951
|
Affiliation(s)
- Nadia Rosenthal
- Mouse Biology Programme, European Molecular Biology Laboratory Mouse Biology Programme, Rome, Italy.
| |
Collapse
|
952
|
Abstract
The use of stem cells in cell replacement therapy for neurodegenerative diseases has received a great deal of scientific and public interest in recent years. This is due to the remarkable pace at which paradigm-changing discoveries have been made regarding the neurogenic potential of embryonic, fetal, and adult cells. Over the last decade, clinical fetal tissue transplants have demonstrated that dopaminergic neurons can survive long term and provide functional clinical benefits for patients with Parkinson's disease. Pluripotent embryonic stem cells and multipotent neural stem cells may provide renewable sources that could replace these primary fetal grafts. Considerable advancement has been made in generating cultures with high numbers of neurons in general and of dopaminergic neurons using a varied array of techniques. However, much of this encouraging progress still remains to be tested on long-term expanded human cultures. Further problems include the low survival rate of these cells following transplantation and the tumorigenic tendencies of embryo-derived cells. However, pre-differentiation or genetic modification of stem cell cultures prior to transplantation may help lead to the generation of high numbers of cells of the desired phenotype following grafting. Boosting particular factors or substrates in the culture media may also protect grafted neurons from oxidative and metabolic stress, and provide epigenetic trophic support. Possible endogenous sources of cells for brain repair include the transdifferentiation of various types of adult cells into neurons. Despite the excitement generated by examples of this phenomenon, further work is needed in order to identify the precise instructive cues that generate neural cells from many other tissue types, and whether or not the new cells are functionally normal. Furthermore, issues such as cell homogeneity and fusion need to be addressed further before the true potential of transdifferentiation can be known. Endogenous stem cells also reside in the neurogenic zones of the adult brain (ventricle lining and hippocampus). Further elucidation of the mechanisms that stimulate cell division and migration are required in order to learn how to amplify the small amount of new cells generated by the adult brain and to direct these cells to areas of injury or degeneration. Finally, a more fundamental understanding of brain injury and disease is required in order to circumvent local brain environmental restrictions on endogenous cell differentiation and survival.
Collapse
Affiliation(s)
- Janel E Le Belle
- Cambridge Centre for Brain Repair, Forvie Site, University of Cambridge, Cambridge, UK.
| | | |
Collapse
|
953
|
Abstract
PURPOSE OF REVIEW Chronic diseases are common and deadly. Stem cell therapies have received intense interest for the repopulation of damaged or diseased tissues. A detailed understanding of the similarities and differences between embryonic stem cells and somatic stem cells will enhance our understanding of mechanisms of tissue repair or cellular augmentation. In addition, emerging technologies will be useful in the definition of the molecular regulation of the respective stem cell populations. RECENT FINDINGS A number of postnatal tissues have a population of somatic stem cells, which function in the maintenance and repair of tissues. Using molecular technologies these somatic stem cell populations have been shown to be pluripotent when placed in a permissive environment. Recent studies have utilized emerging technologies to define a molecular signature of embryonic stem cells and selected somatic stem cell populations. These strategies will be useful for the definition of a molecular program that promotes a stem cell phenotype (i.e. stemness phenotype). SUMMARY Recent studies suggest that embryonic and somatic stem cell populations hold promise as sources for tissue engineering. The use of cell biological and molecular technologies will enhance our understanding of embryonic and somatic stem cell populations and their molecular regulatory events that promote multipotentiation.
Collapse
Affiliation(s)
- Daniel J Garry
- Departments of Internal Medicine and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | | | | | | |
Collapse
|
954
|
Zimmermann S, Voss M, Kaiser S, Kapp U, Waller CF, Martens UM. Lack of telomerase activity in human mesenchymal stem cells. Leukemia 2003; 17:1146-9. [PMID: 12764382 DOI: 10.1038/sj.leu.2402962] [Citation(s) in RCA: 197] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Telomerase activity and telomere maintenance have been associated with immortality in tumor and embryonic stem cells. Whereas most normal somatic cells are telomerase negative, low levels of this enzyme have been found in adult stem cells from the skin, gut and the hematopoietic system. Here, we show that telomerase activity is not detectable in human mesenchymal stem cells (hMSCs), which have the phenotype SH2+, SH3+, SH4+, CD29+, CD44+, CD14-, CD34- and CD45-, and have the capacity to differentiate into adipocytes, chondrocytes and osteoblasts. These data suggest that hMSCs have a different telomere biology compared to other adult stem cells. Alternatively, true mesenchymal stem cells might be a very rare subpopulation that have a detection level that is below the sensitivity of the TRAP assay.
Collapse
Affiliation(s)
- S Zimmermann
- Department of Hematology/Oncology, Freiburg University Medical Center, Hugstetterstrasse 55, D-79106 Freiburg, Germany
| | | | | | | | | | | |
Collapse
|
955
|
Amit M, Margulets V, Segev H, Shariki K, Laevsky I, Coleman R, Itskovitz-Eldor J. Human feeder layers for human embryonic stem cells. Biol Reprod 2003; 68:2150-6. [PMID: 12606388 DOI: 10.1095/biolreprod.102.012583] [Citation(s) in RCA: 355] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Human embryonic stem (hES) cells hold great promise for future use in various research areas, such as human developmental biology and cell-based therapies. Traditionally, these cells have been cultured on mouse embryonic fibroblast (MEF) feeder layers, which permit continuous growth in an undifferentiated stage. To use these unique cells in human therapy, an animal-free culture system must be used, which will prevent exposure to mouse retroviruses. Animal-free culture systems for hES cells enjoy three major advantages in the basic culture conditions: 1). the ability to grow these cells under serum-free conditions, 2). maintenance of the cells in an undifferentiated state on Matrigel matrix with 100% MEF-conditioned medium, and 3). the use of either human embryonic fibroblasts or adult fallopian tube epithelial cells as feeder layers. In the present study, we describe an additional animal-free culture system for hES cells, based on a feeder layer derived from foreskin and a serum-free medium. In this culture condition, hES cells maintain all embryonic stem cell features (i.e., pluripotency, immortality, unlimited undifferentiated proliferation capability, and maintenance of normal karyotypes) after prolonged culture of 70 passages (>250 doublings). The major advantage of foreskin feeders is their ability to be continuously cultured for more than 42 passages, thus enabling proper analysis for foreign agents, genetic modification such as antibiotic resistance, and reduction of the enormous workload involved in the continuous preparation of new feeder lines.
Collapse
Affiliation(s)
- M Amit
- Department of Obstetrics and Gynecology, Rambam Medical Center, Rambam Medical Center, Haifa, Isreal
| | | | | | | | | | | | | |
Collapse
|
956
|
Affiliation(s)
- Sharon Gerecht-Nir
- Biotechnology Interdisciplinary Unit, Technion-Israel Institute of Technology, Haifa, Israel
| | | | | |
Collapse
|
957
|
Brivanlou AH, Gage FH, Jaenisch R, Jessell T, Melton D, Rossant J. Stem cells. Setting standards for human embryonic stem cells. Science 2003; 300:913-6. [PMID: 12738841 DOI: 10.1126/science.1082940] [Citation(s) in RCA: 279] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
958
|
Sone M, Itoh H, Yamashita J, Yurugi-Kobayashi T, Suzuki Y, Kondo Y, Nonoguchi A, Sawada N, Yamahara K, Miyashita K, Park K, Shibuya M, Nito S, Nishikawa SI, Nakao K. Different differentiation kinetics of vascular progenitor cells in primate and mouse embryonic stem cells. Circulation 2003; 107:2085-8. [PMID: 12707232 DOI: 10.1161/01.cir.0000070022.78747.1b] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND We demonstrated that vascular endothelial growth factor receptor 2 (VEGF-R2)-positive cells derived from mouse embryonic stem (ES) cells can differentiate into both endothelial cells and mural cells to suffice as vascular progenitor cells (VPCs). Here we examined whether VPCs occur in primate ES cells and investigated the differences in VPC differentiation kinetics between primate and mouse ES cells. METHODS AND RESULTS In contrast to mouse ES cells, undifferentiated monkey ES cells expressed VEGF-R2. By culturing these undifferentiated ES cells for 4 days on OP9 feeder layer, VEGF-R2 expression disappeared, and then reappeared after 8 days of differentiation. We then isolated these VEGF-R2-positive and vascular endothelial cadherin (VEcadherin)-negative cells by flow cytometry sorting. Additional 5-day reculture of these VEGF-R2+ VEcadherin- cells on OP9 feeder layer resulted in the appearance of platelet endothelial cell adhesion molecule-1 (PECAM1)-positive, VEcadherin-positive, endothelial nitric oxide synthase (eNOS)-positive endothelial cells. On a collagen IV-coated dish in the presence of serum, these cells differentiated into smooth muscle actin (SMA)-positive and calponin-positive mural cells (pericytes or vascular smooth muscle cells). Addition of 50 ng/mL VEGF to the culture on a collagen IV-coated dish resulted in the appearance of PECAM1+ cells surrounded by SMA+ cells. In addition, these differentiated VEGF-R2+ cells can form tube-like structures in a 3-dimensional culture. CONCLUSIONS Our findings indicate that differentiation kinetics of VPCs derived from primate and mouse ES cells were different. Differentiated VEGF-R2+ VEcadherin- cells can act as VPCs in primates. To seek the clinical potential of VPCs for vascular regeneration, investigations of primate ES cells are indispensable.
Collapse
Affiliation(s)
- Masakatsu Sone
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507 Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
959
|
He Q, Li J, Bettiol E, Jaconi ME. Embryonic stem cells: new possible therapy for degenerative diseases that affect elderly people. J Gerontol A Biol Sci Med Sci 2003; 58:279-87. [PMID: 12634295 DOI: 10.1093/gerona/58.3.m279] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The capacity of embryonic stem (ES) cells for virtually unlimited self renewal and differentiation has opened up the prospect of widespread applications in biomedical research and regenerative medicine. The use of these cells would overcome the problems of donor tissue shortage and implant rejection, if the cells are made immunocompatible with the recipient. Since the derivation in 1998 of human ES cell lines from preimplantation embryos, considerable research is centered on their biology, on how differentiation can be encouraged toward particular cell lineages, and also on the means to enrich and purify derivative cell types. In addition, ES cells may be used as an in vitro system not only to study cell differentiation but also to evaluate the effects of new drugs and the identification of genes as potential therapeutic targets. This review will summarize what is known about animal and human ES cells with particular emphasis on their application in four animal models of human diseases. Present studies of mouse ES cell transplantation reveal encouraging results but also technical barriers that have to be overcome before clinical trials can be considered.
Collapse
Affiliation(s)
- Qing He
- Biology of Aging Laboratory, Department of Geriatrics, Geneva University Hospitals, Switzerland
| | | | | | | |
Collapse
|
960
|
Abstract
UNLABELLED Although stem cells have held the fascination of scientists for years, the attention of the general public has recently been captured by the derivation of human embryonic stem cells. In this review we describe the historical experiments leading up to the isolation of human embryonic stem cells and discuss recent advances in our understanding of both embryonic and somatic stem cells. Select examples are used to illustrate the potential of stem cells, both in the sense of their ability to differentiate into specific cell types and in the sense of their power to treat various diseases and conditions. Also discussed are recent studies describing current progress toward the treatment of Parkinson disease, spinal cord injuries, diabetes, and cardiac disease. TARGET AUDIENCE Obstetricians & Gynecologists, Family Physicians LEARNING OBJECTIVES After completion of this article, the reader will be able to describe the various types of stem cells, outline potential clinical uses of stem cells, and summarize the somatic cell transdifferentiation debate.
Collapse
Affiliation(s)
- Kristina C Pfendler
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, California, USA.
| | | |
Collapse
|
961
|
Zwaka TP, Thomson JA. Homologous recombination in human embryonic stem cells. Nat Biotechnol 2003; 21:319-21. [PMID: 12577066 DOI: 10.1038/nbt788] [Citation(s) in RCA: 550] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2002] [Accepted: 12/20/2002] [Indexed: 02/06/2023]
Abstract
Homologous recombination applied to mouse embryonic stem (ES) cells has revolutionized the study of gene function in mammals. Although most often used to generate knockout mice, homologous recombination has also been applied in mouse ES cells allowed to differentiate in vitro. Homologous recombination is an essential technique if human ES cells are to fulfill their promise as a basic research tool. It also has important implications for ES cell-based transplantation and gene therapies. Significant differences between mouse and human ES cells have hampered the development of homologous recombination in human ES cells. High, stable transfection efficiencies in human ES cells have been difficult to achieve, and, in particular, electroporation protocols established for mouse ES cells work poorly in human ES cells. Also, in contrast to their murine counterparts, human ES cells cannot be cloned efficiently from single cells, making it difficult to screen for rare recombination events. Here we report an electroporation approach, based on the physical characteristics of human ES cells, that we used to successfully target HPRT1, the gene encoding hypoxanthine phosphoribosyltransferase-1 (HPRT1), and POU5F1, the gene encoding octamer-binding transcription factor 4 (Oct4; also known as POU domain, class 5, transcription factor 1 (POU5F1)).
Collapse
Affiliation(s)
- Thomas P Zwaka
- National Primate Research Center and the Department of Anatomy, University of Wisconsin-Madison Medical School, Madison, WI 53715, USA
| | | |
Collapse
|
962
|
Abstract
Pluripotential embryonic stem (ES) cells have been derived very efficiently from spare human embryos produced by IVF and grown in culture to the nascent blastocyst stage. The inner cell mass (ICM) is isolated by immunosurgery and grown on selected embryonic fibroblast monolayer cultures. ICM cells lose their memory for axis during formation of ES cell colonies and are then unable to integrate tissue formation with a body plan. ES cells form teratomas in vivo with cells and tissues representative of the three major embryonic lineages (ectoderm, mesoderm, endoderm). The ES cells are continuously renewable and can be directed to differentiate into early progenitors of neural stem cells (Noggin cells) and from there into mature neurons and glia (astrocytes and oligodendrocytes). The neural stem cells formed from human ES cells repopulate the brains of newborn mice when injected into the lateral cerebral ventricles, forming astrocytes dominantly in the parenchyma. The human neural cells can be observed migrating from the subventricular areas along the rostral migratory stream. Human neurons can be found in the olfactory bulb. Human ES cells can also be directed into cardiomyocytes when co-cultured with visceral endoderm-like cells (END-2). These observations provide further scope to explore stem cell therapies, gene therapies and drug discovery. For compatible transplantation, ES may need to be derived with a range of HLA types or by nuclear transplantation or stem cell fusion.
Collapse
Affiliation(s)
- Alan Trounson
- Institute of Reproduction and Development, Monash University, 27-31 Wright Street, Clayton, Victoria 3168, Australia.
| |
Collapse
|
963
|
Gerecht-Nir S, Fishman B, Itskovitz-Eldor J. Cardiovascular potential of embryonic stem cells. ACTA ACUST UNITED AC 2003; 276:58-65. [PMID: 14699634 DOI: 10.1002/ar.a.10136] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Initial events involved in the process of heart formation consist of myocardial differentiation as well as development of endothelial and endocardial tissues. As only limited means are allocated to the studying of cardiovascular system development, embryonic stem cells (ESCs) isolated from the inner cell mass (ICM) of developing mice or human blastocysts offer the first step toward the understanding of these complex and intriguing events. ESCs are able to differentiate into a wide range of cell types, including various vascular cells and cardiomyocytes, and their self-renewal capability renders them a unique, homogeneous, and unlimited preliminary population of cells for the investigation of early developmental events of cardiovascular system and lineage commitment. This review summarizes the accumulated knowledge of the cellular and molecular mechanisms involved in the development of the cardiovascular system.
Collapse
Affiliation(s)
- Sharon Gerecht-Nir
- Biotechnology Interdisciplinary Unit, Technion, Israel Institute of Technology, Haifa, Israel
| | | | | |
Collapse
|
964
|
Mayani H. A glance into somatic stem cell biology: basic principles, new concepts, and clinical relevance. Arch Med Res 2003; 34:3-15. [PMID: 12604368 DOI: 10.1016/s0188-4409(02)00450-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Somatic stem cells are undifferentiated cells with a high capacity for self-renewal that can give rise to one or more specialized cell types with specific functions in the body. Profound characterization of these cells has been difficult due to the fact that their frequency in different tissues of the body is extremely low; furthermore, their identification is not based on their morphology but on immunophenotypic and functional assays. Nevertheless, significant advances in the study of these cells at both cellular and molecular levels have been achieved during the last decade. The majority of what we know concerning somatic stem cell biology has come from work on hematopoietic stem cells. More recently, however, there has been a great amount of information on neural and epithelial stem cells. The importance of stem cell research has gone beyond basic biology and is currently contributing to the development of new medical approaches for treatment of hematologic, neurologic, autoimmune, and metabolic disorders (cellular therapy).
Collapse
Affiliation(s)
- Héctor Mayani
- Unidad de Investigación Médica en Oncología, Hospital de Oncología, Centro Médico Nacional Siglo XXI (CMN-SXXI), Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico.
| |
Collapse
|
965
|
Kehat I, Amit M, Gepstein A, Huber I, Itskovitz-Eldor J, Gepstein L. Development of Cardiomyocytes from Human ES Cells. Methods Enzymol 2003; 365:461-73. [PMID: 14696365 DOI: 10.1016/s0076-6879(03)65032-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Affiliation(s)
- Izhak Kehat
- Cardiovascular Research Laboratory, Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 2 Efron Street, POB 9649, Haifa 31096, Israel
| | | | | | | | | | | |
Collapse
|
966
|
Pera MF, Filipczyk AA, Hawes SM, Laslett AL. Isolation, characterization, and differentiation of human embryonic stem cells. Methods Enzymol 2003; 365:429-46. [PMID: 14696363 DOI: 10.1016/s0076-6879(03)65030-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Martin F Pera
- Monash Institute of Reproduction and Development, Monash University, 246 Clayton Road, Clayton, Victoria 3168, Australia
| | | | | | | |
Collapse
|
967
|
Oh BK, Seong JK, Lee JE, Chae KJ, Roh KJ, Park C, Park YN. Induction of telomerase activity during an early burst of proliferation in pancreatic regeneration. Cancer Lett 2002; 186:93-8. [PMID: 12183080 DOI: 10.1016/s0304-3835(02)00320-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Telomerase activity (TA) and telomerase reverse transcriptase (rTERT) were investigated in Sprague-Dawley rats, killed at 6 and 12 h, and 1, 2, 3, 7, and 14 days after 90% pancreatectomy (px), by TRAPeze enzyme-linked immunosorbent assay telomerase detection and reverse transcriptase-polymerase chain reaction. TA increased at 2 days and reached a maximum at 3 days after px, when the small ductules showed the highest proliferation activity. After 3 days, TA decreased to basal levels as the ductules differentiated into new endocrine and exocrine pancreas. The expression of rTERT showed a correlation with TA. These results suggest telomerase is actively regulated during pancreatic regeneration.
Collapse
Affiliation(s)
- Bong-Kyeong Oh
- Department of Pathology and Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, CPO Box 8044, Seoul, South Korea
| | | | | | | | | | | | | |
Collapse
|
968
|
Abstract
Embryonic stem cells are pluripotent cell lines that are derived from the blastocyst-stage early mammalian embryo. These unique cells are characterized by their capacity for prolonged undifferentiated proliferation in culture while maintaining the potential to differentiate into derivatives of all three germ layers. During in vitro differentiation, embryonic stem cells can develop into specialized somatic cells, including cardiomyocytes, and have been shown to recapitulate many processes of early embryonic development. The present review describes the derivation and unique properties of the recently described human embryonic stem cells as well as the properties of cardiomyocytes derived using this unique differentiating system. The possible applications of this system in several cardiac research areas, including developmental biology, functional genomics, pharmacological testing, cell therapy, and tissue engineering, are discussed. Because of their combined ability to proliferate indefinitely and to differentiate to mature tissue types, human embryonic stem cells can potentially provide an unlimited supply of cardiomyocytes for cell therapy procedures aiming to regenerate functional myocardium. However, many obstacles must still be overcome on the way to successful clinical utilization of these cells.
Collapse
Affiliation(s)
- Lior Gepstein
- Cardiovascular Research Laboratory, the Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, and Rambam Medical Center, Haifa, Israel.
| |
Collapse
|
969
|
Abstract
Cell replacement therapy is a promising approach for the treatment of cardiac diseases, but is challenged by a limited supply of appropriate cells. We have investigated whether functional cardiomyocytes can be efficiently generated from human embryonic stem (hES) cells. Cardiomyocyte differentiation was evaluated using 3 parent (H1, H7, and H9) hES cell lines and 2 clonal (H9.1 and H9.2) hES cell lines. All cell lines examined differentiated into cardiomyocytes, even after long-term culture (50 passages or approximately 260 population doublings). Upon differentiation, beating cells were observed after one week in differentiation conditions, increased in numbers with time, and could retain contractility for over 70 days. The beating cells expressed markers characteristic of cardiomyocytes, such as cardiac alpha-myosin heavy chain, cardiac troponin I and T, atrial natriuretic factor, and cardiac transcription factors GATA-4, Nkx2.5, and MEF-2. In addition, cardiomyocyte differentiation could be enhanced by treatment of cells with 5-aza-2'-deoxycytidine but not DMSO or retinoic acid. Furthermore, the differentiated cultures could be dissociated and enriched by Percoll density centrifugation to give a population containing 70% cardiomyocytes. The enriched population was proliferative and showed appropriate expression of cardiomyocyte markers. The extended replicative capacity of hES cells and the ability to differentiate and enrich for functional human cardiomyocytes warrant further development of these cells for clinical application in heart diseases.
Collapse
Affiliation(s)
- Chunhui Xu
- Geron Corporation, Menlo Park, Calif 94025, USA.
| | | | | | | |
Collapse
|
970
|
Abstract
Recent progress in deriving human embryonic stem (hES) cells and defining their capacity to differentiate has inspired hope that they could become a source of replacement cells for damaged or diseased tissues. We review the immunological barriers to transplanting hES cells and consider several potential solutions, including stem-cell banking, modification of the immunogenicity of donor cells and induction of tolerance to the graft. We evaluate the probable efficacy of these approaches with a view to facilitating the use of hES cells in clinical practice.
Collapse
Affiliation(s)
- J Andrew Bradley
- Department of Surgery, Cambridge University Clinical School, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK.
| | | | | |
Collapse
|
971
|
Nakatsuji N. [Embryonic stem cell lines for regenerative therapy and pharmaceutical research]. Nihon Yakurigaku Zasshi 2002; 120:295-302. [PMID: 12491805 DOI: 10.1254/fpj.120.295] [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: 10/26/2022]
Abstract
The establishment of human embryonic stem (ES) cell lines has brought great potential and expectations for regenerative medicine and pharmaceutical research, because many types of human cells could be produced by their unlimited growth and differentiation in culture. Primate and human ES cell lines have been established from blastocysts of monkey and surplus human blastocysts from fertility clinics. They showed several differences compared to mouse ES cells, including a tendency to produce the trophectoderm lineage and a different expression pattern of surface antigens. This may reflect species-specific differences, or these primate ES cells could represent earlier stages of development than mouse ES cells. Also, they show no response to the LIF and gp130 signals, which are widely used to repress spontaneous differentiation of mouse ES cell colonies. We have established several ES cell lines from blastocysts of the cynomolgus monkey. They can be maintained in culture as stem cell colonies, and they produce several differentiated cell types in culture. When such ES cells were transplanted into SCID mice, they produced teratomas containing many differentiated tissues.
Collapse
Affiliation(s)
- Norio Nakatsuji
- Department of Development and Differentiation and Stem Cell Research Center, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawaharacho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
972
|
Abstract
Pluripotent stem cells are undifferentiated cells that are capable of differentiating to all three embryonic germ layers and their differentiated derivatives. They are transiently found during embryogenesis, in preimplantation embryos and fetal gonads, or as established cell lines. These unique cell types are distinguished by their wide developmental potential and by their ability to be propagated in culture indefinitely, without loosing their undifferentiated phenotype. This short review intends to give a general overview on the pluripotent nature of embryo-derived stem cells with a focus on human embryonic stem cells.
Collapse
Affiliation(s)
- Rachel Eiges
- Department of Genetics, The Life Sciences Institute, The Hebrew University, 91904, Jerusalem, Israel
| | | |
Collapse
|
973
|
Strom TB, Field LJ, Ruediger M. Allogeneic stem cells, clinical transplantation and the origins of regenerative medicine. Curr Opin Immunol 2002; 14:601-5. [PMID: 12183159 DOI: 10.1016/s0952-7915(02)00387-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The therapeutic use of stem cells is a popular topic that is often in the news. Stem cells are of particular relevance to the transplant community, as stem cell-based therapies will probably be derived from allogeneic sources. Stem cells may be obtained from somatic (adult) cell or embryonic cell origin. The ability to establish stem cell lines in vitro means that we can standardize therapeutic grafting applications, which is a prerequisite for the widespread application of stem cell therapy. This attribute also means that we can produce large batches of allogeneic terminally differentiated cells, which can be administered to patients. Three important barriers to the deployment of this therapy currently exist; in addition to the many ethical issues facing the widespread use of stem cell therapy, it is also important to develop methods to derive pure terminally differentiated cells for transplantation and to induce immune tolerance to these allogeneic cells, as daily anti-rejection therapy cannot be justified for many potential applications.
Collapse
Affiliation(s)
- Terry B Strom
- Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA.
| | | | | |
Collapse
|
974
|
Goldstein RS, Drukker M, Reubinoff BE, Benvenisty N. Integration and differentiation of human embryonic stem cells transplanted to the chick embryo. Dev Dyn 2002; 225:80-6. [PMID: 12203723 DOI: 10.1002/dvdy.10108] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Human embryonic stem (ES) cells are pluripotent cells that can differentiate into a large array of cell types and, thus, hold promise for advancing our understanding of human embryology and for contributing to transplantation medicine. In this study, differentiation of human ES cells was examined in vivo by in ovo transplantation to organogenesis-stage embryos. Colonies of human ES cells were grafted into or in place of epithelial-stage somites of chick embryos of 1.5 to 2 days of development. The grafted human ES cells survived in the chick host and were identified by vital staining with carboxyfluorescein diacetate or use of a green fluorescent protein-expressing cells. Histologic analysis showed that human ES cells are easily distinguished from host cells by their larger, more intensely staining nuclei. Some grafted cells differentiated en masse into epithelia, whereas others migrated and mingled with host tissues, including the dorsal root ganglion. Colonies grafted directly adjacent to the host neural tube produced primarily structures with the morphology and molecular characteristics of neural rosettes. These structures contain differentiated neurons as shown by beta-3-tubulin and neurofilament expression in axons and cell bodies. Axons derived from the grafted cells penetrate the host nervous system, and host axons enter the structures derived from the graft. Our results show that human ES cells transplanted in ovo survive, divide, differentiate, and integrate with host tissues and that the host embryonic environment may modulate their differentiation. The chick embryo, therefore, may serve as an accessible and unique experimental system for the study of in vivo development of human ES cells.
Collapse
Affiliation(s)
- Ronald S Goldstein
- Gonda Research Center, Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
| | | | | | | |
Collapse
|
975
|
|
976
|
Abstract
The capacity of embryonic stem cells for virtually unlimited self-renewal and differentiation capacity has opened up the prospect of widespread applications in biomedical research and regenerative medicine. For the latter, the cells provide hope that it will be possible to overcome the problems of donor tissue shortage and also, by making the cells immunocompatible with the recipient, implant rejection. Four years after the first derivation of human pluripotent cell lines from pre-implantation embryos, a great deal has been learnt about their biology and how differentiation can be encouraged towards particular cell lineages. However, considerable research is needed, not least into means to enrich and purify derivative cell lineages, before clinical trials can be considered.
Collapse
Affiliation(s)
- Anne E Bishop
- Tissue Engineering Centre, Imperial College Faculty of Medicine, Chelsea & Westminster Hospital, London, UK.
| | | | | |
Collapse
|
977
|
Abstract
Augmentation of myocardial performance in experimental models of therapeutic infarction and heart failure has been achieved by the transplantation of exogenous cells into damaged myocardium, a procedure known as cellular cardiomyoplasty (CCM). Historically, a wide range of cell types have been used for CCM, including rat and human fetal ventricular myocytes, but the availability of human fetal donor cells for clinical purposes is limited. The quest for suitable alternative donor cells has prompted research into the use of both embryonic stem (ES) cells and adult somatic stem cells, but the optimal choice of donor cell source is not yet known. Recently, there has been a growing body of evidence that multipotent somatic stem cells in adult bone marrow exhibit tremendous functional plasticity and can reprogramme in a new environmental tissue niche to give rise to cell lineages specific for the new organ site. This phenomenon has made a huge impact on myocardial biology and has captured the imagination of scientists who have recently discovered that multipotent adult bone marrow haematopoeitic stem cells and mesenchymal stem cells can repopulate infarcted rodent myocardium and differentiate into both cardiomyocytes and new blood vessels. These data, coupled with the identification of a putative primitive cardiac stem cell population in the adult human heart, may pave the way for novel therapeutic modalities for enhancing myocardial performance and treating end-stage cardiac disease.
Collapse
Affiliation(s)
- Siân Hughes
- St George's Hospital Medical School, London, UK.
| |
Collapse
|
978
|
Prelle K, Zink N, Wolf E. Pluripotent stem cells--model of embryonic development, tool for gene targeting, and basis of cell therapy. Anat Histol Embryol 2002; 31:169-86. [PMID: 12479360 DOI: 10.1046/j.1439-0264.2002.00388.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Embryonic stem (ES) cells are pluripotent cell lines with the capacity of self-renewal and a broad differentiation plasticity. They are derived from pre-implantation embryos and can be propagated as a homogeneous, uncommitted cell population for an almost unlimited period of time without losing their pluripotency and their stable karyotype. Murine ES cells are able to reintegrate fully into embryogenesis when returned into an early embryo, even after extensive genetic manipulation. In the resulting chimeric offspring produced by blastocyst injection or morula aggregation, ES cell descendants are represented among all cell types, including functional gametes. Therefore, mouse ES cells represent an important tool for genetic engineering, in particular via homologous recombination, to introduce gene knock-outs and other precise genomic modifications into the mouse germ line. Because of these properties ES cell technology is of high interest for other model organisms and for livestock species like cattle and pigs. However, in spite of tremendous research activities, no proven ES cells colonizing the germ line have yet been established for vertebrate species other than the mouse (Evans and Kaufman, 1981; Martin, 1981) and chicken (Pain et al., 1996). The in vitro differentiation capacity of ES cells provides unique opportunities for experimental analysis of gene regulation and function during cell commitment and differentiation in early embryogenesis. Recently, pluripotent stem cells were established from human embryos (Thomson et al., 1998) and early fetuses (Shamblott et al., 1998), opening new scenarios both for research in human developmental biology and for medical applications, i.e. cell replacement strategies. At about the same time, research activities focused on characteristics and differentiation potential of somatic stem cells, unravelling an unexpected plasticity of these cell types. Somatic stem cells are found in differentiated tissues and can renew themselves in addition to generating the specialized cell types of the tissue from which they originate. Additional to discoveries of somatic stem cells in tissues that were previously not thought to contain these kinds of cells, they also appear to be capable of developing into cell types of other tissues, but have a reduced differentiation potential as compared to embryo-derived stem cells. Therefore, somatic stem cells are referred to as multipotent rather than pluripotent. This review summarizes characteristics of pluripotent stem cells in the mouse and in selected livestock species, explains their use for genetic engineering and basic research on embryonic development, and evaluates their potential for cell therapy as compared to somatic stem cells.
Collapse
Affiliation(s)
- Katja Prelle
- Department of Molecular Animal Breeding and Biotechnology, Ludwig Maximilian University Munich, Hackerstrasse 27, 85764 Oberschleissheim, Germany.
| | | | | |
Collapse
|
979
|
Abstract
Established lines of human pluripotent stem cells provide a convenient tool for investigating cell differentiation in a way that is pertinent to human embryonic development, providing insights into the causes of birth defects and diseases such as cancer that involve aberrant cell proliferation and differentiation. In principle, human pluripotent stem cells, including embryonic stem and embryonic germ cells, are capable of differentiating into all of the cell types that are present in the adult human. They therefore have the potential to provide a source of tissues for replacement in diseases in which native cell types are inactivated or destroyed. Intense media and public interest has surrounded the announcement of human pluripotent stem cells derivation, focusing on the ethical implications of embryo-related work and on the prospects of an unlimited source of tissues for transplantation-based treatments. Recent studies have focused on identifying method for culture of these cells and inducing their differentiation into specific cell types.
Collapse
Affiliation(s)
- Jonathan S Draper
- Department of Biomedical Science, University of Sheffield, Sheffield, UK.
| | | |
Collapse
|
980
|
|
981
|
Abstract
The recent derivation of human embryonic stem (ES) cell lines, together with results suggesting an unexpected degree of plasticity in later, seemingly more restricted, stem cells (so-called adult stem cells), have combined to focus attention on new opportunities for regenerative medicine, as well as for understanding basic aspects of embryonic development and diseases such as cancer. Many of the ideas that are now discussed have a long history and much has been underpinned by the earlier studies of teratocarcinomas, and their embryonal carcinoma (EC) stem cells, which present a malignant surrogate for the normal stem cells of the early embryo. Nevertheless, although the potential of EC and ES cells to differentiate into a wide range of tissues is now well attested, little is understood of the key regulatory mechanisms that control their differentiation. Apart from the intrinsic biological interest in elucidating these mechanisms, a clear understanding of the molecular process involved will be essential if the clinical potential of these cells is to be realized. The recent observations of stem-cell plasticity suggest that perhaps our current concepts about the operation of cell regulatory pathways are inadequate, and that new approaches for analysing complex regulatory networks will be essential.
Collapse
Affiliation(s)
- Peter W Andrews
- Department of Biomedical Science, Western Bank, University of Sheffield, Sheffield S10 2TN, UK.
| |
Collapse
|
982
|
Yamada T, Yoshikawa M, Takaki M, Torihashi S, Kato Y, Nakajima Y, Ishizaka S, Tsunoda Y. In vitro functional gut-like organ formation from mouse embryonic stem cells. Stem Cells 2002; 20:41-9. [PMID: 11796921 DOI: 10.1634/stemcells.20-1-41] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND AIMS Embryonic stem (ES) cells have a pluripotent ability to differentiate into a variety of cell lineages in vitro. We have recently found that ES cells can give rise to a functional gut-like unit, which forms a three-dimensional dome-like structure with lumen and exhibits mechanical activity, such as spontaneous contraction and peristalsis. The aim of the present study was to investigate the electrophysiological and morphological properties of ES cell-derived contracting clusters. METHODS Electrical activity was examined by an extracellular recording. Morphology and cellular components were investigated by immunohistochemistry and electron microscopy. RESULTS Clusters with rhythmic contractions displayed electrical slow waves at a regular rhythm, and clusters with highly coordinated peristalsis showed regular slow waves and spontaneous spike action potentials. Immunoreactivity for c-Kit, a marker of interstitial cells of Cajal (ICC), was observed in dense network structures. Neuronal marker PGP9.5 immunoreactivity was observed only in clusters with peristalsis. The topographical structure of the wall was organized by an inner epithelial layer and outer smooth muscle layer. The smooth muscle layer was provided with an ICC network and innervated with enteric neurons. CONCLUSIONS ES cells can differentiate into a functional gut-like organ in vitro that exhibits physiological and morphological properties characteristic of the gastrointestinal (GI) tract. This ES cell-derived gut provides a powerful tool for studying GI motility and gut development in vitro, and has potential for elucidating and treating a variety of motility disorders.
Collapse
Affiliation(s)
- Takatsugu Yamada
- Division of Developmental Biology, Department of Parasitology, Nara Medical University, Nara, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
983
|
Abstract
Embryonic stem (ES) cells are pluripotent stem cells that differentiate both in vitro and in vivo into cell types derived from each of the three embryonic germ layers. ES cells and their close relatives, embryonal carcinoma (EC) cells and embryonic germ (EG) cells, have been used extensively as model systems for studying early mammalian development. This work has led to important insights into the mechanisms that control embryogenesis at the molecular and cellular levels. This chapter focuses on the use of ES cells as an in vitro model system for studying cellular differentiation and reviews several areas where important progress has been made. Impressive progress has been made in the isolation and characterization of ES cells from many species, including humans. Significant progress has also been made in the development of culture conditions that help direct the differentiation of ES cells to specific cell types that form during myogenesis, angiogenesis, hematopoiesis, neurogenesis, and cardiogenesis. The ability to inactivate virtually any gene in ES cells by gene targeting has vastly improved our understanding of the roles played by specific genes at the cellular and organismic levels. Moreover, ES cells and EC cells have been used widely to investigate how specific genes are turned on and turned off in the course of differentiation. In this connection, DNA array technology has been used to identify genes regulated when ES cells differentiate. The final section of this chapter discusses how work with ES cells is shaping our understanding of stem cells, mammalian development, and cell replacement therapy.
Collapse
Affiliation(s)
- Angie Rizzino
- Eppley Institute for Cancer Research and Allied Diseases, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, Nebraska 68198-6805, USA
| |
Collapse
|
984
|
Levenberg S, Golub JS, Amit M, Itskovitz-Eldor J, Langer R. Endothelial cells derived from human embryonic stem cells. Proc Natl Acad Sci U S A 2002; 99:4391-6. [PMID: 11917100 PMCID: PMC123658 DOI: 10.1073/pnas.032074999] [Citation(s) in RCA: 602] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human embryonic stem cells have the potential to differentiate into various cell types and, thus, may be useful as a source of cells for transplantation or tissue engineering. We describe here the differentiation steps of human embryonic stem cells into endothelial cells forming vascular-like structures. The human embryonic-derived endothelial cells were isolated by using platelet endothelial cell-adhesion molecule-1 (PECAM1) antibodies, their behavior was characterized in vitro and in vivo, and their potential in tissue engineering was examined. We show that the isolated embryonic PECAM1+ cells, grown in culture, display characteristics similar to vessel endothelium. The cells express endothelial cell markers in a pattern similar to human umbilical vein endothelial cells, their junctions are correctly organized, and they have high metabolism of acetylated low-density lipoprotein. In addition, the cells are able to differentiate and form tube-like structures when cultured on matrigel. In vivo, when transplanted into SCID mice, the cells appeared to form microvessels containing mouse blood cells. With further studies, these cells could provide a source of human endothelial cells that could be beneficial for potential applications such as engineering new blood vessels, endothelial cell transplantation into the heart for myocardial regeneration, and induction of angiogenesis for treatment of regional ischemia.
Collapse
Affiliation(s)
- Shulamit Levenberg
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | |
Collapse
|
985
|
|
986
|
Abstract
The most common causes of death and suffering, even in most underdeveloped nations, are age-related diseases. These diseases share fundamental and often unappreciated pathology at the cellular and genetic levels, through cell senescence. In cancer, enforcing cell senescence permits us to kill cancer cells without significantly harming normal cells. In other age-related diseases, cell senescence plays a direct role, and we may be able to prevent and reverse much of the pathology. While aging is attributed to "wear and tear," genetic studies show that these effects are avoidable (as is the case in germ cell lines) and occur only when cells down-regulate active (and sufficient) repair mechanisms, permitting degradation to occur. Aging occurs when cells permit accumulative damage by wear and tear, by altering their gene expression rather than vice versa. Using telomerase in laboratory settings, we can currently reset this pattern and its consequences both within cells and between cells. Doing so resets not only cell behavior but the pathological consequences within tissues comprising such cells. We can currently grow histologically young, reconstituted human skin using old human skin cells (keratinocytes and fibroblasts). Technically we could now test this approach in joints, vessels, the immune system, and other tissues. This model is consistent with all available laboratory data and known aging pathology. Within the next decade, we will be able to treat age-related diseases more effectively than ever before.
Collapse
|
987
|
Abstract
Embryonic stem (ES) cells are unique cells derived from the inner cell mass of the mammalian blastocyst. These cells are immortal and pluripotent, retain their developmental potential after prolonged culture, and can be continuously cultured in an undifferentiated state. Many in vitro differentiation systems have been developed for mouse ES cells, including reproducible methods for mouse ES cell differentiation into haematopoietic and neural precursors, cardiomyocytes, insulin-secreting cells, endothelial cells and various other cell types. The derivation of new human ES cell lines provides the opportunity to develop unique models for developmental research and for cell therapies. In this review we consider the derivation and spontaneous differentiation of human ES cells.
Collapse
Affiliation(s)
- Michal Amit
- Department of Obstetrics and Gynecology, Rambam Medical CenterHaifa, Israel
- The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of TechnologyHaifa, Israel
| | - Joseph Itskovitz-Eldor
- Department of Obstetrics and Gynecology, Rambam Medical CenterHaifa, Israel
- The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of TechnologyHaifa, Israel
| |
Collapse
|
988
|
Abstract
Production of chimaeras with embryonal carcinoma and embryonic stem cells enabled a very thorough investigation of the potency of these cells in the mouse. Human embryonal carcinoma and embryonic stem cell differ from their murine counterparts in a number of respects and, for obvious reasons, their potency is more difficult to assess. Recently, findings attesting to a surprising degree of plasticity of cells from adults have begun to emerge, which, aside from offering a possible further route to stem cell therapy, raise intriguing questions about the importance of lineage in the process of cellular diversification. Biomedical research is widely perceived to be advancing too fast to allow proper consideration of the implications of its clinical applications. Whilst this was clearly not true in the case of human in vitro fertilization, it has some validity regarding stem cell therapy, even though many of the issues are common to both. Casual use of the term 'embryo' proved unhelpful in the past debate on whether research on early stages of human development should be permitted. Likewise, introduction of the term 'therapeutic cloning' has complicated the present one regarding extension of such research to stem cell therapy.
Collapse
Affiliation(s)
- R L Gardner
- Mammalian Development Laboratory, University of Oxford, Department of Zoology, UK.
| |
Collapse
|
989
|
Mummery C, Ward D, van den Brink CE, Bird SD, Doevendans PA, Opthof T, Brutel de la Riviere A, Tertoolen L, van der Heyden M, Pera M. Cardiomyocyte differentiation of mouse and human embryonic stem cells. J Anat 2002; 200:233-42. [PMID: 12033727 PMCID: PMC1570681 DOI: 10.1046/j.1469-7580.2002.00031.x] [Citation(s) in RCA: 222] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Ischaemic heart disease is the leading cause of morbidity and mortality in the western world. Cardiac ischaemia caused by oxygen deprivation and subsequent oxygen reperfusion initiates irreversible cell damage, eventually leading to widespread cell death and loss of function. Strategies to regenerate damaged cardiac tissue by cardiomyocyte transplantation may prevent or limit post-infarction cardiac failure. We are searching for methods for inducing pluripotent stem cells to differentiate into transplantable cardiomyocytes. We have already shown that an endoderm-like cell line induced the differentiation of embryonal carcinoma cells into immature cardiomyocytes. Preliminary results show that human and mouse embryonic stem cells respond in a similar manner. This study presents initial characterization of these cardiomyocytes and the mouse myocardial infarction model in which we will test their ability to restore cardiac function.
Collapse
Affiliation(s)
- C Mummery
- Hubrecht Laboratory, Utrecht, The Netherland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
990
|
Abstract
Stem cells undergo self-renewal and differentiate into multiple lineages of mature cells. The identification of stem cells in diverse adult tissues and the findings that human embryonic stem cells can be proliferated and differentiated has kindled the imagination of both scientists and the public regarding future stem cell technology. These cells could constitute an unlimited supply of diverse cell types that can be used for cell transplantation or drug discovery. The new options raise several fundamental ethical issues. This review gives an overview of the scientific basis underlying the hope generated by stem cell research and discusses current ethical and funding regulations.
Collapse
Affiliation(s)
- Gesine Paul
- Section for Neuronal Survival, Wallenberg Neuroscience Center, Lund University, BMC A10, 221 84 Lund, Sweden.
| | | | | |
Collapse
|
991
|
Abstract
End-stage liver disease is treated by liver transplantation, but donor organ shortages remain a serious problem. This has prompted the design of bioartificial liver devices to "bridge" patients until they either recover or receive a liver transplant. In these devices, patient plasma is circulated extracorporeally through a bioreactor that houses liver cells (hepatocytes) sandwiched between artificial plates or capillaries.
Collapse
Affiliation(s)
- Alastair J Strain
- Schools of Biosciences and Medicine, University of Birmingham, and Liver and Hepatobiliary Unit, University Hospital Birmingham, Edgbaston, Birmingham B15 2TH, UK.
| | | |
Collapse
|
992
|
Kawasaki H, Suemori H, Mizuseki K, Watanabe K, Urano F, Ichinose H, Haruta M, Takahashi M, Yoshikawa K, Nishikawa SI, Nakatsuji N, Sasai Y. Generation of dopaminergic neurons and pigmented epithelia from primate ES cells by stromal cell-derived inducing activity. Proc Natl Acad Sci U S A 2002; 99:1580-5. [PMID: 11818560 PMCID: PMC122233 DOI: 10.1073/pnas.032662199] [Citation(s) in RCA: 340] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We previously identified a stromal cell-derived inducing activity (SDIA), which induces differentiation of neural cells, including midbrain tyrosine hydroxylase-positive (TH(+)) dopaminergic neurons, from mouse embryonic stem cells. We report here that SDIA induces efficient neural differentiation also in primate embryonic stem cells. Induced neurons contain TH(+) neurons at a frequency of 35% and produce a significant amount of dopamine. Interestingly, differentiation of TH(+) neurons from undifferentiated embryonic cells occurs much faster in vitro (10 days) than it does in the embryo (approximately 5 weeks). In addition, 8% of the colonies contain large patches of Pax6(+)-pigmented epithelium of the retina. The SDIA method provides an unlimited source of primate cells for the study of pathogenesis, drug development, and transplantation in degenerative diseases such as Parkinson's disease and retinitis pigmentosa.
Collapse
Affiliation(s)
- Hiroshi Kawasaki
- Department of Medical Embryology and Neurobiology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
993
|
Abstract
In the decade since the telomere hypothesis of cellular aging was proposed, the two essential genes for human telomerase were cloned and characterized, allowing experimental proof of the causal relationships between telomere loss and replicative senescence, and telomerase activation and immortalization. These relationships were established using a variety of cultured human cell types from both normal and tumor tissues, and were largely confirmed in the telomerase knockout mouse. Taken together, the data provide strong support for the potential utility of telomerase detection and inhibition for cancer, and telomerase activation for degenerative diseases. The specificity of the promoter for the telomerase catalytic gene and the antigenicity of the protein product, hTERT, provide additional strategies for killing telomerase-positive tumor cells. Unfortunately, the strong link between telomerase and cancer has led some to confuse telomerase activation with cancer, and others to overstate the cancer risk of telomerase activation therapies for degenerative diseases. This review clarifies the difference between telomerase, which does not cause growth deregulation, and oncogenes, which do. It also addresses the concept of telomerase repression as a tumor suppressor mechanism early in life, with detrimental tissue degeneration and tumor-promoting consequences late in life. This extended view of the telomere hypothesis helps explain how telomerase inhibition can be therapeutic in cancer patients, while controlled telomerase activation for degenerative diseases may actually reduce, rather than increase, the frequency of age-related tumorigenesis.
Collapse
|
994
|
Abstract
Mouse embryonic stem cells are continuous cell lines derived directly from the fetal founder tissue of the preimplantation embryo. They can be expanded in culture while retaining the functional attributes of pluripotent early embryo cells. In particular, they can participate fully in fetal development when reintroduced into the embryo. The capacity for multilineage differentiation is reproduced in culture where embryonic stem cells can produce a wide range of well-defined cell types. This has stimulated interest in the isolation of analogous cells of human origin. Such human pluripotent stem cells could constitute a renewable source of more differentiated cells that could be employed to replace diseased or damaged tissue by cellular transplantation. In this review, the relationships between mouse embryonic stem cells, resident pluripotent cells in the embryo, and human embryo-derived cell lines are evaluated, and the prospects and challenges of embryo stem cell research are considered.
Collapse
Affiliation(s)
- A G Smith
- Centre for Genome Research, University of Edinburgh, The King's Buildings, West Mains Road, Edinburgh, Scotland, EH9 3JQ, United Kingdom.
| |
Collapse
|
995
|
Abstract
Early differentiation of the mammalian embryo leads to the development of two distinct lineages-the inner cell mass (ICM) and the trophectoderm. Cells of the ICM are pluripotent and give rise to all tissues of the fetus, while trophectoderm cells are restricted in their potential to the trophoblast cell layers of the placenta. In the mouse, apparently immortal stem cell lines can be obtained from both cell types. These cell lines, embryonic stem (ES) cells and trophoblast stem (TS) cells, are morphologically and molecularly distinct and depend on different signaling pathways for their maintenance. They also show different cell fates when introduced into early embryos to generate chimeras. However, a change in the levels of expression of a key regulator of pluripotency, Oct4, can push ES cells towards the TS phenotype, when grown in TS cell conditions. Stem cell potential in the early embryo thus appears to depend on a combination of the levels of expression of key intrinsic regulators and the appropriate extrinsic environmental factors. Manipulation of both intrinsic and extrinsic regulators may be needed to reveal the full potential of stem cells from other stages of development and the adult.
Collapse
Affiliation(s)
- J Rossant
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario, Canada M5G 1X5.
| |
Collapse
|
996
|
Verfaillie CM, Pera MF, Lansdorp PM. Stem cells: hype and reality. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2002; 2002:369-391. [PMID: 12446433 DOI: 10.1182/asheducation-2002.1.369] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
This update discusses what is known regarding embryonic and adult tissue-derived pluripotent stem cells, including the mechanisms underlying self-renewal without senescence, differentiation in multiple cell types both in vitro and in vivo, and future potential clinical uses of such stem cells. In Section I, Dr. Lansdorp reviews the structure and function of telomerase, the enzyme that restores telomeric ends of chromosomes upon cell division, highly present in embryonic stem cells but not adult stem cells. He discusses the structure and function of telomerase and signaling pathways activated by the enzyme, with special emphasis on normal and leukemic hematopoietic stem cells. In Section II, Dr. Pera reviews the present understanding of mammalian pluripotent embryonic stem cells. He discusses the concept of pluripotentiality in its embryonic context, derivation of stem cells from embryonic or fetal tissue, the basic properties of the stem cells, and methods to produce specific types of differentiated cell from stem cells. He examines the potential applications of stem cells in research and medicine and some of the barriers that must be crossed to achieve these goals. In Section III, Dr. Verfaillie reviews the present understanding of pluripotency of adult stem cells. She discusses the concept of stem cell plasticity, a term used to describe the greater potency described by several investigators of adult tissue-derived stem cells, critically reviews the published studies demonstrating stem cell plasticity, and possible mechanisms underlying such plasticity, and examines the possible role of pluripotent adult stem cells in research and medicine.
Collapse
|
997
|
Carpenter MK, Inokuma MS, Denham J, Mujtaba T, Chiu CP, Rao MS. Enrichment of neurons and neural precursors from human embryonic stem cells. Exp Neurol 2001; 172:383-97. [PMID: 11716562 DOI: 10.1006/exnr.2001.7832] [Citation(s) in RCA: 336] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Human embryonic stem (hES) cells proliferate and maintain their pluripotency for over a year in vitro (M. Amit, M. K. Carpenter, M. S. Inokuma, C. P. Chiu, C. P., Harris, M. A. Waknitz, J. Itskovitz-Eldor, and J. A. Thomson. 2000. Dev. Biol. 227: 271-278) and may therefore provide a cell source for cell therapies. hES cells were maintained for over 6 months in vitro (over 100 population doublings) before their ability to differentiate into the neural lineage was evaluated. Differentiation was induced by the formation of embryoid bodies that were subsequently plated onto appropriate substrates in defined medium containing mitogens. These populations contained cells that showed positive immunoreactivity to nestin, polysialylated neural cell adhesion molecule (PS-NCAM) and A2B5. After further maturation, these cells expressed additional neuron-specific antigens (such as MAP-2, synaptophysin, and various neurotransmitters). Calcium imaging demonstrated that these cells responded to neurotransmitter application. Electrophysiological analyses showed that cell membranes contained voltage-dependent channels and that action potentials were triggered by current injection. PS-NCAM and A2B5 immunoselection or culture conditions could be used to produce enriched populations (60-90%) which could be further differentiated into mature neurons. The properties of the hES-derived progenitors and neurons were found to be similar to those of cells derived from primary tissue. These data indicate that hES cells could provide a cell source for the neural progenitor cells and mature neurons for therapeutic and toxicological uses.
Collapse
Affiliation(s)
- M K Carpenter
- Geron Corporation, 230 Constitution Drive, Menlo Park, California 94025, USA.
| | | | | | | | | | | |
Collapse
|
998
|
Zhang SC, Wernig M, Duncan ID, Brüstle O, Thomson JA. In vitro differentiation of transplantable neural precursors from human embryonic stem cells. Nat Biotechnol 2001; 19:1129-33. [PMID: 11731781 DOI: 10.1038/nbt1201-1129] [Citation(s) in RCA: 1341] [Impact Index Per Article: 55.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The remarkable developmental potential and replicative capacity of human embryonic stem (ES) cells promise an almost unlimited supply of specific cell types for transplantation therapies. Here we describe the in vitro differentiation, enrichment, and transplantation of neural precursor cells from human ES cells. Upon aggregation to embryoid bodies, differentiating ES cells formed large numbers of neural tube-like structures in the presence of fibroblast growth factor 2 (FGF-2). Neural precursors within these formations were isolated by selective enzymatic digestion and further purified on the basis of differential adhesion. Following withdrawal of FGF-2, they differentiated into neurons, astrocytes, and oligodendrocytes. After transplantation into the neonatal mouse brain, human ES cell-derived neural precursors were incorporated into a variety of brain regions, where they differentiated into both neurons and astrocytes. No teratoma formation was observed in the transplant recipients. These results depict human ES cells as a source of transplantable neural precursors for possible nervous system repair.
Collapse
Affiliation(s)
- S C Zhang
- Department of Anatomy, University of Wisconsin 1500 Highland Avenue, Madison, WI 53705, USA.
| | | | | | | | | |
Collapse
|
999
|
Abstract
Embryonic stem cells are derived from the inner cell mass of the pre-implantation blastocyst, and can both self-renew and differentiate into all the cells and tissues of the body. The embryonic stem cell is an unsurpassed starting material to begin to understand a critical, largely inaccessible, period of development, as well as an important source of cells for transplantation and gene therapy. Despite their potential, attempts to obtain specific cell types from embryonic stem cells have been only partially successful because many of the growth factor combinations and developmental control genes involved in cell type restricted differentiation are unknown. This article summarizes some of the recent advances in promoting lineage restricted differentiation of embryonic stem cells, focusing on growth factor manipulation, or genetically altering embryonic stem cells to produce a desired phenotype. The two approaches epitomize current scientific concerns regarding the therapeutic use of these cells; genetic alterations will produce more pure cells with the risk of increasing the likelihood of malignant transformation; epigenetic methods for the manipulation of stem cell phenotype are often incomplete and remaining pluripotent cells are likely to form teratomas. As more is known about lineage specification during development, it will be possible to more precisely control cell type specification.
Collapse
Affiliation(s)
- K S O'Shea
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-0616, USA.
| |
Collapse
|
1000
|
Abstract
Pluripotent stem cells can be expanded seemingly indefinitely in culture, maintain a normal karyotype and have the potential to generate any cell type in the body. As such they represent an incredible resource for the repair of diseased or damaged tissues in our bodies. These cells also promise to open a new window into the embryonic development of our species.
Collapse
Affiliation(s)
- P J Donovan
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA.
| | | |
Collapse
|