2801
|
Abstract
Stem cell-based regenerative medicine holds great promise for repair of diseased tissue. Modern directions in the field of epigenetic research aimed to decipher the epigenetic signals that give stem cells their unique ability to self-renew and differentiate into different cell types. However, this research is only the tip of the iceberg when it comes to writing an 'epigenetic instruction manual' for the ramification of molecular details of cell commitment and differentiation. In this review, we discuss the impact of the epigenetic research on our understanding of stem cell biology.
Collapse
Affiliation(s)
- Victoria V Lunyak
- Buck Institute for Age Research, 8001 Redwood Blvd, Novato, CA 94945, USA.
| | | |
Collapse
|
2802
|
McElroy SL, Kee K, Tran N, Menses J, Giudice LC, Reijo Pera RA. Developmental competence of immature and failed/abnormally fertilized human oocytes in nuclear transfer. Reprod Biomed Online 2008; 16:684-93. [PMID: 18492373 DOI: 10.1016/s1472-6483(10)60483-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Somatic cell nuclear transfer holds great promise for basic studies of reprogramming human somatic cells and for the potential development of novel cell-based therapeutics. The aim of this study was to examine experimental aspects of human nuclear transfer via use of an abundant source of oocytes, those that are routinely discarded from assisted reproduction clinics. The results suggest and reinforce several findings based on the analysis of multiple parameters: first, it was observed that supplementation of commercial culture media with hormones promoted embryo development after parthenogenetic activation. Second, the use of the chemical activation reagent puromycin resulted in significant differences in cleavage rates in oocytes that were failed/abnormally fertilized after intracytoplasmic sperm injection relative to those from IVF (P < 0.05). Third, cycloheximide promoted cleavage rates >/=40% in both groups of oocytes; moreover, two blastocysts were produced following cycloheximide treatment. Finally, the use of a subset of oocytes for nuclear transfer resulted in cleaved embryos that expressed green fluorescent protein from a transgene in donor nuclei from human embryonic stem cells. In light of these results, it is suggested that the discarded oocytes can be used to investigate new human nuclear transfer protocols for embryonic stem cell derivation.
Collapse
Affiliation(s)
- Sohyun Lee McElroy
- Institute for Stem Cell Biology and Regenerative Medicine, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, Palo Alto, CA 94304-5542, USA
| | | | | | | | | | | |
Collapse
|
2803
|
Dorsomorphin, a selective small molecule inhibitor of BMP signaling, promotes cardiomyogenesis in embryonic stem cells. PLoS One 2008; 3:e2904. [PMID: 18682835 PMCID: PMC2483414 DOI: 10.1371/journal.pone.0002904] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Accepted: 07/13/2008] [Indexed: 01/13/2023] Open
Abstract
Background Pluripotent embryonic stem (ES) cells, which have the capacity to give rise to all tissue types in the body, show great promise as a versatile source of cells for regenerative therapy. However, the basic mechanisms of lineage specification of pluripotent stem cells are largely unknown, and generating sufficient quantities of desired cell types remains a formidable challenge. Small molecules, particularly those that modulate key developmental pathways like the bone morphogenetic protein (BMP) signaling cascade, hold promise as tools to study in vitro lineage specification and to direct differentiation of stem cells toward particular cell types. Methodology/ Principal Findings We describe the use of dorsomorphin, a selective small molecule inhibitor of BMP signaling, to induce myocardial differentiation in mouse ES cells. Cardiac induction is very robust, increasing the yield of spontaneously beating cardiomyocytes by at least 20 fold. Dorsomorphin, unlike the endogenous BMP antagonist Noggin, robustly induces cardiomyogenesis when treatment is limited to the initial 24-hours of ES cell differentiation. Quantitative-PCR analyses of differentiating ES cells indicate that pharmacological inhibition of BMP signaling during the early critical stage promotes the development of the cardiomyocyte lineage, but reduces the differentiation of endothelial, smooth muscle, and hematopoietic cells. Conclusions/ Significance Administration of a selective small molecule BMP inhibitor during the initial stages of ES cell differentiation substantially promotes the differentiation of primitive pluripotent cells toward the cardiomyocytic lineage, apparently at the expense of other mesodermal lineages. Small molecule modulators of developmental pathways like dorsomorphin could become versatile pharmacological tools for stem cell research and regenerative medicine.
Collapse
|
2804
|
Abstract
Stem cells are defined by their capacity of self-renewal and multilineage differentiation, which make them uniquely situated to treat a broad spectrum of human diseases. Based on a series of remarkable studies in several fields of regenerative medicine, their application is not too far from the clinical practice. Full-thickness burns and severe traumas can injure skin and its appendages, which protect animals from water loss, temperature change, radiation, trauma and infection. In adults, the normal outcome of repair of massive full-thickness burns is fibrosis and scarring without any appendages, such as hair follicles, sweat and sebaceous glands. Perfect skin regeneration has been considered impossible due to the limited regenerative capacity of epidermal keratinocytes, which are generally thought to be the key source of the epidermis and skin appendages. Currently, researches on stem cells, such as epidermal stem cells, dermal stem cells, mesenchymal stem cells from bone marrow, and embryonic stem cells, bring promise to functional repair of skin after severe burn injury, namely, complete regeneration of skin and its appendages. In this study, we present an overview of the most recent advances in skin repair and regeneration by using stem cells.
Collapse
|
2805
|
Woodland HR, Zorn AM. The core endodermal gene network of vertebrates: combining developmental precision with evolutionary flexibility. Bioessays 2008; 30:757-65. [DOI: 10.1002/bies.20785] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
2806
|
|
2807
|
Aoi T, Yae K, Nakagawa M, Ichisaka T, Okita K, Takahashi K, Chiba T, Yamanaka S. Generation of Pluripotent Stem Cells from Adult Mouse Liver and Stomach Cells. Science 2008; 321:699-702. [DOI: 10.1126/science.1154884] [Citation(s) in RCA: 814] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
2808
|
Fenno LE, Ptaszek LM, Cowan CA. Human embryonic stem cells: emerging technologies and practical applications. Curr Opin Genet Dev 2008; 18:324-9. [DOI: 10.1016/j.gde.2008.06.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 06/11/2008] [Accepted: 06/14/2008] [Indexed: 10/21/2022]
|
2809
|
Das Schicksal kardialer Zelltransplantate. ZEITSCHRIFT FUR HERZ THORAX UND GEFASSCHIRURGIE 2008. [DOI: 10.1007/s00398-008-0646-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
2810
|
Yuasa S, Fukuda K. Recent advances in cardiovascular regenerative medicine: the induced pluripotent stem cell era. Expert Rev Cardiovasc Ther 2008; 6:803-10. [PMID: 18570618 DOI: 10.1586/14779072.6.6.803] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Induced pluripotent stem (iPS) cells have recently been established by transfecting mouse and human fibroblasts with the transcription factors Oct3/4, Sox2, Klf4 and c-Myc, known to be expressed at high levels in embryonic stem (ES) cells. These cells have great potential in regenerative medicine as they have the capacity to differentiate into all three germ layer-derived cells and are syngeneic. The differentiation of ES cells into cardiomyocytes mimics the early processes involved in heart development. Recent studies describe the contribution of various growth factors and corresponding inhibitors to heart development during embryogenesis. Bone morphogenetic proteins, Wnt protein and Notch signals play critical roles in heart development in a context- and time-dependent manner. Consistent with ES cells, the exposure of iPS cells to such growth factors is hypothesized to augment differentiation into cardiomyocytes. The combination of iPS cells and appropriate developmental signal information has the potential for providing the foundations for future regenerative medicine.
Collapse
Affiliation(s)
- Shinsuke Yuasa
- Cardiology Division, Department of Medicine, Keio University School of Medicine, 35-Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | | |
Collapse
|
2811
|
Abstract
The development of personalized pluripotent stem cells for research and for possible therapies holds out great hope for patients. However, such cells will face significant technical and regulatory challenges before they can be used as therapeutic reagents. Here we consider two possible sources of personalized pluripotent stem cells: embryonic stem cells derived from nuclear transfer (NT-ESCs) and induced pluripotent stem cells (iPSCs) derived from direct reprogramming of adult somatic cells. Both sources of personalized pluripotent stem cells face unique regulatory hurdles that are in some ways significantly higher than those facing stem cells derived from embryos produced by fertilization (ESCs). However, the outstanding long-term potential of iPSCs and their relative freedom from the ethical concerns raised by both ESCs and NT-ESCs makes direct reprogramming an exceptionally promising approach to advancing research and providing therapies in the field of regenerative medicine.
Collapse
Affiliation(s)
- Maureen L Condic
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, Utah 84132-3401, USA.
| | | |
Collapse
|
2812
|
Dimos JT, Rodolfa KT, Niakan KK, Weisenthal LM, Mitsumoto H, Chung W, Croft GF, Saphier G, Leibel R, Goland R, Wichterle H, Henderson CE, Eggan K. Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science 2008; 321:1218-21. [PMID: 18669821 DOI: 10.1126/science.1158799] [Citation(s) in RCA: 1386] [Impact Index Per Article: 86.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The generation of pluripotent stem cells from an individual patient would enable the large-scale production of the cell types affected by that patient's disease. These cells could in turn be used for disease modeling, drug discovery, and eventually autologous cell replacement therapies. Although recent studies have demonstrated the reprogramming of human fibroblasts to a pluripotent state, it remains unclear whether these induced pluripotent stem (iPS) cells can be produced directly from elderly patients with chronic disease. We have generated iPS cells from an 82-year-old woman diagnosed with a familial form of amyotrophic lateral sclerosis (ALS). These patient-specific iPS cells possess properties of embryonic stem cells and were successfully directed to differentiate into motor neurons, the cell type destroyed in ALS.
Collapse
Affiliation(s)
- John T Dimos
- Harvard Stem Cell Institute, Stowers Medical Institute, Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2813
|
Chiou SH, Yu CC, Huang CY, Lin SC, Liu CJ, Tsai TH, Chou SH, Chien CS, Ku HH, Lo JF. Positive correlations of Oct-4 and Nanog in oral cancer stem-like cells and high-grade oral squamous cell carcinoma. Clin Cancer Res 2008; 14:4085-95. [PMID: 18593985 DOI: 10.1158/1078-0432.ccr-07-4404] [Citation(s) in RCA: 507] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Oral squamous cell carcinoma (OSCC), like many solid tumors, contains a heterogeneous population of cancer cells. Recent data suggest that a rare subpopulation of cancer cells, termed cancer stem cells (CSC), is capable of initiating, maintaining, and expanding the growth of tumor. Identification and characterization of CSC from OSCC facilitates the monitoring, therapy, or prevention of OSCC. EXPERIMENTAL DESIGN We enriched oral cancer stem-like cells (OC-SLC) through sphere formation by cultivating OSCC cells from established OSCC cell lines or primary cultures of OSCC patients within defined serum-free medium. Differential expression profile of stemness genes between enriched OC-SLC and parental OSCC was elucidated. Furthermore, immunohistochemical staining of stemness markers on OSCC patient tissues was examined to evaluate the association between stemness genes and prognosis of OSCC. RESULTS Enriched OC-SLC highly expressed the stem/progenitor cell markers and ABC transporter gene (Oct-4, Nanog, CD117, Nestin, CD133, and ABCG2) and also displayed induced differentiation abilities and enhanced migration/invasion/malignancy capabilities in vitro and in vivo. Elevated expression of CD133 was shown in the enriched OC-SLC from OSCC patients' tumors. Positive correlations of Oct-4, Nanog, or CD133 expression on tumor stage were shown on 52 OSCC patient tissues. Kaplan-Meier analyses exhibited that Nanog/Oct-4/CD133 triple-positive patients predicted the worst survival prognosis of OSCC patients. CONCLUSION We enriched a subpopulation of cancer stem-like cell from OSCC by sphere formation. The enriched OC-SLC possesses the characteristics of both stem cells and malignant tumors. Additionally, expression of stemness markers (Nanog/Oct-4/CD133) contradicts the survival prognosis of OSCC patients.
Collapse
Affiliation(s)
- Shih-Hwa Chiou
- Institutes of Oral Biology, Traditional Medicine, and Anatomy and Cell Biology, National Yang-Ming University, Taiwan, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
2814
|
Mauritz C, Schwanke K, Reppel M, Neef S, Katsirntaki K, Maier LS, Nguemo F, Menke S, Haustein M, Hescheler J, Hasenfuss G, Martin U. Generation of Functional Murine Cardiac Myocytes From Induced Pluripotent Stem Cells. Circulation 2008; 118:507-17. [DOI: 10.1161/circulationaha.108.778795] [Citation(s) in RCA: 380] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background—
The recent breakthrough in the generation of induced pluripotent stem (iPS) cells, which are almost indistinguishable from embryonic stem (ES) cells, facilitates the generation of murine disease– and human patient–specific stem cell lines. The aim of this study was to characterize the cardiac differentiation potential of a murine iPS cell clone in comparison to a well-established murine ES cell line.
Methods and Results—
With the use of a standard embryoid body–based differentiation protocol for ES cells, iPS cells as well as ES cells were differentiated for 24 days. Although the analyzed iPS cell clone showed a delayed and less efficient formation of beating embryoid bodies compared with the ES cell line, the differentiation resulted in an average of 55% of spontaneously contracting iPS cell embryoid bodies. Analyses on molecular, structural, and functional levels demonstrated that iPS cell–derived cardiomyocytes show typical features of ES cell–derived cardiomyocytes. Reverse transcription polymerase chain reaction analyses demonstrated expression of marker genes typical for mesoderm, cardiac mesoderm, and cardiomyocytes including Brachyury, mesoderm posterior factor 1 (Mesp1), friend of GATA2 (FOG-2), GATA-binding protein 4 (GATA4), NK2 transcription factor related, locus 5 (Nkx2.5), T-box 5 (Tbx5), T-box 20 (Tbx20), atrial natriuretic factor (ANF), myosin light chain 2 atrial transcripts (MLC2a), myosin light chain 2 ventricular transcripts (MLC2v), α-myosin heavy chain (α-MHC), and cardiac troponin T in differentiation cultures of iPS cells. Immunocytology confirmed expression of cardiomyocyte-typical proteins including sarcomeric α-actinin, titin, cardiac troponin T, MLC2v, and connexin 43. iPS cell cardiomyocytes displayed spontaneous rhythmic intracellular Ca
2+
fluctuations with amplitudes of Ca
2+
transients comparable to ES cell cardiomyocytes. Simultaneous Ca
2+
release within clusters of iPS cell–derived cardiomyocytes indicated functional coupling of the cells. Electrophysiological studies with multielectrode arrays demonstrated functionality and presence of the β-adrenergic and muscarinic signaling cascade in these cells.
Conclusions—
iPS cells differentiate into functional cardiomyocytes. In contrast to ES cells, iPS cells allow derivation of autologous functional cardiomyocytes for cellular cardiomyoplasty and myocardial tissue engineering.
Collapse
Affiliation(s)
- Christina Mauritz
- From the Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation, and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany (C.M., K.S., K.K., S.M., U.M.); Institute of Neurophysiology, University of Cologne, Cologne, Germany (M.R., F.N., M.H., J.H.); Department of Cardiology, University of Luebeck, Luebeck, Germany (M.R.); and Department of Cardiology and Pneumology, University Medical Center
| | - Kristin Schwanke
- From the Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation, and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany (C.M., K.S., K.K., S.M., U.M.); Institute of Neurophysiology, University of Cologne, Cologne, Germany (M.R., F.N., M.H., J.H.); Department of Cardiology, University of Luebeck, Luebeck, Germany (M.R.); and Department of Cardiology and Pneumology, University Medical Center
| | - Michael Reppel
- From the Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation, and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany (C.M., K.S., K.K., S.M., U.M.); Institute of Neurophysiology, University of Cologne, Cologne, Germany (M.R., F.N., M.H., J.H.); Department of Cardiology, University of Luebeck, Luebeck, Germany (M.R.); and Department of Cardiology and Pneumology, University Medical Center
| | - Stefan Neef
- From the Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation, and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany (C.M., K.S., K.K., S.M., U.M.); Institute of Neurophysiology, University of Cologne, Cologne, Germany (M.R., F.N., M.H., J.H.); Department of Cardiology, University of Luebeck, Luebeck, Germany (M.R.); and Department of Cardiology and Pneumology, University Medical Center
| | - Katherina Katsirntaki
- From the Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation, and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany (C.M., K.S., K.K., S.M., U.M.); Institute of Neurophysiology, University of Cologne, Cologne, Germany (M.R., F.N., M.H., J.H.); Department of Cardiology, University of Luebeck, Luebeck, Germany (M.R.); and Department of Cardiology and Pneumology, University Medical Center
| | - Lars S. Maier
- From the Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation, and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany (C.M., K.S., K.K., S.M., U.M.); Institute of Neurophysiology, University of Cologne, Cologne, Germany (M.R., F.N., M.H., J.H.); Department of Cardiology, University of Luebeck, Luebeck, Germany (M.R.); and Department of Cardiology and Pneumology, University Medical Center
| | - Filomain Nguemo
- From the Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation, and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany (C.M., K.S., K.K., S.M., U.M.); Institute of Neurophysiology, University of Cologne, Cologne, Germany (M.R., F.N., M.H., J.H.); Department of Cardiology, University of Luebeck, Luebeck, Germany (M.R.); and Department of Cardiology and Pneumology, University Medical Center
| | - Sandra Menke
- From the Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation, and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany (C.M., K.S., K.K., S.M., U.M.); Institute of Neurophysiology, University of Cologne, Cologne, Germany (M.R., F.N., M.H., J.H.); Department of Cardiology, University of Luebeck, Luebeck, Germany (M.R.); and Department of Cardiology and Pneumology, University Medical Center
| | - Moritz Haustein
- From the Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation, and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany (C.M., K.S., K.K., S.M., U.M.); Institute of Neurophysiology, University of Cologne, Cologne, Germany (M.R., F.N., M.H., J.H.); Department of Cardiology, University of Luebeck, Luebeck, Germany (M.R.); and Department of Cardiology and Pneumology, University Medical Center
| | - Juergen Hescheler
- From the Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation, and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany (C.M., K.S., K.K., S.M., U.M.); Institute of Neurophysiology, University of Cologne, Cologne, Germany (M.R., F.N., M.H., J.H.); Department of Cardiology, University of Luebeck, Luebeck, Germany (M.R.); and Department of Cardiology and Pneumology, University Medical Center
| | - Gerd Hasenfuss
- From the Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation, and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany (C.M., K.S., K.K., S.M., U.M.); Institute of Neurophysiology, University of Cologne, Cologne, Germany (M.R., F.N., M.H., J.H.); Department of Cardiology, University of Luebeck, Luebeck, Germany (M.R.); and Department of Cardiology and Pneumology, University Medical Center
| | - Ulrich Martin
- From the Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation, and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany (C.M., K.S., K.K., S.M., U.M.); Institute of Neurophysiology, University of Cologne, Cologne, Germany (M.R., F.N., M.H., J.H.); Department of Cardiology, University of Luebeck, Luebeck, Germany (M.R.); and Department of Cardiology and Pneumology, University Medical Center
| |
Collapse
|
2815
|
Abstract
Since its inception 20 years ago, the animal pharming industry has promoted transgenic animals as a cost-effective method of biopharmaceutical production. However, it took until 2006 for the first therapeutic product to gain regulatory approval. This was an important milestone, but scepticism still abounds. Can pharming regain investor confidence, and will society accept transgenic livestock as a production method? There is some cause for optimism, biopharmaceuticals are a large, expanding market and animal pharming has already made considerable strides. A novel production platform has been established, groundbreaking technologies developed, a necessary regulatory framework put in place. Nevertheless, despite cost advantages, pharming has become a niche production method and its long term success may depend on products unique to transgenic animals.
Collapse
|
2816
|
Pei DS, Sun YH, Zhu ZY. Identification of a novel gene K23 over-expressed in fish cross-subfamily cloned embryos. Mol Biol Rep 2008; 36:1375-80. [PMID: 18654838 DOI: 10.1007/s11033-008-9323-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Accepted: 07/14/2008] [Indexed: 10/21/2022]
Abstract
A novel gene-K23, differentially expressed in cross-subfamily cloned embryos, was isolated by RACE-PCR technique. It had 2580 base pairs (bp) in length, with a 1,425 bp open reading frame (ORF) encoding a putative protein of 474 amino acids (aa). Bioinformatic analysis indicated that K23 had 22 phosphorylation sites, but it had no signal peptides. Developmental expression analysis in zebrafish showed that K23 transcripts were maternally expressed in ovum and the amount of K23 transcripts increased gradually from zygote to pharyngula period. Subcellular localization analysis revealed that K23 protein was homogeneously distributed both in nuclei and cytoplasm. Taken together, our findings indicate that K23 gene is a novel gene differentially expressed in fish cross-subfamily cloned embryos.
Collapse
Affiliation(s)
- De-Sheng Pei
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | | | | |
Collapse
|
2817
|
Egli D, Birkhoff G, Eggan K. Mediators of reprogramming: transcription factors and transitions through mitosis. Nat Rev Mol Cell Biol 2008; 9:505-16. [PMID: 18568039 DOI: 10.1038/nrm2439] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
It is thought that most cell types of the human body share the same genetic information as that contained in the zygote from which they originate. Consistent with this view, animal cloning studies demonstrated that the intact genome of a differentiated cell can be reprogrammed to support the development of an entire organism and allow the production of pluripotent stem cells. Recent progress in reprogramming research now points to an important role for transcription factors in the establishment and the maintenance of cellular phenotypes, and to cell division as a mediator of transitions between different states of gene expression.
Collapse
Affiliation(s)
- Dieter Egli
- The Stowers Medical Institute, Harvard Stem Cell Institute and the Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | | | | |
Collapse
|
2818
|
Abstract
The Foxd3 forkhead transcription factor is required for maintaining pluripotent cells in the early mouse embryo and for the establishment of murine embryonic stem cell (ESC) lines. To begin to understand the role of Foxd3 in ESC maintenance, we derived ESC lines from blastocysts that carried two conditional Foxd3 alleles and a tamoxifen-inducible Cre transgene. Tamoxifen treatment produced a rapid and near complete loss of Foxd3 mRNA and protein. Foxd3-deficient ESCs maintained a normal proliferation rate but displayed increased apoptosis, and clonally dispersed ESCs showed a decreased ability to self-renew. Under either self-renewal or differentiation-promoting culture conditions we observed a strong, precocious differentiation of Foxd3 mutant ESCs along multiple lineages, including trophectoderm, endoderm, and mesendoderm. This profound alteration in biological behavior occurred in the face of continued expression of factors known to induce pluripotency, including Oct4, Sox2, and Nanog. We present a model for the role of Foxd3 in repressing differentiation, promoting self-renewal, and maintaining survival of mouse ESCs. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Ying Liu
- Department of Cell and Developmental Biology, Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0494, USA.
| | | |
Collapse
|
2819
|
Parisi S, Passaro F, Aloia L, Manabe I, Nagai R, Pastore L, Russo T. Klf5 is involved in self-renewal of mouse embryonic stem cells. J Cell Sci 2008; 121:2629-34. [PMID: 18653541 DOI: 10.1242/jcs.027599] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Self-renewal of embryonic stem cells (ESCs) is maintained by a complex regulatory mechanism involving transcription factors Oct3/4 (Pou5f1), Nanog and Sox2. Here, we report that Klf5, a Zn-finger transcription factor of the Kruppel-like family, is involved in ESC self-renewal. Klf5 is expressed in mouse ESCs, blastocysts and primordial germ cells, and its knockdown by RNA interference alters the molecular phenotype of ESCs, thereby preventing their correct differentiation. The ability of Klf5 to maintain ESCs in the undifferentiated state is supported by the finding that differentiation of ESCs is prevented when Klf5 is constitutively expressed. Maintenance of the undifferentiated state by Klf5 is, at least in part, due to the control of Nanog and Oct3/4 transcription, because Klf5 directly binds to the promoters of these genes and regulates their transcription.
Collapse
|
2820
|
Stiles CD, Rowitch DH. Glioma stem cells: a midterm exam. Neuron 2008; 58:832-46. [PMID: 18579075 DOI: 10.1016/j.neuron.2008.05.031] [Citation(s) in RCA: 247] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 05/22/2008] [Accepted: 05/23/2008] [Indexed: 02/06/2023]
Abstract
Several years ago, the discovery of a highly tumorigenic subpopulation of stem-like cells embedded within fresh surgical isolates of malignant gliomas lent support to a new paradigm in cancer biology--the cancer stem cell hypothesis. At the same time, these "glioma stem cells" seemed to resolve a long-standing conundrum on the cell of origin for primary cancers of the brain. However, central tenets of the cancer stem cell hypothesis have recently been challenged, and the cellular origins of stem-like cells within malignant glioma are still contended. Here, we summarize the issues that are still in play with respect to the cancer stem cell hypothesis, and we revisit the developmental origins of malignant glioma. Do glioma stem cells arise from developmentally stalled neural progenitors or from dedifferentiated astrocytes? Five separate predictions of a neural progenitor cell of origin are put to the test.
Collapse
Affiliation(s)
- Charles D Stiles
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA.
| | | |
Collapse
|
2821
|
Sakurada K, McDonald F, Shimada F. Regenerative Medicine and Stem Cell Based Drug Discovery. Angew Chem Int Ed Engl 2008; 47:5718-38. [DOI: 10.1002/anie.200700724] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
2822
|
|
2823
|
Kiel ME, Chen CP, Sadowski D, McKinnon RD. Stem cell-derived therapeutic myelin repair requires 7% cell replacement. Stem Cells 2008; 26:2229-36. [PMID: 18635868 DOI: 10.1634/stemcells.2008-0218] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Embryonic stem cells (ESCs) hold great potential for therapeutic regeneration and repair in many diseases. However, many challenges remain before this can be translated into effective therapy. A principal and significant limit for outcome evaluations of clinical trials is to define the minimal graft population necessary for functional repair. Here we used a preclinical model for quantitative analysis of stem cell grafts, with wild-type ESC grafted into myelin mutant shiverer hosts, to determine minimum graft levels for therapeutic benefit. Using a timed motor function test we identified three groups, including recipients indistinguishable from nongrafted shiverer controls (time [t] = 20.1 +/- 1.1 seconds), mice with marginal improvement (t = 15.7 +/- 1 seconds), and mice with substantial phenotype rescue (t = 5.7 +/- 0.9 seconds). The motor function rescued chimeras also had a considerably extended life span (T(50) > 128 days) relative to both shiverer (T(50) = 108 days) and the nonrescued chimeras. Retrospective genotype analysis identified a strong correlation (r(2) = 0.85) between motor function and ESC-derived chimerism, with > 7% chimerism required for rescue in this murine model of central nervous system myelin pathology. These results establish the minimal levels of engraftment to anticipate therapeutic repair of a cell-autonomous defect by cell transplant therapy.
Collapse
Affiliation(s)
- Mary E Kiel
- UMDNJ-Robert Wood Johnson Medical School, 675 Hoes Lane, S-225, Piscataway NJ 08854, USA
| | | | | | | |
Collapse
|
2824
|
Eminli S, Utikal J, Arnold K, Jaenisch R, Hochedlinger K. Reprogramming of neural progenitor cells into induced pluripotent stem cells in the absence of exogenous Sox2 expression. Stem Cells 2008; 26:2467-74. [PMID: 18635867 DOI: 10.1634/stemcells.2008-0317] [Citation(s) in RCA: 239] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Expression of the transcription factors Oct4, Sox2, Klf4, and c-Myc in mesodermal and endodermal derivatives, including fibroblasts, lymphocytes, liver, stomach, and beta cells, generates induced pluripotent stem (iPS) cells. It remains unknown, however, whether cell types of the ectodermal lineage are equally amenable to reprogramming into iPS cells by the same combination of factors. To test this, we have isolated genetically marked neural progenitor cells (NPCs) from neonatal mouse brains and infected them with viral vectors expressing Oct4, Sox2, Klf4, and c-Myc. Infected NPCs gave rise to iPS cells that expressed markers of embryonic stem cells, showed demethylation of pluripotency genes, formed teratomas, and contributed to viable chimeras. In contrast to other somatic cell types, NPCs expressed high levels of endogenous Sox2 and thus did not require viral Sox2 expression for reprogramming into iPS cells. Our data show that in addition to mesoderm- and endoderm-derived cell types, neural progenitor cells of the ectodermal lineage can be reprogrammed into iPS cells, suggesting that in vitro reprogramming is a universal process. These results also imply that the combination of factors necessary for reprogramming is dependent on cellular context. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Sarah Eminli
- Massachusetts General Hospital Cancer Center and Center for Regenerative Medicine, Boston, Massachusetts 02114, USA
| | | | | | | | | |
Collapse
|
2825
|
Werbowetski-Ogilvie TE, Bhatia M. Pluripotent human stem cell lines: what we can learn about cancer initiation. Trends Mol Med 2008; 14:323-32. [PMID: 18635398 DOI: 10.1016/j.molmed.2008.06.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Revised: 06/04/2008] [Accepted: 06/04/2008] [Indexed: 12/30/2022]
Abstract
Although the cancer stem cell (CSC) hypothesis has become an attractive model to account for tumor recurrence, failure to define a cell of origin has created the need to explore alternative models for cancer initiation and maintenance. Recent studies have linked an embryonic stem cell (ESC)-like gene signature with poorly defined high-grade tumors. Here, we review advances in the ESC field with an emphasis on how human pluripotent stem cells (hPSCs) can be used to define early tumorigenic events, including potential miRNA and epigenetic targets, as well as proto-oncogene and tumor suppressor networks that might facilitate hierarchal transformation. These studies allow for investigation of cancer initiation in a manner that cannot be achieved using primary tumors, where only retrospective evaluation of CSC development is possible. By comparing transformed hPSCs with their normal counterparts, we hope to develop novel cell-specific therapies that selectively target CSCs.
Collapse
|
2826
|
Differentiation of embryonic stem cells towards hematopoietic cells: progress and pitfalls. Curr Opin Hematol 2008; 15:312-8. [PMID: 18536568 DOI: 10.1097/moh.0b013e328302f429] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Hematopoietic development from embryonic stem cells has been one of the most productive areas of stem cell biology. Recent studies have progressed from work with mouse to human embryonic stem cells. Strategies to produce defined blood cell populations can be used to better understand normal and abnormal hematopoiesis, as well as potentially improve the generation of hematopoietic cells with therapeutic potential. RECENT FINDINGS Molecular profiling, phenotypic and functional analyses have all been utilized to demonstrate that hematopoietic cells derived from embryonic stem cells most closely represent a stage of hematopoiesis that occurs at embryonic/fetal developmental stages. Generation of hematopoietic stem/progenitor cells comparable to hematopoietic stem cells found in the adult sources, such as bone marrow and cord blood, still remains challenging. However, genetic manipulation of intrinsic factors during hematopoietic differentiation has proven a suitable approach to induce adult definitive hematopoiesis from embryonic stem cells. SUMMARY Concrete evidence has shown that embryonic stem cells provide a powerful approach to study the early stage of hematopoiesis. Multiple hematopoietic lineages can be generated from embryonic stem cells, although most of the evidence suggests that hematopoietic development from embryonic stem cells mimics an embryonic/fetal stage of hematopoiesis.
Collapse
|
2827
|
Yuhki N, Mullikin JC, Beck T, Stephens R, O'Brien SJ. Sequences, annotation and single nucleotide polymorphism of the major histocompatibility complex in the domestic cat. PLoS One 2008; 3:e2674. [PMID: 18629345 PMCID: PMC2453318 DOI: 10.1371/journal.pone.0002674] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2007] [Accepted: 06/12/2008] [Indexed: 11/19/2022] Open
Abstract
Two sequences of major histocompatibility complex (MHC) regions in the domestic cat, 2.976 and 0.362 Mbps, which were separated by an ancient chromosome break (55-80 MYA) and followed by a chromosomal inversion were annotated in detail. Gene annotation of this MHC was completed and identified 183 possible coding regions, 147 human homologues, possible functional genes and 36 pseudo/unidentified genes) by GENSCAN and BLASTN, BLASTP RepeatMasker programs. The first region spans 2.976 Mbp sequence, which encodes six classical class II antigens (three DRA and three DRB antigens) lacking the functional DP, DQ regions, nine antigen processing molecules (DOA/DOB, DMA/DMB, TAPASIN, and LMP2/LMP7,TAP1/TAP2), 52 class III genes, nineteen class I genes/gene fragments (FLAI-A to FLAI-S). Three class I genes (FLAI-H, I-K, I-E) may encode functional classical class I antigens based on deduced amino acid sequence and promoter structure. The second region spans 0.362 Mbp sequence encoding no class I genes and 18 cross-species conserved genes, excluding class I, II and their functionally related/associated genes, namely framework genes, including three olfactory receptor genes. One previously identified feline endogenous retrovirus, a baboon retrovirus derived sequence (ECE1) and two new endogenous retrovirus sequences, similar to brown bat endogenous retrovirus (FERVmlu1, FERVmlu2) were found within a 140 Kbp interval in the middle of class I region. MHC SNPs were examined based on comparisons of this BAC sequence and MHC homozygous 1.9x WGS sequences and found that 11,654 SNPs in 2.84 Mbp (0.00411 SNP per bp), which is 2.4 times higher rate than average heterozygous region in the WGS (0.0017 SNP per bp genome), and slightly higher than the SNP rate observed in human MHC (0.00337 SNP per bp).
Collapse
Affiliation(s)
- Naoya Yuhki
- Laboratory of Genomic Diversity, National Cancer Institute at Frederick, Frederick, Maryland, United States of America.
| | | | | | | | | |
Collapse
|
2828
|
Dejosez M, Krumenacker JS, Zitur LJ, Passeri M, Chu LF, Songyang Z, Thomson JA, Zwaka TP. Ronin is essential for embryogenesis and the pluripotency of mouse embryonic stem cells. Cell 2008; 133:1162-74. [PMID: 18585351 DOI: 10.1016/j.cell.2008.05.047] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2008] [Revised: 04/24/2008] [Accepted: 05/22/2008] [Indexed: 10/21/2022]
Abstract
Pluripotency is a unique biological state that allows cells to differentiate into any tissue type. Here we describe a candidate pluripotency factor, Ronin, that possesses a THAP domain, which is associated with sequence-specific DNA binding and epigenetic silencing of gene expression. Ronin is expressed primarily during the earliest stages of murine embryonic development, and its deficiency in mice produces periimplantational lethality and defects in the inner cell mass. Conditional knockout of Ronin prevents the growth of ES cells while forced expression of Ronin allows ES cells to proliferate without differentiation under conditions that normally do not promote self-renewal. Ectopic expression also partly compensates for the effects of Oct4 knockdown. We demonstrate that Ronin binds directly to HCF-1, a key transcriptional regulator. Our findings identify Ronin as an essential factor underlying embryogenesis and ES cell pluripotency. Its association with HCF-1 suggests an epigenetic mechanism of gene repression in pluripotent cells.
Collapse
Affiliation(s)
- Marion Dejosez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | | | | | | | |
Collapse
|
2829
|
Kwon YJ, Peng CA. Differential interaction of retroviral vector with target cell: quantitative effect of cellular receptor, soluble proteoglycan, and cell type on gene delivery efficiency. Tissue Eng Part A 2008; 14:1497-506. [PMID: 18620488 DOI: 10.1089/ten.tea.2007.0436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Retroviral vectors are powerful tools for gene therapy and stem cell engineering. To improve efficiency of retroviral gene delivery, quantitative understanding of interactions of a retroviral vector and a cell is crucial. Effects of nonspecific adsorption of retrovirus on a cell via proteoglycans and receptor-mediated binding of retrovirus to a cell on overall transduction efficiency were quantified by combining a mathematical model and experimental data. Results represented by transduction rate constant, a lumped parameter of overall transduction efficiency, delineated that chondroitin sulfate C (CSC) plays dual roles as either enhancer or inhibitor of retroviral transduction, depending on its concentrations in the retroviral supernatant. At the concentration of 20 microg/mL, CSC enhanced the transduction efficiency up to threefold but inhibited more than sevenfold at the concentration of 100 microg/mL. Transduction rate constants for amphotropic retroviral infection of NIH 3T3 cells under phosphate-depleted culture condition showed a proportional relationship between cellular receptor density on a cell and transduction efficiency. It was finally shown that amphotropic retrovirus transduced human fibroblast HT1080 cells more efficiently than NIH 3T3 cells. On the contrary, the transduction efficiency of NIH 3T3 cells by vesicular stomatitis virus G protein pseudotyped retroviruses was eightfold higher than that of HT1080 cells. This study implies usefulness of using quantitative analysis of retroviral transduction in understanding and optimizing retroviral gene delivery systems for therapeutic approaches to tissue engineering.
Collapse
Affiliation(s)
- Young Jik Kwon
- Department of Chemical Engineering, University of Southern California, Los Angeles, California 92697, USA.
| | | |
Collapse
|
2830
|
Narazaki G, Uosaki H, Teranishi M, Okita K, Kim B, Matsuoka S, Yamanaka S, Yamashita JK. Directed and systematic differentiation of cardiovascular cells from mouse induced pluripotent stem cells. Circulation 2008; 118:498-506. [PMID: 18625891 DOI: 10.1161/circulationaha.108.769562] [Citation(s) in RCA: 390] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Induced pluripotent stem (iPS) cells are a novel stem cell population induced from mouse and human adult somatic cells through reprogramming by transduction of defined transcription factors. However, detailed differentiation properties and the directional differentiation system of iPS cells have not been demonstrated. METHODS AND RESULTS Previously, we established a novel mouse embryonic stem (ES) cell differentiation system that can reproduce the early differentiation processes of cardiovascular cells. We applied our ES cell system to iPS cells and examined directional differentiation of mouse iPS cells to cardiovascular cells. Flk1 (also designated as vascular endothelial growth factor receptor-2)-expressing mesoderm cells were induced from iPS cells after approximately 4-day culture for differentiation. Purified Flk1(+) cells gave rise to endothelial cells and mural cells by addition of vascular endothelial growth factor and serum. Arterial, venous, and lymphatic endothelial cells were also successfully induced. Self-beating cardiomyocytes could be induced from Flk1(+) cells by culture on OP9 stroma cells. Time course and efficiency of the differentiation were comparable to those of mouse ES cells. Occasionally, reexpression of transgene mRNAs, including c-myc, was observed in long-term differentiation cultures. CONCLUSIONS Various cardiovascular cells can be systematically induced from iPS cells. The differentiation properties of iPS cells are almost completely identical to those of ES cells. This system would greatly contribute to a novel understanding of iPS cell biology and the development of novel cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- Genta Narazaki
- Laboratory of Stem Cell Differentiation, Stem Cell Research Center, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | |
Collapse
|
2831
|
Oct-4 expression maintained cancer stem-like properties in lung cancer-derived CD133-positive cells. PLoS One 2008; 3:e2637. [PMID: 18612434 PMCID: PMC2440807 DOI: 10.1371/journal.pone.0002637] [Citation(s) in RCA: 373] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Accepted: 06/08/2008] [Indexed: 02/06/2023] Open
Abstract
CD133 (prominin-1), a 5-transmembrane glycoprotein, has recently been considered to be an important marker that represents the subset population of cancer stem-like cells. Herein we report the isolation of CD133-positive cells (LC-CD133+) and CD133-negative cells (LC-CD133−) from tissue samples of ten patients with non-small cell lung cancer (LC) and five LC cell lines. LC-CD133+ displayed higher Oct-4 expressions with the ability to self-renew and may represent a reservoir with proliferative potential for generating lung cancer cells. Furthermore, LC-CD133+, unlike LC-CD133−, highly co-expressed the multiple drug-resistant marker ABCG2 and showed significant resistance to chemotherapy agents (i.e., cisplatin, etoposide, doxorubicin, and paclitaxel) and radiotherapy. The treatment of Oct-4 siRNA with lentiviral vector can specifically block the capability of LC-CD133+ to form spheres and can further facilitate LC-CD133+ to differentiate into LC-CD133−. In addition, knock-down of Oct-4 expression in LC-CD133+ can significantly inhibit the abilities of tumor invasion and colony formation, and increase apoptotic activities of caspase 3 and poly (ADP-ribose) polymerase (PARP). Finally, in vitro and in vivo studies further confirm that the treatment effect of chemoradiotherapy for LC-CD133+ can be improved by the treatment of Oct-4 siRNA. In conclusion, we demonstrated that Oct-4 expression plays a crucial role in maintaining the self-renewing, cancer stem-like, and chemoradioresistant properties of LC-CD133+. Future research is warranted regarding the up-regulated expression of Oct-4 in LC-CD133+ and malignant lung cancer.
Collapse
|
2832
|
Wakayama S, Cummins JM, Wakayama T. Nuclear reprogramming to produce cloned mice and embryonic stem cells from somatic cells. Reprod Biomed Online 2008; 16:545-52. [PMID: 18413064 DOI: 10.1016/s1472-6483(10)60462-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cloning methods in mice are now well described and are becoming routine. However, the frequency at which cloned mice are produced remains below 5%, irrespective of the nucleus donor species or cell type. Only a few laboratories have made clones from adult mouse somatic cells and most strains have never produced cloned mice. On the other hand, nuclear transfer can be used to generate human embryonic stem (ntES) cell lines from a patient's own somatic cells. It has been shown that such cells can be generated relatively easily from a variety of mouse genotypes and cell types of both sexes, even though it may be more difficult to generate clones directly. This technique could be used in regenerative medicine and, in theory, in infertility clinics to treat completely infertile individuals. However, these results suggest that the reprogramming integrity of each cloned embryo differs: some cloned embryos can be converted to ntES cells, but these embryos cannot achieve full term development. This review outlines the nature of genomic reprogramming potential and its application, and suggests new approaches to avoid the ethical problems of creating embryos by nuclear transfer.
Collapse
Affiliation(s)
- Sayaka Wakayama
- Centre for Developmental Biology RIKEN Kobe, 2-2-3 Minatojima-minamimachi, Kobe 650-0047, Japan
| | | | | |
Collapse
|
2833
|
Abstract
Hanna et al. (2008) report in a recent issue of Cell that a defined set of transcription factors can reprogram mature B cells back to pluripotent stem cells.
Collapse
Affiliation(s)
- Thomas Graf
- Center for Genomic Regulation, Carrer Dr. Aiguader 88, E-08003 Barcelona, Spain.
| | | |
Collapse
|
2834
|
Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 2008; 133:1106-17. [PMID: 18555785 DOI: 10.1016/j.cell.2008.04.043] [Citation(s) in RCA: 1919] [Impact Index Per Article: 119.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Revised: 03/11/2008] [Accepted: 04/21/2008] [Indexed: 12/18/2022]
Abstract
Transcription factors (TFs) and their specific interactions with targets are crucial for specifying gene-expression programs. To gain insights into the transcriptional regulatory networks in embryonic stem (ES) cells, we use chromatin immunoprecipitation coupled with ultra-high-throughput DNA sequencing (ChIP-seq) to map the locations of 13 sequence-specific TFs (Nanog, Oct4, STAT3, Smad1, Sox2, Zfx, c-Myc, n-Myc, Klf4, Esrrb, Tcfcp2l1, E2f1, and CTCF) and 2 transcription regulators (p300 and Suz12). These factors are known to play different roles in ES-cell biology as components of the LIF and BMP signaling pathways, self-renewal regulators, and key reprogramming factors. Our study provides insights into the integration of the signaling pathways into the ES-cell-specific transcription circuitries. Intriguingly, we find specific genomic regions extensively targeted by different TFs. Collectively, the comprehensive mapping of TF-binding sites identifies important features of the transcriptional regulatory networks that define ES-cell identity.
Collapse
|
2835
|
|
2836
|
Abramovich A, Muradian KK, Fraifeld VE. Have we reached the point for in vivo rejuvenation? Rejuvenation Res 2008; 11:489-92. [PMID: 18442329 DOI: 10.1089/rej.2008.0658] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Stem cells are the major factor ensuring mammalian regeneration. Cell replacement therapy is an attempt to follow this natural process. Another strategy suggests a controlled de-differentiation of somatic cells to a stem-like state with subsequent re-differentiation. Indeed, the cultured mammalian somatic cells may be reprogrammed to a pluripotent state by the induction of a specific set of genes. The next logical step toward the goal of organism rejuvenation is to test the possibility of inducing the pluripotent state in somatic cells in vivo. Such an approach has the potential to improve upon and overcome several obstacles facing today's cell replacement therapy.
Collapse
Affiliation(s)
- Amir Abramovich
- The Shraga Segal Department of Microbiology and Immunology, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | |
Collapse
|
2837
|
Gu P, Reid JG, Gao X, Shaw CA, Creighton C, Tran PL, Zhou X, Drabek RB, Steffen DL, Hoang DM, Weiss MK, Naghavi AO, El-daye J, Khan MF, Legge GB, Wheeler DA, Gibbs RA, Miller JN, Cooney AJ, Gunaratne PH. Novel microRNA candidates and miRNA-mRNA pairs in embryonic stem (ES) cells. PLoS One 2008; 3:e2548. [PMID: 18648548 PMCID: PMC2481296 DOI: 10.1371/journal.pone.0002548] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2007] [Accepted: 05/22/2008] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND MicroRNAS (miRNAS: a class of short non-coding RNAs) are emerging as important agents of post transcriptional gene regulation and integral components of gene networks. MiRNAs have been strongly linked to stem cells, which have a remarkable dual role in development. They can either continuously replenish themselves (self-renewal), or differentiate into cells that execute a limited number of specific actions (pluripotence). METHODOLOGY/PRINCIPAL FINDINGS In order to identify novel miRNAs from narrow windows of development we carried out an in silico search for micro-conserved elements (MCE) in adult tissue progenitor transcript sequences. A plethora of previously unknown miRNA candidates were revealed including 545 small RNAs that are enriched in embryonic stem (ES) cells over adult cells. Approximately 20% of these novel candidates are down-regulated in ES (Dicer(-/-)) ES cells that are impaired in miRNA maturation. The ES-enriched miRNA candidates exhibit distinct and opposite expression trends from mmu-mirs (an abundant class in adult tissues) during retinoic acid (RA)-induced ES cell differentiation. Significant perturbation of trends is found in both miRNAs and novel candidates in ES (GCNF(-/-)) cells, which display loss of repression of pluripotence genes upon differentiation. CONCLUSION/SIGNIFICANCE Combining expression profile information with miRNA target prediction, we identified miRNA-mRNA pairs that correlate with ES cell pluripotence and differentiation. Perturbation of these pairs in the ES (GCNF(-/-)) mutant suggests a role for miRNAs in the core regulatory networks underlying ES cell self-renewal, pluripotence and differentiation.
Collapse
Affiliation(s)
- Peili Gu
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Cancer Genetics, M.D. Anderson Cancer Center, University of Texas , Houston, Texas, United States of America
| | - Jeffrey G. Reid
- Department of Chemistry, University of Houston, Houston, Texas, United States of America
- Department of Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- W. M. Keck Center for Interdisciplinary Bioscience Training, Houston, Texas, United States of America
| | - Xiaolian Gao
- Department of Biology & Biochemistry, University of Houston, Houston, Texas, United States of America
- Department of Chemistry, University of Houston, Houston, Texas, United States of America
| | - Chad A. Shaw
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Chad Creighton
- Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Peter L. Tran
- Department of Biology & Biochemistry, University of Houston, Houston, Texas, United States of America
| | | | - Rafal B. Drabek
- Department of Biology & Biochemistry, University of Houston, Houston, Texas, United States of America
- Department of Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - David L. Steffen
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Bioinformatics Research Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - David M. Hoang
- Department of Biology & Biochemistry, University of Houston, Houston, Texas, United States of America
| | - Michelle K. Weiss
- Department of Biology & Biochemistry, University of Houston, Houston, Texas, United States of America
| | - Arash O. Naghavi
- Department of Biology & Biochemistry, University of Houston, Houston, Texas, United States of America
| | - Jad El-daye
- Department of Biology & Biochemistry, University of Houston, Houston, Texas, United States of America
| | - Mahjabeen F. Khan
- Department of Biology & Biochemistry, University of Houston, Houston, Texas, United States of America
| | - Glen B. Legge
- Department of Biology & Biochemistry, University of Houston, Houston, Texas, United States of America
| | - David A. Wheeler
- Department of Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Richard A. Gibbs
- Department of Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jonathan N. Miller
- Department of Biochemistry & Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Austin J. Cooney
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Preethi H. Gunaratne
- Department of Biology & Biochemistry, University of Houston, Houston, Texas, United States of America
- Department of Pathology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
2838
|
A drug-inducible transgenic system for direct reprogramming of multiple somatic cell types. Nat Biotechnol 2008; 26:916-24. [PMID: 18594521 DOI: 10.1038/nbt1483] [Citation(s) in RCA: 356] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Accepted: 06/25/2008] [Indexed: 12/11/2022]
Abstract
The study of induced pluripotency is complicated by the need for infection with high-titer retroviral vectors, which results in genetically heterogeneous cell populations. We generated genetically homogeneous 'secondary' somatic cells that carry the reprogramming factors as defined doxycycline (dox)-inducible transgenes. These cells were produced by infecting fibroblasts with dox-inducible lentiviruses, reprogramming by dox addition, selecting induced pluripotent stem cells and producing chimeric mice. Cells derived from these chimeras reprogram upon dox exposure without the need for viral infection with efficiencies 25- to 50-fold greater than those observed using direct infection and drug selection for pluripotency marker reactivation. We demonstrate that (i) various induction levels of the reprogramming factors can induce pluripotency, (ii) the duration of transgene activity directly correlates with reprogramming efficiency, (iii) cells from many somatic tissues can be reprogrammed and (iv) different cell types require different induction levels. This system facilitates the characterization of reprogramming and provides a tool for genetic or chemical screens to enhance reprogramming.
Collapse
|
2839
|
Abstract
Pluripotent ES (embryonic stem) cells can be expanded in culture and induced to differentiate into a wide range of cell types. Self-renewal of ES cells involves proliferation with concomitant suppression of differentiation. Some critical and conserved pathways regulating self-renewal in both human and mouse ES cells have been identified, but there is also evidence suggesting significant species differences. Cytoplasmic and receptor tyrosine kinases play important roles in proliferation, survival, self-renewal and differentiation in stem, progenitor and adult cells. The present review focuses on the role of tyrosine kinase signalling for maintenance of the undifferentiated state, proliferation, survival and early differentiation of ES cells.
Collapse
Affiliation(s)
- Cecilia Annerén
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, 751 23 Uppsala, Sweden
| |
Collapse
|
2840
|
Evans PM, Liu C. Roles of Krüpel-like factor 4 in normal homeostasis, cancer and stem cells. Acta Biochim Biophys Sin (Shanghai) 2008; 40:554-64. [PMID: 18604447 DOI: 10.1111/j.1745-7270.2008.00439.x] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Krüpel-like factor 4 (KLF4) is a zinc finger-type transcription factor expressed in a variety of tissues, including the epithelium of the intestine and the skin, and it plays an important role in differentiation and cell cycle arrest. Depending on the gene targeted, KLF4 can both activate and repress transcription. Moreover, in certain cellular contexts, KLF4 can function as a tumor suppressor or an oncogene. Finally, KLF4 is important in reprogramming differentiated fibroblasts into inducible pluripotent stem cells, which highly resemble embryonic stem cells. This review summarizes what is known about the diverse functions of KLF4 as well as their molecular mechanisms.
Collapse
Affiliation(s)
- Paul M Evans
- Department of Biochemistry and Molecular Biology, Sealy Center for Cancer Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555-1448, USA
| | | |
Collapse
|
2841
|
Huangfu D, Maehr R, Guo W, Eijkelenboom A, Snitow M, Chen AE, Melton DA. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat Biotechnol 2008; 26:795-7. [PMID: 18568017 PMCID: PMC6334647 DOI: 10.1038/nbt1418] [Citation(s) in RCA: 1183] [Impact Index Per Article: 73.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2008] [Accepted: 06/05/2008] [Indexed: 12/16/2022]
Abstract
Reprogramming of mouse and human somatic cells can be achieved by ectopic expression of transcription factors, but with low efficiencies. We report that DNA methyltransferase and histone deacetylase (HDAC) inhibitors improve reprogramming efficiency. In particular, valproic acid (VPA), an HDAC inhibitor, improves reprogramming efficiency by more than 100-fold, using Oct4-GFP as a reporter. VPA also enables efficient induction of pluripotent stem cells without introduction of the oncogene c-Myc.
Collapse
Affiliation(s)
- Danwei Huangfu
- Department of Stem Cell and Regenerative Biology, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, Massachusetts 02138, USA
| | | | | | | | | | | | | |
Collapse
|
2842
|
Kastenberg ZJ, Odorico JS. Alternative sources of pluripotency: science, ethics, and stem cells. Transplant Rev (Orlando) 2008; 22:215-22. [PMID: 18631882 DOI: 10.1016/j.trre.2008.04.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Zachary J Kastenberg
- Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health and WiCell Research Institute, Madison, WI 53792, USA
| | | |
Collapse
|
2843
|
Alder JK, Georgantas RW, Hildreth RL, Kaplan IM, Morisot S, Yu X, McDevitt M, Civin CI. Kruppel-like factor 4 is essential for inflammatory monocyte differentiation in vivo. THE JOURNAL OF IMMUNOLOGY 2008; 180:5645-52. [PMID: 18390749 DOI: 10.4049/jimmunol.180.8.5645] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Several members of the Kruppel-like factor (KLF) family of transcription factors play important roles in differentiation, survival, and trafficking of blood and immune cell types. We demonstrate in this study that hematopoietic cells from KLF4(-/-) fetal livers (FL) contained normal numbers of functional hematopoietic progenitor cells, were radioprotective, and performed as well as KLF4(+/+) cells in competitive repopulation assays. However, hematopoietic "KLF4(-/-) chimeras" generated by transplantation of KLF4(-/-) fetal livers cells into lethally irradiated wild-type mice completely lacked circulating inflammatory (CD115(+)Gr1(+)) monocytes, and had reduced numbers of resident (CD115(+)Gr1(-)) monocytes. Although the numbers and function of peritoneal macrophages were normal in KLF4(-/-) chimeras, bone marrow monocytic cells from KLF4(-/-) chimeras expressed lower levels of key trafficking molecules and were more apoptotic. Thus, our in vivo loss-of-function studies demonstrate that KLF4, previously shown to mediate proinflammatory signaling in human macrophages in vitro, is essential for differentiation of mouse inflammatory monocytes, and is involved in the differentiation of resident monocytes. In addition, inducible expression of KLF4 in the HL60 human acute myeloid leukemia cell line stimulated monocytic differentiation and enhanced 12-O-tetradecanoylphorbol 13-acetate induced macrophage differentiation, but blocked all-trans-retinoic acid induced granulocytic differentiation of HL60 cells. The inflammation-selective effects of loss-of-KLF4 and the gain-of-KLF4-induced monocytic differentiation in HL60 cells identify KLF4 as a key regulator of monocytic differentiation and a potential target for translational immune modulation.
Collapse
Affiliation(s)
- Jonathan K Alder
- Divisions of Immunology and Hematopoiesis, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | | | | | | | | | | | | | | |
Collapse
|
2844
|
Pluripotent stem cells induced from adult neural stem cells by reprogramming with two factors. Nature 2008; 454:646-50. [PMID: 18594515 DOI: 10.1038/nature07061] [Citation(s) in RCA: 766] [Impact Index Per Article: 47.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2008] [Accepted: 05/08/2008] [Indexed: 02/08/2023]
Abstract
Reprogramming of somatic cells is a valuable tool to understand the mechanisms of regaining pluripotency and further opens up the possibility of generating patient-specific pluripotent stem cells. Reprogramming of mouse and human somatic cells into pluripotent stem cells, designated as induced pluripotent stem (iPS) cells, has been possible with the expression of the transcription factor quartet Oct4 (also known as Pou5f1), Sox2, c-Myc and Klf4 (refs 1-11). Considering that ectopic expression of c-Myc causes tumorigenicity in offspring and that retroviruses themselves can cause insertional mutagenesis, the generation of iPS cells with a minimal number of factors may hasten the clinical application of this approach. Here we show that adult mouse neural stem cells express higher endogenous levels of Sox2 and c-Myc than embryonic stem cells, and that exogenous Oct4 together with either Klf4 or c-Myc is sufficient to generate iPS cells from neural stem cells. These two-factor iPS cells are similar to embryonic stem cells at the molecular level, contribute to development of the germ line, and form chimaeras. We propose that, in inducing pluripotency, the number of reprogramming factors can be reduced when using somatic cells that endogenously express appropriate levels of complementing factors.
Collapse
|
2845
|
Kato K, Hasui K, Wang J, Kawano Y, Aikou T, Murata F. Homeostatic mass control in gastric non-neoplastic epithelia under infection of Helicobacter pylori: an immunohistochemical analysis of cell growth, stem cells and programmed cell death. Acta Histochem Cytochem 2008; 41:23-38. [PMID: 18636110 PMCID: PMC2447913 DOI: 10.1267/ahc.07021] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2007] [Accepted: 03/04/2008] [Indexed: 12/13/2022] Open
Abstract
We evaluated homeostatic mass control in non-neoplastic gastric epithelia under Helicobacter pylori (HP) infection in the macroscopically normal-appearing mucosa resected from the stomach with gastric cancer, immunohistochemically analyzing the proliferation, kinetics of stem cells and programmed cell death occurring in them. Ki67 antigen-positive proliferating cells were found dominantly in the elongated neck portion, sparsely in the fundic areas and sporadically in the stroma with chronic infiltrates. CD117 could monitor the kinetics of gastric stem cells and showed its expression in two stages of gastric epithelial differentiation, namely, in transient cells from the gastric epithelial stem cells to the foveolar and glandular cells in the neck portion and in what are apparently progenitor cells from the gastric stem cells in the stroma among the infiltrates. Most of the nuclei were positive for ssDNA in the almost normal mucosa, suggesting DNA damage. Cleaved caspase-3-positive foveolar cells were noted under the surface, suggesting the suppression of apoptosis in the surface foveolar cells. Besides such apoptosis of the foveolar cells, in the severely inflamed mucosa apoptotic cells were found in the neck portion where most of the cells were Ki67 antigen-positive proliferating cells. Beclin-1 was recognized in the cytoplasm and in a few nuclei of the fundic glandular cells, suggesting their autophagic cell death and mutated beclin-1 in the nuclei. Taken together, the direct and indirect effects of HP infection on the gastric epithelial proliferation, differentiation and programmed cell death suggested the in-situ occurrence of gastric cancer under HP infection.
Collapse
Affiliation(s)
- Kenji Kato
- Department of Surgical Oncology and Digestive Surgery, Field of Oncology, Course of Advanced Therapeutics, Kagoshima University Graduate School of Medical and Dental Sciences
| | - Kazuhisa Hasui
- Department of Immunology (Previous Second Department of Anatomy), Field of Infection and Immunity, Course of Health Research, Kagoshima University Graduate School of Medical and Dental Sciences
- Division of Persistent & Oncogenic Viruses, Center for Chronic Viral Diseases, Field of Infection and Immunity, Course of Health Research, Kagoshima University Graduate School of Medical and Dental Sciences
| | - Jia Wang
- Department of Immunology (Previous Second Department of Anatomy), Field of Infection and Immunity, Course of Health Research, Kagoshima University Graduate School of Medical and Dental Sciences
- Division of Persistent & Oncogenic Viruses, Center for Chronic Viral Diseases, Field of Infection and Immunity, Course of Health Research, Kagoshima University Graduate School of Medical and Dental Sciences
| | - Yoshifumi Kawano
- Department of Pediatrics, Field of Developmental Medicine, Course of Health Research, Kagoshima University Graduate School of Medical and Dental Sciences
| | - Takashi Aikou
- Department of Surgical Oncology and Digestive Surgery, Field of Oncology, Course of Advanced Therapeutics, Kagoshima University Graduate School of Medical and Dental Sciences
| | - Fusayoshi Murata
- Department of Structural Cell Biology (Previous Second Department of Anatomy), Course of Advanced Therapeutics, Kagoshima University Graduate School of Medical and Dental Sciences
- honorary emeritus Professor of Kagoshima University, vice-President of Kagoshima Medical Technology College
| |
Collapse
|
2846
|
Moore H, Udayashankar R, Aflatoonian B. Stem cells for reproductive medicine. Mol Cell Endocrinol 2008; 288:104-10. [PMID: 18406514 DOI: 10.1016/j.mce.2008.02.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 02/19/2008] [Accepted: 02/25/2008] [Indexed: 10/22/2022]
Abstract
The generation of various pluripotent stem cell lines provides a new route to investigate developmental process of germ cell and embryo development, which until now was difficult to access in the human. In the future these cells may be used for new therapies in reproductive medicine. This brief review outlines the development of germ cells and their pluripotent capabilities, how embryonic and germline stem cells can mimic developmental processes in vitro and generate gamete and trophoblast phenotypes for research and potential treatments.
Collapse
Affiliation(s)
- Harry Moore
- Centre for Stem Cell Biology, University of Sheffield, Sheffield S10 2UH, UK.
| | | | | |
Collapse
|
2847
|
Best M, Carroll M, Hanley NA, Piper Hanley K. Embryonic stem cells to beta-cells by understanding pancreas development. Mol Cell Endocrinol 2008; 288:86-94. [PMID: 18487011 DOI: 10.1016/j.mce.2008.03.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2007] [Revised: 03/18/2008] [Accepted: 03/18/2008] [Indexed: 12/25/2022]
Abstract
Insulin injections treat but do not cure Type 1 diabetes (T1DM). The success of islet transplantation suggests cell replacement therapies may offer a curative strategy. However, cadaver islets are of insufficient number for this to become a widespread treatment. To address this deficiency, the production of beta-cells from pluripotent stem cells offers an ambitious far-sighted opportunity. Recent progress in generating insulin-producing cells from embryonic stem cells has shown promise, highlighting the potential of trying to mimic normal developmental pathways. Here, we provide an overview of the current methodology that has been used to differentiate stem cells toward a beta-cell fate. Parallels are drawn with what is known about normal development, especially regarding the human pancreas.
Collapse
Affiliation(s)
- Marie Best
- Centre for Human Development, Stem Cells & Regeneration, UK
| | | | | | | |
Collapse
|
2848
|
Abstract
An improved understanding of stem-cell and regenerative biology, as well as a better control of stem-cell fate, is likely to produce treatments for many devastating diseases and injuries. Chemical approaches are starting to have an increasingly important role in this young field. Attention has focused on chemical approaches that allow the precise manipulation of cells in vitro to obtain homogeneous cell types for cell-based therapies. Another promising approach is the development of conventional chemical and biological therapeutics to stimulate endogenous cells to regenerate. Such therapeutics can act on target cells or their niches in vivo to promote cell survival, proliferation, differentiation, reprogramming and homing.
Collapse
Affiliation(s)
- Yue Xu
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | | | | |
Collapse
|
2849
|
Abstract
There is much interest in using embryonic stem cells to regenerate tissues and organs. For this approach to succeed, these stem cells or their derivatives must engraft in patients over the long term. Unless a cell transplant is derived from the patient's own cells, however, the cells will be targeted for rejection by the immune system. Although standard methods for suppressing the immune system achieve some success, rejection of the transplant is inevitable. Emerging approaches to address this issue include 're-educating' the immune system to induce tolerance to foreign cells and reducing the immune targeting of the transplant by administering 'self stem cells' instead of foreign cells, but each of these approaches has associated challenges.
Collapse
|
2850
|
Abstract
Derivation of human embryonic stem (ES) cells from preimplantation embryos ten years ago raised great hopes that they may be an excellent source of cells for cell replacement therapy. However, serious ethical concerns and the risk of immune rejection of allotransplanted cells have hindered the translation of ES cell-based therapies into the clinic. In an attempt to circumvent these barriers, a number of methods have been developed for converting adult somatic cells into a pluripotent state from which ethically acceptable patient-specific mature cells of interest could be derived. These efforts, backed by advances in elucidating the molecular basis of pluripotency, have culminated in successful reprogramming of fibroblasts into ES cell-like cells, termed induced pluripotent stem (iPS) cells, by ectopic expression of only a handful of "stemness" factors. iPS cells possess morphological, molecular and developmental features of conventional blastocyst-derived ES cells and have the potential to serve as a source of therapeutic cells for customized tissue repair, gene therapy, drug discovery, toxicological testing and for studying the molecular basis of human disease. The goal of this review is to provide the current state-of-the-art in this very exciting and dynamic field and to discuss barriers that remain to be removed before the therapeutic potential of iPS cells can be fully realized.
Collapse
Affiliation(s)
- Tomo Saric
- Medical Center, Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | | |
Collapse
|