201
|
Sofikitis N, Pappas E, Kawatani A, Baltogiannis D, Loutradis D, Kanakas N, Giannakis D, Dimitriadis F, Tsoukanelis K, Georgiou I, Makrydimas G, Mio Y, Tarlatzis V, Melekos M, Miyagawa I. Efforts to create an artificial testis: culture systems of male germ cells under biochemical conditions resembling the seminiferous tubular biochemical environment. Hum Reprod Update 2005; 11:229-59. [PMID: 15817525 DOI: 10.1093/humupd/dmi007] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Induction of meiotic and post-meiotic alterations of male germ cells in vitro has been the target of several research efforts since 1960. However, to date, the establishment of an ideal culture system in which spermatogonial stem cells can be maintained and directed to proliferate and undergo meiosis and complete spermiogenesis does not exist. This is attributed to the difficulties concerning the isolation and purification of defined subpopulations of germ cells and the establishment of male germ cell lines. In addition, there is no adequate knowledge regarding the optimal biochemical conditions that promote the survival and differentiation of germ cells in long-term cultures. This review focuses on the methodologies that have been proved sufficient to achieve differentiation of cultured male germ cells. Furthermore, the factors regulating spermatogenesis and the technical prerequisites to achieve differentiation of cultured male germ cells are described. Finally, the role of in vitro cultures of immature diploid germ cells in the therapeutic management of men negative for haploid cells in their testes and the subsequent potential genetic and epigenetic risks are discussed.
Collapse
Affiliation(s)
- N Sofikitis
- Laboratory for Molecular Urology and Genetics of Human Reproduction, Department of Urology, Ioannina University School of Medicine, Ioannina, Greece.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
202
|
Kehler J, Tolkunova E, Koschorz B, Pesce M, Gentile L, Boiani M, Lomelí H, Nagy A, McLaughlin KJ, Schöler HR, Tomilin A. Oct4 is required for primordial germ cell survival. EMBO Rep 2005; 5:1078-83. [PMID: 15486564 PMCID: PMC1299174 DOI: 10.1038/sj.embor.7400279] [Citation(s) in RCA: 421] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2004] [Revised: 09/20/2004] [Accepted: 09/21/2004] [Indexed: 11/09/2022] Open
Abstract
Previous studies have shown that Oct4 has an essential role in maintaining pluripotency of cells of the inner cell mass (ICM) and embryonic stem cells. However, Oct4 null homozygous embryos die around the time of implantation, thus precluding further analysis of gene function during development. We have used the conditional Cre/loxP gene targeting strategy to assess Oct4 function in primordial germ cells (PGCs). Loss of Oct4 function leads to apoptosis of PGCs rather than to differentiation into a trophectodermal lineage, as has been described for Oct4-deficient ICM cells. These new results suggest a previously unknown function of Oct4 in maintaining viability of mammalian germline.
Collapse
Affiliation(s)
- James Kehler
- Germline Development, Center for Animal Transgenesis and Germ Cell Research, New Bolton Center, University of Pennsylvania, 382 W. Street Road, Kennett Square, Pennsylvania 19348, USA
| | - Elena Tolkunova
- Department of Developmental Biology, Max Planck Institute of Immunobiology, Stübeweg 51, 79108 Freiburg, Germany
| | - Birgit Koschorz
- Department of Developmental Biology, Max Planck Institute of Immunobiology, Stübeweg 51, 79108 Freiburg, Germany
| | - Maurizio Pesce
- Laboratorio di Biologia Vascolare e Terapia Genica, Centro Cardiologico, Fondazione I Monzino, Parea 4, I-20138, Milan, Italy
| | - Luca Gentile
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Mendelstrasse 7, Münster 48149, Germany
| | - Michele Boiani
- Germline Development, Center for Animal Transgenesis and Germ Cell Research, New Bolton Center, University of Pennsylvania, 382 W. Street Road, Kennett Square, Pennsylvania 19348, USA
- Present address: Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Mendelstrasse 7, Münster 48149, Germany
| | - Hilda Lomelí
- Departamento de Genética y Fisiologia Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de Mexico, A.P. 510-3, Cuernavaca, Morelos 62271, Mexico
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, 381–600 University Avenue, Toronto, Ontario, Canada M5G 1X5
| | - Andras Nagy
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, 381–600 University Avenue, Toronto, Ontario, Canada M5G 1X5
| | - K John McLaughlin
- Developmental Epigenetics Group, Center for Animal Transgenesis and Germ Cell Research, New Bolton Center, University of Pennsylvania, W. Street Road, Kennett Square, Pennsylvania 19348, USA
| | - Hans R Schöler
- Germline Development, Center for Animal Transgenesis and Germ Cell Research, New Bolton Center, University of Pennsylvania, 382 W. Street Road, Kennett Square, Pennsylvania 19348, USA
- Present address: Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Mendelstrasse 7, Münster 48149, Germany
- Tel: +49 251 980 2866; Fax: +49 251 980 2894; E-mail:
| | - Alexey Tomilin
- Germline Development, Center for Animal Transgenesis and Germ Cell Research, New Bolton Center, University of Pennsylvania, 382 W. Street Road, Kennett Square, Pennsylvania 19348, USA
- Department of Developmental Biology, Max Planck Institute of Immunobiology, Stübeweg 51, 79108 Freiburg, Germany
| |
Collapse
|
203
|
Abstract
The restricted potential of a differentiated cell can be reverted back to a pluripotent state by cell fusion; totipotency can even be regained after somatic cell nuclear transfer. To identify factors involved in resetting the genetic program of a differentiated cell, we fused embryonic stem cells (ESCs) with neurosphere cells (NSCs). The fusion activated Oct4, a gene essential for pluripotency, in NSCs. To further identify whether cytoplasmic or nuclear factors are responsible for its reactivation, we fused either karyoplasts or cytoplasts of ESCs with NSCs. Our results show that ESC karyoplasts could induce Oct4 expression in the somatic genome, but cytoplasts lacked this ability. In addition, mitomycin C-treated ESCs, although incapable of DNA replication and cell division, could reprogram 5-azacytidine-treated NSCs. We therefore conclude that the Oct4 reprogramming capacity resides in the ESC karyoplast and that gene reactivation is independent of DNA replication and cell division.
Collapse
Affiliation(s)
- Jeong Tae Do
- Center for Animal Transgenesis and Germ Cell Research, School of Veterinary Medicine, University of Pennsylvania, USA
| | | |
Collapse
|
204
|
Webster KE, O'Bryan MK, Fletcher S, Crewther PE, Aapola U, Craig J, Harrison DK, Aung H, Phutikanit N, Lyle R, Meachem SJ, Antonarakis SE, de Kretser DM, Hedger MP, Peterson P, Carroll BJ, Scott HS. Meiotic and epigenetic defects in Dnmt3L-knockout mouse spermatogenesis. Proc Natl Acad Sci U S A 2005; 102:4068-73. [PMID: 15753313 PMCID: PMC552976 DOI: 10.1073/pnas.0500702102] [Citation(s) in RCA: 207] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The production of mature germ cells capable of generating totipotent zygotes is a highly specialized and sexually dimorphic process. The transition from diploid primordial germ cell to haploid spermatozoa requires genome-wide reprogramming of DNA methylation, stage- and testis-specific gene expression, mitotic and meiotic division, and the histone-protamine transition, all requiring unique epigenetic control. Dnmt3L, a DNA methyltransferase regulator, is expressed during gametogenesis, and its deletion results in sterility. We found that during spermatogenesis, Dnmt3L contributes to the acquisition of DNA methylation at paternally imprinted regions, unique nonpericentric heterochromatic sequences, and interspersed repeats, including autonomous transposable elements. We observed retrotransposition of an LTR-ERV1 element in the DNA from Dnmt3L-/- germ cells, presumably as a result of hypomethylation. Later in development, in Dnmt3L-/- meiotic spermatocytes, we detected abnormalities in the status of biochemical markers of heterochromatin, implying aberrant chromatin packaging. Coincidentally, homologous chromosomes fail to align and form synaptonemal complexes, spermatogenesis arrests, and spermatocytes are lost by apoptosis and sloughing. Because Dnmt3L expression is restricted to gonocytes, the presence of defects in later stages reveals a mechanism whereby early genome reprogramming is linked inextricably to changes in chromatin structure required for completion of spermatogenesis.
Collapse
Affiliation(s)
- Kylie E Webster
- Genetics and Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
205
|
Isotani A, Nakanishi T, Kobayashi S, Lee J, Chuma S, Nakatsuji N, Ishino F, Okabe M. Genomic imprinting of XX spermatogonia and XX oocytes recovered from XX<-->XY chimeric testes. Proc Natl Acad Sci U S A 2005; 102:4039-44. [PMID: 15746241 PMCID: PMC554793 DOI: 10.1073/pnas.0406769102] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We produced XX<-->XY chimeras by using embryos whose X chromosomes were tagged with EGFP (X*), making the fluorescent green female (XX*) germ cells easily distinguishable from their nonfluorescent male (XY) counterparts. Taking advantage of tagging with EGFP, the XX* "prospermatogonia" were isolated from the testes, and the status of their genomic imprinting was examined. It was shown that these XX cells underwent a paternal imprinting, despite their chromosomal constitution. As previously indicated in sex-reversal XXsxr testes, we also found a few green XX* germ cells developed as "eggs" within the seminiferous tubules of XX*<-->XY chimeric testes. These cells were indistinguishable from XX* prospermatogonia at birth but resumed oogenesis in a testicular environment. The biological nature of the "testicular eggs" was examined by recovering the eggs from chimeric testes. The testicular eggs not only formed an egg-specific structure, the zona pellucida, but also were able to fuse with sperm. The collected testicular eggs were indicated to undergo maternal imprinting, despite the testicular environment. The genomic imprinting did not always follow the environmental conditions of where the germ cells resided; rather, it was defined by the sex that was chosen by the germ cells at early embryonic stage.
Collapse
Affiliation(s)
- Ayako Isotani
- Genome Information Research Center and Graduate School of Pharmaceutical Sciences, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
206
|
Abstract
The germ-cell tumours are a fascinating group of neoplasms because of their unusual biology and the spectacular therapeutic results that have been obtained in these tumours. Traditionally, this group of neoplasms is presented in an organ-oriented approach. However, recent clinical and experimental data convincingly demonstrate that these neoplasms are one disease with separate entities that can manifest themselves in different anatomical sites. We propose five entities, in which the developmental potential is determined by the maturation stage and imprinting status of the originating germ cell. Recent progress begins to explain the apparent unpredictable development of germ-cell tumours and offers a basis for understanding their exquisite sensitivity to therapy.
Collapse
Affiliation(s)
- J Wolter Oosterhuis
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Daniel den Hoed Cancer Center, Josephine Nefkens Institute, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands.
| | | |
Collapse
|
207
|
Ranganath RM. Harnessing the developmental potential of nucellar cells: barriers and opportunities. Trends Biotechnol 2005; 22:504-10. [PMID: 15450743 DOI: 10.1016/j.tibtech.2004.08.010] [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: 10/26/2022]
Abstract
Angiosperm nucellar cells can either use or avoid meiosis in vivo, depending on the developmental context. This unique ability contrasts with the conditions required in vitro, either for a reconstituted oocyte to avoid meiosis and produce clones by somatic cell nuclear transfer (SCNT), or for mammalian stem cells to undergo meiosis and produce synthetic sex cells (gametes). Current biotechnological initiatives to harness the potential of nucellar cells are based on the transfer of apomixis genes to sexual crop plants with the aim of producing clones through seeds. The elusive genetic basis of apomixis compels us to examine whether this process involves epigenetic factors. The elegant and versatile developmental platform available in nucellar cells should be explored as a genome-scale science and compared with mammalian stem cell biology for a holistic understanding of developmental programming and reprogramming in eukaryotes.
Collapse
Affiliation(s)
- R M Ranganath
- Cytogenetics and Developmental Biology Laboratory, Department of Botany, Bangalore University, Jnanabharathi Campus, Bangalore 560056, India.
| |
Collapse
|
208
|
Kanatsu-Shinohara M, Inoue K, Lee J, Yoshimoto M, Ogonuki N, Miki H, Baba S, Kato T, Kazuki Y, Toyokuni S, Toyoshima M, Niwa O, Oshimura M, Heike T, Nakahata T, Ishino F, Ogura A, Shinohara T. Generation of pluripotent stem cells from neonatal mouse testis. Cell 2005; 119:1001-12. [PMID: 15620358 DOI: 10.1016/j.cell.2004.11.011] [Citation(s) in RCA: 564] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2004] [Revised: 10/07/2004] [Accepted: 11/02/2004] [Indexed: 12/12/2022]
Abstract
Although germline cells can form multipotential embryonic stem (ES)/embryonic germ (EG) cells, these cells can be derived only from embryonic tissues, and such multipotent cells have not been available from neonatal gonads. Here we report the successful establishment of ES-like cells from neonatal mouse testis. These ES-like cells were phenotypically similar to ES/EG cells except in their genomic imprinting pattern. They differentiated into various types of somatic cells in vitro under conditions used to induce the differentiation of ES cells and produced teratomas after inoculation into mice. Furthermore, these ES-like cells formed germline chimeras when injected into blastocysts. Thus, the capacity to form multipotent cells persists in neonatal testis. The ability to derive multipotential stem cells from the neonatal testis has important implications for germ cell biology and opens the possibility of using these cells for biotechnology and medicine.
Collapse
Affiliation(s)
- Mito Kanatsu-Shinohara
- Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
209
|
Xu Y, Zhang JJ, Grifo JA, Krey LC. DNA methylation patterns in human tripronucleate zygotes. Mol Hum Reprod 2005; 11:167-71. [PMID: 15695773 DOI: 10.1093/molehr/gah145] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
In mammals, the dynamic reprogramming of DNA methylation begins during gametogenesis and continues through embryogenesis. Recently, immunofluorescence staining with an antibody against 5-methylcytosine (anti-5-MeC) has revealed active demethylation of the male pronucleus in zygotes beginning at 4-6 h after fertilization. In this study, we characterized the DNA methylation patterns in mouse zygotes and in human tripronucleate (3 PN) zygotes discarded after conventional fertilization or following ICSI. Pronuclei were subjected to fluorescence in-situ hybridization to identify the X and/or Y chromosomes and then stained with anti-5-MeC. In diandric 3 PN zygotes from conventional IVF, we consistently observed one strongly and two weakly stained pronuclei. In contrast, the majority of 3 PN ICSI zygotes, mainly digynic zygotes, displayed two strongly and one weakly stained pronuclei. Two zygotes from ICSI failed to show any staining difference among the three pronuclei. Our results indicate that the active demethylation of male pronuclei occurs in both mouse and human zygotes. It is possible that the abnormal methylation patterns resulting from a dysfunctional cytoplasm may occur in a small number of oocytes and may affect embryonic viability.
Collapse
MESH Headings
- Animals
- Cell Nucleus/genetics
- Cell Nucleus/metabolism
- Cell Nucleus/ultrastructure
- Chromosomes, Human, X/chemistry
- Chromosomes, Human, X/genetics
- Chromosomes, Human, X/metabolism
- Chromosomes, Human, Y/chemistry
- Chromosomes, Human, Y/genetics
- Chromosomes, Human, Y/metabolism
- DNA/analysis
- DNA/genetics
- DNA/metabolism
- DNA Methylation
- Embryo, Mammalian/cytology
- Embryo, Mammalian/metabolism
- Female
- Fertilization/genetics
- Humans
- In Situ Hybridization, Fluorescence
- Male
- Mice
- Mice, Inbred Strains
- Sperm Injections, Intracytoplasmic
- Zygote/chemistry
- Zygote/cytology
- Zygote/metabolism
Collapse
Affiliation(s)
- Yanwen Xu
- Program for In Vitro Fertilization, Reproductive Surgery and Infertility, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | |
Collapse
|
210
|
Irmak MK, Topal T, Oter S. Melatonin seems to be a mediator that transfers the environmental stimuli to oocytes for inheritance of adaptive changes through epigenetic inheritance system. Med Hypotheses 2005; 64:1138-43. [PMID: 15823703 DOI: 10.1016/j.mehy.2004.12.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2004] [Accepted: 12/21/2004] [Indexed: 10/25/2022]
Abstract
Possibility of inheritance of epigenetic modifications have led us to consider that adaptive geographic variations in humans may result from interactions between environmental factors and epigenetic inheritance system. In this system melatonin seems to be a mediator that transfers the environmental stimuli to germ cells (oocytes). While environmental factors produce modifications in the body, they simultaneously induce epigenetic modifications in the oocytes with the help of melatonin, and these changes are inherited to offspring. In this way, adaptive changes could be passed on to the next generation. This kind of heritable long-term changes is generally labeled biological adaptation. But, how can melatonin cause epigenetic changes in oocytes? We suggest that melatonin induces epigenetic modifications by affecting the nuclear melatonin receptors that can in turn change the superstructure of DNA. It was previously suggested that biological adaptation is limited to neural crest derivatives such as, craniofacial tissues, melanocytes, and structures related to stature, hair form and body proportions. Thus, inheritance of adaptive changes is possible only where environmental factors affect the neural crest derivatives, including the cells that produce the next generation.
Collapse
MESH Headings
- Acetylcholine/physiology
- Adaptation, Physiological/genetics
- Altitude
- Cell Lineage
- Cell Nucleus/metabolism
- DNA Methylation
- DNA-Binding Proteins/physiology
- Environmental Exposure
- Epigenesis, Genetic/genetics
- Epigenesis, Genetic/physiology
- Female
- Germ-Line Mutation
- Humans
- Light
- Melatonin/metabolism
- Melatonin/physiology
- Models, Biological
- Models, Genetic
- Neural Crest/cytology
- Norepinephrine/physiology
- Nuclear Receptor Subfamily 6, Group A, Member 1
- Oocytes/physiology
- Ovarian Follicle/chemistry
- Pineal Gland/metabolism
- Receptors, Cytoplasmic and Nuclear
- Receptors, Melatonin/physiology
- Receptors, Retinoic Acid/physiology
- Retina/radiation effects
- Seasons
- Secretory Rate/drug effects
- Secretory Rate/radiation effects
- Skin Pigmentation
- Temperature
Collapse
Affiliation(s)
- M K Irmak
- Department of Histology and Embryology, School of Medicine, Gulhane Military Medical Academy, 06018-Etlik Ankara, Turkey.
| | | | | |
Collapse
|
211
|
Kimura T, Murayama K, Nakamura T, Watanabe S, Umehara H, Tomooka M, Nakano T. Testicular teratomas: back to pluripotent stem cells. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2005; 40:133-50. [PMID: 17153483 DOI: 10.1007/3-540-27671-8_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Affiliation(s)
- Tohru Kimura
- Department of Pathology, Osaka University Medical School, Japan
| | | | | | | | | | | | | |
Collapse
|
212
|
Affiliation(s)
- Shyh-Jou Shieh
- Division of Plastic and Reconstructive Surgery, Department of Surgery, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | |
Collapse
|
213
|
Chuma S, Kanatsu-Shinohara M, Inoue K, Ogonuki N, Miki H, Toyokuni S, Hosokawa M, Nakatsuji N, Ogura A, Shinohara T. Spermatogenesis from epiblast and primordial germ cells following transplantation into postnatal mouse testis. Development 2005; 132:117-22. [PMID: 15576408 DOI: 10.1242/dev.01555] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Primordial germ cells (PGCs) are derived from a population of pluripotent epiblast cells in mice. However, little is known about when and how PGCs acquire the capacity to differentiate into functional germ cells, while keeping the potential to derive pluripotent embryonic germ cells and teratocarcinomas. In this investigation, we show that epiblast cells and PGCs can establish colonies of spermatogenesis after transfer into postnatal seminiferous tubules of surrogate infertile mice. Furthermore, we obtained normal fertile offspring by microinsemination using spermatozoa or spermatids derived from PGCs harvested from fetuses as early as 8.5 days post coitum. Thus, fetal male germ cell development is remarkably flexible, and the maturation process, from epiblast cells through PGCs to postnatal spermatogonia, can occur in the postnatal testicular environment. Primordial germ cell transplantation techniques will also provide a novel tool to assess the developmental potential of PGCs, such as those manipulated in vitro or recovered from embryos harboring lethal mutations.
Collapse
Affiliation(s)
- Shinichiro Chuma
- Department of Development and Differentiation, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
214
|
Abstract
Currently, behavioral development is thought to result from the interplay among genetic inheritance, congenital characteristics, cultural contexts, and parental practices as they directly impact the individual. Evolutionary ecology points to another contributor, epigenetic inheritance, the transmission to offspring of parental phenotypic responses to environmental challenges-even when the young do not experience the challenges themselves. Genetic inheritance is not altered, gene expression is. Organismic pathways for such transmission exist. Maternal stress during the latter half of a daughter's gestation may affect not only the daughter's but also grand-offspring's physical growth. The author argues that temperamental variation may be influenced in the same way. Implications for theory and research design are presented along with testable predictions.
Collapse
Affiliation(s)
- Lawrence V Harper
- Department of Human and Community Development, University of California, Davis, CA 95616, USA.
| |
Collapse
|
215
|
Dindot SV, Kent KC, Evers B, Loskutoff N, Womack J, Piedrahita JA. Conservation of genomic imprinting at the XIST, IGF2, and GTL2 loci in the bovine. Mamm Genome 2004; 15:966-74. [PMID: 15599555 DOI: 10.1007/s00335-004-2407-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2004] [Accepted: 08/20/2004] [Indexed: 10/24/2022]
Abstract
Genomic imprinting is theorized to exist in all placental mammals and some marsupials; however, extensive comparative analysis of animals aside from humans and mice remains incomplete. Here we report conservation of genomic imprinting in the bovine at the X chromosome inactivation-specific transcript (XIST), insulin-like growth factor 2 (IGF2), and gene trap locus 2 (GTL2) loci. Coding single nucleotide polymorphisms (SNPs) between Bos gaurus and Bos taurus were detected at the XIST, IGF2, and GTL2 loci, which have previously been identified as imprinted in either humans, mice, or sheep. Expression patterns of parental alleles in F1 hybrids indicated preferential paternal expression at the XIST locus solely in the chorion of females, whereas analysis of the IGF2 and GTL2 loci indicated preferential paternal and maternal expression of alleles, respectively, in both fetal and placental tissues. Comparative sequence analysis of the XIST locus and adjacent regions suggests that repression of the maternal allele in the bovine is controlled by a different mechanism than in mice, further reinforcing the importance of comparative analysis of imprinting.
Collapse
Affiliation(s)
- Scott V Dindot
- Department of Veterinary Anatomy and Public Health, College of Veterinary Medicine, Texas A&M University, College Station, Texas 77843, USA
| | | | | | | | | | | |
Collapse
|
216
|
Abstract
Many insights into mammalian germ cell development have been gained through genetic engineering and in vivo studies, but the lack of an in vitro system for deriving germ cells has hindered potential advances in germ cell biology. Recent studies have demonstrated embryonic stem cell differentiation into germ cells and more mature gametes, although significant unanswered questions remain about the functionality of these cells. The derivation of germ cells from embryonic stem cells in vitro provides an invaluable assay both for the genetic dissection of germ cell development and for epigenetic reprogramming, and may one day facilitate nuclear transfer technology and infertility treatments.
Collapse
Affiliation(s)
- Jason A West
- Graduate Program of Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
217
|
Reyftmann L, Dechaud H, Hamamah S, Pucéat M, Hédon B. [Fetal and umbilical blood cord stem cells: a room for the obstetrician and gynaecologist. Part two]. ACTA ACUST UNITED AC 2004; 32:969-75. [PMID: 15567687 DOI: 10.1016/j.gyobfe.2004.07.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Stem cells are undifferentiated cells, with the ability to self renew and to differentiate into specialised cells. Besides embryonic stem cells, adult, fetal and umbilical cord blood (UB) stem cells are to be distinguished. These cells are multipotent. Embryonic germ cells (EG) that also are fetal stem cells have proven to be truly pluripotent, since they are able to give derivatives of the three primitive embryonic layers. EG cells have a normal karyotype, and exhibit remarkable long-term proliferative potential. Fetal stem cells and UB cells have already been used in cell therapy trials (e.g., Parkinson's disease, congenital immunodeficiencies and hemopathies). The applications in the field of reproductive biology will lead to a better understanding of genomic imprinting with EG cells. The obstetrician and gynaecologist could act a central part in the production and study of fetal stem cells, using tissues from aborted fetuses or collecting cord blood stem cells.
Collapse
Affiliation(s)
- L Reyftmann
- Service de gynécologie-obstétrique B et médecine de la reproduction, faculté de médecine, université Montpellier-I, hôpital universitaire Arnaud-de-Villeneuve, 371, avenue du Doyen-Gaston-Giraud, 34295 Montpellier 5, France.
| | | | | | | | | |
Collapse
|
218
|
Manoharan H, Babcock K, Pitot HC. Changes in the DNA methylation profile of the rat H19 gene upstream region during development and transgenic hepatocarcinogenesis and its role in the imprinted transcriptional regulation of the H19 gene. Mol Carcinog 2004; 41:1-16. [PMID: 15352122 DOI: 10.1002/mc.20036] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Monoallelic expression of the imprinted H19 and insulin-like growth factor-2 (Igf2) genes depends on the hypomethylation of the maternal allele and hypermethylation of the paternal allele of the H19 upstream region. Previous studies from our laboratory on liver carcinogenesis in the F1 hybrid of Fischer 344 (F344) and Sprague-Dawley Alb SV40 T Ag transgenic rat (SD) strains revealed the biallelic expression of H19 in hepatomas. We undertook a comparative study of the DNA methylation status of the upstream region of H19 in fetal, adult, and neoplastic liver. Bisulfite DNA sequencing analysis of a 3.745-kb DNA segment extending from 2950 to 6695 bp of the H19 upstream region revealed marked variations in the methylation patterns in fetal, adult, and neoplastic liver. In the fetal liver, equal proportions of hyper- and hypomethylated strands revealed the differentially methylated status of the parental alleles, but in neoplastic liver a pronounced change in the pattern of methylation was observed with a distinct change to hypomethylation in the short segments between 2984 and 3301 bp, 6033-6123 bp, and 6518-6548 bp. These results indicated that methylation of all cytosines in this region may contribute to the imprinting status of the rat H19 gene. This phenomenon of differential methylation-related epigenetic alteration in the key cis-regulatory domains of the H19 promoter influences switching to biallelic expression in hepatocellular carcinogenesis. Similar to mouse and human, we showed that the zinc-finger CCTCC binding factor (CTCF) binds to the unmethylated CTCF binding site in the upstream region to influence monoallelic imprinted expression in fetal liver. CTCF does not appear to be rate limiting in fetal, normal, and neoplastic liver. 3' to the CTCF binding sites, another DNA region exhibits methylation of CpG's in both DNA strands in adult liver, retention of the imprint in fetal liver, and complete demethylation in neoplastic liver. In this region is also a putative binding site for a basic helix-loop-helix leucine-zipper transcription factor, TFEB. The differential CpG methylation seen in the adult that involves the TFEB binding site may explain the lack of expression of the H19 gene in adult normal liver. Furthermore, these findings demonstrate that the loss of imprinting of the H19 gene in hepatic neoplasms of the SD Alb SV40 T Ag transgenic rat is directly correlated with and probably the result of differential methylation of CpG dinucleotides in two distinct regions of the gene that are within 4 kb 5' of the transcription start site. Cytogenetic analysis of hepatocytes in the transgenic animal prior to the appearance of nodules or neoplasms indicates a role of such loss of imprinting in the very early period of neoplastic development, possibly the transition from the stage of promotion to that of progression.
Collapse
Affiliation(s)
- Herbert Manoharan
- McArdle Laboratory for Cancer Research, Medical School, University of Wisconsin, Madison, Wisconsin 53706-1599, USA
| | | | | |
Collapse
|
219
|
Hansis C, Barreto G, Maltry N, Niehrs C. Nuclear reprogramming of human somatic cells by xenopus egg extract requires BRG1. Curr Biol 2004; 14:1475-80. [PMID: 15324664 DOI: 10.1016/j.cub.2004.08.031] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2004] [Revised: 06/08/2004] [Accepted: 06/21/2004] [Indexed: 10/26/2022]
Abstract
Animal cloning by nuclear transplantation in amphibia was demonstrated almost half a century ago and raised the question of the mechanisms and genes involved in nuclear reprogramming. Here, we demonstrate nuclear reprogramming of permeabilized human cells using extracts from Xenopus laevis eggs and early embryos. We show upregulation of pluripotency markers Oct-4 and germ cell alkaline phosphatase (GCAP) in 293T cells and human primary leukocytes. Reprogrammed leukocytes had a limited life span and did not express surface antigens characteristic of pluripotent cells, indicating that reprogramming was incomplete. Reprogramming activity was detected in egg and early embryo extracts until early blastula stage. Late blastula-stage extracts were not only inactive but also inhibitory to reprogramming. Screening for factors required for reprogramming identified the chromatin remodeling ATPase BRG1. Antibody depletion of BRG1 protein or expression of dominant-negative BRG1 abolished the reprogramming ability of amphibian extracts. Conversely, overexpression of BRG1 in Xenopus animal caps extended their competence from blastula to gastrula stage to respond to basic fibroblast growth factor (bFGF) treatment with induction of the mesodermal marker Xbra. Dissection of the molecular machinery using a simplified assay system may aid in achieving complete nuclear reprogramming of somatic cells for regenerative medicine.
Collapse
Affiliation(s)
- Christoph Hansis
- Division of Molecular Embryology, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | | | | | | |
Collapse
|
220
|
Lotem J, Benjamin H, Netanely D, Domany E, Sachs L. Induction in myeloid leukemic cells of genes that are expressed in different normal tissues. Proc Natl Acad Sci U S A 2004; 101:16022-7. [PMID: 15505217 PMCID: PMC528762 DOI: 10.1073/pnas.0406966101] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Using DNA microarray and cluster analysis of expressed genes in a cloned line (M1-t-p53) of myeloid leukemic cells, we have analyzed the expression of genes that are preferentially expressed in different normal tissues. Clustering of 547 highly expressed genes in these leukemic cells showed 38 genes preferentially expressed in normal hematopoietic tissues and 122 other genes preferentially expressed in different normal nonhematopoietic tissues, including neuronal tissues, muscle, liver, and testis. We have also analyzed the genes whose expression in the leukemic cells changed after activation of WT p53 and treatment with the cytokine IL-6 or the calcium mobilizer thapsigargin. Of 620 such genes in the leukemic cells that were differentially expressed in normal tissues, clustering showed 80 genes that were preferentially expressed in hematopoietic tissues and 132 genes in different normal nonhematopoietic tissues that also included neuronal tissues, muscle, liver, and testis. Activation of p53 and treatment with IL-6 or thapsigargin induced different changes in the genes preferentially expressed in these normal tissues. These myeloid leukemic cells thus express genes that are expressed in normal nonhematopoietic tissues, and various treatments can reprogram these cells to induce other such nonhematopoietic genes. The results indicate that these leukemic cells share with normal hematopoietic stem cells the plasticity of differentiation to different cell types. It is suggested that this reprogramming to induce in malignant cells genes that are expressed in different normal tissues may be of clinical value in therapy.
Collapse
Affiliation(s)
- Joseph Lotem
- Departments of Molecular Genetics and Physics of Complex Systems, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | |
Collapse
|
221
|
Murakami Y, Isogai K, Tomita H, Sakurai-Yageta M, Maruyama T, Hidaka A, Nose K, Sugano K, Kaneko A. Detection of allelic imbalance in the gene expression of hMSH2 or RB1 in lymphocytes from pedigrees of hereditary, nonpolyposis, colorectal cancer and retinoblastoma by an RNA difference plot. J Hum Genet 2004; 49:635-641. [PMID: 15480874 DOI: 10.1007/s10038-004-0201-0] [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] [Received: 07/07/2004] [Accepted: 08/24/2004] [Indexed: 10/26/2022]
Abstract
A number of phenotypes in hereditary disorders or common diseases are associated with specific genotypes. However, little is known about the molecular basis of phenotypic variation among individuals carrying the same mutation or polymorphism. Here, a highly quantitative approach was taken to examine a relative amount of mRNA from two polymorphic alleles with a coefficient of variation of less than 10% using an RNA difference plot (RDP). RDP analysis revealed that most genes examined were expressed in equal amount from the two alleles in normal lymphocytes. In contrast, the relative amounts of hMSH2 or RB1 mRNAs carrying premature termination codons were significantly reduced compared with those of wild-type mRNAs in lymphocytes from carriers of hereditary, nonpolyposis, colorectal cancer and hereditary retinoblastoma. The balance of allelic expression of the RB1 was also significantly impaired in a pedigree of retinoblastoma carrying a missense mutation in codon 661. The relative expression of the mutant to the wild-type RB1 alleles among the carriers varied from 0.40 to 2.39. The analysis of the expression diversity of a disease-associated allele by RDP could provide a novel approach to elucidating the mechanisms underlying phenotypic variation among individuals carrying an identical mutation or polymorphism at a single locus.
Collapse
Affiliation(s)
- Yoshinori Murakami
- Tumor Suppression and the Functional Genomics Project, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | - Kana Isogai
- Tumor Suppression and the Functional Genomics Project, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
- Department of Microbiology, Showa University School of Pharmacy, Tokyo, Japan
| | - Hiroyuki Tomita
- Tumor Suppression and the Functional Genomics Project, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
- Bio-system Research Department, Central Research Laboratory, Hitachi Ltd, Tokyo, Japan
| | - Mika Sakurai-Yageta
- Tumor Suppression and the Functional Genomics Project, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Tomoko Maruyama
- Tumor Suppression and the Functional Genomics Project, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Akio Hidaka
- Clinical Laboratory, National Cancer Center Hospital, Tokyo, Japan
| | - Kiyoshi Nose
- Department of Microbiology, Showa University School of Pharmacy, Tokyo, Japan
| | - Kokichi Sugano
- Clinical Laboratory, National Cancer Center Hospital, Tokyo, Japan
- Oncogene Research Unit/Cancer Prevention Unit, Tochigi Cancer Center Research Institute, Utsunomiya, Japan
| | - Akihiro Kaneko
- Department of Ophthalmology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
222
|
Vaiserman AM, Koshel NM, Mechova LV, Voitenko VP. Cross-life stage and cross-generational effects of ? irradiations at the egg stage on Drosophila melanogaster life histories. Biogerontology 2004; 5:327-37. [PMID: 15547320 DOI: 10.1007/s10522-004-2571-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The long-term effects of X-irradiation with 0.25, 0.5, 0.75 and 1 Gy of 1 h eggs on the fitness-related life history traits in adult Drosophila melanogaster fruit flies and their offspring were investigated. Following irradiation with 0.25, 0.5 and 0.75 Gy, both F0 and F1 flies have decreased adult body weight and increased locomotor (photo- and geotactic) activity, whereas metabolic rate measured as the rate of CO2 production was unchanged or even increased, and female fecundity was slightly reduced compared to appropriate controls. In some cases, irradiation resulted in hormetic effects increased resistance to both starvation and heat shock stresses as well as life extension. An explanation of the beneficial long lasting effects induced by early irradiation is offered, which suggests that these effects are due to cross-life stage and cross-generational adaptive phenotypic plasticity.
Collapse
Affiliation(s)
- Alexander M Vaiserman
- Laboratory of Mathematical Modelling of Aging Processes, Institute of Gerontology, Vyshgorodskaya st. 67, Kiev 04114, Ukraine.
| | | | | | | |
Collapse
|
223
|
Simonsson S, Gurdon J. DNA demethylation is necessary for the epigenetic reprogramming of somatic cell nuclei. Nat Cell Biol 2004; 6:984-90. [PMID: 15448701 DOI: 10.1038/ncb1176] [Citation(s) in RCA: 216] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2004] [Accepted: 08/20/2004] [Indexed: 11/09/2022]
Abstract
Nuclear transplantation experiments in amphibia and mammals have shown that oocyte and egg cytoplasm can extensively reprogram somatic cell nuclei with new patterns of gene expression and new pathways of cell differentiation; however, very little is known about the molecular mechanism of nuclear reprogramming. Here we have used nuclear and DNA transfer from mammalian somatic cells to analyse the mechanism of activation of the stem cell marker gene oct4 by Xenopus oocytes. We find that the removal of nuclear protein accelerates the rate of reprogramming, but even more important is the demethylation of somatic cell DNA. DNA demethylation seems to precede gene reprogramming, and is absolutely necessary for oct4 transcription. Reprogramming by oocytes occurs in the absence of DNA replication and RNA/protein synthesis. It is also selective, operating only on the promoter, but not enhancers, of oct4; both a putative Sp1/Sp3 and a GGGAGGG binding site are required for demethylation and transcription. We conclude that the demethylation of promoter DNA may be a necessary step in the epigenetic reprogramming of somatic cell nuclei.
Collapse
Affiliation(s)
- Stina Simonsson
- Wellcome Trust/Cancer Research UK Gurdon Institute and Department of Zoology, University of Cambridge, Cambridge CB2 1QR, UK
| | | |
Collapse
|
224
|
Morgan HD, Dean W, Coker HA, Reik W, Petersen-Mahrt SK. Activation-induced cytidine deaminase deaminates 5-methylcytosine in DNA and is expressed in pluripotent tissues: implications for epigenetic reprogramming. J Biol Chem 2004; 279:52353-60. [PMID: 15448152 DOI: 10.1074/jbc.m407695200] [Citation(s) in RCA: 370] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
DNA deaminases of the Aid/Apobec family convert cytosine into uracil and play key roles in acquired and innate immunity. The epigenetic modification by methylation of cytosine in CpG dinucleotides is also mutagenic, but this is thought to occur by spontaneous deamination. Here we show that Aid and Apobec1 are 5-methylcytosine deaminases resulting in a thymine base opposite a guanine. Their action can thus lead to C --> T transition mutations in methylated DNA, or in conjunction with repair of the T:G mismatch, to demethylation. The Aid and Apobec1 genes are located in a cluster of pluripotency genes including Nanog and Stella and are co-expressed with these genes in oocytes, embryonic germ cells, and embryonic stem cells. These results suggest that Aid and perhaps some of its family members may have roles in epigenetic reprogramming and cell plasticity. Transition in CpG dinucleotides is the most frequent mutation in human genetic diseases, and sequence context analysis of CpG transitions in the APC tumor suppressor gene suggests that DNA deaminases may play a significant role in tumor etiology.
Collapse
Affiliation(s)
- Hugh D Morgan
- Laboratory of Developmental Genetics and Imprinting, Developmental Genetics Programme, The Babraham Institute, Cambridge CB2 4AT, United Kingdom
| | | | | | | | | |
Collapse
|
225
|
Mattioli M, Gioia L, Turriani M, Capacchietti G, Loi P, Ptak G. Oocyte maturation is required for correct sperm chromatin rearrangement. Vet Res Commun 2004; 28 Suppl 1:205-8. [PMID: 15372958 DOI: 10.1023/b:verc.0000045407.33906.c9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- M Mattioli
- Dipartimento di Scienze Biomediche Comparate--Sezione di Fisiologia--Facoltà di Medicina Veterinaria--Università di Teramo, Italy.
| | | | | | | | | | | |
Collapse
|
226
|
Lee JH, Hart SRL, Skalnik DG. Histone deacetylase activity is required for embryonic stem cell differentiation. Genesis 2004; 38:32-8. [PMID: 14755802 DOI: 10.1002/gene.10250] [Citation(s) in RCA: 203] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mammalian development requires commitment of cells to restricted lineages, which requires epigenetic regulation of chromatin structure. Epigenetic modifications were examined during in vitro differentiation of murine embryonic stem (ES) cells. Global histone acetylation, a euchromatin marker, declines dramatically within 1 day of differentiation induction and partially rebounds by day 2. Histone H3-Lys9 methylation, a heterochromatin marker, increases during in vitro differentiation. Conversely, the euchromatin marker H3-Lys4 methylation transiently decreases, then increases to undifferentiated levels by day 4, and decreases by day 6. Global cytosine methylation, another heterochromatin marker, increases slightly during ES cell differentiation. Chromatin structure of the Oct4 and Brachyury gene promoters is modulated in concert with their pattern of expression during ES cell differentiation. Importantly, prevention of global histone deacetylation by treatment with trichostatin A prevents ES cell differentiation. Hence, ES cells undergo functionally important global and gene-specific remodeling of chromatin structure during in vitro differentiation. genesis 38:32-38, 2004.
Collapse
Affiliation(s)
- Jeong-Heon Lee
- Herman B Wells Center for Pediatric Research, Section of Pediatric Hematology/Oncology, Department of Pediatrics and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | |
Collapse
|
227
|
Waterland RA, Jirtle RL. Early nutrition, epigenetic changes at transposons and imprinted genes, and enhanced susceptibility to adult chronic diseases. Nutrition 2004; 20:63-8. [PMID: 14698016 DOI: 10.1016/j.nut.2003.09.011] [Citation(s) in RCA: 509] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Robert A Waterland
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | |
Collapse
|
228
|
Ma D, Shield JPH, Dean W, Leclerc I, Knauf C, Burcelin R RÉM, Rutter GA, Kelsey G. Impaired glucose homeostasis in transgenic mice expressing the human transient neonatal diabetes mellitus locus, TNDM. J Clin Invest 2004. [PMID: 15286800 DOI: 10.1172/jci200419876] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Transient neonatal diabetes mellitus (TNDM) is a rare inherited diabetic syndrome apparent in the first weeks of life and again during early adulthood. The relative contributions of reduced islet beta cell number and impaired beta cell function to the observed hypoinsulinemia are unclear. The inheritance pattern of this imprinted disorder implicates overexpression of one or both genes within the TNDM locus: ZAC, which encodes a proapoptotic zinc finger protein, and HYMAI, which encodes an untranslated mRNA. To investigate the consequences for pancreatic function, we have developed a high-copy transgenic mouse line, TNDM29, carrying the human TNDM locus. TNDM29 neonates display hyperglycemia, and older adults, impaired glucose tolerance. Neonatal hyperglycemia occurs only on paternal transmission, analogous to paternal dependence of TNDM in humans. Embryonic pancreata of TNDM29 mice showed reductions in expression of endocrine differentiation factors and numbers of insulin-staining structures. By contrast, beta cell mass was normal or elevated at all postnatal stages, whereas pancreatic insulin content in neonates and peak serum insulin levels after glucose infusion in adults were reduced. Expression of human ZAC and HYMAI in these transgenic mice thus recapitulates key features of TNDM and implicates impaired development of the endocrine pancreas and beta cell function in disease pathogenesis.
Collapse
Affiliation(s)
- Dan Ma
- Developmental Genetics Programme, The Babraham Institute, Cambridge, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
229
|
Ma D, Shield JPH, Dean W, Leclerc I, Knauf C, Burcelin R RÉM, Rutter GA, Kelsey G. Impaired glucose homeostasis in transgenic mice expressing the human transient neonatal diabetes mellitus locus, TNDM. J Clin Invest 2004; 114:339-48. [PMID: 15286800 PMCID: PMC484972 DOI: 10.1172/jci19876] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2003] [Accepted: 05/25/2004] [Indexed: 02/06/2023] Open
Abstract
Transient neonatal diabetes mellitus (TNDM) is a rare inherited diabetic syndrome apparent in the first weeks of life and again during early adulthood. The relative contributions of reduced islet beta cell number and impaired beta cell function to the observed hypoinsulinemia are unclear. The inheritance pattern of this imprinted disorder implicates overexpression of one or both genes within the TNDM locus: ZAC, which encodes a proapoptotic zinc finger protein, and HYMAI, which encodes an untranslated mRNA. To investigate the consequences for pancreatic function, we have developed a high-copy transgenic mouse line, TNDM29, carrying the human TNDM locus. TNDM29 neonates display hyperglycemia, and older adults, impaired glucose tolerance. Neonatal hyperglycemia occurs only on paternal transmission, analogous to paternal dependence of TNDM in humans. Embryonic pancreata of TNDM29 mice showed reductions in expression of endocrine differentiation factors and numbers of insulin-staining structures. By contrast, beta cell mass was normal or elevated at all postnatal stages, whereas pancreatic insulin content in neonates and peak serum insulin levels after glucose infusion in adults were reduced. Expression of human ZAC and HYMAI in these transgenic mice thus recapitulates key features of TNDM and implicates impaired development of the endocrine pancreas and beta cell function in disease pathogenesis.
Collapse
Affiliation(s)
- Dan Ma
- Developmental Genetics Programme, The Babraham Institute, Cambridge, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
230
|
Abstract
The past decade has witnessed immense progress in research into the molecular basis behind the developmental regulation of genes. Sets of genes functioning under hierarchical control have been identified, evolutionary conserved systems of genes effecting the cell-to-cell transmission of transmembrane signals and assigned a central role in morphogenesis have been intensively studied; the concept of genomic regulatory networks coordinating expression of many genes has been introduced, to mention some of the major breakthroughs. It should be noted that the temporal and tissue-specific parameters of gene expression are correctly regulated in development only in the context of the chromosome and that they are to a great extent dependent on the position of the gene on the chromosome or the interphase nucleus. Moreover epigenetic inheritance of the gene states through successive cell generations has been conducted exclusively at the chromosome level by virtue of cell or chromosome memory. The ontogenetic memory is an inherent property of the chromosome and cis-regulation has a crucial role in its maintenance.
Collapse
Affiliation(s)
- Oleg Serov
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-900, Brasil.
| | | |
Collapse
|
231
|
Liu L, Czerwiec E, Keefe DL. Effect of ploidy and parental genome composition on expression of Oct-4 protein in mouse embryos. Gene Expr Patterns 2004; 4:433-41. [PMID: 15183310 DOI: 10.1016/j.modgep.2004.01.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2003] [Revised: 01/06/2004] [Accepted: 01/07/2004] [Indexed: 11/24/2022]
Abstract
The transcription factor Oct-4 is expressed in germ cells and also is considered as a marker for pluripotency of stem cells. We first examined dynamics of Oct-4 protein expression during preimplantation development using both Western blot analysis, and immunofluorescence staining. We show that intact Oct-4 protein is not detected in either ovulated mature oocytes, or in zygotes and 2-4-cell embryos, which are the only known totipotent cell types in mammals. This finding is unexpected, since Oct-4 has been proposed to play a role in the control of totipotency. The results suggest that Oct-4 is not indispensable for fertilization and early cleavage. Rather, expression of Oct-4 protein is first detected in the nuclei of 8-16 cell morula, increases in early blastocysts, and declines in late blastocysts, in which most Oct-4 protein is confined to the inner cell mass (ICM) region, consistent with previous findings. We further compared Oct-4 protein expression in diploid and tetraploid blastocysts derived from normal fertilization or parthenogenesis, as well as expression in diploid androgenetic blastocysts. Expression levels and localization of Oct-4 protein are similar in both diploid and tetraploid early blastocysts, regardless of whether blastocysts are derived from fertilization or parthenogenesis. Androgenetic diploid blastocysts also express similar levels of Oct-4. Late blastocysts generated by both fertilization and parthenogenesis show a similar pattern of Oct-4 expression, suggesting that paternal genome activation is not required for Oct-4 expression. Expression of Oct-4 protein does not differ between diploid and tetraploid embryos, indicating that tetraploidy does not influence Oct-4 expression. Thus, expression of Oct-4 protein is initiated at morula stage in preimplantation embryos and completely controlled by a mechanism activated in oocytes. Downregulation of Oct-4 expression coincides with differentiation of trophectoderm. Similar profiles of Oct-4 expression observed in embryos with different ploidy and genome composition, are suggestive of Oct-4 being necessary but not sufficient for developmental potency.
Collapse
Affiliation(s)
- Lin Liu
- Department of Obstetrics/Gynecology, Women and Infants Hospital, Brown Medical School, Providence, RI 02905, USA.
| | | | | |
Collapse
|
232
|
von Wangenheim KH, Peterson HP. Aberrant endosperm development in interploidy crosses reveals a timer of differentiation. Dev Biol 2004; 270:277-89. [PMID: 15183714 DOI: 10.1016/j.ydbio.2004.03.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2003] [Revised: 12/19/2003] [Accepted: 03/03/2004] [Indexed: 01/09/2023]
Abstract
The common assumption that the seed failure in interploidy crosses of flowering plants is due to parental genomic imprinting is based on vague interpretations and needs reevaluation since the general question is involved, how differentiation is timed so that cell progenies, while specializing, pass through proper numbers of amplification divisions before proliferation ceases. As recently confirmed, endosperm differentiation is accelerated or de-accelerated, depending upon whether polyploid females are crossed with diploid males, or vice-versa. Unlike the zygote, the first cell of the endosperm is determined to produce a tissue that successively induces growth of maternal tissues, stimulates and nourishes the embryo, and finally ceases cell cycling. Altered timing of endosperm differentiation, thus, perturbs seed development. During fertilization, only the female genomes contribute cytoplasmic equivalents to endosperm development so that in interploidy crosses, the initial amount of cytoplasm per chromosome set is altered, and due to semi-autonomy of cytoplasmic growth, altered numbers of division cycles are needed to provide the amount of cytoplasmic organelles required for differentiation. Cytoplasmic semi-autonomy and dependence of differentiation on an increase in cytoplasm has been shown in other tissues of plants and animals, thus, revealing a common mechanism for intracellular timing of differentiation. As demonstrated, imprinted genes can alter the extent of cell proliferation by interfering with this mechanism.
Collapse
|
233
|
Abstract
Epigenetics refers to covalent modifications of DNA and core histones that regulate gene activity without altering DNA sequence. To date, the best-characterized DNA modification associated with the modulation of gene activity is methylation of cytosine residues within CpG dinucleotides. Human disorders associated with epigenetic abnormalities include rare imprinting diseases, molar pregnancies, and childhood cancers. Germ cell development and early embryo development are critical times when epigenetic patterns are initiated or maintained. This review focuses on the epigenetic modification DNA methylation and discusses recent progress that has been made in understanding when and how epigenetic patterns are differentially established in the male and female germlines, the mouse, and human disorders associated with abnormalities in epigenetic programming in germ cells and early embryos, as well as genetic and other modulators (e.g. nutrition and drugs) of reproductive epigenetic events.
Collapse
Affiliation(s)
- T L J Kelly
- McGill University-Montreal Children's Hospital Research Institute and Department of Paediatrics, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
234
|
Nishino K, Hattori N, Tanaka S, Shiota K. DNA methylation-mediated control of Sry gene expression in mouse gonadal development. J Biol Chem 2004; 279:22306-13. [PMID: 14978045 DOI: 10.1074/jbc.m309513200] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
DNA methylation at CpG sequences is involved in tissue-specific and developmentally regulated gene expression. The Sry (sex-determining region on the Y chromosome) gene encodes a master protein for initiating testis differentiation in mammals, and its expression is restricted to gonadal somatic cells at 10.5-12.5 days post-coitum (dpc) in the mouse. We found that in vitro methylation of the 5'-flanking region of the Sry gene caused suppression of reporter activity, implying that Sry gene expression could be regulated by DNA methylation-mediated gene silencing. Bisulfite restriction mapping and sodium bisulfite sequencing revealed that the 5'-flanking region of the Sry gene was hypermethylated in the 8.5-dpc embryos in which the Sry gene was not expressed. Importantly, this region was specifically hypomethylated in the gonad at 11.5 dpc, while the hypermethylated status was maintained in tissues that do not express the Sry gene. We concluded that expression of the Sry gene is under the control of an epigenetic mechanism mediated by DNA methylation.
Collapse
Affiliation(s)
- Koichiro Nishino
- Department of Animal Resource Sciences/Veterinary Medical Sciences, The University of Tokyo, Yayoi 1-1-1, Bukyo-ku, Tokyo 113-8657
| | | | | | | |
Collapse
|
235
|
Alison MR, Poulsom R, Otto WR, Vig P, Brittan M, Direkze NC, Lovell M, Fang TC, Preston SL, Wright NA. Recipes for adult stem cell plasticity: fusion cuisine or readymade? J Clin Pathol 2004; 57:113-20. [PMID: 14747430 PMCID: PMC1770217 DOI: 10.1136/jcp.2003.010074] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2003] [Indexed: 01/25/2023]
Abstract
A large body of evidence supports the idea that certain adult stem cells, particularly those of bone marrow origin, can engraft at alternative locations, particularly when the recipient organ is damaged. Under strong and positive selection pressure these cells will clonally expand/differentiate, making an important contribution to tissue replacement. Similarly, bone marrow derived cells can be amplified in vitro and differentiated into many types of tissue. Despite seemingly irrefutable evidence for stem cell plasticity, a veritable chorus of detractors has emerged, some doubting its very existence, motivated perhaps by more than a little self interest. The issues that have led to this situation include the inability to reproduce certain quite startling observations, and extrapolation from the behaviour of embryonic stem cells to suggest that adult bone marrow cells simply fuse with other cells and adopt their phenotype. Although these issues need resolving and, accepting that cell fusion does appear to allow reprogramming of haemopoietic cells in special circumstances, criticising this whole new field because some areas remain unclear is not good science.
Collapse
Affiliation(s)
- M R Alison
- Histopathology Unit, Cancer Research (UK), London WC2A 3PX, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
236
|
Kinoshita T, Miura A, Choi Y, Kinoshita Y, Cao X, Jacobsen SE, Fischer RL, Kakutani T. One-Way Control of FWA Imprinting in Arabidopsis Endosperm by DNA Methylation. Science 2004; 303:521-3. [PMID: 14631047 DOI: 10.1126/science.1089835] [Citation(s) in RCA: 437] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The Arabidopsis FWA gene was initially identified from late-flowering epigenetic mutants that show ectopic FWA expression associated with heritable hypomethylation of repeats around transcription starting sites. Here, we show that wild-type FWA displays imprinted (maternal origin-specific) expression in endosperm. The FWA imprint depends on the maintenance DNA methyltransferase MET1, as is the case in mammals. Unlike mammals, however, the FWA imprint is not established by allele-specific de novo methylation. It is established by maternal gametophyte-specific gene activation, which depends on a DNA glycosylase gene, DEMETER. Because endosperm does not contribute to the next generation, the activated FWA gene need not be silenced again. Double fertilization enables plants to use such "one-way" control of imprinting and DNA methylation in endosperm.
Collapse
Affiliation(s)
- Tetsu Kinoshita
- Integrated Genetics, National Institute of Genetics, Mishima 411-8540, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
237
|
N/A, 赵 文, 王 宇. N/A. Shijie Huaren Xiaohua Zazhi 2004; 12:205-208. [DOI: 10.11569/wcjd.v12.i1.205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
|
238
|
Abstract
The study of embryo stem cells began in 1963, initially using disaggregates of cleaving rabbit and mouse embryos. Their differentiation in vitro was modest, and usually curtailed at best to the formation of trophectoderm cells, which attached to plastic. Rabbit morulae and blastocysts adhered more readily, trophectoderm forming a sheet of cells which was overgrown by stem cells from inner cell mass. Whole-blastocyst cultures on collagen-coated surfaces produced a pile of cells, and its outgrowths included neural, blood, neuronal, phagocytic and many other types of cell. When inner cell mass was freed and cultured intact or as cell disaggregates, lines of embryo stem cells (ES) were established which possessed good rates of cleavage, and immense stability in their secretion of enzymes, morphology and chromosomal complement. Developmental capacities of single mouse embryo stem cells were measured by injecting one or more into a recipient blastocyst, and extent of colonization in resulting chimaeras measured their pluripotency. In mouse, cell clumps were termed embryoid bodies, which produced similar outgrowths as in rabbit. Component cells again differentiated widely, depending to a limited extent on their exposure to various cytokines or substrates. Markers for differentiation or pluripotency were established, which revealed how neural, cardiac, haematological and other ES lines could be established in vitro. These have proved useful to study early differentiation and their use in grafting to sick recipients. Displaying similar properties, human ES cells emerged in the late 1990s. Models for the clinical use of ES cells showed how they colonized rapidly, travelled to target tissues via fetal pathways, differentiated and colonized target organs. No signs of inflammation or tissue damage were noted; injured tissues could be repaired including remyelination, and no cancers were formed. ES cells offer wide therapeutic potentials for humans, although extensive clinical trials are still awaited.
Collapse
Affiliation(s)
- R G Edwards
- Reproductive BioMedicine Online, Duck End Farm, Dry Drayton, Cambridge CB3 8DB, UK.
| |
Collapse
|
239
|
|
240
|
Mogi C, Miyai S, Nishimura Y, Fukuro H, Yokoyama K, Takaki A, Inoue K. Differentiation of skeletal muscle from pituitary folliculo-stellate cells and endocrine progenitor cells. Exp Cell Res 2004; 292:288-94. [PMID: 14697336 DOI: 10.1016/j.yexcr.2003.09.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
We previously reported the ectopic differentiation of skeletal muscle cells in a pituitary gland transplanted beneath a kidney capsule. Morphological observation suggested that the skeletal muscle cells may have differentiated from folliculo-stellate (FS) cells in the anterior pituitary gland. However, at that time, we did not confirm this directly with an in vitro system. To obtain direct evidence, we used the Tpit/F1 cell line. The Tpit/F1 cell line was recently established from the pituitary gland of a temperature-sensitive T antigen transgenic mouse and has the characters of pituitary FS cells. Using Tpit/F1 cells, we have found that FS cells of the pituitary are able to differentiate into muscle cells in vitro. Additionally, we showed that the cells have some characteristics of pituitary FS cells and also express pituitary endocrine cell-specific transcription factor (pit-1) and prolactin genes, and can differentiate into striated muscle cells. The anterior pituitary gland is known to be of ectodermal origin, so the differentiation of its cells into striated muscle is completely unexpected. This is the first report of direct evidence of ectopic differentiation of skeletal muscle cells from pituitary cells.
Collapse
MESH Headings
- Actin Cytoskeleton/metabolism
- Actin Cytoskeleton/ultrastructure
- Animals
- Antimetabolites, Antineoplastic/pharmacology
- Azacitidine/pharmacology
- Biomarkers
- Cell Differentiation/physiology
- Cell Line
- Cell Line, Tumor/metabolism
- Cell Line, Tumor/ultrastructure
- Choristoma/metabolism
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Ectoderm/metabolism
- Ectoderm/ultrastructure
- Embryonic Induction/genetics
- Gene Expression Regulation, Developmental/physiology
- Mice
- Microscopy, Electron
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/ultrastructure
- Muscle, Skeletal/embryology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/ultrastructure
- MyoD Protein/genetics
- MyoD Protein/metabolism
- Myogenin/genetics
- Myogenin/metabolism
- Myoglobin/genetics
- Myoglobin/metabolism
- Pituitary Gland, Anterior/embryology
- Pituitary Gland, Anterior/metabolism
- Pituitary Gland, Anterior/ultrastructure
- Prolactin/genetics
- Prolactin/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Stem Cells/metabolism
- Stem Cells/ultrastructure
- Transcription Factor Pit-1
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Chihiro Mogi
- Department of Regulation Biology, Faculty of Science, Saitama University, Saitama 338-8570, Japan
| | | | | | | | | | | | | |
Collapse
|
241
|
Adams IR, McLaren A. Identification and characterisation of mRif1: A mouse telomere-associated protein highly expressed in germ cells and embryo-derived pluripotent stem cells. Dev Dyn 2004; 229:733-44. [PMID: 15042697 DOI: 10.1002/dvdy.10471] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
We have identified a mouse ortholog of the yeast Rif1 family of telomere-associated proteins on the basis of its high expression in primordial germ cells and embryo-derived pluripotent stem cell lines. mRif1 is also highly expressed in totipotent and pluripotent cells during early mouse development, and in male and female germ cells in adult mice. mRif1 expression is induced during derivation of embryonic stem cells and is rapidly down-regulated upon differentiation of embryonic stem cells in vitro. Furthermore, we show that mRif1 physically interacts with the telomere-associated protein mTrf2 and can be cross-linked to telomeric repeat DNA in mouse embryonic stem cells. mRif1 may be involved in the maintenance of telomere length or pluripotency in the germline and during early mouse development.
Collapse
Affiliation(s)
- Ian R Adams
- Wellcome Trust/Cancer Research UK Institute of Cancer and Developmental Biology, Cambridge, United Kingdom
| | | |
Collapse
|
242
|
Abstract
Somatic cloning by nuclear transfer returns a differentiated cell to a totipotent stage, a process termed nuclear reprogramming. During this de-differentiation process, genes inactivated during tissue differentiation are re-activated in a temporal and spatial special manner. It is believed that tissue differentiation occurs through epigenetic mechanisms, genetic inheritance that does not involve changes in DNA sequences. Developmental abnormalities and a high mortality rate in cloned offspring have frequently been observed and probably result from incomplete nuclear reprogramming. In this review, the reprogramming of two epigenetic mechanisms, imprinting and X chromosome inactivation, as well as recent attempts to modify pre-existing epigenetic marks in donor cells to improve nuclear transfer efficacy, are discussed.
Collapse
Affiliation(s)
- X Cindy Tian
- Centre for Regenerative Biology/Department of Animal Science, University of Connecticut, Storrs, CT 06269-4243, USA.
| |
Collapse
|
243
|
Abstract
Cell plasticity is a central issue in stem cell biology. Differentiated somatic nuclei have the flexibility to dedifferentiate when transferred into oocytes or when fused to pluripotent embryonic stem cells. Recent publications also claim that somatic stem cells can convert into developmentally unrelated cell types both in vivo and ex vivo without such drastic cell manipulations. Some of these claims are still controversial, making it difficult for us to determine the reality of somatic stem cell plasticity. Indeed, we have heard enough about the "potentials" of cell plasticity; how much do we know about mechanisms? A fundamental issue in current stem cell biology is to understand the mechanisms underlying cell plasticity. In this short review, we overview three research fields related to cell plasticity: nuclear transfer, transdifferentiation, and cell fusion, with an emphasis on studies of molecular mechanisms underlying cell plasticity.
Collapse
Affiliation(s)
- Michael S Rutenberg
- Department of Pathology, University of Florida College of Medicine, Gainesville, Florida 32610, USA
| | | | | | | |
Collapse
|
244
|
Abstract
For many years, adult haemopoietic stem cells (HSCs) have been considered 'plastic' in their proliferative and differentiation capacities. Recently, evidence that supports newer concepts of adult stem cell plasticity has been reported. In particular, stem cells from haemopoietic tissues seem to have 'extraordinary' abilities to generate or switch between haemopoietic and nonhaemopoietic lineages, exhibiting an unexpected degree of developmental or differentiation potential. The mechanisms by which cell fate reprogramming occurs are still poorly understood. Nevertheless, an increasing number of studies is challenging one of the main dogmas in biology, namely that mammalian cell differentiation follows established programmes in a hierarchical fashion, and once committed to a particular somatic cell lineage, cells do not change into another somatic lineage. The 'nonhierarchical', 'reversible' phenotype of stem cells in haemopoietic tissues, if it exists, would be an advantage that could be exploited in regenerative medicine. Here, we review the recent advances in HSC biology and discuss the general concepts of adult stem cell plasticity with respect to these cells and how these might be exploited clinically.
Collapse
Affiliation(s)
- E Martin-Rendon
- Stem Cell Research Laboratory, National Blood Service, Oxford Centre, Oxford, UK
| | | |
Collapse
|
245
|
Tian XC, Kubota C, Enright B, Yang X. Cloning animals by somatic cell nuclear transfer--biological factors. Reprod Biol Endocrinol 2003; 1:98. [PMID: 14614770 PMCID: PMC521203 DOI: 10.1186/1477-7827-1-98] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2003] [Accepted: 11/13/2003] [Indexed: 01/25/2023] Open
Abstract
Cloning by nuclear transfer using mammalian somatic cells has enormous potential application. However, somatic cloning has been inefficient in all species in which live clones have been produced. High abortion and fetal mortality rates are commonly observed. These developmental defects have been attributed to incomplete reprogramming of the somatic nuclei by the cloning process. Various strategies have been used to improve the efficiency of nuclear transfer, however, significant breakthroughs are yet to happen. In this review we will discuss studies conducted, in our laboratories and those of others, to gain a better understanding of nuclear reprogramming. Because cattle are a species widely used for nuclear transfer studies, and more laboratories have succeeded in cloning cattle than any other species, this review will be focused on somatic cell cloning of cattle.
Collapse
Affiliation(s)
- X Cindy Tian
- Center for Regenerative Biology/Department of Animal Science, University of Connecticut, Storrs, Connecticut 06269-4243, USA
| | - Chikara Kubota
- Kagoshima Prefectural Cattle Breeding and Genetic Institute, Kagoshima, Japan
| | - Brian Enright
- Center for Regenerative Biology/Department of Animal Science, University of Connecticut, Storrs, Connecticut 06269-4243, USA
| | - Xiangzhong Yang
- Center for Regenerative Biology/Department of Animal Science, University of Connecticut, Storrs, Connecticut 06269-4243, USA
- Evergen Biotechnologies, Inc. Incubator Program at the University of Connecticut, 1392 Storrs Road, Storrs, CT06269-USA
| |
Collapse
|
246
|
Vaiserman AM, Voitenko VP. Early programming of adult longevity: demographic and experimental studies. JOURNAL OF ANTI-AGING MEDICINE 2003; 6:11-20. [PMID: 12941179 DOI: 10.1089/109454503765361542] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
It is supposed that longevity might be programmed by early life exposures. We had carried out demographic and experimental researches for the examination of the possibility of longevity programming. In demographic study, the recorded deaths in Kiev (Ukraine) between 1990 and 2000 (51,503 men and 50,131 women) were used. Age at death was strongly associated with month of birth. Subjects born in the middle of year (April-July) had the lowest longevity. Increasing longevity was observed with each successive birth-month in the second half of the year, with a peak longevity for births in December. To research of the mechanisms responsible for longevity programming, study of adult D. melanogaster DNA repair capacity after irradiation at the egg stage was carried out, using marker such as DNA strand breaks. Insects irradiated in low doses (0.50 and 0.75 Gy) had extended life span and increased stability to S1 nuclease treatment. The probable explanation of observed postponed effects might be the long-term modulation of certain (possibly repair) genes activity. We hypothesize that life-extending effects of different anti-aging treatments might be a consequence of their unspecific (hormetic) action, rather then specific (geroprotector) action on the some aging-related processes, and induction an "transcriptional reprogramming" may be a key mechanism of the longevity programming and artificial life extension.
Collapse
Affiliation(s)
- Alexander M Vaiserman
- Laboratory of Mathematical Modeling of Aging Processes, Institute of Gerontology, Kiev, Ukraine.
| | | |
Collapse
|
247
|
Abstract
Primordial germ cells (PGCs), the embryonic precursors of the gametes of the adult animal, can give rise to two types of pluripotent stem cells. In vivo, PGCs can give rise to embryonal carcinoma cells, the pluripotent stem cells of testicular tumors. Cultured PGCs exposed to a specific cocktail of growth factors give rise to embryonic germ cells, pluripotent stem cells that can contribute to all the lineages of chimeric embryos including the germline. The conversion of PGCs into pluripotent stem cells is a remarkably similar process to nuclear reprogramming in which a somatic nucleus is reprogrammed in the egg cytoplasm. Understanding the genetics of embryonal carcinoma cell formation and the growth factor signaling pathways controlling embryonic germ cell derivation could tell us much about the molecular controls on developmental potency in mammals.
Collapse
Affiliation(s)
- Peter J Donovan
- Kimmel Cancer Center, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10th Street, Philadelphia, Pennsylvania 19107, USA.
| | | |
Collapse
|
248
|
Yamazaki Y, Mann MRW, Lee SS, Marh J, McCarrey JR, Yanagimachi R, Bartolomei MS. Reprogramming of primordial germ cells begins before migration into the genital ridge, making these cells inadequate donors for reproductive cloning. Proc Natl Acad Sci U S A 2003; 100:12207-12. [PMID: 14506296 PMCID: PMC218737 DOI: 10.1073/pnas.2035119100] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Germ cells undergo epigenetic modifications as they develop, which suggests that they may be ideal donors for nuclear transfer (cloning). In this study, nuclei from confirmed embryonic germ cells were used as donors to determine whether they are competent for cloning and at which stage they are most competent. Embryos cloned from migrating 10.5-days-postcoitum (dpc) primordial germ cells (PGCs) showed normal morphological development to midgestation but died shortly thereafter. In contrast, embryos cloned from later-stage germ cells were developmentally delayed at midgestation. Thus, donor germ cell age inversely correlated with the developmental stage attained by cloned embryos. The methylation status of the H19- and Snrpn-imprinting control regions in germ cell clones paralleled that of the donors, and revealed that demethylation, or erasure of imprints, was already initiated in PGCs at 10.5 dpc and was complete by 13.5 dpc. Similarly, clones derived from male 15.5-dpc germ cells showed increased methylation correlating with the initiation of de novo methylation that resets imprints at this stage, and clones from neonatal germ cells showed nearly complete methylation in the H19 imprinting control region. These results indicate that the epigenetic state of the donor nucleus is retained in cloned embryos, and that germ cells are therefore inadequate nuclear donors for cloning because they are either erasing or resetting epigenetic patterns.
Collapse
Affiliation(s)
- Yukiko Yamazaki
- Institute for Biogenesis Research, Department of Anatomy and Reproductive Biology, University of Hawaii Medical School, Honolulu, HI 96822, USA
| | | | | | | | | | | | | |
Collapse
|
249
|
Erhardt S, Su IH, Schneider R, Barton S, Bannister AJ, Perez-Burgos L, Jenuwein T, Kouzarides T, Tarakhovsky A, Surani MA. Consequences of the depletion of zygotic and embryonic enhancer of zeste 2 during preimplantation mouse development. Development 2003; 130:4235-48. [PMID: 12900441 DOI: 10.1242/dev.00625] [Citation(s) in RCA: 249] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Enhancer of zeste 2 (Ezh2), a SET domain-containing protein, is crucial for development in many model organisms, including early mouse development. In mice, Ezh2 is detected as a maternally inherited protein in the oocyte but its function at the onset of development is unknown. We have used a conditional allele of Ezh2 to deplete the oocyte of this maternal inheritance. We show that the loss of maternal Ezh2 has a long-term effect causing severe growth retardation of neonates despite 'rescue' through embryonic transcription from the paternal allele. This phenotypic effect on growth could be attributed to the asymmetric localisation of the Ezh2/Eed complex and the associated histone methylation pattern to the maternal genome, which is disrupted in Ezh2 mutant zygotes. During subsequent development, we detect distinct histone methylation patterns in the trophectoderm and the pluripotent epiblast. In the latter where Oct4 expression continues from the zygote onwards, the Ezh2/Eed complex apparently establishes a unique epigenetic state and plasticity, which probably explains why loss of Ezh2 is early embryonic lethal and obligatory for the derivation of pluripotent embryonic stem cells. By contrast, in the differentiating trophectoderm cells where Oct4 expression is progressively downregulated Ezh2/Eed complex is recruited transiently to one X chromosome in female embryos at the onset of X-inactivation. This accumulation and the associated histone methylation are also lost in Ezh2 mutants, suggesting a role in X inactivation. Thus, Ezh2 has significant and diverse roles during early development, as well as during the establishment of the first differentiated cells, the trophectoderm, and of the pluripotent epiblast cells.
Collapse
Affiliation(s)
- Sylvia Erhardt
- Wellcome Trust/Cancer Research UK Institute, University of Cambridge, Cambridge CB2 1QR, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
250
|
Gómez MC, Jenkins JA, Giraldo A, Harris RF, King A, Dresser BL, Pope CE. Nuclear transfer of synchronized african wild cat somatic cells into enucleated domestic cat oocytes. Biol Reprod 2003; 69:1032-41. [PMID: 12773426 DOI: 10.1095/biolreprod.102.014449] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The African wild cat is one of the smallest wild cats and its future is threatened by hybridization with domestic cats. Nuclear transfer, a valuable tool for retaining genetic variability, offers the possibility of species continuation rather than extinction. The aim of this study was to investigate the ability of somatic cell nuclei of the African wild cat (AWC) to dedifferentiate within domestic cat (DSH) cytoplasts and to support early development after nuclear transplantation. In experiment 1, distributions of AWC and DSH fibroblasts in each cell-cycle phase were assessed by flow cytometry using cells cultured to confluency and disaggregated with pronase, trypsin, or mechanical separation. Trypsin (89.0%) and pronase (93.0%) yielded higher proportions of AWC nuclei in the G0/G1 phase than mechanical separation (82.0%). In contrast, mechanical separation yielded higher percentages of DSH nuclei in the G0/G1 phase (86.6%) than pronase (79.7%) or trypsin (74.2%) treatments. In both species, pronase induced less DNA damage than trypsin. In experiment 2, the effects of serum starvation, culture to confluency, and exposure to roscovitine on the distribution of AWC and DSH fibroblasts in various phases of the cell cycle were determined. Flow cytometry analyses revealed that the dynamics of the cell cycle varied as culture conditions were modified. Specifically, a higher percentage of AWC and DSH nuclei were in the G0/G1 phase after cells were serum starved (83% vs. 96%) than were present in cycling cells (50% vs. 64%), after contact inhibition (61% vs. 88%), or after roscovitine (56% vs. 84%) treatment, respectively. In experiment 3, we evaluated the effects of cell synchronization and oocyte maturation (in vivo vs. in vitro) on the reconstruction and development of AWC-DSH- and DSH-DSH-cloned embryos. The method of cell synchronization did not affect the fusion and cleavage rate because only a slightly higher percentage of fused couplets cleaved when donor nuclei were synchronized by serum starvation (83.0%) than after roscovitine (80.0%) or contact-inhibition (80.0%). The fusion efficiency of in vivo and in vitro matured oocytes used as recipient cytoplasts of AWC donor nuclei (86.6% vs. 85.2%) was similar to the rates obtained with DSH donor nuclei, 83.7% vs. 73.0%, respectively. The only significant effect of source of donor nucleus (AWC vs. DSH) was on the rate of blastocyst formation in vitro. A higher percentage of the embryos derived from AWC nuclei developed to the blastocyst stage than did embryos produced from DSH nuclei, 24.2% vs. 3.3%, respectively (P < 0.05). In experiment 4, the effect of calcium in the fusion medium on induction of oocyte activation and development of AWC-DSH-cloned embryos was determined. The presence of calcium in the fusion medium induced a high incidence of cleavage of DSH oocytes (54.3%), while oocyte cleavage frequency was much lower in the absence of calcium (16.6%). The presence or absence of calcium in the fusion medium did not affect the fusion, cleavage, and blastocyst development of AWC-DSH-cloned embryos. In experiment 5, AWC-DSH-cloned embryos were transferred to the uteri of 11 synchronized domestic cat recipients on Day 6 or 7 after oocyte aspiration. Recipients were assessed by ultrasonography on Day 21 postovulation, but no pregnancies were observed. In the present study, after NT, AWC donor nuclei were able to dedifferentiate in DSH cytoplasts and support high rates of blastocyst development in vitro. Incomplete reprogramming of the differentiated nucleus may be a major constraint to the in vivo developmental potential of the embryos.
Collapse
Affiliation(s)
- Martha C Gómez
- Department of Animal Science, Louisiana State University, Baton Rouge, Louisiana 70808, USA.
| | | | | | | | | | | | | |
Collapse
|