251
|
Pluripotency factors flick the switch. Nat Rev Genet 2008. [DOI: 10.1038/nrg2469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
252
|
Wen S, Li H, Liu J. Epigenetic background of neuronal fate determination. Prog Neurobiol 2008; 87:98-117. [PMID: 19007844 DOI: 10.1016/j.pneurobio.2008.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Revised: 09/03/2008] [Accepted: 10/15/2008] [Indexed: 01/07/2023]
Abstract
The development of the central nervous system (CNS) starts from neural stem cells (NSCs). During this process, NSCs are specified in space- and time-related fashions, becoming spatially heterogeneous and generating a progressively restricted repertoire of cell types: neurons, astrocytes and oligodendrocytes. The processes of neurodevelopment are determined reciprocally by intrinsic and external factors which interface to program and re-program the profiling of fate-determination gene expression. Multiple signaling pathways act in a dynamic web mode to determine the fate of NSCs through modulating the activity of a distinct set of transcription factors which in turn trigger the transcription of neural fate-determination genes. Accumulating evidence reveals that during CNS development, multiple epigenetic factors regulate the activities of extracellular signaling and corresponding transcription factors in a coordinative manner, leading to the formation of a system with sophisticated structure and magic functions. This review aims to introduce recent advances in the epigenetic background of neural cell fate determination.
Collapse
Affiliation(s)
- Shu Wen
- Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, 116044 Dalian, Liaoning, PR China
| | | | | |
Collapse
|
253
|
Golob JL, Paige SL, Muskheli V, Pabon L, Murry CE. Chromatin remodeling during mouse and human embryonic stem cell differentiation. Dev Dyn 2008; 237:1389-98. [PMID: 18425849 DOI: 10.1002/dvdy.21545] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Embryonic stem cell (ESC) differentiation is an excellent model to study chromatin changes at developmentally regulated loci. Differentiating mouse and human ESCs increase genome-wide acetylation (euchromatic) and tri-methylation (heterochromatic) of lysine 9 on histone H3. The Oct4 locus is euchromatic when expressed in undifferentiated ESCs and heterochromatic after differentiation. Brachyury T, a mesoderm-specific transcription factor, is not yet expressed in undifferentiated cells, where its locus has "bivalent" tri-methyl lysine 4 and lysine 27 modifications. During directed differentiation to pre-cardiac mesoderm, the activated brachyury locus has high levels of tri-methyl lysine 4 (euchromatin), switching to heterochromatin after gene silencing. Thus, ESC differentiation is accompanied by genome-wide commitment to euchromatin or heterochromatin. Undifferentiated hESCs bivalently modify the brachyury locus, activate it to euchromatin during mesoderm induction, and subsequently repress it to heterochromatin, demonstrating, to our knowledge, the first analysis of chromatin dynamics at a locus essential for mesoderm and endoderm differentiation.
Collapse
Affiliation(s)
- Jonathan L Golob
- Department of Pathology, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA
| | | | | | | | | |
Collapse
|
254
|
Li X, Barkho BZ, Luo Y, Smrt RD, Santistevan NJ, Liu C, Kuwabara T, Gage FH, Zhao X. Epigenetic regulation of the stem cell mitogen Fgf-2 by Mbd1 in adult neural stem/progenitor cells. J Biol Chem 2008; 283:27644-27652. [PMID: 18689796 PMCID: PMC2562066 DOI: 10.1074/jbc.m804899200] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2008] [Revised: 08/08/2008] [Indexed: 11/06/2022] Open
Abstract
Whether and how mechanisms intrinsic to stem cells modulate their proliferation and differentiation are two central questions in stem cell biology. Although exogenous basic fibroblast growth factor 2 (FGF-2/Fgf-2) is commonly used to expand adult neural stem/progenitor cells (NSPCs) in vitro, we do not yet understand the functional significance or the molecular regulation of Fgf-2 expressed endogenously by adult NSPCs. We previously demonstrated that methylated CpG binding protein 1 (MBD1/Mbd1) is a transcriptional repressor of Fgf-2 and is enriched in adult brains. Mbd1 deficiency in mice selectively affected adult neurogenesis and the differentiation of NSPCs. Here we show that an Mbd1 and DNA methylation-mediated epigenetic mechanism regulated the expression of stem cell mitogen Fgf-2 in adult NSPCs. Mbd1 bound to the Fgf-2 promoter and regulates its expression in adult NSPCs. In the absence of functional Mbd1, the Fgf-2 promoter was hypomethylated, and treatment with a DNA methylation inhibitor resulted in increased Fgf-2 expression in adult NSPCs. We further demonstrated that both acute knockdown of Mbd1 or overexpression of Fgf-2 in adult NSPCs inhibited their neuronal differentiation, which could be responsible for the neurogenic deficits observed in Mbd1-deficient mice. These data indicate that intrinsic epigenetic mechanisms play critical roles in the regulation of adult NSPC functions.
Collapse
Affiliation(s)
- Xuekun Li
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131 and the
| | - Basam Z Barkho
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131 and the
| | - Yuping Luo
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131 and the
| | - Richard D Smrt
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131 and the
| | - Nicholas J Santistevan
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131 and the
| | - Changmei Liu
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131 and the
| | - Tomoko Kuwabara
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California 92037
| | - Fred H Gage
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California 92037
| | - Xinyu Zhao
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131 and the.
| |
Collapse
|
255
|
Guven-Ozkan T, Nishi Y, Robertson SM, Lin R. Global transcriptional repression in C. elegans germline precursors by regulated sequestration of TAF-4. Cell 2008; 135:149-60. [PMID: 18854162 PMCID: PMC2652481 DOI: 10.1016/j.cell.2008.07.040] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Revised: 04/18/2008] [Accepted: 07/16/2008] [Indexed: 10/21/2022]
Abstract
In C. elegans, four asymmetric divisions, beginning with the zygote (P0), generate transcriptionally repressed germline blastomeres (P1-P4) and somatic sisters that become transcriptionally active. The protein PIE-1 represses transcription in the later germline blastomeres but not in the earlier germline blastomeres P0 and P1. We show here that OMA-1 and OMA-2, previously shown to regulate oocyte maturation, repress transcription in P0 and P1 by binding to and sequestering in the cytoplasm TAF-4, a component critical for assembly of TFIID and the pol II preinitiation complex. OMA-1/2 binding to TAF-4 is developmentally regulated, requiring phosphorylation by the DYRK kinase MBK-2, which is activated at meiosis II after fertilization. OMA-1/2 are normally degraded after the first mitosis, but ectopic expression of wild-type OMA-1 is sufficient to repress transcription in both somatic and later germline blastomeres. We propose that phosphorylation by MBK-2 serves as a developmental switch, converting OMA-1/2 from oocyte to embryo regulators.
Collapse
Affiliation(s)
- Tugba Guven-Ozkan
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Scott M. Robertson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rueyling Lin
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
256
|
Bibikova M, Laurent LC, Ren B, Loring JF, Fan JB. Unraveling epigenetic regulation in embryonic stem cells. Cell Stem Cell 2008; 2:123-34. [PMID: 18371433 DOI: 10.1016/j.stem.2008.01.005] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Embryonic stem (ES) cells can replicate indefinitely while retaining the capacity to differentiate into functionally distinct cell types. ES cells proliferate and differentiate without detectable genetic changes, indicating that these processes are controlled by epigenetic factors. Here we describe what is known about the epigenetics of ES cells and speculate that a dynamic balance among at least three epigenetic elements (chromatin structure, DNA methylation, and microRNAs), in conjunction with transcription factors, contributes to the maintenance of pluripotence. Understanding the interactions among these factors will be critical to the development of improved strategies to reprogram differentiated cells or direct differentiation of pluripotent cells.
Collapse
|
257
|
Abstract
Acute myeloid leukemia (AML) is a disease characterized by uncontrolled proliferation of clonal neoplastic hematopoietic precursor cells. This leads to the disruption of normal hematopoiesis and bone marrow failure. Major breakthroughs in the past have contributed to our understanding of the genetic failures and the changed biology in AML cells that underlie the initiation and progression of the disease. It is now recognized that not only genetic but also epigenetic alterations are similarly important in this process. Since these alterations do not change the DNA sequences and are pharmacologically reversible, they have been regarded as optimal targets for what is now known as epigenetic therapy. In this review, we will discuss our current understanding of normal epigenetic processes, outline our knowledge of epigenetic alterations in AML, and discuss how this information is being used to improve current therapy of this disease.
Collapse
|
258
|
The promise of human induced pluripotent stem cells for research and therapy. Nat Rev Mol Cell Biol 2008; 9:725-9. [PMID: 18698329 DOI: 10.1038/nrm2466] [Citation(s) in RCA: 302] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Induced pluripotent stem (iPS) cells are human somatic cells that have been reprogrammed to a pluripotent state. There are several hurdles to be overcome before iPS cells can be considered as a potential patient-specific cell therapy, and it will be crucial to characterize the developmental potential of human iPS cell lines. As a research tool, iPS-cell technology provides opportunities to study normal development and to understand reprogramming. iPS cells can have an immediate impact as models for human diseases, including cancer
Collapse
|
259
|
Thorne JL, Campbell MJ, Turner BM. Transcription factors, chromatin and cancer. Int J Biochem Cell Biol 2008; 41:164-75. [PMID: 18804550 DOI: 10.1016/j.biocel.2008.08.029] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2008] [Revised: 08/15/2008] [Accepted: 08/18/2008] [Indexed: 01/26/2023]
Abstract
Transcription factors, chromatin and chromatin-modifying enzymes are key components in a complex network through which the genome interacts with its environment. For many transcription factors, binding motifs are found adjacent to the promoter regions of a large proportion of genes, requiring mechanisms that confer binding specificity in any given cell type. These include association of the factor with other proteins and packaging of DNA, as chromatin, at the binding sequence so as to inhibit or facilitate binding. Recent evidence suggests that specific post-translational modifications of the histones packaging promoter DNA can help guide transcription factors to selected sites. The enzymes that put such modifications in place are dependent on metabolic components (e.g. acetyl CoA, S-adenosyl methionine) and susceptible to inhibition or activation by environmental factors. Local patterns of histone modification can be altered or maintained through direct interaction between the transcription factor and histone modifying enzymes. The functional consequences of transcription factor binding are also dependent on protein modifying enzymes, particularly those that alter lysine methylation at selected residues. Remarkably, the role of these enzymes is not limited to promoter-proximal events, but can be linked to changes in the intranuclear location of target genes. In this review we describe results that begin to define how transcription factors, chromatin and environmental variables interact and how these interactions are subverted in cancer. We focus on the nuclear receptor family of transcription factors, where binding of ligands such as steroid hormones and dietary derived factors provides an extra level of environmental input.
Collapse
Affiliation(s)
- James L Thorne
- University of Birmingham Medical School, Edgbaston, Birmingham, B15 2TT, UK
| | | | | |
Collapse
|
260
|
Fleuriel C, Touka M, Boulay G, Guérardel C, Rood BR, Leprince D. HIC1 (Hypermethylated in Cancer 1) epigenetic silencing in tumors. Int J Biochem Cell Biol 2008; 41:26-33. [PMID: 18723112 DOI: 10.1016/j.biocel.2008.05.028] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 05/19/2008] [Accepted: 05/19/2008] [Indexed: 12/27/2022]
Abstract
HIC1 (Hypermethylated in Cancer 1), as it name implied, was originally isolated as a new candidate tumor suppressor gene located at 17p13.3 because it resides in a CpG island that is hypermethylated in many types of human cancers. HIC1 encodes a transcription factor associating an N-terminal BTB/POZ domain to five C-terminal Krüppel-like C(2)H(2) zinc finger motifs. In this review, we will begin by providing an overview of the current knowledge on HIC1 function, mainly gained from in vitro studies, as a sequence-specific transcriptional repressor interacting with a still growing range of HDAC-dependent and HDAC-independent corepressor complexes. We will then summarize the studies that have demonstrated frequent hypermethylation changes or losses of heterozygosity of the HIC1 locus in human cancers. Next, we will review animal models which have firmly established HIC1 as a bona fide tumor suppressor gene epigenetically silenced and functionally cooperating notably with p53 within a complex HIC1-p53-SIRT1 regulatory loop. Finally, we will discuss how this epigenetic inactivation of HIC1 might "addict" cancer cells to altered survival and signaling pathways or to lineage-specific transcription factors during the early stages of tumorigenesis.
Collapse
Affiliation(s)
- Capucine Fleuriel
- Université de Lille 1 et de Lille 2, Institut PASTEUR de LILLE, 59017 Lille Cedex, France
| | | | | | | | | | | |
Collapse
|
261
|
A new coactivator function for Zac1's C2H2 zinc finger DNA-binding domain in selectively controlling PCAF activity. Mol Cell Biol 2008; 28:6078-93. [PMID: 18663001 DOI: 10.1128/mcb.00842-08] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The generally accepted paradigm of transcription by regulated recruitment defines sequence-specific transcription factors and coactivators as separate categories that are distinguished by their abilities to bind DNA autonomously. The C(2)H(2) zinc finger protein Zac1, with an established role in canonical DNA binding, also acts as a coactivator. Commensurate with this function, p73, which is related to p53, is here shown to recruit Zac1, together with the coactivators p300 and PCAF, to the p21(Cip1) promoter during the differentiation of embryonic stem cells into neurons. In the absence of autonomous DNA binding, Zac1's zinc fingers stabilize the association of PCAF with p300, suggesting its scaffolding function. Furthermore, Zac1 regulates the affinities of PCAF substrates as well as the catalytic activities of PCAF to induce a selective switch in favor of histone H4 acetylation and thereby the efficient transcription of p21(Cip1). These results are consistent with an authentic coactivator function of Zac1's C(2)H(2) zinc finger DNA-binding domain and suggest coactivation by sequence-specific transcription factors as a new facet of transcriptional control.
Collapse
|
262
|
Transcriptional regulation of neuronal differentiation: the epigenetic layer of complexity. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2008; 1779:432-7. [PMID: 18674649 DOI: 10.1016/j.bbagrm.2008.07.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2008] [Revised: 07/11/2008] [Accepted: 07/18/2008] [Indexed: 11/22/2022]
Abstract
The transcriptional programs of neural progenitor cells change dynamically during neurogenesis, a process regulated by both intrinsic and extrinsic factors. Although many of the transcription factors required for neuronal differentiation have long been identified, we are only at the brink of understanding how epigenetic mechanisms influence transcriptional activity and the accessibility of transcription factors to bind consensus cis-elements. Herein, we delineate the chief epigenetic modifications and the machinery responsible for these alterations. Further, we review the epigenetic modifications presently known to participate in the maintenance of the neural progenitor cell state and in the regulation of neuronal differentiation.
Collapse
|
263
|
Fazzio TG, Huff JT, Panning B. An RNAi screen of chromatin proteins identifies Tip60-p400 as a regulator of embryonic stem cell identity. Cell 2008; 134:162-74. [PMID: 18614019 DOI: 10.1016/j.cell.2008.05.031] [Citation(s) in RCA: 347] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 03/04/2008] [Accepted: 05/01/2008] [Indexed: 01/30/2023]
Abstract
Proper regulation of chromatin structure is necessary for the maintenance of cell type-specific gene expression patterns. The embryonic stem cell (ESC) expression pattern governs self-renewal and pluripotency. Here, we present an RNAi screen in mouse ESCs of 1008 loci encoding chromatin proteins. We identified 68 proteins that exhibit diverse phenotypes upon knockdown (KD), including seven subunits of the Tip60-p400 complex. Phenotypic analyses revealed that Tip60-p400 is necessary to maintain characteristic features of ESCs. We show that p400 localization to the promoters of both silent and active genes is dependent upon histone H3 lysine 4 trimethylation (H3K4me3). Furthermore, the Tip60-p400 KD gene expression profile is enriched for developmental regulators and significantly overlaps with that of the transcription factor Nanog. Depletion of Nanog reduces p400 binding to target promoters without affecting H3K4me3 levels. Together, these data indicate that Tip60-p400 integrates signals from Nanog and H3K4me3 to regulate gene expression in ESCs.
Collapse
Affiliation(s)
- Thomas G Fazzio
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94158, USA.
| | | | | |
Collapse
|
264
|
Häyry V, Tanner M, Blom T, Tynninen O, Roselli A, Ollikainen M, Sariola H, Wartiovaara K, Nupponen NN. Copy number alterations of the polycomb gene BMI1 in gliomas. Acta Neuropathol 2008; 116:97-102. [PMID: 18427816 DOI: 10.1007/s00401-008-0376-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Revised: 04/01/2008] [Accepted: 04/04/2008] [Indexed: 01/08/2023]
Abstract
Gliomas are heterogeneous tumours that grow in an uninhibited fashion, and these brain tumour cells share numerous characteristics with neural stem cells. The BMI1 gene encodes a component of the polycomb protein complex regulating epigenetically gene activity via histone modification. It functions for instance during the development of the central nervous system and maturation of neural cells. BMI-1 protein expression is deregulated in several forms of cancer and gene amplification has been identified in mantle cell lymphomas. Since BMI1 is located at chromosome 10p, a region implicated frequently in brain tumourigenesis, we investigated the genetic status and the corresponding expression patterns of BMI1 in a series of 100 low- and high-grade primary and recurrent gliomas. Chromogenic in situ hybridisation (CISH) with probes directed against BMI1 at 10p13 and the centromere of chromosome 10 was used in the analyses. Of all gliomas, 59% demonstrated aberrant copy numbers of BMI1. Deletions of the BMI1 locus were found in most types of tumours, and in a univariate survival analysis these cases had poor prognosis. Increased copy numbers of the BMI1 locus (3-5 copies) were found in all histological types, especially in high-grade astrocytomas. No difference in prognosis between cases with normal copy numbers and cases with increased copy numbers could be observed. This data suggests that BMI1 gene is aberrant at the chromosomal level in a subset of gliomas, and possibly contributes to brain tumour pathogenesis.
Collapse
|
265
|
Farthing CR, Ficz G, Ng RK, Chan CF, Andrews S, Dean W, Hemberger M, Reik W. Global mapping of DNA methylation in mouse promoters reveals epigenetic reprogramming of pluripotency genes. PLoS Genet 2008; 4:e1000116. [PMID: 18584034 PMCID: PMC2432031 DOI: 10.1371/journal.pgen.1000116] [Citation(s) in RCA: 260] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2008] [Accepted: 06/04/2008] [Indexed: 12/11/2022] Open
Abstract
DNA methylation patterns are reprogrammed in primordial germ cells and in preimplantation embryos by demethylation and subsequent de novo methylation. It has been suggested that epigenetic reprogramming may be necessary for the embryonic genome to return to a pluripotent state. We have carried out a genome-wide promoter analysis of DNA methylation in mouse embryonic stem (ES) cells, embryonic germ (EG) cells, sperm, trophoblast stem (TS) cells, and primary embryonic fibroblasts (pMEFs). Global clustering analysis shows that methylation patterns of ES cells, EG cells, and sperm are surprisingly similar, suggesting that while the sperm is a highly specialized cell type, its promoter epigenome is already largely reprogrammed and resembles a pluripotent state. Comparisons between pluripotent tissues and pMEFs reveal that a number of pluripotency related genes, including Nanog, Lefty1 and Tdgf1, as well as the nucleosome remodeller Smarcd1, are hypomethylated in stem cells and hypermethylated in differentiated cells. Differences in promoter methylation are associated with significant differences in transcription levels in more than 60% of genes analysed. Our comparative approach to promoter methylation thus identifies gene candidates for the regulation of pluripotency and epigenetic reprogramming. While the sperm genome is, overall, similarly methylated to that of ES and EG cells, there are some key exceptions, including Nanog and Lefty1, that are highly methylated in sperm. Nanog promoter methylation is erased by active and passive demethylation after fertilisation before expression commences in the morula. In ES cells the normally active Nanog promoter is silenced when targeted by de novo methylation. Our study suggests that reprogramming of promoter methylation is one of the key determinants of the epigenetic regulation of pluripotency genes. Epigenetic reprogramming in the germline prior to fertilisation and the reprogramming of key pluripotency genes in the early embryo is thus crucial for transmission of pluripotency.
Collapse
Affiliation(s)
- Cassandra R. Farthing
- Laboratory of Developmental Genetics and Imprinting, The Babraham Institute, Cambridge, United Kingdom
| | - Gabriella Ficz
- Laboratory of Developmental Genetics and Imprinting, The Babraham Institute, Cambridge, United Kingdom
| | - Ray Kit Ng
- Laboratory of Developmental Genetics and Imprinting, The Babraham Institute, Cambridge, United Kingdom
| | - Chun-Fung Chan
- Laboratory of Developmental Genetics and Imprinting, The Babraham Institute, Cambridge, United Kingdom
| | - Simon Andrews
- Bioinformatics Group, The Babraham Institute, Cambridge, United Kingdom
| | - Wendy Dean
- Laboratory of Developmental Genetics and Imprinting, The Babraham Institute, Cambridge, United Kingdom
| | - Myriam Hemberger
- Laboratory of Developmental Genetics and Imprinting, The Babraham Institute, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Wolf Reik
- Laboratory of Developmental Genetics and Imprinting, The Babraham Institute, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
266
|
Parthenogenesis as an approach to pluripotency: advantages and limitations involved. ACTA ACUST UNITED AC 2008; 4:127-35. [PMID: 18548354 DOI: 10.1007/s12015-008-9027-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2008] [Indexed: 10/22/2022]
Abstract
Embryonic stem cells (ESCs) are invaluable cells derived from the inner cell mass of the mammalian blastocyst. They have nearly indefinite self-renewal, retain their developmental potential after prolonged periods in culture and display great plasticity that allow them to differentiate into all cell types of the body. They provide exciting opportunities to develop unique models for developmental research and hold great potential for cell and tissue replacement therapy. However, these unique cells cannot be obtained without destroying an embryo and, despite the potential therapeutic usefulness, their derivation in the human raises substantial ethical as well as legal and political concerns because it unavoidably involves the destruction of viable embryos. In the recent years a number of scientific proposals that do not require the generation and subsequent destruction of human embryos have been put forward in an attempt to fill the gap between ethical questions and potential scientific and medical benefits. In this review we briefly summarize data obtained from the literature related to these different alternative approaches and focus in more details on our experience in the derivation of parthenothes, as a possible alternative source for pluripotent cells, discussing the advantages as well as the limits of these cell lines.
Collapse
|
267
|
Sauer S, Bruno L, Hertweck A, Finlay D, Leleu M, Spivakov M, Knight ZA, Cobb BS, Cantrell D, O'Connor E, Shokat KM, Fisher AG, Merkenschlager M. T cell receptor signaling controls Foxp3 expression via PI3K, Akt, and mTOR. Proc Natl Acad Sci U S A 2008; 105:7797-802. [PMID: 18509048 PMCID: PMC2409380 DOI: 10.1073/pnas.0800928105] [Citation(s) in RCA: 696] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Indexed: 12/13/2022] Open
Abstract
Regulatory T (Treg) cells safeguard against autoimmunity and immune pathology. Because determinants of the Treg cell fate are not completely understood, we have delineated signaling events that control the de novo expression of Foxp3 in naive peripheral CD4 T cells and in thymocytes. We report that premature termination of TCR signaling and inibition of phosphatidyl inositol 3-kinase (PI3K) p110alpha, p110delta, protein kinase B (Akt), or mammalian target of rapamycin (mTOR) conferred Foxp3 expression and Treg-like gene expression profiles. Conversely, continued TCR signaling and constitutive PI3K/Akt/mTOR activity antagonised Foxp3 induction. At the chromatin level, di- and trimethylation of lysine 4 of histone H3 (H3K4me2 and -3) near the Foxp3 transcription start site (TSS) and within the 5' untranslated region (UTR) preceded active Foxp3 expression and, like Foxp3 inducibility, was lost upon continued TCR stimulation. These data demonstrate that the PI3K/Akt/mTOR signaling network regulates Foxp3 expression.
Collapse
Affiliation(s)
| | | | | | - David Finlay
- Division of Cell Biology and Immunology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom; and
| | | | | | - Zachary A. Knight
- Department of Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, CA 94143
| | | | - Doreen Cantrell
- Division of Cell Biology and Immunology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom; and
| | - Eric O'Connor
- Flow Cytometry Facility, Medical Research Council Clinical Sciences Centre, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - Kevan M. Shokat
- Department of Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, CA 94143
| | | | | |
Collapse
|
268
|
Collas P, Noer A, Sørensen AL. Epigenetic Basis for the Differentiation Potential of Mesenchymal and Embryonic Stem Cells. ACTA ACUST UNITED AC 2008; 35:205-215. [PMID: 21547118 DOI: 10.1159/000127449] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Accepted: 02/06/2008] [Indexed: 12/13/2022]
Abstract
SUMMARY: Stem cells have the ability to self-renew, and give rise to one or more differentiated cell types. Embryonic stem cells can differentiate into all cell types of the body and have unlimited self-renewal capacity. Somatic stem cells are found in many adult tissues. They have an extensive but finite lifespan and can differentiate into a more restricted range of cell types. Increasing evidence indicates that the multilineage differentiation ability of stem cells is defined by the potential for expression of developmentally regulated transcription factors and of lineage specification genes. Gene expression, or as emphasized here, the potential for gene expression, is largely controlled by epigenetic modifications of DNA (DNA methylation) and chromatin (such as post-translational histone modifications) in the regulatory regions of specific genes. Epigenetic modifications can also influence the timing of DNA replication. We highlight here how mechanisms by which genes are poised for transcription in undifferentiated stem cells are being uncovered through the mapping of DNA methylation profiles on differentiation-regulated promoters and at the genome-wide level, histone modifications, and transcription factor binding. Epigenetic marks on developmentally regulated and lineage specification genes in stem cells seem to define a state of pluripotency.
Collapse
Affiliation(s)
- Philippe Collas
- Institute of Basic Medical Sciences, Department of Biochemistry, Faculty of Medicine, University of Oslo, Norway
| | | | | |
Collapse
|
269
|
Wu X, Gong Y, Yue J, Qiang B, Yuan J, Peng X. Cooperation between EZH2, NSPc1-mediated histone H2A ubiquitination and Dnmt1 in HOX gene silencing. Nucleic Acids Res 2008; 36:3590-9. [PMID: 18460542 PMCID: PMC2441805 DOI: 10.1093/nar/gkn243] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
An intricate interplay between DNA methylation and polycomb-mediated gene silencing has been highlighted recently. Here we provided evidence that Nervous System Polycomb 1 (NSPc1), a BMI1 homologous polycomb protein, plays important roles in promoting H2A ubiquitination and cooperates with DNA methylation in HOX gene silencing. We showed that NSPc1 stimulates H2A ubiquitination in vivo and in vitro through direct interaction with both RING2 and H2A. RT-PCR analysis revealed that loss of NSPc1, EZH2 or DNA methyltransferase 1 (Dnmt1), or inhibition of DNA methylation in HeLa cells de-represses the expression of HOXA7. Chromatin immunoprecipitation (ChIP) assays demonstrated that NSPc1, EZH2 and Dnmt1 bind to the promoter of HOXA7, which is frequently hypermethylated in tumors. Knockdown of NSPc1 results in significant reduction of H2A ubiquitination and DNA demethylation as well as Dnmt1 dissociation in the HOXA7 promoter. Meanwhile Dnmt1 deficiency affects NSPc1 recruitment and H2A ubiquitination, whereas on both cases EZH2-mediated H3K27 trimethylation remains unaffected. When EZH2 was depleted, however, NSPc1 and Dnmt1 enrichment was abolished concomitant with local reduction of H3K27 trimethylation, H2A ubiquitination and DNA methylation. Taken together, our findings indicated that NSPc1-mediated H2A ubiquitination and DNA methylation, both being directed by EZH2, are interdependent in long-term target gene silencing within cancer cells.
Collapse
Affiliation(s)
- Xudong Wu
- School of Basic Medicine, Peking Union Medical College; National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, China
| | | | | | | | | | | |
Collapse
|
270
|
Gurtner A, Fuschi P, Magi F, Colussi C, Gaetano C, Dobbelstein M, Sacchi A, Piaggio G. NF-Y dependent epigenetic modifications discriminate between proliferating and postmitotic tissue. PLoS One 2008; 3:e2047. [PMID: 18431504 PMCID: PMC2295263 DOI: 10.1371/journal.pone.0002047] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Accepted: 03/13/2008] [Indexed: 02/03/2023] Open
Abstract
The regulation of gene transcription requires posttranslational modifications of histones that, in concert with chromatin remodeling factors, shape the structure of chromatin. It is currently under intense investigation how this structure is modulated, in particular in the context of proliferation and differentiation. Compelling evidence suggests that the transcription factor NF-Y acts as a master regulator of cell cycle progression, activating the transcription of many cell cycle regulatory genes. However, the underlying molecular mechanisms are not yet completely understood. Here we show that NF-Y exerts its effect on transcription through the modulation of the histone "code". NF-Y colocalizes with nascent RNA, while RNA polymerase II is I phosphorylated on serine 2 of the YSPTSPS repeats within its carboxyterminal domain and histones are carrying modifications that represent activation signals of gene expression (H3K9ac and PAN-H4ac). Comparing postmitotic muscle tissue from normal mice and proliferating muscles from mdx mice, we demonstrate by chromatin immunoprecipitation (ChIP) that NF-Y DNA binding activity correlates with the accumulation of acetylated histones H3 and H4 on promoters of key cell cycle regulatory genes, and with their active transcription. Accordingly, p300 is recruited onto the chromatin of NF-Y target genes in a NF-Y-dependent manner, as demonstrated by Re-ChIP. Conversely, the loss of NF-Y binding correlates with a decrease of acetylated histones, the recruitment of HDAC1, and a repressed heterochromatic state with enrichment of histones carrying modifications known to mediate silencing of gene expression (H3K9me3, H3K27me2 and H4K20me3). As a consequence, NF-Y target genes are downregulated in this context. In conclusion, our data indicate a role of NF-Y in modulating the structure and transcriptional competence of chromatin in vivo and support a model in which NF-Y-dependent histone "code" changes contribute to the proper discrimination between proliferating and postmitotic cells in vivo and in vitro.
Collapse
Affiliation(s)
- Aymone Gurtner
- Molecular Oncogenesis Laboratory, Experimental Oncology Department, Regina Elena Cancer Institute, Rome, Italy
| | - Paola Fuschi
- Molecular Oncogenesis Laboratory, Experimental Oncology Department, Regina Elena Cancer Institute, Rome, Italy
| | - Fiorenza Magi
- Molecular Oncogenesis Laboratory, Experimental Oncology Department, Regina Elena Cancer Institute, Rome, Italy
| | - Claudia Colussi
- Laboratory of Vascular Pathology, Istituto Dermopatico dell'Immacolata, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Carlo Gaetano
- Laboratory of Vascular Pathology, Istituto Dermopatico dell'Immacolata, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Matthias Dobbelstein
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), Göttingen, Germany
| | - Ada Sacchi
- Molecular Oncogenesis Laboratory, Experimental Oncology Department, Regina Elena Cancer Institute, Rome, Italy
| | - Giulia Piaggio
- Molecular Oncogenesis Laboratory, Experimental Oncology Department, Regina Elena Cancer Institute, Rome, Italy
- Rome Oncogenomic Center, Experimental Oncology Department, Regina Elena Cancer Institute, Rome, Italy
- * E-mail:
| |
Collapse
|
271
|
Yeo GW, Coufal N, Aigner S, Winner B, Scolnick JA, Marchetto MC, Muotri AR, Carson C, Gage FH. Multiple layers of molecular controls modulate self-renewal and neuronal lineage specification of embryonic stem cells. Hum Mol Genet 2008; 17:R67-75. [DOI: 10.1093/hmg/ddn065] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
272
|
Karantzali E, Schulz H, Hummel O, Hubner N, Hatzopoulos A, Kretsovali A. Histone deacetylase inhibition accelerates the early events of stem cell differentiation: transcriptomic and epigenetic analysis. Genome Biol 2008; 9:R65. [PMID: 18394158 PMCID: PMC2643936 DOI: 10.1186/gb-2008-9-4-r65] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Revised: 01/14/2008] [Accepted: 04/04/2008] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Epigenetic mechanisms regulate gene expression patterns affecting cell function and differentiation. In this report, we examine the role of histone acetylation in gene expression regulation in mouse embryonic stem cells employing transcriptomic and epigenetic analysis. RESULTS Embryonic stem cells treated with the histone deacetylase inhibitor Trichostatin A (TSA), undergo morphological and gene expression changes indicative of differentiation. Gene profiling utilizing Affymetrix microarrays revealed the suppression of important pluripotency factors, including Nanog, a master regulator of stem cell identity, and the activation of differentiation-related genes. Transcriptional and epigenetic changes induced after 6-12 hours of TSA treatment mimic those that appear during embryoid body differentiation. We show here that the early steps of stem cell differentiation are marked by the enhancement of bulk activatory histone modifications. At the individual gene level, we found that transcriptional reprogramming triggered by histone deacetylase inhibition correlates with rapid changes in activating K4 trimethylation and repressive K27 trimethylation of histone H3. The establishment of H3K27 trimethylation is required for stable gene suppression whereas in its absence, genes can be reactivated upon TSA removal. CONCLUSION Our data suggest that inhibition of histone deacetylases accelerates the early events of differentiation by regulating the expression of pluripotency- and differentiation-associated genes in an opposite manner. This analysis provides information about genes that are important for embryonic stem cell function and the epigenetic mechanisms that regulate their expression.
Collapse
Affiliation(s)
- Efthimia Karantzali
- Institute of Molecular Biology and Biotehnology, FORTH, Heraklion 71110 Greece
| | | | | | | | | | | |
Collapse
|
273
|
Graumann J, Hubner NC, Kim JB, Ko K, Moser M, Kumar C, Cox J, Schöler H, Mann M. Stable Isotope Labeling by Amino Acids in Cell Culture (SILAC) and Proteome Quantitation of Mouse Embryonic Stem Cells to a Depth of 5,111 Proteins. Mol Cell Proteomics 2008; 7:672-83. [DOI: 10.1074/mcp.m700460-mcp200] [Citation(s) in RCA: 245] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
274
|
Li Z, Barron MR, Lough J, Zhao M. Rapid Single-Step Separation of Pluripotent Mouse Embryonic Stem Cells from Mouse Feeder Fibroblasts. Stem Cells Dev 2008; 17:383-7. [DOI: 10.1089/scd.2007.0138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Zhixin Li
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Matthew R. Barron
- Department of Cell Biology, Neurobiology & Anatomyz, Medical College of Wisconsin, Milwaukee, WI 53226
| | - John Lough
- Department of Cell Biology, Neurobiology & Anatomyz, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Ming Zhao
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226
| |
Collapse
|
275
|
|
276
|
Epigenetic events in mammalian germ-cell development: reprogramming and beyond. Nat Rev Genet 2008; 9:129-40. [PMID: 18197165 DOI: 10.1038/nrg2295] [Citation(s) in RCA: 613] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The epigenetic profile of germ cells, which is defined by modifications of DNA and chromatin, changes dynamically during their development. Many of the changes are associated with the acquisition of the capacity to support post-fertilization development. Our knowledge of this aspect has greatly increased- for example, insights into how the re-establishment of parental imprints is regulated. In addition, an emerging theme from recent studies is that epigenetic modifiers have key roles in germ-cell development itself--for example, epigenetics contributes to the gene-expression programme that is required for germ-cell development, regulation of meiosis and genomic integrity. Understanding epigenetic regulation in germ cells has implications for reproductive engineering technologies and human health.
Collapse
|
277
|
Exner V, Hennig L. Chromatin rearrangements in development. CURRENT OPINION IN PLANT BIOLOGY 2008; 11:64-9. [PMID: 18024147 DOI: 10.1016/j.pbi.2007.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2007] [Revised: 10/02/2007] [Accepted: 10/03/2007] [Indexed: 05/22/2023]
Abstract
Chromatin states change dramatically during plant development. Globally, cytologically defined heterochromatin increases during cell differentiation and organ maturation, while it decreases during callus formation and protoplastization. Interestingly, around the time of bolting, heterochromatin content of leaf nuclei decreases transiently. Locally, chromatin compactness of the regulatory gene GLABRA2 is controlled by positional cues and correlates with transcriptional activity. In the case of the flowering time regulator FLC, chromatin compactness and histone modifications are controlled by environmental cues and ensure faithful maintenance of gene repression after vernalization. The combination of cytological studies, locus-specific analyses, and novel genome-wide profiling techniques should soon lead to a more detailed understanding of the mechanisms coupling intranuclear architecture and development.
Collapse
Affiliation(s)
- Vivien Exner
- Institute of Plant Sciences & Zurich-Basel Plant Science Center, ETH Zurich, CH-8092 Zurich, Switzerland.
| | | |
Collapse
|
278
|
Orford KW, Scadden DT. Deconstructing stem cell self-renewal: genetic insights into cell-cycle regulation. Nat Rev Genet 2008; 9:115-28. [DOI: 10.1038/nrg2269] [Citation(s) in RCA: 670] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
279
|
Flintoft L. Safeguarding pluripotency. Nat Rev Genet 2008. [DOI: 10.1038/nrg2306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
280
|
Lewitzky M, Yamanaka S. Reprogramming somatic cells towards pluripotency by defined factors. Curr Opin Biotechnol 2008; 18:467-73. [PMID: 18024106 DOI: 10.1016/j.copbio.2007.09.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Revised: 09/24/2007] [Accepted: 09/26/2007] [Indexed: 02/07/2023]
Abstract
The recent years have seen great advances in reversal of programming of differentiated somatic cells towards pluripotency by methods not involving nuclear transfer. Some of these may present a first step on the way to individual-based cell therapy without the problems connected to collection of mammalian unfertilised oocytes. Although differentiation of cells involves complex genetic and epigenetic changes, it is now possible to generate cells with many properties of pluripotent embryonic stem cells by retroviral transduction of differentiated cells with only four transcription factors: Oct3/4, Sox2, Klf4 and c-Myc. The re-programmed cells contribute to live chimeric mice and are transmitted via the germline.
Collapse
Affiliation(s)
- Marc Lewitzky
- Department of Stem Cell Biology, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | | |
Collapse
|
281
|
Schneider R, Grosschedl R. Dynamics and interplay of nuclear architecture, genome organization, and gene expression. Genes Dev 2008; 21:3027-43. [PMID: 18056419 DOI: 10.1101/gad.1604607] [Citation(s) in RCA: 316] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The organization of the genome in the nucleus of a eukaryotic cell is fairly complex and dynamic. Various features of the nuclear architecture, including compartmentalization of molecular machines and the spatial arrangement of genomic sequences, help to carry out and regulate nuclear processes, such as DNA replication, DNA repair, gene transcription, RNA processing, and mRNA transport. Compartmentalized multiprotein complexes undergo extensive modifications or exchange of protein subunits, allowing for an exquisite dynamics of structural components and functional processes of the nucleus. The architecture of the interphase nucleus is linked to the spatial arrangement of genes and gene clusters, the structure of chromatin, and the accessibility of regulatory DNA elements. In this review, we discuss recent studies that have provided exciting insight into the interplay between nuclear architecture, genome organization, and gene expression.
Collapse
|
282
|
Abstract
ES cell research represents an exploding field of exploration. Initially predicted to provide rapid cures for numerous human diseases, the clinical usefulness of ES cell-derived cells remains untested in humans. However, ES cells have rapidly expanded our knowledge of human development and the molecular details of differentiation. Our ability to generate relatively pure populations of specifically differentiated cells for transplantation has markedly improved. It is hoped that soon researchers will overcome the biologic impediments to successful treatment of human disease with ES cell-derived cells.
Collapse
|
283
|
|
284
|
Stock JK, Giadrossi S, Casanova M, Brookes E, Vidal M, Koseki H, Brockdorff N, Fisher AG, Pombo A. Ring1-mediated ubiquitination of H2A restrains poised RNA polymerase II at bivalent genes in mouse ES cells. Nat Cell Biol 2007; 9:1428-35. [PMID: 18037880 DOI: 10.1038/ncb1663] [Citation(s) in RCA: 493] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Accepted: 10/29/2007] [Indexed: 12/16/2022]
Abstract
Changes in phosphorylation of the carboxy-terminal domain (CTD) of RNA polymerase II (RNAP) are associated with transcription initiation, elongation and termination. Sites of active transcription are generally characterized by hyperphosphorylated RNAP, particularly at Ser 2 residues, whereas inactive or poised genes may lack RNAP or may bind Ser 5-phosphorylated RNAP at promoter proximal regions. Recent studies have demonstrated that silent developmental regulator genes have an unusual histone modification profile in ES cells, being simultaneously marked with Polycomb repressor-mediated histone H3K27 methylation, and marks normally associated with gene activity. Contrary to the prevailing view, we show here that this important subset of developmental regulator genes, termed bivalent genes, assemble RNAP complexes phosphorylated on Ser 5 and are transcribed at low levels. We provide evidence that this poised RNAP configuration is enforced by Polycomb Repressor Complex (PRC)-mediated ubiquitination of H2A, as conditional deletion of Ring1A and Ring1B leads to the sequential loss of ubiquitination of H2A, release of poised RNAP, and subsequent gene de-repression. These observations provide an insight into the molecular mechanisms that allow ES cells to self-renew and yet retain the ability to generate multiple lineage outcomes.
Collapse
Affiliation(s)
- Julie K Stock
- Nuclear Organisation, MRC Clinical Sciences Centre, Imperial College School of Medicine, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
285
|
RNA sequence analysis defines Dicer's role in mouse embryonic stem cells. Proc Natl Acad Sci U S A 2007; 104:18097-102. [PMID: 17989215 DOI: 10.1073/pnas.0709193104] [Citation(s) in RCA: 248] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Short RNA expression was analyzed from Dicer-positive and Dicer-knockout mouse embryonic [corrected] stem (ES) cells, using high-throughput pyrosequencing. A correlation of miRNA quantification with sequencing frequency estimates that there are 110,000 miRNAs per ES cell, the majority of which can be accounted for by six distinct miRNA loci. Four of these miRNA loci or their human homologues have demonstrated roles in cell cycle regulation or oncogenesis, suggesting that a major function of the miRNA pathway in ES cells may be to shape their distinct cell cycle. Forty-six previously uncharacterized miRNAs were identified, most of which are expressed at low levels and are less conserved than the set of known miRNAs. Low-abundance short RNAs matching all classes of repetitive elements were present in cells lacking Dicer, although the production of some SINE- and simple repeat-associated short RNAs appeared to be Dicer-dependent. These and other Dicer-dependent sequences resembled miRNAs. At a depth of sequencing that approaches the total number of 5' phosphorylated short RNAs per cell, miRNAs appeared to be Dicer's only substrate. The results presented suggest a model in which repeat-associated miRNAs serve as host defenses against repetitive elements, a function canonically ascribed to other classes of short RNA.
Collapse
|
286
|
Abstract
In opposition to terminally differentiated cells, stem cells can self-renew and give rise to multiple cell types. Embryonic stem cells retain the ability of the inner cell mass of blastocysts to differentiate into all cell types of the body and have acquired in culture unlimited self-renewal capacity. Somatic stem cells are found in many adult tissues, have an extensive but finite lifespan and can differentiate into a more restricted array of cell types. A growing body of evidence indicates that multi-lineage differentiation ability of stem cells can be defined by the potential for expression of lineage-specification genes. Gene expression, or as emphasized here, potential for gene expression, is largely controlled by epigenetic modifications of DNA and chromatin on genomic regulatory and coding regions. These modifications modulate chromatin organization not only on specific genes but also at the level of the whole nucleus; they can also affect timing of DNA replication. This review highlights how mechanisms by which genes are poised for transcription in undifferentiated stem cells are being uncovered through primarily the mapping of DNA methylation, histone modifications and transcription factor binding throughout the genome. The combinatorial association of epigenetic marks on developmentally regulated and lineage-specifying genes in undifferentiated cells seems to define a pluripotent state.
Collapse
Affiliation(s)
- Philippe Collas
- Institute of Basic Medical Sciences, Department of Biochemistry, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway.
| | | | | |
Collapse
|
287
|
Matushansky I, Hernando E, Socci ND, Mills JE, Matos TA, Edgar MA, Singer S, Maki RG, Cordon-Cardo C. Derivation of sarcomas from mesenchymal stem cells via inactivation of the Wnt pathway. J Clin Invest 2007; 117:3248-57. [PMID: 17948129 PMCID: PMC2030456 DOI: 10.1172/jci31377] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2006] [Accepted: 08/03/2007] [Indexed: 01/26/2023] Open
Abstract
Malignant fibrous histiocytoma (MFH), now termed high-grade undifferentiated pleomorphic sarcoma, is a commonly diagnosed mesenchymal tumor, yet both the underlying molecular mechanisms of tumorigenesis and cell of origin remain unidentified. We present evidence demonstrating that human mesenchymal stem cells (hMSCs) are the progenitors of MFH. DKK1, a Wnt inhibitor and mediator of hMSC proliferation, is overexpressed in MFH. Using recombinant proteins, antibody depletion, and siRNA knockdown strategies of specific Wnt elements, we show that DKK1 inhibits hMSC commitment to differentiation via Wnt2/beta-catenin canonical signaling and that Wnt5a/JNK noncanonical signaling regulates a viability checkpoint independent of Dkk1. Finally, we illustrate that hMSCs can be transformed via inhibition of Wnt signaling to form MFH-like tumors in nude mice, and conversely, MFH cells in which Wnt signaling is appropriately reestablished can differentiate along mature connective tissue lineages. Our results provide mechanistic insights regarding the cell of origin of MFH, establish what we believe is a novel tumor suppressor role for Wnt signaling, and identify a potential therapeutic differentiation strategy for sarcomas.
Collapse
Affiliation(s)
- Igor Matushansky
- Department of Medicine, Columbia University, New York, New York, USA.
Department of Pathology, New York University School of Medicine, New York, New York, USA.
Computational Biology Center, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Department of Pathology, Columbia University, New York, New York, USA.
Department of Pathology,
Department of Surgery, and
Department of Medicine, MSKCC, New York, New York, USA
| | - Eva Hernando
- Department of Medicine, Columbia University, New York, New York, USA.
Department of Pathology, New York University School of Medicine, New York, New York, USA.
Computational Biology Center, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Department of Pathology, Columbia University, New York, New York, USA.
Department of Pathology,
Department of Surgery, and
Department of Medicine, MSKCC, New York, New York, USA
| | - Nicholas D. Socci
- Department of Medicine, Columbia University, New York, New York, USA.
Department of Pathology, New York University School of Medicine, New York, New York, USA.
Computational Biology Center, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Department of Pathology, Columbia University, New York, New York, USA.
Department of Pathology,
Department of Surgery, and
Department of Medicine, MSKCC, New York, New York, USA
| | - Joslyn E. Mills
- Department of Medicine, Columbia University, New York, New York, USA.
Department of Pathology, New York University School of Medicine, New York, New York, USA.
Computational Biology Center, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Department of Pathology, Columbia University, New York, New York, USA.
Department of Pathology,
Department of Surgery, and
Department of Medicine, MSKCC, New York, New York, USA
| | - Tulio A. Matos
- Department of Medicine, Columbia University, New York, New York, USA.
Department of Pathology, New York University School of Medicine, New York, New York, USA.
Computational Biology Center, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Department of Pathology, Columbia University, New York, New York, USA.
Department of Pathology,
Department of Surgery, and
Department of Medicine, MSKCC, New York, New York, USA
| | - Mark A. Edgar
- Department of Medicine, Columbia University, New York, New York, USA.
Department of Pathology, New York University School of Medicine, New York, New York, USA.
Computational Biology Center, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Department of Pathology, Columbia University, New York, New York, USA.
Department of Pathology,
Department of Surgery, and
Department of Medicine, MSKCC, New York, New York, USA
| | - Samuel Singer
- Department of Medicine, Columbia University, New York, New York, USA.
Department of Pathology, New York University School of Medicine, New York, New York, USA.
Computational Biology Center, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Department of Pathology, Columbia University, New York, New York, USA.
Department of Pathology,
Department of Surgery, and
Department of Medicine, MSKCC, New York, New York, USA
| | - Robert G. Maki
- Department of Medicine, Columbia University, New York, New York, USA.
Department of Pathology, New York University School of Medicine, New York, New York, USA.
Computational Biology Center, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Department of Pathology, Columbia University, New York, New York, USA.
Department of Pathology,
Department of Surgery, and
Department of Medicine, MSKCC, New York, New York, USA
| | - Carlos Cordon-Cardo
- Department of Medicine, Columbia University, New York, New York, USA.
Department of Pathology, New York University School of Medicine, New York, New York, USA.
Computational Biology Center, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Department of Pathology, Columbia University, New York, New York, USA.
Department of Pathology,
Department of Surgery, and
Department of Medicine, MSKCC, New York, New York, USA
| |
Collapse
|
288
|
Rajasekhar VK, Begemann M. Concise Review: Roles of Polycomb Group Proteins in Development and Disease: A Stem Cell Perspective. Stem Cells 2007; 25:2498-510. [PMID: 17600113 DOI: 10.1634/stemcells.2006-0608] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The acquisition and maintenance of cell fate are essential for metazoan growth and development. A strict coordination between genetic and epigenetic programs regulates cell fate determination and maintenance. Polycomb group (PcG) genes are identified as essential in these epigenetic developmental processes. These genes encode components of multimeric transcriptional repressor complexes that are crucial in maintaining cell fate. PcG proteins have also been shown to play a central role in stem cell maintenance and lineage specification. PcG proteins, together with a battery of components including sequence-specific DNA binding/accessory factors, chromatin remodeling factors, signaling pathway intermediates, noncoding small RNAs, and RNA interference machinery, generally define a dynamic cellular identity through tight regulation of specific gene expression patterns. Epigenetic modification of chromatin structure that results in expression silencing of specific genes is now emerging as an important molecular mechanism in this process. In embryonic stem (ES) cells and adult stem cells, such specific genes represent those associated with differentiation and development, and silencing of these genes in a PcG protein-dependent manner confers stemness. ES cells also contain novel chromatin motifs enriched in epigenetic modifications associated with both activation and repression of genes, suggesting that certain genes are poised for activation or repression. Interestingly, these chromatin domains are highly coincident with the promoters of developmental regulators, which are also found to be occupied by PcG proteins. The epigenetic integrity is compromised, however, by mutations or other alterations that affect the function of PcG proteins in stem cells leading to aberrant cell proliferation and tissue transformation, a hallmark of cancer. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Vinagolu K Rajasekhar
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Rockefeller Research Laboratories, Room #945, New York, New York 10021, USA.
| | | |
Collapse
|