMID: 22962584 PMCID: PMC3433473 DOI: 10.1371/journal.pone.0043395] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 07/19/2012] [Indexed: 12/16/2022] Open
Abstract
Human mesenchymal stromal/stem cells (MSC) isolated from fetal tissues hold promise for use in tissue engineering applications and cell-based therapies, but their collection is restricted ethically and technically. In contrast, the placenta is a potential source of readily-obtainable stem cells throughout pregnancy. In fetal tissues, early gestational stem cells are known to have advantageous characteristics over neonatal and adult stem cells. Accordingly, we investigated whether early fetal placental chorionic stem cells (e-CSC) were physiologically superior to their late gestation fetal chorionic counterparts (l-CSC). We showed that e-CSC shared a common phenotype with l-CSC, differentiating down the osteogenic, adipogenic and neurogenic pathways, and containing a subset of cells endogenously expressing NANOG, SOX2, c-MYC, and KLF4, as well as an array of genes expressed in pluripotent stem cells and primordial germ cells, including CD24, NANOG, SSEA4, SSEA3, TRA-1-60, TRA-1-81, STELLA, FRAGILIS, NANOS3, DAZL and SSEA1. However, we showed that e-CSC have characteristics of an earlier state of stemness compared to l-CSC, such as smaller size, faster kinetics, uniquely expressing OCT4A variant 1 and showing higher levels of expression of NANOG, SOX2, c-MYC and KLF4 than l-CSC. Furthermore e-CSC, but not l-CSC, formed embryoid bodies containing cells from the three germ layer lineages. Finally, we showed that e-CSC demonstrate higher tissue repair in vivo; when transplanted in the osteogenesis imperfecta mice, e-CSC, but not l-CSC increased bone quality and plasticity; and when applied to a skin wound, e-CSC, but not l-CSC, accelerated healing compared to controls. Our results provide insight into the ontogeny of the stemness phenotype during fetal development and suggest that the more primitive characteristics of early compared to late gestation fetal chorionic stem cells may be translationally advantageous.
Collapse
Affiliation(s)
- Gemma N. Jones
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Dafni Moschidou
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | | | - Katarzyna Kuleszewicz
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Maximilien Vanleene
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | | | - George Bou-Gharios
- Kennedy Institute of Rheumatology, University of Oxford, London, United Kingdom
| | - Nicholas M. Fisk
- UQ Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
| | - Anna L. David
- Prenatal Cell and Gene Therapy Group, Institute for Women's Health, University College London, London, United Kingdom
| | - Paolo De Coppi
- Surgery Unit, UCL Institute of Child Health, London, United Kingdom
| | - Pascale V. Guillot
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
39
|
Kramer J, Granier CJ, Davis S, Piso K, Hand J, Rabson AB, Sabaawy HE. PDCD2 controls hematopoietic stem cell differentiation during development. Stem Cells Dev 2012; 22:58-72. [PMID: 22800338 DOI: 10.1089/scd.2012.0074] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Programmed cell death 2 (Pdcd2) is a highly conserved protein of undefined function, and is widely expressed in embryonic and adult tissues. The observations that knockout of Pdcd2 in the mouse is embryonic lethal at preimplantation stages, and that in Drosophila, Zfrp8, the ortholog of Pdcd2, is required for normal lymph gland development suggest that Pdcd2 is important for regulating hematopoietic development. Through genetic and functional studies, we investigated pdcd2 function during the zebrafish ontogeny. Knockdown of pdcd2 expression in zebrafish embryos resulted in defects in embryonic hematopoietic development. Loss of pdcd2 function caused increased expression of progenitor markers, and accumulation of erythroid progenitors during primitive hematopoiesis. Additionally, hematopoietic stem cells (HSCs) failed to appear in the aorta-gonad mesonephros, and were not able to terminally differentiate or reconstitute hematopoiesis. Pdcd2 effects on HSC emergence were cell autonomous and P53-independent, and loss of pdcd2 function was associated with mitotic defects and apoptosis. Restoration of runx1 function(s) and modulation of apoptosis through the inhibition of Jak/Stat signaling rescued the hematopoietic and erythroid defects resulting from pdcd2 knockdown. Our studies suggest that pdcd2 plays a critical role in regulating the transcriptional hierarchy controlling hematopoietic lineage determination. Furthermore, the effects of pdcd2 in regulating mitotic cell death may contribute to its role(s) in directing hematopoietic differentiation during development.
Collapse
Affiliation(s)
- Joseph Kramer
- Department of Medicine, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Picard D, Miller S, Hawkins CE, Bouffet E, Rogers HA, Chan TSY, Kim SK, Ra YS, Fangusaro J, Korshunov A, Toledano H, Nakamura H, Hayden JT, Chan J, Lafay-Cousin L, Hu PX, Fan X, Muraszko KM, Pomeroy SL, Lau CC, Ng HK, Jones C, Meter TV, Clifford SC, Eberhart C, Gajjar A, Pfister SM, Grundy RG, Huang A. Markers of survival and metastatic potential in childhood CNS primitive neuro-ectodermal brain tumours: an integrative genomic analysis. Lancet Oncol 2012; 13:838-48. [PMID: 22691720 PMCID: PMC3615440 DOI: 10.1016/s1470-2045(12)70257-7] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Childhood CNS primitive neuro-ectodermal brain tumours (PNETs) are very aggressive brain tumours for which the molecular features and best treatment approaches are unknown. We assessed a large cohort of these rare tumours to identify molecular markers to enhance clinical management of this disease. METHODS We obtained 142 primary hemispheric CNS PNET samples from 20 institutions in nine countries and examined transcriptional profiles for a subset of 51 samples and copy number profiles for a subset of 77 samples. We used clustering, gene, and pathway enrichment analyses to identify tumour subgroups and group-specific molecular markers, and applied immunohistochemical and gene-expression analyses to validate and assess the clinical significance of the subgroup markers. FINDINGS We identified three molecular subgroups of CNS PNETs that were distinguished by primitive neural (group 1), oligoneural (group 2), and mesenchymal lineage (group 3) gene-expression signatures with differential expression of cell-lineage markers LIN28 and OLIG2. Patients with group 1 tumours were most often female (male:female ratio 0·61 for group 1 vs 1·25 for group 2 and 1·63 for group 3; p=0·043 [group 1 vs groups 2 and 3]), youngest (median age at diagnosis 2·9 years [95% CI 2·4-5·2] for group 1 vs 7·9 years [6·0-9·7] for group 2 and 5·9 years [4·9-7·8] for group 3; p=0·005), and had poorest survival (median survival 0·8 years [95% CI 0·5-1·2] in group 1, 1·8 years [1·4-2·3] in group 2 and 4·3 years [0·8-7·8] in group 3; p=0·019). Patients with group 3 tumours had the highest incidence of metastases at diagnosis (no distant metastasis:metastasis ratio 0·90 for group 3 vs 2·80 for group 1 and 5·67 for group 2; p=0·037). INTERPRETATION LIN28 and OLIG2 are promising diagnostic and prognostic molecular markers for CNS PNET that warrant further assessment in prospective clinical trials. FUNDING Canadian Institute of Health Research, Brainchild/SickKids Foundation, and the Samantha Dickson Brain Tumour Trust.
Collapse
Affiliation(s)
- Daniel Picard
- Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumour Research Centre, Dept of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Suzanne Miller
- Children's Brain Tumour Research Centre, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | | | - Eric Bouffet
- Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumour Research Centre, Dept of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Hazel A Rogers
- Children's Brain Tumour Research Centre, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Tiffany SY Chan
- Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumour Research Centre, Dept of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Seung-Ki Kim
- Dept of Neurosurgery, Seoul National University Children's Hospital, Seoul, South Korea
| | - Young-Shin Ra
- Dept of Neurosurgery, Asan Medical Center, Seoul, Korea
| | - Jason Fangusaro
- Division of Pediatric Hematology/Oncology and Stem Cell Transplantation, Children's Memorial Hospital, Chicago, USA
| | - Andrey Korshunov
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center, Heidelberg, Germany
| | | | | | - James T Hayden
- Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Jennifer Chan
- Dept of Pathology & Laboratory Medicine, University of Calgary, Calgary, Canada
| | - Lucie Lafay-Cousin
- Dept of Pediatric Oncology, Alberta Children's Hospital, Calgary, Canada
| | - Ping X Hu
- The Centre for Applied Genomics, Hospital for Sick Children, Toronto, Canada
| | - Xing Fan
- Dept of Neurosurgery, University of Michigan Medical School, Ann Arbor, USA
| | - Karin M Muraszko
- Dept of Neurosurgery, University of Michigan Medical School, Ann Arbor, USA
| | | | - Ching C Lau
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, USA
| | - Ho-Keung Ng
- Dept of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China
| | - Chris Jones
- Dept of Paediatric Molecular Pathology, Institute of Cancer Research, Sutton, United Kingdom
| | | | - Steven C Clifford
- Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Charles Eberhart
- Division of Pathology, John Hopkins University School of Medicine, Baltimore, USA
| | - Amar Gajjar
- Neuro-oncology Division, St. Jude Children's Research Hospital, Memphis, USA
| | - Stefan M Pfister
- German Cancer Research Centre, and Paediatric, Haematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Richard G Grundy
- Children's Brain Tumour Research Centre, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Annie Huang
- Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumour Research Centre, Dept of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Canada
| |
Collapse
|
41
|
Jääger K, Islam S, Zajac P, Linnarsson S, Neuman T. RNA-seq analysis reveals different dynamics of differentiation of human dermis- and adipose-derived stromal stem cells. PLoS One 2012; 7:e38833. [PMID: 22723894 PMCID: PMC3378616 DOI: 10.1371/journal.pone.0038833] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 05/15/2012] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Tissue regeneration and recovery in the adult body depends on self-renewal and differentiation of stem and progenitor cells. Mesenchymal stem cells (MSCs) that have the ability to differentiate into various cell types, have been isolated from the stromal fraction of virtually all tissues. However, little is known about the true identity of MSCs. MSC populations exhibit great tissue-, location- and patient-specific variation in gene expression and are heterogeneous in cell composition. METHODOLOGY/PRINCIPAL FINDINGS Our aim was to analyze the dynamics of differentiation of two closely related stromal cell types, adipose tissue-derived MSCs (AdMSCs) and dermal fibroblasts (FBs) along adipogenic, osteogenic and chondrogenic lineages using multiplex RNA-seq technology. We found that undifferentiated donor-matched AdMSCs and FBs are distinct populations that stay different upon differentiation into adipocytes, osteoblasts and chondrocytes. The changes in lineage-specific gene expression occur early in differentiation and persist over time in both AdMSCs and FBs. Further, AdMSCs and FBs exhibit similar dynamics of adipogenic and osteogenic differentiation but different dynamics of chondrogenic differentiation. CONCLUSIONS/SIGNIFICANCE Our findings suggest that stromal stem cells including AdMSCs and dermal FBs exploit different molecular mechanisms of differentiation to reach a common cell fate. The early mechanisms of differentiation are lineage-specific and are similar for adipogenic and osteogenic differentiation but are distinct for chondrogenic differentiation between AdMSCs and FBs.
Collapse
Affiliation(s)
- Kersti Jääger
- Institute of Gene Technology, Tallinn University of Technology, Tallinn, Estonia.
| | | | | | | | | |
Collapse
|
42
|
Laser-Based Propagation of Human iPS and ES Cells Generates Reproducible Cultures with Enhanced Differentiation Potential. Stem Cells Int 2012; 2012:926463. [PMID: 22701128 PMCID: PMC3369526 DOI: 10.1155/2012/926463] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 02/03/2012] [Accepted: 02/03/2012] [Indexed: 12/18/2022] Open
Abstract
Proper maintenance of stem cells is essential for successful utilization of ESCs/iPSCs as tools in developmental and drug discovery studies and in regenerative medicine. Standardization is critical for all future applications of stem cells and necessary to fully understand their potential. This study reports a novel approach for the efficient, consistent expansion of human ESCs and iPSCs using laser sectioning, instead of mechanical devices or enzymes, to divide cultures into defined size clumps for propagation. Laser-mediated propagation maintained the pluripotency, quality, and genetic stability of ESCs/iPSCs and led to enhanced differentiation potential. This approach removes the variability associated with ESC/iPSC propagation, significantly reduces the expertise, labor, and time associated with manual passaging techniques and provides the basis for scalable delivery of standardized ESC/iPSC lines. Adoption of standardized protocols would allow researchers to understand the role of genetics, environment, and/or procedural effects on stem cells and would ensure reproducible production of stem cell cultures for use in clinical/therapeutic applications.
Collapse
|
43
|
Zhao W, Ji X, Zhang F, Li L, Ma L. Embryonic stem cell markers. Molecules 2012; 17:6196-236. [PMID: 22634835 PMCID: PMC6268870 DOI: 10.3390/molecules17066196] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 03/31/2012] [Accepted: 05/04/2012] [Indexed: 02/07/2023] Open
Abstract
Embryonic stem cell (ESC) markers are molecules specifically expressed in ES cells. Understanding of the functions of these markers is critical for characterization and elucidation for the mechanism of ESC pluripotent maintenance and self-renewal, therefore helping to accelerate the clinical application of ES cells. Unfortunately, different cell types can share single or sometimes multiple markers; thus the main obstacle in the clinical application of ESC is to purify ES cells from other types of cells, especially tumor cells. Currently, the marker-based flow cytometry (FCM) technique and magnetic cell sorting (MACS) are the most effective cell isolating methods, and a detailed maker list will help to initially identify, as well as isolate ESCs using these methods. In the current review, we discuss a wide range of cell surface and generic molecular markers that are indicative of the undifferentiated ESCs. Other types of molecules, such as lectins and peptides, which bind to ESC via affinity and specificity, are also summarized. In addition, we review several markers that overlap with tumor stem cells (TSCs), which suggest that uncertainty still exists regarding the benefits of using these markers alone or in various combinations when identifying and isolating cells.
Collapse
Affiliation(s)
- Wenxiu Zhao
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
| | - Xiang Ji
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
- Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, China
| | - Fangfang Zhang
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
- Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, China
| | - Liang Li
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
- Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, China
| | - Lan Ma
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
| |
Collapse
|
44
|
Hernandez-Vargas H, Sincic N, Ouzounova M, Herceg Z. Epigenetic signatures in stem cells and cancer stem cells. Epigenomics 2012; 1:261-80. [PMID: 22122702 DOI: 10.2217/epi.09.19] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The physiological properties of pluripotency in stem cells and the processes of cell specialization are governed by epigenetic mechanisms, as they are inheritable but not dependent on the cell genotype. There is cumulating evidence demonstrating the presence of cells with stem cell properties within tumors, suggesting that these cells are responsible for tumor growth and heterogeneity. As epigenetic control of self-renewal and pluripotency is a hallmark of stem cells, there is increased interest in studying similar epigenetic mechanisms governing these stemness properties in cancer stem cells. Here we will review the evidence supporting a role for epigenetic mechanisms in the induction of cancer stem cells, with an emphasis on the epigenetic regulatory networks involved in the establishment of normal self-renewal and pluripotency, and their potential deregulation in cancer. We will also discuss the data supporting the plasticity of these mechanisms and its potential therapeutic implications.
Collapse
Affiliation(s)
- Hector Hernandez-Vargas
- Epigenetics Group, International Agency for Research on Cancer, 150 cours Albert-Thomas, Lyon cedex 08, France
| | | | | | | |
Collapse
|
45
|
Su Z, Xia J, Zhao Z. Functional complementation between transcriptional methylation regulation and post-transcriptional microRNA regulation in the human genome. BMC Genomics 2011; 12 Suppl 5:S15. [PMID: 22369656 PMCID: PMC3287497 DOI: 10.1186/1471-2164-12-s5-s15] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND DNA methylation in the 5' promoter regions of genes and microRNA (miRNA) regulation at the 3' untranslated regions (UTRs) are two major epigenetic regulation mechanisms in most eukaryotes. Both DNA methylation and miRNA regulation can suppress gene expression and their corresponding protein product; thus, they play critical roles in cellular processes. Although there have been numerous investigations of gene regulation by methylation changes and miRNAs, there is no systematic genome-wide examination of their coordinated effects in any organism. RESULTS In this study, we investigated the relationship between promoter methylation at the transcription level and miRNA regulation at the post-transcription level by taking advantage of recently released human methylome data and high quality miRNA and other gene annotation data. We found methylation level in the promoter regions and expression level was negatively correlated. Then, we showed that miRNAs tended to target the genes with a low DNA methylation level in their promoter regions. We further demonstrated that this observed pattern was not attributed to the gene expression level, expression broadness, or the number of transcription factor binding sites. Interestingly, we found miRNA target sites were significantly enriched in the genes located in differentially methylated regions or partially methylated domains. Finally, we explored the features of DNA methylation and miRNA regulation in cancer genes and found cancer genes tended to have low methylation level and more miRNA target sites. CONCLUSION This is the first genome-wide investigation of the combined regulation of gene expression. Our results supported a complementary regulation between DNA methylation (transcriptional level) and miRNA function (post-transcriptional level) in the human genome. The results were helpful for our understanding of the evolutionary forces towards organisms' complexity beyond traditional sequence level investigation.
Collapse
Affiliation(s)
- Zhixi Su
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | |
Collapse
|
46
|
Lifantseva N, Koltsova A, Krylova T, Yakovleva T, Poljanskaya G, Gordeeva O. Expression patterns of cancer-testis antigens in human embryonic stem cells and their cell derivatives indicate lineage tracks. Stem Cells Int 2011; 2011:795239. [PMID: 21785609 PMCID: PMC3140037 DOI: 10.4061/2011/795239] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 04/25/2011] [Indexed: 11/20/2022] Open
Abstract
Pluripotent stem cells can differentiate into various lineages but undergo genetic and epigenetic changes during long-term cultivation and, therefore, require regular monitoring. The expression patterns of cancer-testis antigens (CTAs) MAGE-A2, -A3, -A4, -A6, -A8, -B2, and GAGE were examined in undifferentiated human embryonic stem (hES) cells, their differentiated derivatives, teratocarcinoma (hEC) cells, and cancer cell lines of neuroectodermal and mesodermal origin. Undifferentiated hES cells and embryoid body cells expressed MAGE-A3, -A6, -A4, -A8, and GAGEs while later differentiated derivatives expressed only MAGE-A8 or MAGE-A4. Likewise, mouse pluripotent stem cells also express CTAs of Magea but not Mageb family. Despite similarity of the hES and hEC cell expression patterns, MAGE-A2 and MAGE-B2 were detected only in hEC cells but not in hES cells. Moreover, our analysis has shown that CTAs are aberrantly expressed in cancer cell lines and display low tissue specificity. The identification of CTA expression patterns in pluripotent stem cells and their derivatives may be useful for isolation of abnormally CTA-expressing cells to improve the safety of stem-cell based therapy.
Collapse
Affiliation(s)
- Nadya Lifantseva
- Kol'tsov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, Moscow 119334, Russia
| | | | | | | | | | | |
Collapse
|
47
|
Nagvenkar P, Pethe P, Pawani H, Telang J, Kumar N, Hinduja I, Zaveri K, Bhartiya D. Evaluating differentiation propensity of in-house derived human embryonic stem cell lines KIND-1 and KIND-2. In Vitro Cell Dev Biol Anim 2011; 47:406-419. [PMID: 21614653 DOI: 10.1007/s11626-011-9420-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Accepted: 04/20/2011] [Indexed: 02/07/2023]
Abstract
Human embryonic stem (hES) cells possess the ability to self-renew indefinitely and provide a potential source of differentiated progeny representing all three embryonic germ layers. Although hES cell lines share the expression of typical pluripotency markers, limited data is available regarding their differentiation capabilities. We have earlier reported the in-house derivation of two hES cell lines, KIND-1 and KIND-2 on human feeders. Here, we describe a comparative study carried out on both these cell lines to better understand the differentiation potential of KIND-1 and KIND-2 by gene expression analysis of representative gene transcripts reflecting pluripotency and the three germ layers viz. ectoderm, mesoderm, and endoderm. Gene expression analysis and immunolocalization studies were undertaken on (a) 7- and 14-d old embryoid bodies (EBs) (b) spontaneously differentiated cells from EBs, (c) cells derived from EBs under the influence of various growth factor treatments and (d) KIND-1 and KIND-2 cells co-cultured on mouse embryonic visceral endoderm-like feeder (END-2). Despite both the cell lines being XX, derived, passaged, and cultured similarly, KIND-1 exhibits preferential differentiation towards endodermal lineage whereas KIND-2 spontaneously forms beating cardiomyocytes. Perhaps the occurrence of discrete epigenetic profile in both the cell lines predisposes them to encompass different developmental potential in vitro. Our data provide evidence for existence of distinct differentiation propensity among hES cell lines and emphasizes the need to derive more hES cell lines for future regenerative medicine.
Collapse
Affiliation(s)
- Punam Nagvenkar
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Parel, Mumbai, 400 012, India
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Melichar H, Li O, Ross J, Haber H, Cado D, Nolla H, Robey EA, Winoto A. Comparative study of hematopoietic differentiation between human embryonic stem cell lines. PLoS One 2011; 6:e19854. [PMID: 21603627 PMCID: PMC3095633 DOI: 10.1371/journal.pone.0019854] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 04/04/2011] [Indexed: 12/27/2022] Open
Abstract
Directed differentiation of human embryonic stem cells (hESCs) into any desired cell type has been hailed as a therapeutic promise to cure many human diseases. However, substantial roadblocks still exist for in vitro differentiation of hESCs into distinct cell types, including T lymphocytes. Here we examined the hematopoietic differentiation potential of six different hESC lines. We compare their ability to develop into CD34+ or CD34+CD45+ hematopoietic precursor populations under several differentiation conditions. Comparison of lymphoid potential of hESC derived- and fetal tissue derived-hematopoietic precursors was also made. We found diverse hematopoietic potential between hESC lines depending on the culture or passage conditions. In contrast to fetal-derived hematopoietic precursors, none of the CD34+ precursors differentiated from hESCs were able to develop further into T cells. These data underscore the difficulties in the current strategy of hESC forward differentiation and highlight distinct differences between CD34+ hematopoietic precursors generated in vitro versus in vivo.
Collapse
Affiliation(s)
- Heather Melichar
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Ou Li
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Jenny Ross
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Hilary Haber
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Dragana Cado
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Hector Nolla
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Ellen A. Robey
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Astar Winoto
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
- * E-mail:
| |
Collapse
|
49
|
Burridge PW, Thompson S, Millrod MA, Weinberg S, Yuan X, Peters A, Mahairaki V, Koliatsos VE, Tung L, Zambidis ET. A universal system for highly efficient cardiac differentiation of human induced pluripotent stem cells that eliminates interline variability. PLoS One 2011; 6:e18293. [PMID: 21494607 PMCID: PMC3072973 DOI: 10.1371/journal.pone.0018293] [Citation(s) in RCA: 318] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 02/23/2011] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The production of cardiomyocytes from human induced pluripotent stem cells (hiPSC) holds great promise for patient-specific cardiotoxicity drug testing, disease modeling, and cardiac regeneration. However, existing protocols for the differentiation of hiPSC to the cardiac lineage are inefficient and highly variable. We describe a highly efficient system for differentiation of human embryonic stem cells (hESC) and hiPSC to the cardiac lineage. This system eliminated the variability in cardiac differentiation capacity of a variety of human pluripotent stem cells (hPSC), including hiPSC generated from CD34(+) cord blood using non-viral, non-integrating methods. METHODOLOGY/PRINCIPAL FINDINGS We systematically and rigorously optimized >45 experimental variables to develop a universal cardiac differentiation system that produced contracting human embryoid bodies (hEB) with an improved efficiency of 94.7±2.4% in an accelerated nine days from four hESC and seven hiPSC lines tested, including hiPSC derived from neonatal CD34(+) cord blood and adult fibroblasts using non-integrating episomal plasmids. This cost-effective differentiation method employed forced aggregation hEB formation in a chemically defined medium, along with staged exposure to physiological (5%) oxygen, and optimized concentrations of mesodermal morphogens BMP4 and FGF2, polyvinyl alcohol, serum, and insulin. The contracting hEB derived using these methods were composed of high percentages (64-89%) of cardiac troponin I(+) cells that displayed ultrastructural properties of functional cardiomyocytes and uniform electrophysiological profiles responsive to cardioactive drugs. CONCLUSION/SIGNIFICANCE This efficient and cost-effective universal system for cardiac differentiation of hiPSC allows a potentially unlimited production of functional cardiomyocytes suitable for application to hPSC-based drug development, cardiac disease modeling, and the future generation of clinically-safe nonviral human cardiac cells for regenerative medicine.
Collapse
Affiliation(s)
- Paul W. Burridge
- Johns Hopkins Institute for Cell Engineering, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
- * E-mail: (ETZ); (PWB)
| | - Susan Thompson
- Department of Biomedical Engineering, The Johns Hopkins University School
of Medicine, Baltimore, Maryland, United States of America
| | - Michal A. Millrod
- Johns Hopkins Institute for Cell Engineering, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
| | - Seth Weinberg
- Department of Biomedical Engineering, The Johns Hopkins University School
of Medicine, Baltimore, Maryland, United States of America
| | - Xuan Yuan
- Johns Hopkins Institute for Cell Engineering, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
| | - Ann Peters
- Johns Hopkins Institute for Cell Engineering, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
| | - Vasiliki Mahairaki
- Division of Neuropathology, Department of Pathology, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
| | - Vassilis E. Koliatsos
- Division of Neuropathology, Department of Pathology, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
- Department of Neurology, The Johns Hopkins University School of Medicine,
Baltimore, Maryland, United States of America
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
| | - Leslie Tung
- Department of Biomedical Engineering, The Johns Hopkins University School
of Medicine, Baltimore, Maryland, United States of America
| | - Elias T. Zambidis
- Johns Hopkins Institute for Cell Engineering, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
- * E-mail: (ETZ); (PWB)
| |
Collapse
|
50
|
Elliott AM, Elliott KAH, Kammesheidt A. High resolution array-CGH characterization of human stem cells using a stem cell focused microarray. Mol Biotechnol 2011; 46:234-42. [PMID: 20524159 DOI: 10.1007/s12033-010-9294-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Human embryonic and induced pluripotent stem cells (ESCs, iPSCs) that are cultured for an extended period of time are susceptible to genomic instability. Chromosomal aberrations decrease the reliability and reproducibility of experiments and could deem the cells unusable for therapeutic purposes. The genetic stability of human ESCs and iPSCs is commonly monitored by karyotype analysis. However, this low-resolution technique can only identify large aneuploidies. A reliable, high-resolution technique to detect genomic aberrations at a cost comparable to karyotyping is needed to better characterize stem cell lines. We have designed a stem cell focused array-comparative genomic hybridization microarray that covers the entire genome at high resolution with increased probe coverage in over 60 stem cell associated genes and more than 195 cancer related genes. Several iPSC lines were analyzed using the focused microarray and compared with either karyotyping or a standard Agilent 44K microarray. In addition to the abnormalities detected by these platforms, the custom microarray identified several small duplications spanning stem cell and/or cancer related genes. Scientists using a stem cell focused microarray to characterize their stem cells will be aware of the structural variants present in their cells and be more confident in their experimental results.
Collapse
|