1
|
Cucinella G, Gullo G, Catania E, Perino A, Billone V, Marinelli S, Napoletano G, Zaami S. Stem Cells and Infertility: A Review of Clinical Applications and Legal Frameworks. J Pers Med 2024; 14:135. [PMID: 38392569 PMCID: PMC10890184 DOI: 10.3390/jpm14020135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Infertility is a condition defined by the failure to establish a clinical pregnancy after 12 months of regular, unprotected sexual intercourse or due to an impairment of a person's capacity to reproduce either as an individual or with their partner. The authors have set out to succinctly investigate, explore, and assess infertility treatments, harnessing the potential of stem cells to effectively and safely treat infertility; in addition, this paper will present the legal and regulatory complexities at the heart of stem cell research, with an overview of the legislative state of affairs in six major European countries. For couples who cannot benefit from assisted reproductive technologies (ART) to treat their infertility, stem-cells-based approaches have been shown to be a highly promising approach. Nonetheless, lingering ethical and immunological uncertainties require more conclusive findings and data before such treatment avenues can become mainstream and be applied on a large scale. The isolation of human embryonic stem cells (ESCs) is ethically controversial, since their collection involves the destruction of human embryonic tissue. Overall, stem cell research has resulted in important new breakthroughs in the treatment of infertility. The effort to untangle the complex web of ethical and legal issues associated with such therapeutic approaches will have to rely on evidence-based, broadly shared standards, guidelines, and best practices to make sure that the procreative rights of patients can be effectively reconciled with the core values at the heart of medical ethics.
Collapse
Affiliation(s)
- Gaspare Cucinella
- IVF Unit, Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, University of Palermo, 90146 Palermo, Italy
| | - Giuseppe Gullo
- IVF Unit, Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, University of Palermo, 90146 Palermo, Italy
| | - Erika Catania
- IVF Unit, Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, University of Palermo, 90146 Palermo, Italy
| | - Antonio Perino
- IVF Unit, Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, University of Palermo, 90146 Palermo, Italy
| | - Valentina Billone
- IVF Unit, Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, University of Palermo, 90146 Palermo, Italy
| | | | - Gabriele Napoletano
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Simona Zaami
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, "Sapienza" University of Rome, 00161 Rome, Italy
| |
Collapse
|
2
|
Kolagar TA, Farzaneh M, Nikkar N, Khoshnam SE. Human Pluripotent Stem Cells in Neurodegenerative Diseases: Potentials, Advances and Limitations. Curr Stem Cell Res Ther 2020; 15:102-110. [PMID: 31441732 DOI: 10.2174/1574888x14666190823142911] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/15/2019] [Accepted: 07/17/2019] [Indexed: 12/14/2022]
Abstract
Neurodegenerative diseases are progressive and uncontrolled gradual loss of motor neurons function or death of neuron cells in the central nervous system (CNS) and the mechanisms underlying their progressive nature remain elusive. There is urgent need to investigate therapeutic strategies and novel treatments for neural regeneration in disorders like Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). Currently, the development and identification of pluripotent stem cells enabling the acquisition of a large number of neural cells in order to improve cell recovery after neurodegenerative disorders. Pluripotent stem cells which consist of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) are characterized by their ability to indefinitely self-renew and the capacity to differentiate into different types of cells. The first human ESC lines were established from donated human embryos; while, because of a limited supply of donor embryos, human ESCs derivation remains ethically and politically controversial. Hence, hiPSCs-based therapies have been shown as an effective replacement for human ESCs without embryo destruction. Compared to the invasive methods for derivation of human ESCs, human iPSCs has opened possible to reprogram patient-specific cells by defined factors and with minimally invasive procedures. Human pluripotent stem cells are a good source for cell-based research, cell replacement therapies and disease modeling. To date, hundreds of human ESC and human iPSC lines have been generated with the aim of treating various neurodegenerative diseases. In this review, we have highlighted the recent potentials, advances, and limitations of human pluripotent stem cells for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Tannaz Akbari Kolagar
- Faculty of Biological Sciences, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Negin Nikkar
- Department of Biology, Faculty of Sciences, Alzahra University, Tehran, Iran
| | - Seyed Esmaeil Khoshnam
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
3
|
Zhang PY, Fan Y, Tan T, Yu Y. Generation of Artificial Gamete and Embryo From Stem Cells in Reproductive Medicine. Front Bioeng Biotechnol 2020; 8:781. [PMID: 32793569 PMCID: PMC7387433 DOI: 10.3389/fbioe.2020.00781] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/19/2020] [Indexed: 12/20/2022] Open
Abstract
In addition to the great growing need for assisted reproduction technologies (ART), additional solutions for patients without functional gametes are strongly needed. Due to ethical restrictions, limited studies can be performed on human gametes and embryos; however, artificial gametes and embryos represent a new hope for clinical application and basic research in the field of reproductive medicine. Here, we provide a review of the research progress and possible application of artificial gametes and embryos from different species, including mice, monkeys and humans. Gametes specification from adult stem cells and embryonic stem cells (ESCs) as well as propagation of stem cells from the reproductive system and from organized embryos, which are similar to blastocysts, have been realized in some nonhuman mammals, but not all achievements can be replicated in humans. This area of research remains noteworthy and requires further study and effort to achieve the reconstitution of the entire cycle of gametogenesis and embryo development in vitro.
Collapse
Affiliation(s)
- Pu-Yao Zhang
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tao Tan
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China.,Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Yang Yu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
4
|
Isolation, Culture, and Functional Characterization of Human Embryonic Stem Cells: Current Trends and Challenges. Stem Cells Int 2018; 2018:1429351. [PMID: 30254679 PMCID: PMC6142731 DOI: 10.1155/2018/1429351] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/19/2018] [Accepted: 07/30/2018] [Indexed: 12/12/2022] Open
Abstract
Human embryonic stem cells (hESCs) hold great potential for the treatment of various degenerative diseases. Pluripotent hESCs have a great ability to undergo unlimited self-renewal in culture and to differentiate into all cell types in the body. The journey of hESC research is not that smooth, as it has faced several challenges which are limited to not only tumor formation and immunorejection but also social, ethical, and political aspects. The isolation of hESCs from the human embryo is considered highly objectionable as it requires the destruction of the human embryo. The issue was debated and discussed in both public and government platforms, which led to banning of hESC research in many countries around the world. The banning has negatively affected the progress of hESC research as many federal governments around the world stopped research funding. Afterward, some countries lifted the ban and allowed the funding in hESC research, but the damage has already been done on the progress of research. Under these unfavorable conditions, still some progress was made to isolate, culture, and characterize hESCs using different strategies. In this review, we have summarized various strategies used to successfully isolate, culture, and characterize hESCs. Finally, hESCs hold a great promise for clinical applications with proper strategies to minimize the teratoma formation and immunorejection and better cell transplantation strategies.
Collapse
|
5
|
HLA and Histo-Blood Group Antigen Expression in Human Pluripotent Stem Cells and their Derivatives. Sci Rep 2017; 7:13072. [PMID: 29026098 PMCID: PMC5638960 DOI: 10.1038/s41598-017-12231-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/06/2017] [Indexed: 01/03/2023] Open
Abstract
One prerequisite for a successful clinical outcome of human pluripotent stem cell (hPSC) based therapies is immune compatibility between grafted cells/tissue and recipient. This study explores immune determinants of human embryonic stem cell lines (hESC) and induced human pluripotent stem cell (hiPSC) lines and hepatocyte- and cardiomyocyte-like cells derived from these cells. HLA class I was expressed on all pluripotent hPSC lines which upon differentiation into hepatocyte-like cells was considerably reduced in contrast to cardiomyocyte-like cells which retained class I antigens. No HLA class II antigens were found in the pluripotent or differentiated cells. Histo-blood group carbohydrate antigens SSEA-3/SSEA-4/SSEA-5, Globo H, A, Lex/Ley and sialyl-lactotetra were expressed on all hPSC lines. Blood group AB(O)H antigen expression was in accordance with ABO genotype. Interestingly, only a subpopulation of A1O1 cells expressed A. During differentiation of hPSC, some histo-blood group antigens showed congruent alteration patterns while expression of other antigens differed between the cell lines. No systematic difference in the hPSC cell surface tissue antigen expression was detected. In conclusion, hPSC and their derivatives express cell surface antigens that may cause an immune rejection. Furthermore, tissue antigen expression must be established for each individual stem cell line prior to clinical application.
Collapse
|
6
|
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
|
7
|
One Standardized Differentiation Procedure Robustly Generates Homogenous Hepatocyte Cultures Displaying Metabolic Diversity from a Large Panel of Human Pluripotent Stem Cells. Stem Cell Rev Rep 2016; 12:90-104. [PMID: 26385115 DOI: 10.1007/s12015-015-9621-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Human hepatocytes display substantial functional inter-individual variation regarding drug metabolizing functions. In order to investigate if this diversity is mirrored in hepatocytes derived from different human pluripotent stem cell (hPSC) lines, we evaluated 25 hPSC lines originating from 24 different donors for hepatic differentiation and functionality. Homogenous hepatocyte cultures could be derived from all hPSC lines using one standardized differentiation procedure. To the best of our knowledge this is the first report of a standardized hepatic differentiation procedure that is generally applicable across a large panel of hPSC lines without any adaptations to individual lines. Importantly, with regard to functional aspects, such as Cytochrome P450 activities, we observed that hepatocytes derived from different hPSC lines displayed inter-individual variation characteristic for primary hepatocytes obtained from different donors, while these activities were highly reproducible between repeated experiments using the same line. Taken together, these data demonstrate the emerging possibility to compile panels of hPSC-derived hepatocytes of particular phenotypes/genotypes relevant for drug metabolism and toxicity studies. Moreover, these findings are of significance for applications within the regenerative medicine field, since our stringent differentiation procedure allows the derivation of homogenous hepatocyte cultures from multiple donors which is a prerequisite for the realization of future personalized stem cell based therapies.
Collapse
|
8
|
Galán A, Simón C. Monitoring Stemness in Long-Term hESC Cultures by Real-Time PCR. Methods Mol Biol 2016; 1307:89-104. [PMID: 25403464 DOI: 10.1007/7651_2014_131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Human embryonic stem cells (hESC) involve long-term cultures that must remain undifferentiated. The real-time PCR (RT-PCR) technique allows the relative quantification of target genes, including undifferentiation and differentiation markers when referred to a housekeeping control with the addition of a calibrator that serves as an internal control to compare different lots of reactions during the time. The main aspects will include a minimal number of cells to be analyzed, genes to be tested, and how to choose the appropriate calibrator sample and the reference gene. In this chapter, we present how to apply the RT-PCR technique, protocols for its performance, experimental setup, and software analysis, as of the gene expression of hESC lines in consecutive passages for long-term culture surveillance.
Collapse
Affiliation(s)
- Amparo Galán
- Gene Expression and RNA Metabolism, Prince Felipe Research Center (CIPF), Valencia, Spain
| | | |
Collapse
|
9
|
Tolosa L, Caron J, Hannoun Z, Antoni M, López S, Burks D, Castell JV, Weber A, Gomez-Lechon MJ, Dubart-Kupperschmitt A. Transplantation of hESC-derived hepatocytes protects mice from liver injury. Stem Cell Res Ther 2015; 6:246. [PMID: 26652177 PMCID: PMC4676869 DOI: 10.1186/s13287-015-0227-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 10/25/2015] [Accepted: 11/06/2015] [Indexed: 12/25/2022] Open
Abstract
Background Hepatic cell therapy has become a viable alternative to liver transplantation for life-threatening liver diseases. However, the supply of human hepatocytes is limited due to the shortage of suitable donor organs required to isolate high-quality cells. Human pluripotent stem cells reflect a potential renewable source for generating functional hepatocytes. However, most differentiation protocols use undefined matrices or factors of animal origin; as such, the resulting hepatocytes are not Good Manufacturing Practice compliant. Moreover, the preclinical studies employed to assess safety and function of human embryonic stem cell (hESC)-derived hepatocytes are generally limited to immunodeficient mice. In the present study, we evaluate the generation of hepatocytes under defined conditions using a European hESC line (VAL9) which was derived under animal-free conditions. The function capacity of VAL9-derived hepatocytes was assessed by transplantation into mice with acetaminophen-induced acute liver failure, a clinically relevant model. Methods We developed a protocol that successfully differentiates hESCs into bipotent hepatic progenitors under defined conditions, without the use of chromatin modifiers such as dimethyl sulphoxide. These progenitors can be cryopreserved and are able to generate both committed precursors of cholangiocytes and neonate-like hepatocytes. Results Thirty days post-differentiation, hESCs expressed hepatocyte-specific markers such as asialoglycoprotein receptor and hepatic nuclear factors including HNF4α. The cells exhibited properties of mature hepatocytes such as urea secretion and UGT1A1 and cytochrome P450 activities. When transplanted into mice with acetaminophen-induced acute liver failure, a model of liver damage, the VAL9-derived hepatocytes efficiently engrafted and proliferated, repopulating up to 10 % of the liver. In these transplanted livers, we observed a significant decrease of liver transaminases and found no evidence of tumourigenicity. Thus, VAL9-derived hepatocytes were able to rescue hepatic function in acetaminophen-treated animals. Conclusions Our study reveals an efficient protocol for differentiating VAL9 hESCs to neonatal hepatocytes which are then able to repopulate livers in vivo without tumour induction. The human hepatocytes are able to rescue liver function in mice with acetaminophen-induced acute toxicity. These results provide proof-of-concept that replacement therapies using hESC-derived hepatocytes are effective for treating liver diseases. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0227-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laia Tolosa
- INSERM, U 1193, Hôpital Paul Brousse, Villejuif, F-94807, France. .,Unidad de Hepatología Experimental, IIS LA Fe, Valencia, S-46026, Spain. .,Univ Paris-Sud, UMR-S 1193, Villejuif, F-94800, France. .,DHU Hepatinov, Villejuif, F-94800, France.
| | - Jérôme Caron
- INSERM, U 1193, Hôpital Paul Brousse, Villejuif, F-94807, France. .,Univ Paris-Sud, UMR-S 1193, Villejuif, F-94800, France. .,DHU Hepatinov, Villejuif, F-94800, France.
| | - Zara Hannoun
- INSERM, U 1193, Hôpital Paul Brousse, Villejuif, F-94807, France. .,Univ Paris-Sud, UMR-S 1193, Villejuif, F-94800, France. .,DHU Hepatinov, Villejuif, F-94800, France.
| | - Marc Antoni
- INSERM, U 1193, Hôpital Paul Brousse, Villejuif, F-94807, France. .,Univ Paris-Sud, UMR-S 1193, Villejuif, F-94800, France. .,DHU Hepatinov, Villejuif, F-94800, France.
| | - Silvia López
- Unidad de Hepatología Experimental, IIS LA Fe, Valencia, S-46026, Spain.
| | - Deborah Burks
- CIBERDEM, Centro de Investigacion Prıncipe Felipe, Valencia, S-46012, Spain.
| | - Jose Vicente Castell
- Unidad de Hepatología Experimental, IIS LA Fe, Valencia, S-46026, Spain. .,CIBERehd, FIS, Barcelona, S-08036, Spain.
| | - Anne Weber
- INSERM, U 1193, Hôpital Paul Brousse, Villejuif, F-94807, France. .,Univ Paris-Sud, UMR-S 1193, Villejuif, F-94800, France. .,DHU Hepatinov, Villejuif, F-94800, France.
| | - Maria-Jose Gomez-Lechon
- Unidad de Hepatología Experimental, IIS LA Fe, Valencia, S-46026, Spain. .,CIBERehd, FIS, Barcelona, S-08036, Spain.
| | - Anne Dubart-Kupperschmitt
- INSERM, U 1193, Hôpital Paul Brousse, Villejuif, F-94807, France. .,Univ Paris-Sud, UMR-S 1193, Villejuif, F-94800, France. .,DHU Hepatinov, Villejuif, F-94800, France.
| |
Collapse
|
10
|
Moreno I, Míguez-Forjan JM, Simón C. Artificial gametes from stem cells. Clin Exp Reprod Med 2015; 42:33-44. [PMID: 26161331 PMCID: PMC4496429 DOI: 10.5653/cerm.2015.42.2.33] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 06/18/2015] [Accepted: 06/18/2015] [Indexed: 11/06/2022] Open
Abstract
The generation of artificial gametes is a real challenge for the scientific community today. In vitro development of human eggs and sperm will pave the way for the understanding of the complex process of human gametogenesis and will provide with human gametes for the study of infertility and the onset of some inherited disorders. However, the great promise of artificial gametes resides in their future application on reproductive treatments for all these people wishing to have genetically related children and for which gamete donation is now their unique option of parenthood. This is the case of infertile patients devoid of suitable gametes, same sex couples, singles and those fertile couples in a high risk of transmitting serious diseases to their progeny. In the search of the best method to obtain artificial gametes, many researchers have successfully obtained human germ cell-like cells from stem cells at different stages of differentiation. In the near future, this field will evolve to new methods providing not only viable but also functional and safe artificial germ cells. These artificial sperm and eggs should be able to recapitulate all the genetic and epigenetic processes needed for the correct gametogenesis, fertilization and embryogenesis leading to the birth of a healthy and fertile newborn.
Collapse
Affiliation(s)
- Inmaculada Moreno
- Department of Research and Development, Igenomix S.L., Paternam, Spain
| | | | - Carlos Simón
- Department of Research and Development, Igenomix S.L., Paternam, Spain. ; Fundación Instituto Valenciano de Infertilidad (FIVI), Valencia, Spain. ; Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
11
|
Ghasemi-Dehkordi P, Allahbakhshian-Farsani M, Abdian N, Mirzaeian A, Saffari-Chaleshtori J, Heybati F, Mardani G, Karimi-Taghanaki A, Doosti A, Jami MS, Abolhasani M, Hashemzadeh-Chaleshtori M. Comparison between the cultures of human induced pluripotent stem cells (hiPSCs) on feeder-and serum-free system (Matrigel matrix), MEF and HDF feeder cell lines. J Cell Commun Signal 2015; 9:233-46. [PMID: 25820945 DOI: 10.1007/s12079-015-0289-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 03/17/2015] [Indexed: 11/27/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are a type of pluripotent stem cells artificially derived from an adult somatic cell (typically human fibroblast) by forced expression of specific genes. In recent years, different feeders like inactivated mouse embryonic fibroblasts (MEFs), human dermal fibroblasts (HDFs), and feeder free system have commonly been used for supporting the culture of stem cells in undifferentiated state. In the present work, the culture of hiPSCs and their characterizations on BD Matrigel (feeder-and serum-free system), MEF and HDF feeders using cell culture methods and molecular techniques were evaluated and compared. The isolated HDFs from foreskin samples were reprogrammed to hiPSCs using gene delivery system. Then, the pluripotency ability of hiPSCs cultured on each layer was determined by teratoma formation and immunohistochemical staining. After EBs generation the expression level of three germ layers genes were evaluated by Q-real-time PCR. Also, the cytogenetic stability of hiPSCs cultured on each condition was analyzed by karyotyping and comet assay. Then, the presence of pluripotency antigens were confirmed by Immunocytochemistry (ICC) test and alkaline phosphatase staining. This study were showed culturing of hiPSCs on BD Matrigel, MEF and HDF feeders had normal morphology and could maintain in undifferentiated state for prolonged expansion. The hiPSCs cultured in each system had normal karyotype without any chromosomal abnormalities and the DNA lesions were not observed by comet assay. Moreover, up-regulation in three germ layers genes in cultured hiPSCs on each layer (same to ESCs) compare to normal HDFs were observed (p < 0.05). The findings of the present work were showed in stem cells culturing especially hiPSCs both MEF and HDF feeders as well as feeder free system like Matrigel are proper despite benefits and disadvantages. Although, MEFs is suitable for supporting of stem cell culturing but it can animal pathogens transferring and inducing immune response. Furthermore, HDFs have homologous source with hiPSCs and can be used as feeder instead of MEF but in therapeutic approaches the cells contamination is a problem. So, this study were suggested feeder free culturing of hiPSCs on Matrigel in supplemented media (without using MEF conditioned medium) resolves these problems and could prepare easy applications of hiPSCs in therapeutic approaches of regenerative medicine such as stem-cell therapy and somatic cell nuclear in further researches.
Collapse
Affiliation(s)
- Payam Ghasemi-Dehkordi
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Rahmatieh, 8813833435, Shahrekord, Iran
| | - Mehdi Allahbakhshian-Farsani
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Narges Abdian
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Rahmatieh, 8813833435, Shahrekord, Iran
| | - Amin Mirzaeian
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Rahmatieh, 8813833435, Shahrekord, Iran
| | | | - Fatemeh Heybati
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Rahmatieh, 8813833435, Shahrekord, Iran
| | - Gashtasb Mardani
- Clinical Biochemistry Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Alireza Karimi-Taghanaki
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Rahmatieh, 8813833435, Shahrekord, Iran
| | - Abbas Doosti
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Mohammad-Saeid Jami
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Rahmatieh, 8813833435, Shahrekord, Iran
| | - Marziyeh Abolhasani
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Rahmatieh, 8813833435, Shahrekord, Iran
| | | |
Collapse
|
12
|
Desai N, Rambhia P, Gishto A. Human embryonic stem cell cultivation: historical perspective and evolution of xeno-free culture systems. Reprod Biol Endocrinol 2015; 13:9. [PMID: 25890180 PMCID: PMC4351689 DOI: 10.1186/s12958-015-0005-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 02/09/2015] [Indexed: 01/23/2023] Open
Abstract
Human embryonic stem cells (hESC) have emerged as attractive candidates for cell-based therapies that are capable of restoring lost cell and tissue function. These unique cells are able to self-renew indefinitely and have the capacity to differentiate in to all three germ layers (ectoderm, endoderm and mesoderm). Harnessing the power of these pluripotent stem cells could potentially offer new therapeutic treatment options for a variety of medical conditions. Since the initial derivation of hESC lines in 1998, tremendous headway has been made in better understanding stem cell biology and culture requirements for maintenance of pluripotency. The approval of the first clinical trials of hESC cells for treatment of spinal cord injury and macular degeneration in 2010 marked the beginning of a new era in regenerative medicine. Yet it was clearly recognized that the clinical utility of hESC transplantation was still limited by several challenges. One of the most immediate issues has been the exposure of stem cells to animal pathogens, during hESC derivation and during in vitro propagation. Initial culture protocols used co-culture with inactivated mouse fibroblast feeder (MEF) or human feeder layers with fetal bovine serum or alternatively serum replacement proteins to support stem cell proliferation. Most hESC lines currently in use have been exposed to animal products, thus carrying the risk of xeno-transmitted infections and immune reaction. This mini review provides a historic perspective on human embryonic stem cell culture and the evolution of new culture models. We highlight the challenges and advances being made towards the development of xeno-free culture systems suitable for therapeutic applications.
Collapse
Affiliation(s)
- Nina Desai
- Department of Obstetrics and Gynecology, Cleveland Clinic, Beachwood, OH, USA.
| | - Pooja Rambhia
- Department of Obstetrics and Gynecology, Cleveland Clinic, Beachwood, OH, USA.
| | - Arsela Gishto
- Department of Obstetrics and Gynecology, Cleveland Clinic, Beachwood, OH, USA.
| |
Collapse
|
13
|
Mas A, Cervelló I, Fernández-Álvarez A, Faus A, Díaz A, Burgués O, Casado M, Simón C. Overexpression of the truncated form of High Mobility Group A proteins (HMGA2) in human myometrial cells induces leiomyoma-like tissue formation. Mol Hum Reprod 2014; 21:330-8. [PMID: 25542836 DOI: 10.1093/molehr/gau114] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 12/17/2014] [Indexed: 12/24/2022] Open
Abstract
The pathogenesis of uterine leiomyomas, the most common benign tumor in women, is still unknown. This lack of basic knowledge limits the development of novel non-invasive therapies. Our group has previously demonstrated that leiomyoma side population (SP) cells are present in tumor lesions and act like putative tumor-initiating stem cells in human leiomyoma. Moreover, accumulated evidence demonstrates that these benign tumors of mesenchymal origin are characterized by rearrangements of the High Mobility Group A proteins (HMGA). In this work, we tested the hypothesis that leiomyoma development may be due to overexpression of HMGA2 (encoding high mobility group AT-hook2) in myometrial stem cells using in vitro and in vivo approaches. Our work demonstrates that the truncated/short form of HMGA2 induces myometrial cell transformation toward putative tumor-initiating leiomyoma cells and opens up new possibilities to understand the origin of leiomyomas and the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Aymara Mas
- Fundación IVI, Instituto Universitario IVI, Department of Obstetrics & Gynecology, School of Medicine, University of Valencia, INCLIVA, Valencia, Spain
| | - Irene Cervelló
- Fundación IVI, Instituto Universitario IVI, Department of Obstetrics & Gynecology, School of Medicine, University of Valencia, INCLIVA, Valencia, Spain
| | | | - Amparo Faus
- Fundación IVI, Instituto Universitario IVI, Department of Obstetrics & Gynecology, School of Medicine, University of Valencia, INCLIVA, Valencia, Spain
| | - Ana Díaz
- Central Service for Experimental Research, University of Valencia, INCLIVA, Valencia, Spain
| | - Octavio Burgués
- Department of Pathology, Hospital Clínico Universitario of Valencia, Valencia, Spain
| | - Marta Casado
- Biomedical Institute of Valencia (IBV-CSIC), Valencia, Spain
| | - Carlos Simón
- Fundación IVI, Instituto Universitario IVI, Department of Obstetrics & Gynecology, School of Medicine, University of Valencia, INCLIVA, Valencia, Spain Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
14
|
Liu CX, Zhang RL, Gao J, Li T, Ren Z, Zhou CQ, Wen AM. Derivation of human embryonic stem cell lines without any exogenous growth factors. Mol Reprod Dev 2014; 81:470-9. [PMID: 24554631 DOI: 10.1002/mrd.22312] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 02/11/2014] [Indexed: 01/16/2023]
Abstract
Human embryonic stem cell (hESC) lines are traditionally derived through immunosurgery. Their maintenance in culture requires the presence of mouse embryonic fibroblasts (MEFs) as feeder cells and media supplemented with basic fibroblast growth factor (bFGF) or other growth factors-both of which might introduce animal-derived culture components. The drawbacks associated with immunosurgery, MEF co-culture, and the cost of growth factors necessitate the exploration of a xeno-free method to maintain the self-renewal capacity of hESCs. Here, we describe an isolation method for the human inner cell mass (ICM), which was then cultured in the absence of exogenous growth factors and in the presence of human foreskin fibroblasts (HFFs) as feeder cells. Three hESC lines were obtained from poor-quality embryos by this near-xeno-free protocol. After culturing for more than 10 months, the hESCs retained normal morphology, expressed all expected cell surface markers, could differentiate to embryoid bodies upon culture in vitro, and formed teratomas in vivo. Furthermore, secretion of bFGF by HFFs was observed. In conclusion, this is the first study to describe an inexpensive, xeno-free culture system for the isolation and maintenance of hESCs that does not require bFGF supplementation.
Collapse
Affiliation(s)
- Cai Xia Liu
- Reproductive Medicine Center, Guangdong Academy of Medical Sciences/Guangdong General Hospital, Guangzhou, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Abbasalizadeh S, Baharvand H. Technological progress and challenges towards cGMP manufacturing of human pluripotent stem cells based therapeutic products for allogeneic and autologous cell therapies. Biotechnol Adv 2013; 31:1600-23. [PMID: 23962714 DOI: 10.1016/j.biotechadv.2013.08.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 06/20/2013] [Accepted: 08/12/2013] [Indexed: 12/16/2022]
Abstract
Recent technological advances in the generation, characterization, and bioprocessing of human pluripotent stem cells (hPSCs) have created new hope for their use as a source for production of cell-based therapeutic products. To date, a few clinical trials that have used therapeutic cells derived from hESCs have been approved by the Food and Drug Administration (FDA), but numerous new hPSC-based cell therapy products are under various stages of development in cell therapy-specialized companies and their future market is estimated to be very promising. However, the multitude of critical challenges regarding different aspects of hPSC-based therapeutic product manufacturing and their therapies have made progress for the introduction of new products and clinical applications very slow. These challenges include scientific, technological, clinical, policy, and financial aspects. The technological aspects of manufacturing hPSC-based therapeutic products for allogeneic and autologous cell therapies according to good manufacturing practice (cGMP) quality requirements is one of the most important challenging and emerging topics in the development of new hPSCs for clinical use. In this review, we describe main critical challenges and highlight a series of technological advances in all aspects of hPSC-based therapeutic product manufacturing including clinical grade cell line development, large-scale banking, upstream processing, downstream processing, and quality assessment of final cell therapeutic products that have brought hPSCs closer to clinical application and commercial cGMP manufacturing.
Collapse
Affiliation(s)
- Saeed Abbasalizadeh
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | |
Collapse
|
16
|
Hannan NRF, Segeritz CP, Touboul T, Vallier L. Production of hepatocyte-like cells from human pluripotent stem cells. Nat Protoc 2013; 8:430-7. [PMID: 23424751 DOI: 10.1038/nprot.2012.153] [Citation(s) in RCA: 235] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Large-scale production of hepatocytes from a variety of genetic backgrounds would be beneficial for drug screening and to provide a source of cells to be used as a substitute for liver transplantation. However, fully functional primary hepatocytes remain difficult to expand in vitro, and circumventing this problem by using an alternative source of cells is desirable. Here we describe a 25-d protocol to direct the differentiation of human pluripotent stem cells into a near-homogenous population of hepatocyte-like cells. As cells progress through this protocol, they express genes in a chronological manner similar to that described during in vivo hepatic development. The protocol relies on culture systems devoid of serum, feeders or complex extracellular matrices, which enable molecular analyses without interference from unknown factors. This approach works efficiently with human embryonic stem cells and human induced pluripotent stem cells and was recently used to model liver diseases in vitro.
Collapse
Affiliation(s)
- Nicholas R F Hannan
- Wellcome Trust–Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
| | | | | | | |
Collapse
|
17
|
Hannan NRF, Segeritz CP, Touboul T, Vallier L. Production of hepatocyte-like cells from human pluripotent stem cells. Nat Protoc 2013; 496:1269-1275. [PMID: 23424751 DOI: 10.1016/j.bbrc.2018.01.186] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 01/30/2018] [Indexed: 12/12/2022]
Abstract
Large-scale production of hepatocytes from a variety of genetic backgrounds would be beneficial for drug screening and to provide a source of cells to be used as a substitute for liver transplantation. However, fully functional primary hepatocytes remain difficult to expand in vitro, and circumventing this problem by using an alternative source of cells is desirable. Here we describe a 25-d protocol to direct the differentiation of human pluripotent stem cells into a near-homogenous population of hepatocyte-like cells. As cells progress through this protocol, they express genes in a chronological manner similar to that described during in vivo hepatic development. The protocol relies on culture systems devoid of serum, feeders or complex extracellular matrices, which enable molecular analyses without interference from unknown factors. This approach works efficiently with human embryonic stem cells and human induced pluripotent stem cells and was recently used to model liver diseases in vitro.
Collapse
Affiliation(s)
- Nicholas R F Hannan
- Wellcome Trust–Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
| | | | | | | |
Collapse
|
18
|
Defining the genomic signature of totipotency and pluripotency during early human development. PLoS One 2013; 8:e62135. [PMID: 23614026 PMCID: PMC3629124 DOI: 10.1371/journal.pone.0062135] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 03/19/2013] [Indexed: 11/25/2022] Open
Abstract
The genetic mechanisms governing human pre-implantation embryo development and the in vitro counterparts, human embryonic stem cells (hESCs), still remain incomplete. Previous global genome studies demonstrated that totipotent blastomeres from day-3 human embryos and pluripotent inner cell masses (ICMs) from blastocysts, display unique and differing transcriptomes. Nevertheless, comparative gene expression analysis has revealed that no significant differences exist between hESCs derived from blastomeres versus those obtained from ICMs, suggesting that pluripotent hESCs involve a new developmental progression. To understand early human stages evolution, we developed an undifferentiation network signature (UNS) and applied it to a differential gene expression profile between single blastomeres from day-3 embryos, ICMs and hESCs. This allowed us to establish a unique signature composed of highly interconnected genes characteristic of totipotency (61 genes), in vivo pluripotency (20 genes), and in vitro pluripotency (107 genes), and which are also proprietary according to functional analysis. This systems biology approach has led to an improved understanding of the molecular and signaling processes governing human pre-implantation embryo development, as well as enabling us to comprehend how hESCs might adapt to in vitro culture conditions.
Collapse
|
19
|
Abstract
Human embryonic stem cells (hESCs) are pluripotent cells derived from the inner cell mass (ICM) of the developing embryo. hESCs culture as cell lines in vitro and possess great potential in such research fields as developmental biology and cell-based therapy, as well as such industrial purposes as drug screening and toxicology. When ESCs were first derived by Thomson and colleagues, traditional methods of immunostaining and culturing, using primary mouse embryonic fibroblasts and medium supplemented by serum were used. Considerable efforts have since led to improved methods for isolating new lines in defined and reproducible conditions. This chapter discusses sources for embryos for ESC isolation, commonly used methods for deriving hESC lines, and a number of possible culture systems.
Collapse
Affiliation(s)
- Michal Amit
- Department of Obstetrics and Gynecology, Technion Israel Institute of Technology, Rambam Medical Center, Haifa, Israel
| |
Collapse
|
20
|
Mas A, Cervelló I, Gil-Sanchis C, Faus A, Ferro J, Pellicer A, Simón C. Identification and characterization of the human leiomyoma side population as putative tumor-initiating cells. Fertil Steril 2012; 98:741-751.e6. [DOI: 10.1016/j.fertnstert.2012.04.044] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 04/26/2012] [Accepted: 04/26/2012] [Indexed: 12/31/2022]
|
21
|
Ma Y, Gu J, Li C, Wei X, Tang F, Shi G, Jiang J, Kuang Y, Li J, Wang Z, Xie X, Jin Y. Human foreskin fibroblast produces interleukin-6 to support derivation and self-renewal of mouse embryonic stem cells. Stem Cell Res Ther 2012; 3:29. [PMID: 22849865 PMCID: PMC3580467 DOI: 10.1186/scrt120] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 07/31/2012] [Indexed: 02/06/2023] Open
Abstract
Introduction Embryonic stem cells (ESCs) provide an attractive cell source for basic research and disease treatment. Currently, the common culture system for mouse ESC requires mouse embryonic fibroblast (MEF) as a feeder layer supplemented with leukemia inhibitory factor (LIF). The drawbacks associated with MEF and the cost of LIF have motivated exploration of new feeder cell types to maintain self-renewal of mouse ESCs without the need of exogenous LIF. However, why these feeder cells could maintain ESCs at the undifferentiated state independent of exogenous LIF is unclear. Methods We derived mouse ESC lines using human foreskin fibroblast (HFF) in the absence of exogenous LIF. We also examined the dependence of HFF on the JAK-Stat3 pathway to maintain ESC identities and explored the potential molecular basis for HFF to support self-renewal of ESCs. Results HFF supported mouse ESC self-renewal superiorly to MEFs. Using the HFF system, multiple lines of mouse ESCs were successfully derived without addition of exogenous LIF and any small molecular inhibitors. These ESCs had capacities to self-renew for a long period of time and to differentiate into various cell types of the three germ layers both in vitro and in vivo. Moreover, the ESCs participated in embryonic development and contributed to germ cell lineages in the chimeric mouse. At a molecular level, HFF was dependent on the JAK-Stat3 pathway to maintain ESC self-renewal. The high level of interleukin-6 (IL-6) produced by HFF might be responsible for the exogenous LIF-independent effect. Conclusion This study describes an efficient, convenient and economic system to establish and maintain mouse ESC lines, and provides insights into the functional difference in the support of ESC culture between MEF and HFF.
Collapse
|
22
|
Fu X, Xu Y. Challenges to the clinical application of pluripotent stem cells: towards genomic and functional stability. Genome Med 2012; 4:55. [PMID: 22741526 PMCID: PMC3698533 DOI: 10.1186/gm354] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human embryonic stem cells (hESCs) can undergo unlimited self-renewal and are pluripotent, retaining the ability to differentiate into all cell types in the body. As a renewable source of various types of human cells, hESCs hold great therapeutic potential. Although significant advances have been achieved in defining the conditions needed to differentiate hESCs into various types of biologically active cells, many challenges remain in the clinical development of hESC-based cell therapy, such as the immune rejection of allogeneic hESC-derived cells by recipients. Breakthroughs in the generation of induced pluripotent stem cells (iPSCs), which are reprogrammed from somatic cells with defined factors, raise the hope that autologous cells derived from patient-specific iPSCs can be transplanted without immune rejection. However, recent genomic studies have revealed epigenetic and genetic abnormalities associated with induced pluripotency, a risk of teratomas, and immunogenicity of some iPSC derivatives. These findings have raised safety concerns for iPSC-based therapy. Here, we review recent advances in understanding the genomic and functional stability of human pluripotent stem cells, current challenges to their clinical application and the progress that has been made to overcome these challenges.
Collapse
Affiliation(s)
- Xuemei Fu
- Chengdu Women's and Children's Central Hospital, Chengdu, Sichuan, China ; Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Yang Xu
- Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
23
|
Krylova TA, Koltsova AM, Zenin VV, Musorina AS, Yakovleva TK, Poljanskaya GG. Comparative characteristics of new lines of mesenchymal stem cells derived from human embryonic stem cells, bone marrow, and foreskin. ACTA ACUST UNITED AC 2012. [DOI: 10.1134/s1990519x12020071] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
24
|
Abstract
In this chapter, we describe the derivation and characterization of nine hIn this chapter, we describe the derivation and characterization of nine human embryonic stem cells (hESC) (VAL-3 to -11B) from different developmental embryo stages (inner cell mass from a blastocyst, morula, and blastomere from a 3-day embryo) under xeno-free conditions providing the necessary protocols and techniques to carry out their derivation, characterization, and propagation.
Collapse
Affiliation(s)
- Amparo Galán
- Valencia Node of the National Stem Cell Bank, Prince Felipe Research Centre (CIPF), Valencia, Spain
| | | |
Collapse
|
25
|
Fu X, Xu Y. Self-renewal and scalability of human embryonic stem cells for human therapy. Regen Med 2011; 6:327-34. [PMID: 21548738 DOI: 10.2217/rme.11.18] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Human embryonic stem cells (hESCs) can undergo unlimited self-renewal and retain the pluripotency to differentiate into all cell types in the body. Therefore, as a renewable source of various cell types, hESCs hold great promise for human cell replacement therapy. While significant progress has been made in establishing the culture conditions for the long-term self-renewal of hESCs, several challenges remain to be overcome for the clinical application of hESCs. One such challenge is to develop strategies to scale-up the production of clinic-grade hESCs in xeno-free and chemically defined medium without inducing genomic instability. To achieve this goal, it is critical to elucidate the molecular pathways required to maintain the self-renewal, survival and genomic stability of hESCs. This article describes recent progress in addressing this challenge and discusses the strategies to improve the scalability of the production of hESCs by inhibiting their apoptosis.
Collapse
Affiliation(s)
- Xuemei Fu
- Chengdu Women's & Children's Central Hospital, Chengdu, Sichuan, China
| | | |
Collapse
|
26
|
Hongisto H, Vuoristo S, Mikhailova A, Suuronen R, Virtanen I, Otonkoski T, Skottman H. Laminin-511 expression is associated with the functionality of feeder cells in human embryonic stem cell culture. Stem Cell Res 2011; 8:97-108. [PMID: 22099024 DOI: 10.1016/j.scr.2011.08.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 08/19/2011] [Accepted: 08/22/2011] [Indexed: 11/30/2022] Open
Abstract
Fibroblast feeder cells play an important role in supporting the derivation and long term culture of undifferentiated, pluripotent human embryonic stem cells (hESCs). The feeder cells secrete various growth factors and extracellular matrix (ECM) proteins into extracellular milieu. However, the roles of the feeder cell-secreted factors are largely unclear. Animal feeder cells and use of animal serum also make current feeder cell culture conditions unsuitable for derivation of clinical grade hESCs. We established xeno-free feeder cell lines using human serum (HS) and studied their function in hESC culture. While human foreskin fibroblast (hFF) feeder cells were clearly hESC supportive, none of the established xeno-free human dermal fibroblast (hDF) feeder cells were able to maintain undifferentiated hESC growth. The two fibroblast types were compared for their ECM protein synthesis, integrin receptor expression profiles and key growth factor secretion. We show that hESC supportive feeder cells produce laminin-511 and express laminin-binding integrins α3ß1, α6ß1 and α7ß1. These results indicate specific laminin isoforms and integrins in maintenance of hESC pluripotency in feeder-dependent cultures. In addition, several genes with a known or possible role for hESC pluripotency were differentially expressed in distinct feeder cells.
Collapse
Affiliation(s)
- Heidi Hongisto
- Regea - Institute for Regenerative Medicine, University of Tampere, Tampere, Finland.
| | | | | | | | | | | | | |
Collapse
|
27
|
Kol’tsova AM, Gordeeva OF, Krylova TA, Lifantseva NV, Musorina AS, Yakovleva TK, Poljanskaya GG. Comparative characteristics of new human embryonic stem cell lines SC5, SC6, SC7, and SC3a. Russ J Dev Biol 2011. [DOI: 10.1134/s1062360411040072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
28
|
Cervelló I, Mas A, Gil-Sanchis C, Peris L, Faus A, Saunders PTK, Critchley HOD, Simón C. Reconstruction of endometrium from human endometrial side population cell lines. PLoS One 2011; 6:e21221. [PMID: 21712999 PMCID: PMC3119688 DOI: 10.1371/journal.pone.0021221] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Accepted: 05/23/2011] [Indexed: 12/23/2022] Open
Abstract
Endometrial regeneration is mediated, at least in part, by the existence of a specialized somatic stem cell (SSC) population recently identified by several groups using the side population (SP) technique. We previously demonstrated that endometrial SP displays genotypic, phenotypic and the functional capability to develop human endometrium after subcutaneous injection in NOD-SCID mice. We have now established seven human endometrial SP (hESP) cell lines (ICE 1-7): four from the epithelial and three from the stromal fraction, respectively. SP cell lines were generated under hypoxic conditions based on their cloning efficiency ability, cultured for 12-15 passages (20 weeks) and cryopreserved. Cell lines displayed normal 46XX karyotype, intermediate telomerase activity pattern and expressed mRNAs encoding proteins that are considered characteristic of undifferentiated cells (Oct-4, GDF3, DNMT3B, Nanog, GABR3) and those of mesodermal origin (WT1, Cardiac Actin, Enolase, Globin, REN). Phenotype analysis corroborated their epithelial (CD9+) or stromal (vimentin+) cell origin and mesenchymal (CD90+, CD73+ and CD45⁻) attributes. Markers considered characteristic of ectoderm or endoderm were not detected. Cells did not express either estrogen receptor alpha (ERα) or progesterone receptor (PR). The hESP cell lines were able to differentiate in vitro into adipocytes and osteocytes, which confirmed their mesenchymal origin. Finally, we demonstrated their ability to generate human endometrium when transplanted beneath the renal capsule of NOD-SCID mice. These findings confirm that SP cells exhibit key features of human endometrial SSC and open up new possibilities for the understanding of gynecological disorders such as endometriosis or Asherman syndrome. Our cell lines can be a valuable model to investigate new targets for endometrium proliferation in endometriosis.
Collapse
Affiliation(s)
- Irene Cervelló
- Fundación IVI-Instituto Universitario IVI, Universidad de Valencia, INCLIVA, Valencia, Spain
| | - Aymara Mas
- Fundación IVI-Instituto Universitario IVI, Universidad de Valencia, INCLIVA, Valencia, Spain
| | - Claudia Gil-Sanchis
- Fundación IVI-Instituto Universitario IVI, Universidad de Valencia, INCLIVA, Valencia, Spain
| | - Laura Peris
- Fundación IVI-Instituto Universitario IVI, Universidad de Valencia, INCLIVA, Valencia, Spain
| | - Amparo Faus
- Fundación IVI-Instituto Universitario IVI, Universidad de Valencia, INCLIVA, Valencia, Spain
| | - Philippa T. K. Saunders
- MRC/University of Edinburgh Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Hilary O. D. Critchley
- MRC/University of Edinburgh Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Carlos Simón
- Fundación IVI-Instituto Universitario IVI, Universidad de Valencia, INCLIVA, Valencia, Spain
- Valencian Node of the Spanish Stem Cell Bank, Prince Felipe Research Centre (CIPF), Unidad Mixta CIPF-UVEG, Valencia, Spain
- * E-mail:
| |
Collapse
|