1
|
Smith L, Quelch-Cliffe R, Liu F, Aguilar AH, Przyborski S. Evaluating Strategies to Assess the Differentiation Potential of Human Pluripotent Stem Cells: A Review, Analysis and Call for Innovation. Stem Cell Rev Rep 2024:10.1007/s12015-024-10793-5. [PMID: 39340737 DOI: 10.1007/s12015-024-10793-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2024] [Indexed: 09/30/2024]
Abstract
Pluripotent stem cells have the ability to differentiate into all cells and tissues within the human body, and as a result they are attractive resources for use in basic research, drug discovery and regenerative medicine. In order to successfully achieve this application, starting cell sources ideally require in-depth characterisation to confirm their pluripotent status and their ability to differentiate into tissues representative of the three developmental germ layers. Many different methods to assess potency are employed, each having its own distinct advantages and limitations. Some aspects of this characterisation process are not always well standardised, particularly techniques used to assess pluripotency as a function. In this article, we consider the methods used to establish cellular pluripotency and subsequently analyse characterisation data for over 1590 human pluripotent cell lines from publicly available repositories in the UK and USA. In particular, we focus on the teratoma xenograft assay, its use and protocols, demonstrating the level of variation and the frequency with which it is used. Finally, we reflect on the implications of the findings, and suggest in vitro alternatives using modern innovative technology as a way forward.
Collapse
Affiliation(s)
- Lucy Smith
- Department of Biosciences, Durham University, Durham, England
| | | | - Felicity Liu
- Department of Biosciences, Durham University, Durham, England
| | | | - Stefan Przyborski
- Department of Biosciences, Durham University, Durham, England.
- Reprocell Europe Ltd, NETPark, Sedgefield, England.
| |
Collapse
|
2
|
Wang WC, Hou TC, Kuo CY, Lai YC. Amplifications of EVX2 and HOXD9-HOXD13 on 2q31 in mature cystic teratomas of the ovary identified by array comparative genomic hybridization may explain teratoma characteristics in chondrogenesis and osteogenesis. J Ovarian Res 2024; 17:129. [PMID: 38907278 PMCID: PMC11193297 DOI: 10.1186/s13048-024-01458-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 06/16/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Teratomas are a common type of germ cell tumor. However, only a few reports on their genomic constitution have been published. The study of teratomas may provide a better understanding of their stepwise differentiation processes and molecular bases, which could prove useful for the development of tissue-engineering technologies. METHODS In the present study, we analyzed the copy number aberrations of nine ovarian mature cystic teratomas using array comparative genomic hybridization in an attempt to reveal their genomic aberrations. RESULTS The many chromosomal aberrations observed on array comparative genomic hybridization analysis reveal the complex genetics of this tumor. Amplifications and deletions of large DNA fragments were observed in some samples, while amplifications of EVX2 and HOXD9-HOXD13 on 2q31.1, NDUFV1 on 11q13.2, and RPL10, SNORA70, DNASE1L1, TAZ, ATP6AP1, and GDI1 on Xq28 were found in all nine mature cystic teratomas. CONCLUSIONS Our results indicated that amplifications of these genes may play an important etiological role in teratoma formation. Moreover, amplifications of EVX2 and HOXD9-HOXD13 on 2q31.1, found on array comparative genomic hybridization, may help to explain the characteristics of teratomas in chondrogenesis and osteogenesis.
Collapse
Affiliation(s)
- Wen-Chung Wang
- Department of Obstetrics and Gynecology, Jen-Ai Hospital, Taichung, Taiwan
| | - Tai-Cheng Hou
- Department of Pathology, Jen-Ai Hospital, Taichung, Taiwan
| | - Chen-Yun Kuo
- Department of Pathology, Jen-Ai Hospital, Taichung, Taiwan
| | - Yen-Chein Lai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, No.110, Sec. 1, Chien Kuo N. Road, Taichung, 402, Taiwan, R.O.C..
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
3
|
Corsini M, Mitola S. Alternative In Vivo Models to Study Teratoma. Methods Mol Biol 2023; 2572:181-189. [PMID: 36161417 DOI: 10.1007/978-1-0716-2703-7_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Embryonic stem cells give rise to teratomas when injected in vivo in experimental animal models. The characterization, the manipulation, and the breaking off of this specific characteristic are doubtlessly the last frontier for the applications of stem cells in translational medicine. Moreover, the urgency to adapt to new scientific demands drives the researcher to find alternative and faster models for testing the teratogenic properties of embryonic stem cells. Here, we compare the emerging model of the chick embryo chorioallantoic membrane (CAM) to the murine model, which represents the gold standard procedure for teratogenesis.
Collapse
Affiliation(s)
- Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
4
|
Wang WC, Lai YC. DUSP5 and PHLDA1 mutations in mature cystic teratomas of the ovary identified on whole-exome sequencing may explain teratoma characteristics. Hum Genomics 2022; 16:50. [PMID: 36289533 PMCID: PMC9609193 DOI: 10.1186/s40246-022-00424-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/19/2022] [Indexed: 11/21/2022] Open
Abstract
Background Mature cystic teratomas of the ovary are the most common type of germ cell tumor, comprising 33% of ovarian tumors. Studying these tumors may result in a better understanding of their stepwise developmental processes and molecular bases and provide useful information for the development of tissue-engineering technologies. Methods In the present study, 9 mature cystic teratomas of the ovary were analyzed by whole-exome sequencing and the results were compared with the Catalogue of Somatic Mutations in Cancer and dbSNP databases. Results Mutations were validated in 15 genes with alterations in all 9 (100%) samples and changes in protein coding. The top 10 mutated genes were FLG, MUC17, MUC5B, RP1L1, NBPF1, GOLGA6L2, SLC29A3, SGK223, PTGFRN, and FAM186A. Moreover, 7 variants in exons with changes in protein coding are likely of importance in the development of mature cystic teratomas of the ovary, namely PTGFRN, DUSP5, MPP2, PHLDA1, PRR21, GOLGA6L2, and KRTAP4-2. Conclusions These genetic alterations may play an important etiological role in teratoma formation. Moreover, novel mutations in DUSP5 and PHLDA1 genes found on whole-exome sequencing may help to explain the characteristics of teratomas. Supplementary Information The online version contains supplementary material available at 10.1186/s40246-022-00424-w.
Collapse
Affiliation(s)
- Wen-Chung Wang
- grid.414969.70000 0004 0642 8534Department of Obstetrics and Gynecology, Jen-Ai Hospital, Taichung, 412 Taiwan
| | - Yen-Chein Lai
- grid.411641.70000 0004 0532 2041Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, No. 110, Sec. 1, Chien Kuo N. Road, Taichung, 402 Taiwan ,grid.411645.30000 0004 0638 9256Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
5
|
Hidalgo Aguilar A, Smith L, Owens D, Quelch R, Przyborski S. Recreating Tissue Structures Representative of Teratomas In Vitro Using a Combination of 3D Cell Culture Technology and Human Embryonic Stem Cells. Bioengineering (Basel) 2022; 9:bioengineering9050185. [PMID: 35621463 PMCID: PMC9138123 DOI: 10.3390/bioengineering9050185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/11/2022] [Accepted: 04/19/2022] [Indexed: 11/22/2022] Open
Abstract
In vitro studies using human embryonic stem cells (hESCs) are a valuable method to study aspects of embryogenesis, avoiding ethical issues when using embryonic materials and species dissimilarities. The xenograft teratoma assay is often traditionally used to establish pluripotency in putative PSC populations, but also has additional applications, including the study of tissue differentiation. The stem cell field has long sought an alternative due to various well-established issues with the in vivo technique, including significant protocol variability and animal usage. We have established a two-step culture method which combines PSC-derived embryoid bodies (EBs) with porous scaffolds to enhance their viability, prolonging the time these structures can be maintained, and therefore, permitting more complex, mature differentiation. Here, we have utilised human embryonic stem cell-derived EBs, demonstrating the formation of tissue rudiments of increasing complexity over time and the ability to manipulate their differentiation through the application of exogenous morphogens to achieve specific lineages. Crucially, these EB-derived tissues are highly reminiscent of xenograft teratoma samples derived from the same cell line. We believe this in vitro approach represents a reproducible, animal-free alternative to the teratoma assay, which can be used to study human tissue development.
Collapse
Affiliation(s)
| | - Lucy Smith
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (A.H.A.); (L.S.); (D.O.); (R.Q.)
| | - Dominic Owens
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (A.H.A.); (L.S.); (D.O.); (R.Q.)
| | - Rebecca Quelch
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (A.H.A.); (L.S.); (D.O.); (R.Q.)
| | - Stefan Przyborski
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (A.H.A.); (L.S.); (D.O.); (R.Q.)
- Reprocell Europe, NETPark, Sedgefield TS21 3FD, UK
- Correspondence:
| |
Collapse
|
6
|
Mousaei Ghasroldasht M, Seok J, Park HS, Liakath Ali FB, Al-Hendy A. Stem Cell Therapy: From Idea to Clinical Practice. Int J Mol Sci 2022; 23:ijms23052850. [PMID: 35269990 PMCID: PMC8911494 DOI: 10.3390/ijms23052850] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/20/2022] [Accepted: 03/03/2022] [Indexed: 11/27/2022] Open
Abstract
Regenerative medicine is a new and promising mode of therapy for patients who have limited or no other options for the treatment of their illness. Due to their pleotropic therapeutic potential through the inhibition of inflammation or apoptosis, cell recruitment, stimulation of angiogenesis, and differentiation, stem cells present a novel and effective approach to several challenging human diseases. In recent years, encouraging findings in preclinical studies have paved the way for many clinical trials using stem cells for the treatment of various diseases. The translation of these new therapeutic products from the laboratory to the market is conducted under highly defined regulations and directives provided by competent regulatory authorities. This review seeks to familiarize the reader with the process of translation from an idea to clinical practice, in the context of stem cell products. We address some required guidelines for clinical trial approval, including regulations and directives presented by the Food and Drug Administration (FDA) of the United States, as well as those of the European Medicine Agency (EMA). Moreover, we review, summarize, and discuss regenerative medicine clinical trial studies registered on the Clinicaltrials.gov website.
Collapse
|
7
|
Inagaki E, Arai E, Hatou S, Sayano T, Taniguchi H, Negishi K, Kanai Y, Sato Y, Okano H, Tsubota K, Shimmura S. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:841-849. [PMID: 35666752 PMCID: PMC9397653 DOI: 10.1093/stcltm/szac036] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 04/09/2022] [Indexed: 11/25/2022] Open
Abstract
Pluripotent stem cell (PSC)-based cell therapies have increased steadily over the past few years, and assessing the risk of tumor formation is a high priority for clinical studies. Current in vivo tumorigenesis studies require several months and depend strongly on the site of grafting. In this study, we report that the anterior eye chamber is preferable to the subcutaneous space for in vivo tumorigenesis studies for several reasons. First, cells can easily be transplanted into the anterior chamber and monitored in real-time without sacrificing the animals due to the transparency of the cornea. Second, tumor formation is faster than with the conventional subcutaneous method. The median tumor formation time in the subcutaneous area was 18.50 weeks (95% CI 10.20-26.29), vs. 4.0 weeks (95% CI 3.34-.67) in the anterior chamber (P = .0089). When hiPSCs were spiked with fibroblasts, the log10TPD50 was 3.26, compared with 4.99 when hiPSCs were transplanted without fibroblasts. There was more than a 40-fold difference in the log10TPD50 values with fibroblasts. Furthermore, the log10TPD50 for HeLa cells was 1.45 and 100% of animals formed tumors at a concentration greater than 0.1%, indicating that the anterior chamber tumorigenesis assays can be applied for cancer cell lines as well. Thus, our method has the potential to become a powerful tool in all areas of tumorigenesis studies and cancer research.
Collapse
Affiliation(s)
- Emi Inagaki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Japanese Society for the Promotion of Science (JSPS), Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Eri Arai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Shin Hatou
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Cellusion Inc., Tokyo, Japan
| | - Tomoko Sayano
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Cellusion Inc., Tokyo, Japan
| | - Hiroko Taniguchi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yasunori Sato
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Shigeto Shimmura
- Corresponding author: Shigeto Shimmura, MD, Department of Ophthalmology, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan. Tel: +81 3 3358 5962; Fax: +81 3 3359 8302;
| |
Collapse
|
8
|
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: 1.0] [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
|
9
|
Smith LA, Hidalgo Aguilar A, Owens DDG, Quelch RH, Knight E, Przyborski SA. Using Advanced Cell Culture Techniques to Differentiate Pluripotent Stem Cells and Recreate Tissue Structures Representative of Teratoma Xenografts. Front Cell Dev Biol 2021; 9:667246. [PMID: 34026759 PMCID: PMC8134696 DOI: 10.3389/fcell.2021.667246] [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: 02/12/2021] [Accepted: 04/12/2021] [Indexed: 11/24/2022] Open
Abstract
Various methods are currently used to investigate human tissue differentiation, including human embryo culture and studies utilising pluripotent stem cells (PSCs) such as in vitro embryoid body formation and in vivo teratoma assays. Each method has its own distinct advantages, yet many are limited due to being unable to achieve the complexity and maturity of tissue structures observed in the developed human. The teratoma xenograft assay allows maturation of more complex tissue derivatives, but this method has ethical issues surrounding animal usage and significant protocol variation. In this study, we have combined three-dimensional (3D) in vitro cell technologies including the common technique of embryoid body (EB) formation with a novel porous scaffold membrane, in order to prolong cell viability and extend the differentiation of PSC derived EBs. This approach enables the formation of more complex morphologically identifiable 3D tissue structures representative of all three primary germ layers. Preliminary in vitro work with the human embryonal carcinoma line TERA2.SP12 demonstrated improved EB viability and enhanced tissue structure formation, comparable to teratocarcinoma xenografts derived in vivo from the same cell line. This is thought to be due to reduced diffusion distances as the shape of the spherical EB transforms and flattens, allowing for improved nutritional/oxygen support to the developing structures over extended periods. Further work with EBs derived from murine embryonic stem cells demonstrated that the formation of a wide range of complex, recognisable tissue structures could be achieved within 2–3 weeks of culture. Rudimentary tissue structures from all three germ layers were present, including epidermal, cartilage and epithelial tissues, again, strongly resembling tissue structure of teratoma xenografts of the same cell line. Proof of concept work with EBs derived from the human embryonic stem cell line H9 also showed the ability to form complex tissue structures within this system. This novel yet simple model offers a controllable, reproducible method to achieve complex tissue formation in vitro. It has the potential to be used to study human developmental processes, as well as offering an animal free alternative method to the teratoma assay to assess the developmental potential of novel stem cell lines.
Collapse
Affiliation(s)
- L A Smith
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - A Hidalgo Aguilar
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - D D G Owens
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - R H Quelch
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - E Knight
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - S A Przyborski
- Department of Biosciences, Durham University, Durham, United Kingdom.,Reprocell Europe, NETPark, Sedgefield, United Kingdom
| |
Collapse
|
10
|
Yang X, Ku T, Sun Z, Liu QS, Yin N, Zhou Q, Faiola F, Liao C, Jiang G. Assessment of the carcinogenic effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin using mouse embryonic stem cells to form teratoma in vivo. Toxicol Lett 2019; 312:139-147. [PMID: 31082521 DOI: 10.1016/j.toxlet.2019.05.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 03/21/2019] [Accepted: 05/09/2019] [Indexed: 12/26/2022]
Abstract
As the most toxic dioxin, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) has gained lots of concerns, due to its diverse deleterious effects. However, the knowledge on carcinogenic risk of TCDD during early stage of development remains scarce. The in vivo teratoma formation model based on the transplantation of embryonic stem cells (ESCs) in immunodeficient mice is appealing for studying pluripotency and tumorigenicity in developmental biology, and also shows promise in environmental toxicology, especially in carcinogenesis researches. In this study, the malignant transformation of mouse embryonic stem cells (mESCs) pretreated with TCDD was investigated during their in vivo differentiation using teratoma formation model. Based on characterization of the pluripotency and differentiation capabilities of mESCs, evil changes in teratomas derived from TCDD-exposed mESCs were systematically studied. The results showed that TCDD significantly up-regulated CYP1A1 transcriptional levels in mESCs, elevated the incidence of malignant change in mESC-derived teratomas, and caused indefinite proliferation capabilities in sequential cultures of tumor tissues. The findings suggested that TCDD could exert carcinogenic effect on mESCs during their differentiation into teratoma in vivo, and more attention should be paid to the adverse health effects of this chemical during gestation or early developmental period.
Collapse
Affiliation(s)
- Xiaoxi Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tingting Ku
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, 030006, China
| | - Zhendong Sun
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qian S Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Nuoya Yin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Qunfang Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China; Institute of Environment and Health, Jianghan University, Wuhan, 430056, China.
| | - Francesco Faiola
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Chunyang Liao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
11
|
Mitchell A, Wanczyk H, Jensen T, Finck C. Assessment of iPSC teratogenicity throughout directed differentiation toward an alveolar-like phenotype. Differentiation 2019; 105:45-53. [PMID: 30711828 DOI: 10.1016/j.diff.2019.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 11/30/2018] [Accepted: 01/18/2019] [Indexed: 12/21/2022]
Abstract
Considerable work has gone into creating cell therapies from induced pluripotent stem cells (iPSCs) since their discovery just over a decade ago. However, comparatively little research has been done concerning the safety of iPSCs and their progeny and specifically the mechanisms governing teratogenicity. The aim of this study was to ascertain at what developmental phase iPSCs undergoing differentiation to an alveolar-like phenotype lose their capacity to form a teratoma and uncover potential mechanisms responsible. iPSCs were differentiated using a previously published directed differentiation protocol mirroring alveolar embryogenesis. At each developmental phase cell phenotype was assessed and cells mixed with Matrigel and injected subcutaneously above the hind limbs of NSG mice to determine teratogenicity. A genetic screen of 42 genes commonly associated with teratoma formation was conducted on all the cells and any resulting teratoma. It was found that neither NKX2-1 lung progenitors nor terminally differentiated alveolar-like cells formed teratomas. As expected the expression of pluripotency markers was diminished over differentiation. However, the expression of two proteoglycans, decorin and lumican, was increased more than 3000x during differentiation. Both decorin and lumican are putative tumor suppressors with additional functions in angiogenesis, fibrosis, inflammation and autophagy. We hypothesize that the increasing expression of these proteoglycans by iPSCs as they differentiate may act to inhibit host endothelial cell recruitment when implanted resulting in the inhibition of any teratoma formation by any remaining undifferentiated iPSCs.
Collapse
Affiliation(s)
- Adam Mitchell
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA.
| | - Heather Wanczyk
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA.
| | - Todd Jensen
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA.
| | - Christine Finck
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA; Department of Surgery, Connecticut Children's Medical Center, Hartford, CT, USA.
| |
Collapse
|
12
|
Al-Yamani A, Kalamegam G, Ahmed F, Abbas M, Sait KHW, Anfinan N, Al-Wasiyah MK, Huwait EA, Gari M, Al-Qahtani M. Evaluation of in vitro chondrocytic differentiation: A stem cell research initiative at the King Abdulaziz University, Kingdom of Saudi Arabia. Bioinformation 2018; 14:53-59. [PMID: 29618900 PMCID: PMC5879944 DOI: 10.6026/97320630014053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 02/01/2018] [Accepted: 02/01/2018] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) from various sources have been used in cartilage differentiation with variable success. Therefore, it is
of interest to evaluate the in vitro differentiation potential of the hWJSCs derived from the human umbilical cords into chondrocytes at
the stem cell research facility at the King Abdulaziz University. hWJSCs are an attractive choice for tissue engineering and regenerative
medical applications including cartilage regeneration. We evaluated the hWJSCs using classical histological and cartilage related gene
expression studies. Some of the known parameters were re-examined for consistency at the current laboratory conditions. Early
passages (P1-P4) showed short fibroblastic morphology and high expression of MSC related surface markers namely CD29 (99.9%),
CD44 (97.8%), CD73 (99.6%), CD90 (95.1%) and CD105 (98.9%). MTT assay showed time dependent increase in hWJSCs proliferation by
61.06% and 206.31% at 48h and 72h respectively. Toluidine blue histology showed that hWJSCs were successfully differentiated into
chondrocytes in chondrocytic differentiation medium for 21 days. Differentiated hWJSCs also showed significantly increased
expression of collagen type II, aggrecan and SOX9 compared to the undifferentiated control. It should be noted that the determination
of the average cell yield, the population doubling time and histological staining wtih alcian blue and/or safronin O is required in future
studies for improved evaluation of differentiation. Painless derivation, abundance of stem cells that are hypo-immunogenic and safety
issues makes this method advantages to MSCs derived from other sources.
Collapse
Affiliation(s)
- Aisha Al-Yamani
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Gauthaman Kalamegam
- Stem Cell Unit, Centre of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Sheikh Salem Bin Mahfouz Scientific Chair for Treatment of Osteoarthritis by Stem Cells, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Farid Ahmed
- Stem Cell Unit, Centre of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Abbas
- Sheikh Salem Bin Mahfouz Scientific Chair for Treatment of Osteoarthritis by Stem Cells, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Orthopaedic Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Khalid Hussein Wali Sait
- Department of Obstetrics and Gynaecology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nisreen Anfinan
- Department of Obstetrics and Gynaecology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Khalid Al-Wasiyah
- Stem Cell Unit, Centre of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Etimad A Huwait
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Mamdouh Gari
- Stem Cell Unit, Centre of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Mohammed Al-Qahtani
- Stem Cell Unit, Centre of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
13
|
Pei Y, Yue L, Zhang W, Xiang J, Ma Z, Han J. Murine pluripotent stem cells that escape differentiation inside teratomas maintain pluripotency. PeerJ 2018; 6:e4177. [PMID: 29312817 PMCID: PMC5756617 DOI: 10.7717/peerj.4177] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/29/2017] [Indexed: 12/02/2022] Open
Abstract
Background Pluripotent stem cells (PSCs) offer immense potential as a source for regenerative therapies. The teratoma assay is widely used in the field of stem cells and regenerative medicine, but the cell composition of teratoma is still elusive. Methods We utilized PSCs expressing enhanced green fluorescent protein (EGFP) under the control of the Pou5f1 promoter to study the persistence of potential pluripotent cells during teratoma formation in vivo. OCT4-MES (mouse embryonic stem cells) were isolated from the blastocysts of 3.5-day OCT4-EGFP mice (transgenic mice express EGFP cDNA under the control of the Pou5f1 promoter) embryos, and TG iPS 1-7 (induced pluripotent stem cells) were generated from mouse embryonic fibroblasts (MEFs) from 13.5-day OCT4-EGFP mice embryos by infecting them with a virus carrying OCT4, SOX2, KLF4 and c-MYC. These pluripotent cells were characterized according to their morphology and expression of pluripotency markers. Their differentiation ability was studied with in vivo teratoma formation assays. Further differences between pluripotent cells were examined by real-time quantitative PCR (qPCR). Results The results showed that several OCT4-expressing PSCs escaped differentiation inside of teratomas, and these escaped cells (MES-FT, GFP-positive cells separated from OCT4-MES-derived teratomas; and iPS-FT, GFP-positive cells obtained from teratomas formed by TG iPS 1-7) retained their pluripotency. Interestingly, a small number of GFP-positive cells in teratomas formed by MES-FT and iPS-FT (MES-ST, GFP-positive cells isolated from MES-FT-derived teratomas; iPS-ST, GFP-positive cells obtained from teratomas formed by iPS-FT) were still pluripotent, as shown by alkaline phosphatase (AP) staining, immunofluorescent staining and PCR. MES-FT, iPS-FT, MES-ST and iPS-ST cells also expressed several markers associated with germ cell formation, such as Dazl, Stella and Stra8. Conclusions In summary, a small number of PSCs escaped differentiation inside of teratomas, and these cells maintained pluripotency and partially developed towards germ cells. Both escaped PSCs and germ cells present a risk of tumor formation. Therefore, medical workers must be careful in preventing tumor formation when stem cells are used to treat specific diseases.
Collapse
Affiliation(s)
- Yangli Pei
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China.,State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Liang Yue
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Wei Zhang
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jinzhu Xiang
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zhu Ma
- Beijing Dairy Cattle Center, Beijing, China
| | - Jianyong Han
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
14
|
Andrews PW, Baker D, Benvinisty N, Miranda B, Bruce K, Brüstle O, Choi M, Choi YM, Crook JM, de Sousa PA, Dvorak P, Freund C, Firpo M, Furue MK, Gokhale P, Ha HY, Han E, Haupt S, Healy L, Hei DJ, Hovatta O, Hunt C, Hwang SM, Inamdar MS, Isasi RM, Jaconi M, Jekerle V, Kamthorn P, Kibbey MC, Knezevic I, Knowles BB, Koo SK, Laabi Y, Leopoldo L, Liu P, Lomax GP, Loring JF, Ludwig TE, Montgomery K, Mummery C, Nagy A, Nakamura Y, Nakatsuji N, Oh S, Oh SK, Otonkoski T, Pera M, Peschanski M, Pranke P, Rajala KM, Rao M, Ruttachuk R, Reubinoff B, Ricco L, Rooke H, Sipp D, Stacey GN, Suemori H, Takahashi TA, Takada K, Talib S, Tannenbaum S, Yuan BZ, Zeng F, Zhou Q. Points to consider in the development of seed stocks of pluripotent stem cells for clinical applications: International Stem Cell Banking Initiative (ISCBI). Regen Med 2015; 10:1-44. [PMID: 25675265 DOI: 10.2217/rme.14.93] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Affiliation(s)
- P W Andrews
- Department of Biomedical Science, The University of Sheffield, Sheffield, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Experimental teratoma: at the crossroad of fetal- and onco-development. Semin Cancer Biol 2014; 29:75-9. [PMID: 25153353 DOI: 10.1016/j.semcancer.2014.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 08/11/2014] [Indexed: 12/17/2022]
Abstract
Xenografting is the so far only available in vivo model for assessing pluripotency of human stem cells. This review describes known biological features of experimental teratoma from human pluripotent stem cells. We focus on the dual nature mimicking both normal and abnormal development, and propose this model system to be particularly interesting for investigations of the relationship between developmentally controlled differentiation and neoplasia of embryonic origin. In resemblance to the wide range of clinical teratomas, pluripotent stem cell (PSC) induced teratoma (PSCT) typically shows a mixture of developing tissues in randomly distributed compartments. The combined literature suggests that for teratomas derived from human diploid bona fide PSC the embryonic development in the separate tissue-niches can show a controlled differentiation into organoid patterns closely mimicking early development. In the experimental situation such PSCT human homologous in vivo tissue-niches have been shown to provide also matching microenvironment for a micrometastatic colonization and outgrowth of embryonic tumors transplanted directly from patients. Single or small clusters of normal and neoplastic cells can easily be visualized together in microscope-based imaging systems, enabling multi-parameter detection of in the scans of tissue slides/specimens.
Collapse
|
16
|
Abstract
Autism and autism spectrum disorders (ASDs) are complex neurodevelopmental disorders. ASDs are clinically defined by deficits in communication, social skills, and repetitive and/or restrictive interests and behaviours. With the prevalence rates for ASDs rapidly increasing, the need for effective therapies for autism is a priority for biomedical research. Currently available medications do not target the core symptoms, can have markedly adverse side-effects, and are mainly palliative for negative behaviours. The development of molecular and regenerative interventions is progressing rapidly, and medicine holds great expectations for stem cell therapies. Cells could be designed to target the observed molecular mechanisms of ASDs, that is, abnormal neurotransmitter regulation, activated microglia, mitochondrial dysfunction, blood-brain barrier disruptions, and chronic intestinal inflammation. Presently, the paracrine, secretome, and immunomodulatory effects of stem cells would appear to be the likely mechanisms of application for ASD therapeutics. This review will focus on the potential use of the various types of stem cells: embryonic, induced pluripotential, fetal, and adult stem cells as targets for ASD therapeutics.
Collapse
|
17
|
Perspectives on the use of stem cells for autism treatment. Stem Cells Int 2013; 2013:262438. [PMID: 24222772 PMCID: PMC3810518 DOI: 10.1155/2013/262438] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 08/22/2013] [Accepted: 09/06/2013] [Indexed: 12/13/2022] Open
Abstract
Autism and autism spectrum disorders (ASDs) are complex neurodevelopmental disorders. ASDs are clinically defined by deficits in communication, social skills, and repetitive and/or restrictive interests and behaviours. With the prevalence rates for ASDs rapidly increasing, the need for effective therapies for autism is a priority for biomedical research. Currently available medications do not target the core symptoms, can have markedly adverse side-effects, and are mainly palliative for negative behaviours. The development of molecular and regenerative interventions is progressing rapidly, and medicine holds great expectations for stem cell therapies. Cells could be designed to target the observed molecular mechanisms of ASDs, that is, abnormal neurotransmitter regulation, activated microglia, mitochondrial dysfunction, blood-brain barrier disruptions, and chronic intestinal inflammation. Presently, the paracrine, secretome, and immunomodulatory effects of stem cells would appear to be the likely mechanisms of application for ASD therapeutics. This review will focus on the potential use of the various types of stem cells: embryonic, induced pluripotential, fetal, and adult stem cells as targets for ASD therapeutics.
Collapse
|
18
|
Turner NJ, Keane TJ, Badylak SF. Lessons from developmental biology for regenerative medicine. ACTA ACUST UNITED AC 2013; 99:149-59. [DOI: 10.1002/bdrc.21040] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 07/27/2013] [Accepted: 07/27/2013] [Indexed: 12/17/2022]
Affiliation(s)
- Neill J. Turner
- McGowan Institute for Regenerative Medicine; University of Pittsburgh, Pittsburgh, Pennsylvania and Department of Surgery, University of Pittsburgh; Pittsburgh Pennsylvania
| | - Timothy J. Keane
- McGowan Institute for Regenerative Medicine; University of Pittsburgh, Pittsburgh, Pennsylvania and Department of Bioengineering, University of Pittsburgh; Pittsburgh Pennsylvania
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, and Department of Bioengineering, University of Pittsburgh; Pittsburgh Pennsylvania
| |
Collapse
|
19
|
Srivastava S, Bedi U, Roy P. Synergistic actions of insulin-sensitive and Sirt1-mediated pathways in the differentiation of mouse embryonic stem cells to osteoblast. Mol Cell Endocrinol 2012; 361:153-64. [PMID: 22542761 DOI: 10.1016/j.mce.2012.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 04/05/2012] [Accepted: 04/10/2012] [Indexed: 12/31/2022]
Abstract
Murine embryonic stem cells (mESCs) have the potential to differentiate into almost any type of cell, and hence, represent a useful biological resource for tissue engineering. The differentiation of mESCs into osteoblasts in vitro is usually dampened by simultaneous differentiation of adipocytes. Insulin exerts a profound effect on bone development through increased differentiation of osteoblasts and concurrent formation of adipocytes. Comparatively, Sirt1, which plays a crucial role in osteoblast differentiation, has been reported to down regulate adipocyte formation during osteoblast differentiation. This study analyzed the combined effects of insulin and Sirt1 on the differentiation of osteoblasts. Osteoblast differentiation was quantified by estimating the accumulation of mineralized matrix and expression of osteogenic genes. The present data show that the simultaneous action of the insulin and Sirt1-mediated pathways increased the efficiency of osteoblast differentiation. When the cells were tested for ALP activity and Alizarin red staining, there was a respective increase of ~180% and ~166% (P<0.05) compared to the control. Furthermore, the mRNA expression patterns of osteoprotegerin, osterix, runx2, and osteopontin were increased by 3.6, 2.3, 1.8, and 1.7-fold, respectively, with a concomitant decrease in the mRNA expression levels of adipocyte marker genes. Interestingly, blocking the effects of both Sirt1 and insulin resulted in decreased osteoblastogenesis (60%) and subsequent increased adipocyte differentiation (195%) (P<0.05). Moreover, immunoblotting analysis demonstrated that this activation was via an Akt-dependent pathway. In conclusion, the present data suggests an enhanced process of osteoblast differentiation that can be exploited further to improve mESC differentiation.
Collapse
Affiliation(s)
- Swati Srivastava
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | | | | |
Collapse
|
20
|
Fong CY, Subramanian A, Gauthaman K, Venugopal J, Biswas A, Ramakrishna S, Bongso A. Human umbilical cord Wharton's jelly stem cells undergo enhanced chondrogenic differentiation when grown on nanofibrous scaffolds and in a sequential two-stage culture medium environment. Stem Cell Rev Rep 2012; 8:195-209. [PMID: 21671058 DOI: 10.1007/s12015-011-9289-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The current treatments used for osteoarthritis from cartilage damage have their disadvantages of donor site morbidity, complicated surgical interventions and risks of infection and graft rejection. Recent advances in tissue engineering have offered much promise in cartilage repair but the best cell source and in vitro system have not as yet been optimised. Human bone marrow mesenchymal stem cells (hBMSCs) have thus far been the cell of choice. However, we derived a unique stem cell from the human umbilical cord Wharton's jelly (hWJSC) that has properties superior to hBMSCs in terms of ready availability, prolonged stemness characteristics in vitro, high proliferation rates, wide multipotency, non-tumorigenicity and tolerance in allogeneic transplantation. We observed enhanced cell attachment, cell proliferation and chondrogenesis of hWJSCs over hBMSCs when grown on PCL/Collagen nanoscaffolds in the presence of a two-stage sequential complex/chondrogenic medium for 21 days. Improvement of these three parameters were confirmed via inverted optics, field emission scanning electron microscopy (FESEM), MTT assay, pellet diameters, Alcian blue histology and staining, glycosaminglycans (GAG) and hyaluronic acid production and expression of key chondrogenic genes (SOX9, Collagen type II, COMP, FMOD) using immunohistochemistry and real-time polymerase chain reaction (qRT-PCR). In separate experiments we demonstrated that the 16 ng/ml of basic fibroblast growth factor (bFGF) present in the complex medium may have contributed to driving chondrogenesis. We conclude that hWJSCs are an attractive stem cell source for inducing chondrogenesis in vitro when grown on nanoscaffolds and exposed sequentially first to complex medium and then followed by chondrogenic medium.
Collapse
Affiliation(s)
- Chui-Yee Fong
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | | | | | | | | | | | | |
Collapse
|
21
|
Stachelscheid H, Wulf-Goldenberg A, Eckert K, Jensen J, Edsbagge J, Björquist P, Rivero M, Strehl R, Jozefczuk J, Prigione A, Adjaye J, Urbaniak T, Bussmann P, Zeilinger K, Gerlach JC. Teratoma formation of human embryonic stem cells in three-dimensional perfusion culture bioreactors. J Tissue Eng Regen Med 2012; 7:729-41. [PMID: 22438087 DOI: 10.1002/term.1467] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 10/14/2011] [Accepted: 01/05/2012] [Indexed: 11/10/2022]
Abstract
Teratoma formation in mice is today the most stringent test for pluripotency that is available for human pluripotent cells, as chimera formation and tetraploid complementation cannot be performed with human cells. The teratoma assay could also be applied for assessing the safety of human pluripotent cell-derived cell populations intended for therapeutic applications. In our study we examined the spontaneous differentiation behaviour of human embryonic stem cells (hESCs) in a perfused 3D multi-compartment bioreactor system and compared it with differentiation of hESCs and human induced pluripotent cells (hiPSCs) cultured in vitro as embryoid bodies and in vivo in an experimental mouse model of teratoma formation. Results from biochemical, histological/immunohistological and ultrastuctural analyses revealed that hESCs cultured in bioreactors formed tissue-like structures containing derivatives of all three germ layers. Comparison with embryoid bodies and the teratomas revealed a high degree of similarity of the tissues formed in the bioreactor to these in the teratomas at the histological as well as transcriptional level, as detected by comparative whole-genome RNA expression profiling. The 3D culture system represents a novel in vitro model that permits stable long-term cultivation, spontaneous multi-lineage differentiation and tissue formation of pluripotent cells that is comparable to in vivo differentiation. Such a model is of interest, e.g. for the development of novel cell differentiation strategies. In addition, the 3D in vitro model could be used for teratoma studies and pluripotency assays in a fully defined, controlled environment, alternatively to in vivo mouse models.
Collapse
Affiliation(s)
- H Stachelscheid
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Obokata H, Yamato M, Tsuneda S, Okano T. Reproducible subcutaneous transplantation of cell sheets into recipient mice. Nat Protoc 2011; 6:1053-9. [PMID: 21720318 DOI: 10.1038/nprot.2011.356] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Perfecting tissue engineering and cell sheet transplantation is an important step toward realizing regenerative medicine and is a growing area of research. Before being applied to clinical settings, it is important that these approaches are evaluated in vivo. Here we provide a detailed protocol for handling thin cell sheets, for a simple and highly reproducible subcutaneous transplantation of cell sheets into mice, and for the histological examination of regenerated tissues. Various aspects of transplants can be assessed, such as maintenance, differentiation and proliferation. An emphasis is placed on surgical precision and reproducibility. The resulting consistency between surgeries helps minimize artifacts from surgical variation and therefore enables researchers to not only observe and compare the interactions between host tissues but also to compare transplants among different host animals. A single transplantation can be carried out within ∼10 min.
Collapse
Affiliation(s)
- Haruko Obokata
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | | | | | | |
Collapse
|
23
|
Ablation of Dido3 compromises lineage commitment of stem cells in vitro and during early embryonic development. Cell Death Differ 2011; 19:132-43. [PMID: 21660050 PMCID: PMC3252825 DOI: 10.1038/cdd.2011.62] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The death inducer obliterator (Dido) locus encodes three protein isoforms, of which Dido3 is the largest and most broadly expressed. Dido3 is a nuclear protein that forms part of the spindle assembly checkpoint (SAC) and is necessary for correct chromosome segregation in somatic and germ cells. Here we report that specific ablation of Dido3 function in mice causes lethal developmental defects at the onset of gastrulation. Although these defects are associated with centrosome amplification, spindle malformation and a DNA damage response, we provide evidence that embryonic lethality of the Dido3 mutation cannot be explained by its impact on chromosome segregation alone. We show that loss of Dido3 expression compromises differentiation of embryonic stem cells in vitro and of epiblast cells in vivo, resulting in early embryonic death at around day 8.5 of gestation. Close analysis of Dido3 mutant embryoid bodies indicates that ablation of Dido3, rather than producing a generalized differentiation blockade, delays the onset of lineage commitment at the primitive endoderm specification stage. The dual role of Dido3 in chromosome segregation and stem cell differentiation supports the implication of SAC components in stem cell fate decisions.
Collapse
|
24
|
Fong CY, Chak LL, Biswas A, Tan JH, Gauthaman K, Chan WK, Bongso A. Human Wharton's jelly stem cells have unique transcriptome profiles compared to human embryonic stem cells and other mesenchymal stem cells. Stem Cell Rev Rep 2011; 7:1-16. [PMID: 20602182 DOI: 10.1007/s12015-010-9166-x] [Citation(s) in RCA: 238] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The human umbilical cord that originates from the embryo is an extra-embryonic membrane and the Wharton's jelly within it is a rich source of stem cells (hWJSCs). It is not definitely known whether these cells behave as human embryonic stem cells (hESCs), human mesenchymal stem cells (hMSC) or both. They have the unique properties of high proliferation rates, wide multipotency, hypoimmunogenicity, do not induce teratomas and have anticancer properties. These advantages are important considerations for their use in cell based therapies and treatment of cancers. In a search for properties that confer these advantages we compared a detailed transcriptome profiling of hWJSCs using DNA microarrays with that of a panel of known hESCs, hMSCs and stromal cells. hWJSCs expressed low levels of the pluripotent embryonic stem cell markers including POUF1, NANOG, SOX2 and LIN28, thus explaining why they do not produce teratomas. Several cytokines were significantly upregulated in hWJSCs including IL12A which is associated with the induction of apoptosis, thus explaining their anticancer properties. When GO Biological Process analysis was compared between the various stem cell types, hWJSCs showed an increased expression of genes associated with the immune system, chemotaxis and cell death. The ability to modulate immune responses makes hWJSCs an important compatible stem cell source for transplantation therapy in allogeneic settings without immunorejection. The data in the present study which is the first detailed report on hWJSC transcriptomes provide a foundation for future functional studies where the exact mechanisms of these unique properties of hWJSCs can be confirmed.
Collapse
Affiliation(s)
- Chui-Yee Fong
- Department of Obstetrics and Gynaecology, National University of Singapore, Kent Ridge, Singapore
| | | | | | | | | | | | | |
Collapse
|
25
|
Handschel J, Naujoks C, Langenbach F, Berr K, Depprich RA, Ommerborn MA, Kübler NR, Brinkmann M, Kögler G, Meyer U. Comparison of ectopic bone formation of embryonic stem cells and cord blood stem cells in vivo. Tissue Eng Part A 2011; 16:2475-83. [PMID: 20214449 DOI: 10.1089/ten.tea.2009.0546] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cell-based reconstruction therapies promise new therapeutic opportunities for bone regeneration. Unrestricted somatic stem cells (USSC) from cord blood and embryonic stem cells (ESCs) can be differentiated into osteogenic cells. The purpose of this in vivo study was to compare their ability to induce ectopic bone formation in vivo. Human USSCs and murine ESCs were cultured as both monolayer cultures and micromasses and seeded on insoluble collagenous bone matrix (ICBM). One week and 1, 2, and 3 months after implanting the constructs in immune-deficient rats, computed tomography scans were performed to detect any calcification. Subsequently, the implanted constructs were examined histologically. The radiological examination showed a steep increase in the mineralized bone-like tissue in the USSC groups. This increase can be considered as statistically significant compared to the basic value. Moreover, the volume and the calcium portion measured by computed tomography scans were about 10 times higher than in the ESC group. The volume of mineralization in the ESC group increased to a much smaller extent over the course of time, and the control group (ICBM without cells) showed almost no alterations during the study. The histological examinations parallel the radiological findings. Cord blood stem cells in combination with ICBM-induced ectopic bone formation in vivo are stronger than ESCs.
Collapse
Affiliation(s)
- Jörg Handschel
- Department of Cranio- and Maxillofacial Surgery, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Castro CA, Ben-Yehudah A, Ozolek JA, Mills PH, Redinger CJ, Mich-Basso JD, McFarland DA, Oliver SL, Ahrens ET, Schatten G. Semiquantitative histopathology and 3D magnetic resonance microscopy as collaborative platforms for tissue identification and comparison within teratomas derived from pedigreed primate embryonic stem cells. Stem Cell Res 2010; 5:201-11. [PMID: 20864427 DOI: 10.1016/j.scr.2010.07.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Revised: 07/28/2010] [Accepted: 07/29/2010] [Indexed: 12/19/2022] Open
Abstract
Teratoma formation in xenografts is a sufficiently stringent pluripotency assay for stem cells. However, little is known about the composition and spatial relationships of tissues within teratomas that may provide clues about development and platforms for studying organ development. Additionally, teratoma formation and analysis lack standards for reporting as assays of pluripotency. Three of 27 total teratomas derived from pedigreed primate embryonic stem cells underwent quantitative three-dimensional high-resolution magnetic resonance microscopy (MRM). Teratomas were subsequently serially sectioned and tissue types identified, semiquantitated, and correlated with MRM images. All teratomas demonstrated tissue derivatives from the three germ layers and approximately 23 different tissue types were identified. Certain tissue groups attempted to form organs more frequently (e.g., trachea/bronchi, small intestine). MRM discriminated some tissues readily (e.g., bone, adipose, cartilage) while other tissue types with like MR intensities could not be distinguished. Semiquantitative histopathological analysis of teratomas demonstrates the ability to delineate multiple tissues as derived from ectoderm, mesoderm, or endoderm and to use this information for comparison to other teratomas. MRM provides rapid quantitative imaging of intact teratomas that complements histology and identifies sites of interest for additional biological studies.
Collapse
Affiliation(s)
- Carlos A Castro
- Pittsburgh Development Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Gerlach JC, Lübberstedt M, Edsbagge J, Ring A, Hout M, Baun M, Rossberg I, Knöspel F, Peters G, Eckert K, Wulf-Goldenberg A, Björquist P, Stachelscheid H, Urbaniak T, Schatten G, Miki T, Schmelzer E, Zeilinger K. Interwoven four-compartment capillary membrane technology for three-dimensional perfusion with decentralized mass exchange to scale up embryonic stem cell culture. Cells Tissues Organs 2010; 192:39-49. [PMID: 20197653 DOI: 10.1159/000291014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2009] [Indexed: 01/31/2023] Open
Abstract
We describe hollow fiber-based three-dimensional (3D) dynamic perfusion bioreactor technology for embryonic stem cells (ESC) which is scalable for laboratory and potentially clinical translation applications. We added 2 more compartments to the typical 2-compartment devices, namely an additional media capillary compartment for countercurrent 'arteriovenous' flow and an oxygenation capillary compartment. Each capillary membrane compartment can be perfused independently. Interweaving the 3 capillary systems to form repetitive units allows bioreactor scalability by multiplying the capillary units and provides decentralized media perfusion while enhancing mass exchange and reducing gradient distances from decimeters to more physiologic lengths of <1 mm. The exterior of the resulting membrane network, the cell compartment, is used as a physically active scaffold for cell aggregation; adjusting intercapillary distances enables control of the size of cell aggregates. To demonstrate the technology, mouse ESC (mESC) were cultured in 8- or 800-ml cell compartment bioreactors. We were able to confirm the hypothesis that this bioreactor enables mESC expansion qualitatively comparable to that obtained with Petri dishes, but on a larger scale. To test this, we compared the growth of 129/SVEV mESC in static two-dimensional Petri dishes with that in 3D perfusion bioreactors. We then tested the feasibility of scaling up the culture. In an 800-ml prototype, we cultured approximately 5 x 10(9) cells, replacing up to 800 conventional 100-mm Petri dishes. Teratoma formation studies in mice confirmed protein expression and gene expression results with regard to maintaining 'stemness' markers during cell expansion.
Collapse
Affiliation(s)
- Jörg C Gerlach
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15203, USA. joerg.gerlach @ cellnet.org
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Ruiz-Vela A, Aguilar-Gallardo C, Simón C. Building a Framework for Embryonic Microenvironments and Cancer Stem Cells. Stem Cell Rev Rep 2009; 5:319-27. [DOI: 10.1007/s12015-009-9096-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
29
|
Fong CY, Chak LL, Subramanian A, Tan JH, Biswas A, Gauthaman K, Choolani M, Chan WK, Bongso A. A three dimensional anchorage independent in vitro system for the prolonged growth of embryoid bodies to study cancer cell behaviour and anticancer agents. Stem Cell Rev Rep 2009; 5:410-9. [PMID: 20058203 DOI: 10.1007/s12015-009-9092-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Accepted: 08/26/2009] [Indexed: 12/31/2022]
Abstract
We describe a three dimensional (3D) anchorage independent in vitro protocol for the prolonged growth of human embryoid bodies (EBs) up to 90 days. We grew hESCs (46XX) in methylcellulose (MC) in motion culture in the presence of EB medium (EB), EB medium with Matrigel (EB + MAT), bulk culture medium (BCM), and BCM medium with Matrigel (BCM + MAT). All four experimental groups produced embryoid bodies (EBs) which with prolonged growth to 90 days acquired blood vessels and tissues from all three germ layers. Based on histology, microarray gene expression profiles and the definition for experimental teratomas, we could classify the EBs into early EBs, mature EBs and teratomas. The EB + MAT group produced the highest number of teratomas and their microarray data suggested the presence of inductive microenvironment niches and activation of pathways for self-organization, morphogenesis and growth. When we microinjected hepatocarcinoma-Green Fluorescent Protein cells (HepG2-GFP) (46XY) into the teratomas, after 10 days the HepG2-GFP cells had grown inside the teratoma as confirmed by confocal microscopy and SRY gene analysis. This 3D-MC-(EB + MAT) in vitro system requires few cells to produce many teratomas, can be used to test pluripotency of potential human embryonic and induced pluripotent stem cell lines (hESC, hiPSC), and is an experimental humanized platform to study cancer cell behavior.
Collapse
Affiliation(s)
- Chui-Yee Fong
- Department of Obstetrics & Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore 119074
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
The Use of Discontinuous Density Gradients in Stem Cell Research and Application. Stem Cell Rev Rep 2009; 5:428-34. [DOI: 10.1007/s12015-009-9090-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Accepted: 08/14/2009] [Indexed: 02/03/2023]
|
31
|
Zhu S, Wurdak H, Wang J, Lyssiotis CA, Peters EC, Cho CY, Wu X, Schultz PG. A Small Molecule Primes Embryonic Stem Cells for Differentiation. Cell Stem Cell 2009; 4:416-26. [DOI: 10.1016/j.stem.2009.04.001] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Revised: 02/24/2009] [Accepted: 04/01/2009] [Indexed: 12/25/2022]
|
32
|
Bongso A, Fong CY, Gauthaman K. Taking stem cells to the clinic: Major challenges. J Cell Biochem 2009; 105:1352-60. [PMID: 18980213 DOI: 10.1002/jcb.21957] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Stem cell therapy offers tremendous promise in the treatment of many incurable diseases. A variety of stem cell types are being studied but human embryonic stem cells (hESCs) appear to be the most versatile as they are pluripotent and can theoretically differentiate into all the tissues of the human body via the three primordial germ layers and the male and female germ lines. Currently, hESCs have been successfully converted in vitro into functional insulin secreting islets, cardiomyocytes, and neuronal cells and transfer of such cells into diabetic, ischaemic, and parkinsonian animal models respectively have shown successful engraftment. However, hESC-derived tissue application in the human is fraught with the problems of ethics, immunorejection, tumorigenesis from rogue undifferentiated hESCs, and inadequate cell numbers because of long population doubling times in hESCs. Human mesenchymal stem cells (hMSC) though not tumorigenic, also have their limitations of multipotency, immunorejection, and are currently confined to autologous transplantation with the genuine benefits in allogeneic settings not conclusively shown in large controlled human trials. Human Wharton's jelly stem cells (WJSC) from the umbilical cord matrix which are of epiblast origin and containing both hESC and hMSC markers appear to be less troublesome in not being an ethically controversial source, widely multipotent, not tumorigenic, maintain "stemness" for several serial passages and because of short population doubling time can be scaled up in large numbers. This report describes in detail the hurdles all these stem cell types have to overcome before stem cell-based therapy becomes a genuine reality.
Collapse
Affiliation(s)
- Ariff Bongso
- Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore 119074, Singapore.
| | | | | |
Collapse
|
33
|
Separation of SSEA-4 and TRA-1-60 labelled undifferentiated human embryonic stem cells from a heterogeneous cell population using magnetic-activated cell sorting (MACS) and fluorescence-activated cell sorting (FACS). Stem Cell Rev Rep 2009; 5:72-80. [PMID: 19184635 DOI: 10.1007/s12015-009-9054-4] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Accepted: 01/21/2009] [Indexed: 10/21/2022]
Abstract
A major concern in human embryonic stem cell (hESC)-derived cell replacement therapy is the risk of tumorigenesis from undifferentiated hESCs residing in the population of hESC-derived cells. Separation of these undifferentiated hESCs from the differentiated derivatives using cell sorting methods may be a plausible approach in overcoming this problem. We therefore explored magnetic activated cell sorting (MACS) and fluorescence activated cell sorting (FACS) to separate labelled undifferentiated hESCs from a heterogeneous population of hESCs and hepatocellular carcinoma cells (HepG2) deliberately mixed respectively at different ratios (10:90, 20:80, 30:70, 40:60 and 50:50) to mimic a standard in vitro differentiation protocol, instead of using a hESC-differentiated cell population, so that we could be sure of the actual number of cells separated. HES-3 and HES-4 cells were labelled in separate experiments for the stem cell markers SSEA-4 and TRA-1-60 using primary antibodies. Anti-PE magnetic microbeads that recognize the PE-conjugated SSEA-4 labelled hESCs was added to the heterogeneous cell mixture and passed through the MACS column. The cells that passed through the column ('flow-through' fraction) and those retained ('labelled' fraction') were subsequently analysed using FACS. The maximum efficacy of hESCs retention using MACS was 81.0 +/- 2.9% (HES-3) and 83.6 +/- 4.2% (HES-4). Using FACS, all the undifferentiated hESCs labelled with the two cell-surface markers could be removed by selective gating. Both hESCs and HepG2 cells in the 'flow-through' fraction following MACS separation were viable in culture whereas by FACS separation only the HepG2 cells were viable. FACS efficiently helps to eliminate the undifferentiated hESCs based on their cell-surface antigens expressed.
Collapse
|
34
|
Quantitative identification of teratoma tissues formed by human embryonic stem cells with TeratomEye. Biotechnol Lett 2009; 31:653-8. [DOI: 10.1007/s10529-009-9928-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Revised: 12/15/2008] [Accepted: 12/19/2008] [Indexed: 10/21/2022]
|
35
|
Denker HW. Induced pluripotent stem cells: how to deal with the developmental potential. Reprod Biomed Online 2009; 19 Suppl 1:34-7. [DOI: 10.1016/s1472-6483(10)60062-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|