251
|
Phosphorylation-dependent interaction of SATB1 and PIAS1 directs SUMO-regulated caspase cleavage of SATB1. Mol Cell Biol 2010; 30:2823-36. [PMID: 20351170 DOI: 10.1128/mcb.01603-09] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Special AT-rich sequence-binding protein 1 (SATB1) is a tissue-restricted genome organizer that provides a key link between DNA loop organization, chromatin modification/remodeling, and transcription factor association at matrix attachment regions (MARs). The SUMO E3 ligase PIAS1 enhances SUMO conjugation to SATB1 lysine-744, and this modification regulates caspase-6 mediated cleavage of SATB1 at promyelocytic leukemia nuclear bodies (PML NBs). Since this regulated caspase cleavage occurs on only a subset of SATB1, and the products are relatively stable, proteolysis likely mediates cellular processes other than programmed cell death. However, the mechanism for the spatial and temporal regulation of SATB1 sumoylation and caspase cleavage is not known. Here we report that these processes are controlled by SATB1 phosphorylation; specifically, PIAS1 interaction with SATB1 is inhibited by phosphorylation. Mutagenesis studies identified interaction of the PIAS SAP (scaffold attachment factor-A/B/acinus/PIAS) motif with SATB1 N-terminal sequences. Notably, phosphorylation of SATB1 at threonine-188 regulates its interaction with PIAS1. Sequences near this phosphorylation site, LXXLL (residues 193 to 197), appear to be conserved among a subset of SUMO substrate proteins. Thus, this motif may be commonly involved in interaction with the PIAS SAP, and phosphorylation may similarly inhibit some of these substrates by preventing their interaction with the ligase.
Collapse
|
252
|
Abstract
Epigenetic regulation is important for stable maintenance of cell identity. For continued function of organs and tissues, illegitimate changes in cell identity must be avoided. Failure to do so can trigger tumour development and disease. How epigenetic patterns are established during cell differentiation has been explored by studying model systems such as X inactivation. Mammals balance the X-linked gene dosage between the sexes by silencing of one of the two X chromosomes in females. This is initiated by expression of the non-coding X-inactive specific transcript (Xist) RNA and depends on specific cellular contexts, in which essential silencing factors are expressed. Normally X inactivation is initiated in early embryogenesis, but recent reports identified instances where Xist is expressed and can initiate gene repression. Here we describe the features that characterize the cellular permissivity to initiation of X inactivation and note that these can also occur in cancer cells and in specific haematopoietic progenitors. We propose that embryonic pathways for epigenetic regulation are re-established in adult progenitor cells and tumour cells. Understanding their reactivation will deepen our understanding of tumourigenesis and may be exploited for cancer therapy.
Collapse
Affiliation(s)
- Ruben Agrelo
- Research Institute of Molecular Pathology, Vienna, Austria.
| | | |
Collapse
|
253
|
Wang TY, Han ZM, Chai YR, Zhang JH. A mini review of MAR-binding proteins. Mol Biol Rep 2010; 37:3553-60. [PMID: 20174991 DOI: 10.1007/s11033-010-0003-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Accepted: 02/08/2010] [Indexed: 02/08/2023]
Abstract
Genomic DNA encompasses several levels of organization, the nuclear matrix mediates the formation of DNA loop domains that are anchored to matrix attachment regions (MARs). By means of specific interaction with MAR binding proteins (MARBPs), MAR plays an important regulation role in enhancing transgene expression, decreasing expression variation among individuals of different transformants and serving as the replication origin. Through these years, some MARBPs have been identified and characterized from humans, plants, animals and algae so far and the list is growing. Most of MARBPs exist in a co-repressor/co-activator complex and involve in chromosome folding, regulation of gene expression, influencing cell development and inducing cell apoptosis. This review covers recent advances that have contributed to our understanding of MARBPs.
Collapse
Affiliation(s)
- Tian-Yun Wang
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Jinsui Road, Xinxiang, Henan, 453003, People's Republic of China.
| | | | | | | |
Collapse
|
254
|
Hammerich-Hille S, Kaipparettu BA, Tsimelzon A, Creighton CJ, Jiang S, Polo JM, Melnick A, Meyer R, Oesterreich S. SAFB1 mediates repression of immune regulators and apoptotic genes in breast cancer cells. J Biol Chem 2010; 285:3608-3616. [PMID: 19901029 PMCID: PMC2823501 DOI: 10.1074/jbc.m109.066431] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 11/04/2009] [Indexed: 12/18/2022] Open
Abstract
The scaffold attachment factors SAFB1 and SAFB2 are paralogs, which are involved in cell cycle regulation, apoptosis, differentiation, and stress response. They have been shown to function as estrogen receptor corepressors, and there is evidence for a role in breast tumorigenesis. To identify their endogenous target genes in MCF-7 breast cancer cells, we utilized a combined approach of chromatin immunoprecipitation (ChIP)-on-chip and gene expression array studies. By performing ChIP-on-chip on microarrays containing 24,000 promoters, we identified 541 SAFB1/SAFB2-binding sites in promoters of known genes, with significant enrichment on chromosomes 1 and 6. Gene expression analysis revealed that the majority of target genes were induced in the absence of SAFB1 or SAFB2 and less were repressed. Interestingly, there was no significant overlap between the genes identified by ChIP-on-chip and gene expression array analysis, suggesting regulation through regions outside the proximal promoters. In contrast to SAFB2, which shared most of its target genes with SAFB1, SAFB1 had many unique target genes, most of them involved in the regulation of the immune system. A subsequent analysis of the estrogen treatment group revealed that 12% of estrogen-regulated genes were dependent on SAFB1, with the majority being estrogen-repressed genes. These were primarily genes involved in apoptosis, such as BBC3, NEDD9, and OPG. Thus, this study confirms the primary role of SAFB1/SAFB2 as corepressors and also uncovers a previously unknown role for SAFB1 in the regulation of immune genes and in estrogen-mediated repression of genes.
Collapse
Affiliation(s)
- Stephanie Hammerich-Hille
- From the Lester and Sue Smith Breast Center, Department of Medicine and Molecular and Cellular Biology, Texas Children's Cancer Center, Houston, Texas 77030
| | - Benny A Kaipparettu
- From the Lester and Sue Smith Breast Center, Department of Medicine and Molecular and Cellular Biology, Texas Children's Cancer Center, Houston, Texas 77030
| | - Anna Tsimelzon
- From the Lester and Sue Smith Breast Center, Department of Medicine and Molecular and Cellular Biology, Texas Children's Cancer Center, Houston, Texas 77030
| | - Chad J Creighton
- From the Lester and Sue Smith Breast Center, Department of Medicine and Molecular and Cellular Biology, Texas Children's Cancer Center, Houston, Texas 77030
| | - Shiming Jiang
- From the Lester and Sue Smith Breast Center, Department of Medicine and Molecular and Cellular Biology, Texas Children's Cancer Center, Houston, Texas 77030
| | - Jose M Polo
- the Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Ari Melnick
- the Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Rene Meyer
- Department of Pediatric Hematology/Oncology, Texas Children's Cancer Center, Houston, Texas 77030 and
| | - Steffi Oesterreich
- From the Lester and Sue Smith Breast Center, Department of Medicine and Molecular and Cellular Biology, Texas Children's Cancer Center, Houston, Texas 77030; Department of Pediatric Hematology/Oncology, Texas Children's Cancer Center, Houston, Texas 77030 and.
| |
Collapse
|
255
|
Notani D, Gottimukkala KP, Jayani RS, Limaye AS, Damle MV, Mehta S, Purbey PK, Joseph J, Galande S. Global regulator SATB1 recruits beta-catenin and regulates T(H)2 differentiation in Wnt-dependent manner. PLoS Biol 2010; 8:e1000296. [PMID: 20126258 PMCID: PMC2811152 DOI: 10.1371/journal.pbio.1000296] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Accepted: 12/16/2009] [Indexed: 12/24/2022] Open
Abstract
Chromatin organizer SATB1 and Wnt transducer β-catenin form a complex and regulate expression of GATA3 and TH2 cytokines in Wnt-dependent manner and orchestrate TH2 lineage commitment. In vertebrates, the conserved Wnt signalling cascade promotes the stabilization and nuclear accumulation of β-catenin, which then associates with the lymphoid enhancer factor/T cell factor proteins (LEF/TCFs) to activate target genes. Wnt/β -catenin signalling is essential for T cell development and differentiation. Here we show that special AT-rich binding protein 1 (SATB1), the T lineage-enriched chromatin organizer and global regulator, interacts with β-catenin and recruits it to SATB1's genomic binding sites. Gene expression profiling revealed that the genes repressed by SATB1 are upregulated upon Wnt signalling. Competition between SATB1 and TCF affects the transcription of TCF-regulated genes upon β-catenin signalling. GATA-3 is a T helper type 2 (TH2) specific transcription factor that regulates production of TH2 cytokines and functions as TH2 lineage determinant. SATB1 positively regulated GATA-3 and siRNA-mediated knockdown of SATB1 downregulated GATA-3 expression in differentiating human CD4+ T cells, suggesting that SATB1 influences TH2 lineage commitment by reprogramming gene expression. In the presence of Dickkopf 1 (Dkk1), an inhibitor of Wnt signalling, GATA-3 is downregulated and the expression of signature TH2 cytokines such as IL-4, IL-10, and IL-13 is reduced, indicating that Wnt signalling is essential for TH2 differentiation. Knockdown of β-catenin also produced similar results, confirming the role of Wnt/β-catenin signalling in TH2 differentiation. Furthermore, chromatin immunoprecipitation analysis revealed that SATB1 recruits β-catenin and p300 acetyltransferase on GATA-3 promoter in differentiating TH2 cells in a Wnt-dependent manner. SATB1 coordinates TH2 lineage commitment by reprogramming gene expression. The SATB1:β-catenin complex activates a number of SATB1 regulated genes, and hence this study has potential to find novel Wnt responsive genes. These results demonstrate that SATB1 orchestrates TH2 lineage commitment by mediating Wnt/β-catenin signalling. This report identifies a new global transcription factor involved in β-catenin signalling that may play a major role in dictating the functional outcomes of this signalling pathway during development, differentiation, and tumorigenesis. In vertebrates the canonical Wnt signalling culminates in β-catenin moving into the nucleus where it activates transcription of target genes. Wnt/β-catenin signalling is essential for the thymic maturation and differentiation of naïve T cells. Here we show that SATB1, a T cell lineage-enriched chromatin organizer and global regulator, binds to β-catenin and recruits it to SATB1's genomic binding sites so that genes formerly repressed by SATB1 are upregulated by Wnt signalling. Some of the genes known to be regulated by SATB1 (such as genes encoding cytokines and the transcription factor GATA3) are required for differentiation of Th2 cells, an important subset of helper T cells. Specifically we show that siRNA-mediated knockdown of SATB1 downregulated GATA-3 expression in differentiating human CD4+ T cells. Inhibiting Wnt signalling led to downregulation of GATA-3 and of signature TH2 cytokines such as IL-4, IL-10, and IL-13. Knockdown of β-catenin also produced similar results, thus together these data confirm the role of Wnt/β-catenin signalling in TH2 differentiation. Our data demonstrate that SATB1 orchestrates TH2 lineage commitment by modulating Wnt/β-catenin signalling.
Collapse
Affiliation(s)
- Dimple Notani
- National Centre for Cell Science, Ganeshkhind, Pune, India
| | | | | | | | | | - Sameet Mehta
- National Centre for Cell Science, Ganeshkhind, Pune, India
- Centre for Modelling and Simulation, University of Pune, Ganeshkhind, Pune, India
| | | | - Jomon Joseph
- National Centre for Cell Science, Ganeshkhind, Pune, India
| | - Sanjeev Galande
- National Centre for Cell Science, Ganeshkhind, Pune, India
- * E-mail:
| |
Collapse
|
256
|
Notani D, Limaye AS, Kumar PP, Galande S. Phosphorylation-dependent regulation of SATB1, the higher-order chromatin organizer and global gene regulator. Methods Mol Biol 2010; 647:317-335. [PMID: 20694677 DOI: 10.1007/978-1-60761-738-9_20] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
The chromatin organizer SATB1 regulates distant genes by selectively tethering matrix attachment regions (MARs) to the nuclear matrix. Post-translational modifications (PTMs) are important regulators of functional activities of proteins. Recently, a phosphorylation-dependent molecular switch that provided insights into the molecular mechanism of transcriptional repression and activation by SATB1 was discovered. SATB1 is specifically phosphorylated by protein kinase C (PKC) at serine 185 in vivo, and this modification leads to repression of transcription by SATB1 via increased association with the histone deacetylase 1 (HDAC1) corepressor. In this chapter, we describe methods for overexpression and purification of full length SATB1 protein and for its in vitro phosphorylation. We also describe method for in vivo phosphorylation of SATB1 upon immunoprecipitation using anti-SATB1. Finally, we describe a functional assay to monitor the effect of phosphorylation on transcription activity of SATB1 in vivo using MAR-linked reporter assay, in the presence and absence of PKC inhibitors.
Collapse
Affiliation(s)
- Dimple Notani
- National Centre for Cell Science, Ganeshkhind, Maharashtra, India
| | | | | | | |
Collapse
|
257
|
Li QQ, Chen ZQ, Xu JD, Cao XX, Chen Q, Liu XP, Xu ZD. Overexpression and involvement of special AT-rich sequence binding protein 1 in multidrug resistance in human breast carcinoma cells. Cancer Sci 2010; 101:80-6. [PMID: 19860849 PMCID: PMC11159454 DOI: 10.1111/j.1349-7006.2009.01372.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Special AT-rich sequence binding protein (SATB) 1 has been proposed to act as a determinant for the acquisition of metastatic activity by controlling expression of a specific set of genes that promote metastatic activity. Here we found that SATB1 expression is upregulated in multidrug-resistant breast cancer cells that exhibit higher invasive potential than the parental cells. Apart from accelerating metastasis and inducing epithelial-mesenchymal transition, SATB1 was demonstrated to confer resistance to both P-glycoprotein-related and P-glycoprotein-non-related drugs on MCF7 cells, which was accompanied by decreasing accumulation of adriamycin in SATB1-overexpressing transfectants. SATB1 depletion could partially reverse the multidrug resistance (MDR) phenotype of MCF7/ADR in vitro and in vivo. The SATB1-induced P-glycoprotein-mediated MDR could be reversed by treatment with anti-P-glycoprotein mAb. Moreover, SATB1 plays an important role in anti-apoptotic activity in MCF7/ADR cells in response to adriamycin treatment, which suggests another mechanism contributing to SATB1-related MDR of breast cancers. These data provide new insights into the mode by which breast tumors acquire the MDR phenotype and also imply a role for SATB1 in this process.
Collapse
Affiliation(s)
- Qing-Quan Li
- Department of Pathology, Shanghai Medical College, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
258
|
Modulation of chromatin boundary activities by nucleosome-remodeling activities in Drosophila melanogaster. Mol Cell Biol 2009; 30:1067-76. [PMID: 19995906 DOI: 10.1128/mcb.00183-09] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Chromatin boundaries facilitate independent gene regulation by insulating genes from the effects of enhancers or organized chromatin. However, the mechanisms of boundary action are not well understood. To investigate whether boundary function depends on a higher order of chromatin organization, we examined the function of several Drosophila melanogaster insulators in cells with reduced chromatin-remodeling activities. We found that knockdown of NURF301 and ISWI, key components of the nucleosome-remodeling factor (NURF), synergistically disrupted the enhancer-blocking function of Fab7 and SF1 and augmented the function of Fab8. Mutations in Nurf301/Ebx and Iswi also affected the function of these boundaries in vivo. We further show that ISWI was localized on the endogenous Fab7 and Fab8 insulators and that NURF knockdown resulted in a marked increase in the nucleosome occupancy at these insulator sites. In contrast to the effect of NURF knockdown, reduction in dMi-2, the ATPase component of the Drosophila nucleosome-remodeling and deacetylation (NuRD) complex, augmented Fab7 and suppressed Fab8. Our results provide the first evidence that higher-order chromatin organization influences the enhancer-blocking activity of chromatin boundaries. In particular, the NURF and NuRD nucleosome-remodeling complexes may regulate Hox expression by modulating the function of boundaries in these complexes. The unique responses by different classes of boundaries to changes in the chromatin environment may be indicative of their distinct mechanisms of action, which may influence their placement in the genome and selection during evolution.
Collapse
|
259
|
Savarese F, Dávila A, Nechanitzky R, De La Rosa-Velazquez I, Pereira CF, Engelke R, Takahashi K, Jenuwein T, Kohwi-Shigematsu T, Fisher AG, Grosschedl R. Satb1 and Satb2 regulate embryonic stem cell differentiation and Nanog expression. Genes Dev 2009; 23:2625-38. [PMID: 19933152 DOI: 10.1101/gad.1815709] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Satb1 and the closely related Satb2 proteins regulate gene expression and higher-order chromatin structure of multigene clusters in vivo. In examining the role of Satb proteins in murine embryonic stem (ES) cells, we find that Satb1(-/-) cells display an impaired differentiation potential and augmented expression of the pluripotency determinants Nanog, Klf4, and Tbx3. Metastable states of self-renewal and differentiation competence have been attributed to heterogeneity of ES cells in the expression of Nanog. Satb1(-/-) cultures have a higher proportion of Nanog(high) cells, and an increased potential to reprogram human B lymphocytes in cell fusion experiments. Moreover, Satb1-deficient ES cells show an increased expression of Satb2, and we find that forced Satb2 expression in wild-type ES cells antagonizes differentiation-associated silencing of Nanog and enhances the induction of NANOG in cell fusions with human B lymphocytes. An antagonistic function of Satb1 and Satb2 is also supported by the almost normal differentiation potential of Satb1(-/-)Satb2(-/-) ES cells. Taken together with the finding that both Satb1 and Satb2 bind the Nanog locus in vivo, our data suggest that the balance of Satb1 and Satb2 contributes to the plasticity of Nanog expression and ES cell pluripotency.
Collapse
Affiliation(s)
- Fabio Savarese
- Max Planck Institute of Immunobiology, Department of Cellular and Molecular Immunology, 79108 Freiburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
260
|
Ng KH, Yu H, Ito T. AGAMOUS controls GIANT KILLER, a multifunctional chromatin modifier in reproductive organ patterning and differentiation. PLoS Biol 2009; 7:e1000251. [PMID: 19956801 PMCID: PMC2774341 DOI: 10.1371/journal.pbio.1000251] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Accepted: 10/16/2009] [Indexed: 11/18/2022] Open
Abstract
The floral, homeotic protein AGAMOUS coordinates multiple downstream genes through direct transcriptional regulation of the nuclear matrix attachment region binding protein GIANT KILLER. The Arabidopsis homeotic protein AGAMOUS (AG), a MADS domain transcription factor, specifies reproductive organ identity during flower development. Using a binding assay and expression analysis, we identified a direct target of AG, GIANT KILLER (GIK), which fine-tunes the expression of multiple genes downstream of AG. The GIK protein contains an AT-hook DNA binding motif that is widely found in chromosomal proteins and that binds to nuclear matrix attachment regions of DNA elements. Overexpression and loss of function of GIK cause wide-ranging defects in patterning and differentiation of reproductive organs. GIK directly regulates the expression of several key transcriptional regulators, including ETTIN/AUXIN RESPONSE FACTOR 3 (ETT/ARF3) that patterns the gynoecium, by binding to the matrix attachment regions of target promoters. Overexpression of GIK causes a swift and dynamic change in repressive histone modification in the ETT promoter. We propose that GIK acts as a molecular node downstream of the homeotic protein AG, regulating patterning and differentiation of reproductive organs through chromatin organization. Multicellular development depends on proper expression of thousands of genes. Master regulators, such as homeotic proteins, code for transcription factors in both plants and animals and are thought to act by regulating other genes. Recent genomic studies in the plant Arabidopsis have shown that over 1,000 genes are regulated by homeotic proteins that directly control various target genes, including different classes of transcriptional regulators. It is not known, however, how expression of so many genes is coordinated by a single homeotic gene to form functional organs and tissues. Here we identified a transcriptional target of the plant homeotic protein AGAMOUS using bioinformatics analysis and showed that AGAMOUS directly controls GIANT KILLER, a multifunctional chromatin modifier. GIANT KILLER then binds to the upstream regions of multiple genes involved in patterning and differentiation in the AGAMOUS pathway and fine-tunes the expression of these genes. These data therefore provide a possible mechanism by which a homeotic gene coordinates multiple downstream targets in plants.
Collapse
Affiliation(s)
- Kian-Hong Ng
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore
| | - Hao Yu
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore
| | - Toshiro Ito
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore
- PRESTO, Japan Science and Technology Agency, Saitama, Japan
- * E-mail:
| |
Collapse
|
261
|
Wang S, Zhou J, Wang XY, Hao JM, Chen JZ, Zhang XM, Jin H, Liu L, Zhang YF, Liu J, Ding YQ, Li JM. Down-regulated expression of SATB2 is associated with metastasis and poor prognosis in colorectal cancer. J Pathol 2009; 219:114-22. [PMID: 19557828 DOI: 10.1002/path.2575] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
To identify novel biomarkers of metastasis of colorectal cancer (CRC), we developed an orthotopic implantation model of murine CRC and selected in vivo M5, a subclone of the SW480 CRC cell line with enhanced potential for metastasis to the liver. We compared the differences in the gene expression profiles between M5 and SW480 cells using gene expression profiling. We found that expression of special AT-rich sequence-binding protein 2 (SATB2) was down-regulated in M5 cells. Immunohistochemical analysis of 146 colorectal tumour samples showed that underexpression of SATB2 was strongly correlated with poor prognosis, tumour invasion, lymph node metastasis, distant metastasis, and Dukes' classification for CRC. Univariate and multivariate survival analyses further showed that SATB2 expression was a potential favourable prognostic factor for CRC. These results demonstrated not only that SATB2 is a potential novel prognostic factor for CRC, but also that selection of a highly metastatic clone of SW480 in vivo coupled with gene expression profiling is a powerful approach to identifying prognostic markers for CRC.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
262
|
Patani N, Jiang W, Mansel R, Newbold R, Mokbel K. The mRNA expression of SATB1 and SATB2 in human breast cancer. Cancer Cell Int 2009; 9:18. [PMID: 19642980 PMCID: PMC2731048 DOI: 10.1186/1475-2867-9-18] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 07/30/2009] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND SATB1 is a nuclear protein that has been recently reported to be a 'genome organizer' which delineates specific epigenetic modifications at target gene loci, directly up-regulating metastasis-associated genes while down-regulating tumor-suppressor genes. In this study, the level of mRNA expression of SATB1 and SATB2 were assessed in normal and malignant breast tissue in a cohort of women with breast cancer and correlated to conventional clinico-pathological parameters. MATERIALS AND METHODS Breast cancer tissues (n = 115) and normal background tissues (n = 31) were collected immediately after excision during surgery. Following RNA extraction, reverse transcription was carried out and transcript levels were determined using real-time quantitative PCR and normalized against beta-actin expression. Transcript levels within the breast cancer specimens were compared to the normal background tissues and analyzed against TNM stage, nodal involvement, tumour grade and clinical outcome over a 10 year follow-up period. RESULTS The levels of SATB1 were higher in malignant compared with normal breast tissue (p = 0.0167). SATB1 expression increased with increasing TNM stage (TNM1 vs. TNM2 p = 0.0264), increasing tumour grade (grade1 vs. grade 3 p = 0.017; grade 2 vs. grade 3 p = 0.0437; grade 1 vs. grade 2&3 p = 0.021) and Nottingham Prognostic Index (NPI) (NPI-1 vs. NPI-3 p = 0.0614; NPI-2 vs. NPI-3 p = 0.0495). Transcript levels were associated with oestrogen receptor (ER) positivity (ER(-) vs. ER(+) p = 0.046). SABT1 expression was also significantly correlated with downstream regulated genes IL-4 and MAF-1 (Pearson's correlation coefficient r = 0.21 and r = 0.162) and SATB2 (r = 0.506). After a median follow up of 10 years, there was a trend for higher SATB1 expression to be associated with shorter overall survival (OS). Higher levels of SATB2 were also found in malignant compared to background tissue (p = 0.049). SATB2 expression increased with increasing tumour grade (grade 1 vs. grade 3 p = 0.035). SATB2 was associated with ER positivity (ER(-) vs. ER(+) p = 0.0283) within ductal carcinomas. Higher transcript levels showed a significant association with poorer OS (p = 0.0433). CONCLUSION SATB1 mRNA expression is significantly associated with poor prognostic parameters in breast cancer, including increasing tumour grade, TNM stage and NPI. SATB2 mRNA expression is significantly associated with increasing tumour grade and poorer OS. These results are consistent with the notion that SATB1 acts as a 'master genome organizer' in human breast carcinogenesis.
Collapse
Affiliation(s)
- Neill Patani
- Department of Breast Surgery, St. George's University of London, London, UK
- The London Breast Institute, The Princess Grace Hospital, London, UK
| | - Wen Jiang
- Metastasis and Angiogenesis Research Group, University Department of Surgery, Cardiff University, Cardiff, UK
| | - Robert Mansel
- Metastasis and Angiogenesis Research Group, University Department of Surgery, Cardiff University, Cardiff, UK
| | - Robert Newbold
- The Brunel Institute of Cancer Genetics and Pharmacogenomics, London, UK
| | - Kefah Mokbel
- Department of Breast Surgery, St. George's University of London, London, UK
- The London Breast Institute, The Princess Grace Hospital, London, UK
- The Brunel Institute of Cancer Genetics and Pharmacogenomics, London, UK
| |
Collapse
|
263
|
Abstract
The packaging of chromosomal DNA by nucleosomes condenses and organizes the genome, but occludes many regulatory DNA elements. However, this constraint also allows nucleosomes and other chromatin components to actively participate in the regulation of transcription, chromosome segregation, DNA replication, and DNA repair. To enable dynamic access to packaged DNA and to tailor nucleosome composition in chromosomal regions, cells have evolved a set of specialized chromatin remodeling complexes (remodelers). Remodelers use the energy of ATP hydrolysis to move, destabilize, eject, or restructure nucleosomes. Here, we address many aspects of remodeler biology: their targeting, mechanism, regulation, shared and unique properties, and specialization for particular biological processes. We also address roles for remodelers in development, cancer, and human syndromes.
Collapse
Affiliation(s)
- Cedric R Clapier
- Howard Hughes Medical Institute, Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| | | |
Collapse
|
264
|
Expression Vector Engineering for Recombinant Protein Production. ACTA ACUST UNITED AC 2009. [DOI: 10.1007/978-90-481-2245-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
265
|
|
266
|
Zhang J, Lu L, Ji L, Yang G, Zheng C. Functional characterization of a tobacco matrix attachment region-mediated enhancement of transgene expression. Transgenic Res 2009; 18:377-85. [PMID: 19043795 DOI: 10.1007/s11248-008-9230-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Accepted: 11/10/2008] [Indexed: 10/21/2022]
Abstract
TM2, a new matrix attachment region (MAR) isolated from tobacco, increases transgene expression in plants. We have carried out a more detailed analysis of the DNA elements in TM2 with the aim of improving its effect on transcription activation. Our study of the location effect of individual MARs on the expression of the adjacent 35S:gusA cassette indicated that the TM2 functions in a bidirectional manner, with the 5'-MAR being more efficient in enhancing beta-glucuronidase expression than the 3'-MAR. The influence of 5'-MAR on different linked mini-promoters in transgenic tobacco cells suggested that the role of TM2 depends on the basic expression of the transgenes. Deletion analysis of one topo II site and two unwinding sites together with one T-box revealed that all these sites contribute most (93.3%) of the transcription activation mediated from the TM2 sequence. Additionally, micrococcal nuclease accessibility of the 35S promoter region can be strengthened by linked TM2, suggesting that the TM2 mediates the spreading of nucleosome opening. Taken together, our results reveal that the TM2 mediates a more open and accessible chromatin DNA structure for promoter-dependent active transcription, which in turn enhances transgene expression.
Collapse
Affiliation(s)
- Jiedao Zhang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Shandong, People's Republic of China
| | | | | | | | | |
Collapse
|
267
|
Cruickshank MN, Fenwick E, Karimi M, Abraham LJ, Ulgiati D. Cell- and stage-specific chromatin structure across the Complement receptor 2 (CR2/CD21) promoter coincide with CBF1 and C/EBP-beta binding in B cells. Mol Immunol 2009; 46:2613-22. [PMID: 19487031 DOI: 10.1016/j.molimm.2009.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2009] [Revised: 05/01/2009] [Accepted: 05/02/2009] [Indexed: 01/19/2023]
Abstract
Stringent developmental transcription requires multiple transcription factor (TF) binding sites, cell-specific expression of signaling molecules, TFs and co-regulators and appropriate chromatin structure. During B-lymphopoiesis, human Complement receptor 2 (CR2/CD21) is detected on immature and mature B cells but not on B cell precursors and plasma cells. We examined cell- and stage-specific human CR2 gene regulation using cell lines modeling B-lymphopoiesis. Chromatin accessibility assays revealed a region between -409 and -262 with enhanced accessibility in mature B cells and pre-B cells, compared to either non-lymphoid or plasma cell-types, however, accessibility near the transcription start site (TSS) was elevated only in CR2-expressing B cells. A correlation between histone acetylation and CR2 expression was observed, while histone H3K4 dimethylation was enriched near the TSS in both CR2-expressing B cells and non-expressing pre-B cells. Candidate sites within the CR2 promoter were identified which could regulate chromatin, including a matrix attachment region associated with CDP, SATB1/BRIGHT and CEBP-beta sites as well as two CBF1 sites. ChIP assays verified that both CBF1 and C/EBP-beta bind the CR2 promoter in B cells raising the possibility that these factors facilitate or respond to alterations in chromatin structure to control the timing and/or level of CR2 transcription.
Collapse
|
268
|
Gautier VW, Gu L, O'Donoghue N, Pennington S, Sheehy N, Hall WW. In vitro nuclear interactome of the HIV-1 Tat protein. Retrovirology 2009; 6:47. [PMID: 19454010 PMCID: PMC2702331 DOI: 10.1186/1742-4690-6-47] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Accepted: 05/19/2009] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND One facet of the complexity underlying the biology of HIV-1 resides not only in its limited number of viral proteins, but in the extensive repertoire of cellular proteins they interact with and their higher-order assembly. HIV-1 encodes the regulatory protein Tat (86-101aa), which is essential for HIV-1 replication and primarily orchestrates HIV-1 provirus transcriptional regulation. Previous studies have demonstrated that Tat function is highly dependent on specific interactions with a range of cellular proteins. However they can only partially account for the intricate molecular mechanisms underlying the dynamics of proviral gene expression. To obtain a comprehensive nuclear interaction map of Tat in T-cells, we have designed a proteomic strategy based on affinity chromatography coupled with mass spectrometry. RESULTS Our approach resulted in the identification of a total of 183 candidates as Tat nuclear partners, 90% of which have not been previously characterised. Subsequently we applied in silico analysis, to validate and characterise our dataset which revealed that the Tat nuclear interactome exhibits unique signature(s). First, motif composition analysis highlighted that our dataset is enriched for domains mediating protein, RNA and DNA interactions, and helicase and ATPase activities. Secondly, functional classification and network reconstruction clearly depicted Tat as a polyvalent protein adaptor and positioned Tat at the nexus of a densely interconnected interaction network involved in a range of biological processes which included gene expression regulation, RNA biogenesis, chromatin structure, chromosome organisation, DNA replication and nuclear architecture. CONCLUSION We have completed the in vitro Tat nuclear interactome and have highlighted its modular network properties and particularly those involved in the coordination of gene expression by Tat. Ultimately, the highly specialised set of molecular interactions identified will provide a framework to further advance our understanding of the mechanisms of HIV-1 proviral gene silencing and activation.
Collapse
Affiliation(s)
- Virginie W Gautier
- UCD-Centre for Research in Infectious Diseases, School of Medicine and Medical Science, University College Dublin (UCD), Belfield, Dublin 4, Ireland.
| | | | | | | | | | | |
Collapse
|
269
|
Silencing of host cell CYBB gene expression by the nuclear effector AnkA of the intracellular pathogen Anaplasma phagocytophilum. Infect Immun 2009; 77:2385-91. [PMID: 19307214 DOI: 10.1128/iai.00023-09] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Coevolution of intracellular bacterial pathogens and their host cells resulted in the appearance of effector molecules that when translocated into the host cell modulate its function, facilitating bacterial survival within the hostile host environment. Some of these effectors interact with host chromatin and other nuclear components. In this report, we show that the AnkA protein of Anaplasma phagocytophilum, which is translocated into the host cell nucleus, interacts with gene regulatory regions of host chromatin and is involved in downregulating expression of CYBB (gp91(phox)) and other key host defense genes. AnkA effector protein rapidly accumulated in nuclei of infected cells coincident with changes in CYBB transcription. AnkA interacted with transcriptional regulatory regions of the CYBB locus at sites where transcriptional regulators bind. AnkA binding to DNA occurred at regions with high AT contents. Mutation of AT stretches at these sites abrogated AnkA binding. Histone H3 acetylation decreased dramatically at the CYBB locus during A. phagocytophilum infection, particularly around AnkA binding sites. Transcription of CYBB and other defense genes was significantly decreased in AnkA-transfected HL-60 cells. These data suggest a mechanism by which intracellular pathogens directly regulate host cell gene expression mediated by nuclear effectors and changes in host chromatin structure.
Collapse
|
270
|
Kim SI, Bultman SJ, Kiefer CM, Dean A, Bresnick EH. BRG1 requirement for long-range interaction of a locus control region with a downstream promoter. Proc Natl Acad Sci U S A 2009; 106:2259-2264. [PMID: 19171905 PMCID: PMC2650142 DOI: 10.1073/pnas.0806420106] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Indexed: 11/18/2022] Open
Abstract
The dynamic packaging of DNA into chromatin is a fundamental step in the control of diverse nuclear processes. Whereas certain transcription factors and chromosomal components promote the formation of higher-order chromatin loops, the co-regulator machinery mediating loop assembly and disassembly is unknown. Using mice bearing a hypomorphic allele of the BRG1 chromatin remodeler, we demonstrate that the Brg1 mutation abrogated a cell type-specific loop between the beta-globin locus control region and the downstream beta major promoter, despite trans-acting factor occupancy at both sites. By contrast, distinct loops were insensitive to the Brg1 mutation. Molecular analysis with a conditional allele of GATA-1, a key regulator of hematopoiesis, in a novel cell-based system provided additional evidence that BRG1 functions early in chromatin domain activation to mediate looping. Although the paradigm in which chromatin remodelers induce nucleosome structural transitions is well established, our results demonstrating an essential role of BRG1 in the genesis of specific chromatin loops expands the repertoire of their functions.
Collapse
Affiliation(s)
- Shin-Il Kim
- Department of Pharmacology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706
| | - Scott J. Bultman
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599; and
| | - Christine M. Kiefer
- Laboratory of Cellular and Developmental Biology, National Institutes of Diabetes and Digestive and Kidney Disorders, National Institutes of Health, Bethesda, MD 20892
| | - Ann Dean
- Laboratory of Cellular and Developmental Biology, National Institutes of Diabetes and Digestive and Kidney Disorders, National Institutes of Health, Bethesda, MD 20892
| | - Emery H. Bresnick
- Department of Pharmacology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706
| |
Collapse
|
271
|
Acetylation-dependent interaction of SATB1 and CtBP1 mediates transcriptional repression by SATB1. Mol Cell Biol 2008; 29:1321-37. [PMID: 19103759 DOI: 10.1128/mcb.00822-08] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Special AT-rich binding protein 1 (SATB1) acts as a global regulator of gene expression by recruiting various corepressor or coactivator complexes, thereby establishing a unique chromatin structure at its genomic targets in a context-dependent manner. Although SATB1 acts predominantly as a repressor via recruitment of histone deacetylase 1 (HDAC1) complexes, the precise mechanism of global repression is not clear. Here we report that SATB1 and C-terminal binding protein 1 (CtBP1) form a repressor complex in vivo. The interaction occurs via the CtBP1 interaction consensus motif PVPLS within the PDZ-like domain of SATB1. The acetylation of SATB1 upon LiCl and ionomycin treatments disrupts its association with CtBP1, resulting in enhanced target gene expression. Chromatin immunoprecipitation analysis indicated that the occupancy of CtBP1 and HDAC1 is gradually decreased and the occupancy of PCAF is elevated at the SATB1 binding sites within the human interleukin-2 and mouse c-Myc promoters. Moreover, gene expression profiling studies using cells in which expression of SATB1 and CtBP1 was silenced indicated commonly targeted genes that may be coordinately repressed by the SATB1-CtBP1 complex. Collectively, these results provide a mechanistic insight into the role of SATB1-CtBP1 interaction in the repression and derepression of SATB1 target genes during Wnt signaling in T cells.
Collapse
|
272
|
Singh K, Sinha S, Malonia SK, Bist P, Tergaonkar V, Chattopadhyay S. Tumor suppressor SMAR1 represses IkappaBalpha expression and inhibits p65 transactivation through matrix attachment regions. J Biol Chem 2008; 284:1267-78. [PMID: 18981184 DOI: 10.1074/jbc.m801088200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Aberrant NF-kappaB activity promotes tumorigenesis. However, NF-kappaB also inhibits tumor growth where tumor suppressor pathways remain unaltered. Thus, its role in tumorigenesis depends upon the function of other cellular factors. Tumor suppressor SMAR1 down-modulated in high grade breast cancers is regulated by p53 and is reported to interact and stabilize p53. Because both SMAR1 and NF-kappaB are involved in tumorigenesis, we investigated the effect of SMAR1 upon NF-kappaB activity. We show that SMAR1 induction by doxorubicin or overexpression produces functional NF-kappaB complexes that are competent for binding to NF-kappaB consensus sequence. However, SMAR1 induced p65-p50 complex is phosphorylation- and transactivation-deficient. Induction of functional NF-kappaB complexes stems from down-regulation of IkappaBalpha transcription through direct binding of SMAR1 to the matrix attachment region site present in IkappaBalpha promoter and recruitment of corepressor complex. Real time PCR array for NF-kappaB target genes revealed that SMAR1 down-regulates a subset of NF-kappaB target genes that are involved in tumorigenesis. We also show that SMAR1 inhibits tumor necrosis factor alpha-induced induction of NF-kappaB suggesting that activation of NF-kappaB by SMAR1 is independent and different from classical pathway. Thus, for the first time we report that a tumor suppressor protein SMAR1 can modulate NF-kappaB transactivation and inhibit tumorigenesis by regulating NF-kappaB target genes.
Collapse
Affiliation(s)
- Kamini Singh
- National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune 411007, Maharastra, India
| | | | | | | | | | | |
Collapse
|
273
|
Liu YI, Chang MV, Li HE, Barolo S, Chang JL, Blauwkamp TA, Cadigan KM. The chromatin remodelers ISWI and ACF1 directly repress Wingless transcriptional targets. Dev Biol 2008; 323:41-52. [PMID: 18786525 DOI: 10.1016/j.ydbio.2008.08.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2008] [Revised: 08/01/2008] [Accepted: 08/09/2008] [Indexed: 02/05/2023]
Abstract
The highly conserved Wingless/Wnt signaling pathway controls many developmental processes by regulating the expression of target genes, most often through members of the TCF family of DNA-binding proteins. In the absence of signaling, many of these targets are silenced, by mechanisms involving TCFs that are not fully understood. Here we report that the chromatin remodeling proteins ISWI and ACF1 are required for basal repression of WG target genes in Drosophila. This regulation is not due to global repression by ISWI and ACF1 and is distinct from their previously reported role in chromatin assembly. While ISWI is localized to the same regions of Wingless target gene chromatin as TCF, we find that ACF1 binds much more broadly to target loci. This broad distribution of ACF1 is dependent on ISWI. ISWI and ACF1 are required for TCF binding to chromatin, while a TCF-independent role of ISWI-ACF1 in repression of Wingless targets is also observed. Finally, we show that Wingless signaling reduces ACF1 binding to WG targets, and ISWI and ACF1 regulate repression by antagonizing histone H4 acetylation. Our results argue that WG signaling activates target gene expression partly by overcoming the chromatin barrier maintained by ISWI and ACF1.
Collapse
Affiliation(s)
- Yan I Liu
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109-1048, USA
| | | | | | | | | | | | | |
Collapse
|
274
|
SATB1 is required for CD8 coreceptor reversal. Mol Immunol 2008; 46:207-11. [PMID: 18722016 DOI: 10.1016/j.molimm.2008.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2008] [Accepted: 07/06/2008] [Indexed: 01/19/2023]
Abstract
Intrathymic signals induce the differentiation of immature CD4(+)CD8(+) double positive (DP) thymocytes into mature CD4(+) or CD8(+) single positive (SP) T cells. The transcriptional mechanism by which CD8 lineage is determined is not fully understood. The best evidence, which favors the kinetic signaling/coreceptor reversal model, indicates that signaled DP thymocytes terminate CD8 transcription prior to their subsequent re-initiation of CD8 transcription and ultimate differentiation into CD8SP T cells. We and others have shown that CD8 lineage commitment is severely perturbed in mice in which expression of the transcription factor SATB1 is either conventionally knocked out or T cell-specifically knocked down. Here, we demonstrate that, as with normal thymocytes, cultured SATB1-deficient DP thymocytes inactivate CD8 coreceptor transcription following receipt of signals (PMA plus ionomycin) that mimic TCR-mediated positive selection. However, this terminated CD8 transcription is not re-initiated by signals (IL-7) conducive to CD8 differentiation in SATB1-deficient DP. We show that SATB1 specifically binds to a cis-regulatory element within the CD8 enhancer (E8(III)) known to be required for coreceptor reversal. A requirement in CD8 coreceptor reversal identifies SATB1 as an essential trans-regulator of CD8 lineage fate, whose action may be mediated via recruitment to the E8(III) DP enhancer.
Collapse
|
275
|
Williams H, Jayaraman PS, Gaston K. DNA wrapping and distortion by an oligomeric homeodomain protein. J Mol Biol 2008; 383:10-23. [PMID: 18755198 DOI: 10.1016/j.jmb.2008.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2008] [Revised: 07/04/2008] [Accepted: 08/01/2008] [Indexed: 12/29/2022]
Abstract
Many transcription factors alter DNA or chromatin structure. Changes in chromatin structure are often brought about by the recruitment of chromatin-binding proteins, chromatin-modifying proteins, or other transcription co-activator or co-repressor proteins. However, some transcription factors form oligomeric assemblies that may themselves induce changes in DNA conformation and chromatin structure. The proline-rich homeodomain (PRH/Hex) protein is a transcription factor that regulates cell differentiation and cell proliferation, and has multiple roles in embryonic development. Earlier, we showed that PRH can repress transcription by multiple mechanisms, including the recruitment of co-repressor proteins belonging to the TLE family of chromatin-binding proteins. Our in vivo crosslinking studies have shown that PRH forms oligomeric complexes in cells and a variety of biophysical techniques suggest that the protein forms octamers. However, as yet we have little knowledge of the role played by PRH oligomerisation in the regulation of promoter activity or of the architecture of promoters that are regulated directly by PRH in cells. Here, we compare the binding of PRH and the isolated PRH homeodomain to DNA fragments with single and multiple PRH sites, using gel retardation assays and DNase I and chemical footprinting. We show that the PRH oligomer binds to multiple sites within the human Goosecoid promoter with high affinity and that the binding of PRH brings about DNA distortion. We suggest that PRH octamers wrap DNA in order to bring about transcriptional repression.
Collapse
Affiliation(s)
- Hannah Williams
- Department of Biochemistry, School of Medical Sciences, University of Bristol, Bristol, UK
| | | | | |
Collapse
|
276
|
Gyorgy AB, Szemes M, de Juan Romero C, Tarabykin V, Agoston DV. SATB2 interacts with chromatin-remodeling molecules in differentiating cortical neurons. Eur J Neurosci 2008; 27:865-73. [PMID: 18333962 DOI: 10.1111/j.1460-9568.2008.06061.x] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
During our search for developmental regulators of neuronal differentiation, we identified special AT-rich sequence-binding protein (SATB)2 that is specifically expressed in the developing rat neocortex and binds to AT-rich DNA elements. Here we investigated whether the regulatory function of SATB2 involves chromatin remodeling at the AT-rich DNA site. In-vitro and in-vivo assays using a DNA affinity pre-incubation specificity test of recognition and chromatin immunoprecipitation showed that SATB2 specifically binds to histone deacetylase 1 and metastasis-associated protein 2, members of the nucleosome-remodeling and histone deacetylase complex. Double immunohistochemistry showed that, in the developing rat neocortex, SATB2 is coexpressed with both proteins. Using a cell culture model, we showed that trichostatin A treatment, which blocks the activities of histone deacetylases, reverses the AT-rich dsDNA-dependent repressor effect of SATB2. These findings suggested that the molecular regulatory function of SATB2 involves modification of the chromatin structure. Semi-quantitative chromatin immunoprecipitation analysis of cortices from SATB2 mutant and wild-type animals indicated that, in the knock-out brains, SATB2 is replaced in the chromatin-remodeling complex by AU-rich element RNA binding protein 1, another AT-rich DNA binding protein also expressed in differentiating cortical neurons. These results suggested that an altered chromatin structure, due to the presence of different AT-rich DNA binding proteins in the chromatin-remodeling complex, may contribute to the developmental abnormalities observed in the SATB2 mutant animals. These findings also raised the interesting possibility that SATB2, along with other AT-rich DNA binding proteins, is involved in mediating epigenetic influences during cortical development.
Collapse
Affiliation(s)
- Andrea B Gyorgy
- Department of Anatomy, Physiology and Genetics, School of Medicine USU, Bethesda, MD 20814, USA
| | | | | | | | | |
Collapse
|
277
|
Zheng J. Is SATB1 a Master Regulator in Breast Cancer Growth and Metastasis? WOMENS HEALTH 2008; 4:329-32. [DOI: 10.2217/17455057.4.4.329] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Evaluation of: Han HJ, Russo J, Kohwi Y et al.: SATB1 reprogrammes gene expression to promote breast tumor growth and metastasis. Nature 452(7184), 187–193 (2008). Metastasis is the most common cause of death in cancer patients. However, the genetic mechanisms involved in the master control genes of metastasis remain unclear. In this study, the authors found that special AT-rich sequence-binding protein 1 (SATB1) expression contributed to breast cancer growth and metastasis. SATB1 expression is detected in aggressive breast cancer cells rather than nonaggressive breast cancer cells. Moreover, by introducing the SATB1 gene into nonmetastatic breast cancer cells, invasive tumors can be induced in mice; whereas, silencing of SATB1 in metastatic cells not only abolishes metastasis and tumor growth in mice, but also returns cells to their normal appearance. These effects are related as SATB1 upregulates metastasis-associated genes while downregulating tumor-suppressor genes through epigenetic modification. The research suggests that SATB1 is a master regulator in the metastasis of breast cancer and, therefore, can be considered as an independent prognostic factor and a potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Jie Zheng
- Southeast University, Department of Pathology, School of Medicine, Nanjing 210009, Jiangsu, The People's Republic of China, Tel.: +86 258 327 2358; Fax: +86 258 332 4887
| |
Collapse
|
278
|
Burgio G, La Rocca G, Sala A, Arancio W, Di Gesù D, Collesano M, Sperling AS, Armstrong JA, van Heeringen SJ, Logie C, Tamkun JW, Corona DFV. Genetic identification of a network of factors that functionally interact with the nucleosome remodeling ATPase ISWI. PLoS Genet 2008; 4:e1000089. [PMID: 18535655 PMCID: PMC2390755 DOI: 10.1371/journal.pgen.1000089] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2008] [Accepted: 05/02/2008] [Indexed: 02/03/2023] Open
Abstract
Nucleosome remodeling and covalent modifications of histones play fundamental roles in chromatin structure and function. However, much remains to be learned about how the action of ATP-dependent chromatin remodeling factors and histone-modifying enzymes is coordinated to modulate chromatin organization and transcription. The evolutionarily conserved ATP-dependent chromatin-remodeling factor ISWI plays essential roles in chromosome organization, DNA replication, and transcription regulation. To gain insight into regulation and mechanism of action of ISWI, we conducted an unbiased genetic screen to identify factors with which it interacts in vivo. We found that ISWI interacts with a network of factors that escaped detection in previous biochemical analyses, including the Sin3A gene. The Sin3A protein and the histone deacetylase Rpd3 are part of a conserved histone deacetylase complex involved in transcriptional repression. ISWI and the Sin3A/Rpd3 complex co-localize at specific chromosome domains. Loss of ISWI activity causes a reduction in the binding of the Sin3A/Rpd3 complex to chromatin. Biochemical analysis showed that the ISWI physically interacts with the histone deacetylase activity of the Sin3A/Rpd3 complex. Consistent with these findings, the acetylation of histone H4 is altered when ISWI activity is perturbed in vivo. These findings suggest that ISWI associates with the Sin3A/Rpd3 complex to support its function in vivo.
Collapse
Affiliation(s)
- Giosalba Burgio
- Dipartimento di Scienze Biochimiche, Universita' degli Studi di Palermo, Palermo, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
279
|
Stinski MF, Isomura H. Role of the cytomegalovirus major immediate early enhancer in acute infection and reactivation from latency. Med Microbiol Immunol 2008; 197:223-231. [PMID: 18097687 DOI: 10.1007/s00430-007-0069-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2007] [Indexed: 01/19/2023]
Abstract
The cytomegalovirus (CMV) major immediate early (MIE) enhancer-containing promoter regulates the expression of the downstream MIE genes, which have critical roles in reactivation from latency and acute infection. The enhancer consists of binding sites for cellular transcription factors that are repeated multiple times. The primate and nonprimate CMV enhancers can substitute for one another. The enhancers are not functionally equivalent, but they do have overlapping activities. The CMV MIE enhancers are located between divergent promoters where the leftward genes are critical and essential for reactivation from latency and acute infection and the rightward gene is nonessential. The rightward transcription unit is controlled by an enhancer for murine CMV. In contrast, human CMV has a set of repressor elements that prevents enhancer effects on the rightward viral promoter. The human CMV enhancer that controls the leftward transcription unit has a distal component that is nonessential at high multiplicity of infection (MOI), but has a significant impact on the MIE gene expression at low MOI. The proximal enhancer influences directly the level of transcription of the MIE genes and contains an essential Sp-1 site. The MIE promoter has a site adjacent to the transcription start site that is essential at the earliest stage of infection. The MIE enhancer-containing promoter responds to signal transduction events and to cellular differentiation. The role of the CMV MIE enhancer-containing promoter in acute infection and reactivation from latency are reviewed.
Collapse
Affiliation(s)
- Mark F Stinski
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | | |
Collapse
|
280
|
Spensberger D, Vermeulen M, Le Guezennec X, Beekman R, van Hoven A, Bindels E, Stunnenberg H, Delwel R. Myeloid transforming protein Evi1 interacts with methyl-CpG binding domain protein 3 and inhibits in vitro histone deacetylation by Mbd3/Mi-2/NuRD. Biochemistry 2008; 47:6418-26. [PMID: 18500823 DOI: 10.1021/bi800267f] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The ecotropic viral integration site 1 ( Evi1) gene encodes a putative transcription regulator, which is aberrantly expressed in acute myeloid leukemias (AML) with chromosomal abnormalities involving the 3q26 locus. Repression and activation of transcriptional control have been reported, but it is currently unclear how Evi1 may evoke these opposing effects. Using a yeast two-hybrid screen, we identified a novel binding partner of Evi1, i.e., methyl binding domain 3b (Mbd3b) protein, a member of the Mi-2/NuRD histone deacetylase complex. Applying in vitro and in vivo assays, we found that Evi1 interacts with Mbd3b but not with other MBD family members Mbd1, -2, and -4 or MeCP2. We show that interaction of Evi1 with Mbd3 requires 40 amino acids that are adjacent and downstream of the methyl binding domain (MBD). We further demonstrate that the first three zinc fingers of Evi1 are needed for Mbd3 interaction. Evi1 acts as a transcriptional repressor when recruited to an active promoter, yet when present in the Mi-2/NuRD complex through Mbd3 interaction, it inhibits the histone deacetylation function of this multiprotein structure. Our data may in part explain how Evi1 could act as a repressor as well as an activator of transcription.
Collapse
Affiliation(s)
- Dominik Spensberger
- Department of Hematology, Erasmus University Medical Center, Rotterdam 3015 GE, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
281
|
Alcamo EA, Chirivella L, Dautzenberg M, Dobreva G, Fariñas I, Grosschedl R, McConnell SK. Satb2 regulates callosal projection neuron identity in the developing cerebral cortex. Neuron 2008; 57:364-77. [PMID: 18255030 DOI: 10.1016/j.neuron.2007.12.012] [Citation(s) in RCA: 482] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2007] [Revised: 10/23/2007] [Accepted: 12/03/2007] [Indexed: 02/07/2023]
Abstract
Satb2 is a DNA-binding protein that regulates chromatin organization and gene expression. In the developing brain, Satb2 is expressed in cortical neurons that extend axons across the corpus callosum. To assess the role of Satb2 in neurons, we analyzed mice in which the Satb2 locus was disrupted by insertion of a LacZ gene. In mutant mice, beta-galactosidase-labeled axons are absent from the corpus callosum and instead descend along the corticospinal tract. Satb2 mutant neurons acquire expression of Ctip2, a transcription factor that is necessary and sufficient for the extension of subcortical projections by cortical neurons. Conversely, ectopic expression of Satb2 in neural stem cells markedly decreases Ctip2 expression. Finally, we find that Satb2 binds directly to regulatory regions of Ctip2 and induces changes in chromatin structure. These data suggest that Satb2 functions as a repressor of Ctip2 and regulatory determinant of corticocortical connections in the developing cerebral cortex.
Collapse
|
282
|
Britanova O, de Juan Romero C, Cheung A, Kwan KY, Schwark M, Gyorgy A, Vogel T, Akopov S, Mitkovski M, Agoston D, Sestan N, Molnár Z, Tarabykin V. Satb2 is a postmitotic determinant for upper-layer neuron specification in the neocortex. Neuron 2008; 57:378-92. [PMID: 18255031 DOI: 10.1016/j.neuron.2007.12.028] [Citation(s) in RCA: 489] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2007] [Revised: 11/13/2007] [Accepted: 12/21/2007] [Indexed: 12/18/2022]
Abstract
Pyramidal neurons of the neocortex can be subdivided into two major groups: deep- (DL) and upper-layer (UL) neurons. Here we report that the expression of the AT-rich DNA-binding protein Satb2 defines two subclasses of UL neurons: UL1 (Satb2 positive) and UL2 (Satb2 negative). In the absence of Satb2, UL1 neurons lose their identity and activate DL- and UL2-specific genetic programs. UL1 neurons in Satb2 mutants fail to migrate to superficial layers and do not contribute to the corpus callosum but to the corticospinal tract, which is normally populated by DL axons. Ctip2, a gene required for the formation of the corticospinal tract, is ectopically expressed in all UL1 neurons in the absence of Satb2. Satb2 protein interacts with the Ctip2 genomic region and controls chromatin remodeling at this locus. Satb2 therefore is required for the initiation of the UL1-specific genetic program and for the inactivation of DL- and UL2-specific genes.
Collapse
Affiliation(s)
- Olga Britanova
- Max-Planck-Institute for Experimental Medicine, Hermann-Rein Strasse 3, 37075 Göttingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
283
|
Lee C, Gyorgy A, Maric D, Sadri N, Schneider RJ, Barker JL, Lawson M, Agoston DV. Members of the NuRD chromatin remodeling complex interact with AUF1 in developing cortical neurons. Cereb Cortex 2008; 18:2909-19. [PMID: 18413351 DOI: 10.1093/cercor/bhn051] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Chromatin remodeling plays an important role in coordinating gene expression during cortical development, however the identity of molecular complexes present in differentiating cortical neurons that mediate the process remains poorly understood. The A + U-rich element-binding factor 1 (AUF1) is a known regulator of messenger RNA stability and also acts as a transcription factor upon binding to AT-rich DNA elements. Here we show that AUF1 is specifically expressed in subsets of proliferating neural precursors and differentiating postmitotic neurons of the developing cerebral cortex. Moreover, AUF1 is coexpressed with histone deacetylase 1 (HDAC1) and metastasis-associated protein 2 (MTA2), members of the nucleosome remodeling and histone deacetylase complex. AUF1 specifically and simultaneously binds to HDAC1, MTA2, and AT-rich DNA element, its gene regulatory function is modulated by the extent of histone acetylation and in animals lacking AUF1, the composition of the complex is modified. These results suggest that AUF1 is involved in integrating genetic and epigenetic signals during cortical development through recruiting HDAC1 and MTA2 to AT-rich DNA elements.
Collapse
Affiliation(s)
- Cheol Lee
- Neuroscience Program, School of Medicine, USUHS, Bethesda, MD 20814, USA
| | | | | | | | | | | | | | | |
Collapse
|
284
|
Beta-globin LCR and intron elements cooperate and direct spatial reorganization for gene therapy. PLoS Genet 2008; 4:e1000051. [PMID: 18404216 PMCID: PMC2271131 DOI: 10.1371/journal.pgen.1000051] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2007] [Accepted: 03/11/2008] [Indexed: 12/15/2022] Open
Abstract
The Locus Control Region (LCR) requires intronic elements within β-globin transgenes to direct high level expression at all ectopic integration sites. However, these essential intronic elements cannot be transmitted through retrovirus vectors and their deletion may compromise the therapeutic potential for gene therapy. Here, we systematically regenerate functional β-globin intron 2 elements that rescue LCR activity directed by 5′HS3. Evaluation in transgenic mice demonstrates that an Oct-1 binding site and an enhancer in the intron cooperate to increase expression levels from LCR globin transgenes. Replacement of the intronic AT-rich region with the Igμ 3′MAR rescues LCR activity in single copy transgenic mice. Importantly, a combination of the Oct-1 site, Igμ 3′MAR and intronic enhancer in the BGT158 cassette directs more consistent levels of expression in transgenic mice. By introducing intron-modified transgenes into the same genomic integration site in erythroid cells, we show that BGT158 has the greatest transcriptional induction. 3D DNA FISH establishes that induction stimulates this small 5′HS3 containing transgene and the endogenous locus to spatially reorganize towards more central locations in erythroid nuclei. Electron Spectroscopic Imaging (ESI) of chromatin fibers demonstrates that ultrastructural heterochromatin is primarily perinuclear and does not reorganize. Finally, we transmit intron-modified globin transgenes through insulated self-inactivating (SIN) lentivirus vectors into erythroid cells. We show efficient transfer and robust mRNA and protein expression by the BGT158 vector, and virus titer improvements mediated by the modified intron 2 in the presence of an LCR cassette composed of 5′HS2-4. Our results have important implications for the mechanism of LCR activity at ectopic integration sites. The modified transgenes are the first to transfer intronic elements that potentiate LCR activity and are designed to facilitate correction of hemoglobinopathies using single copy vectors. Expression of the β-globin gene is regulated by interactions between a distant Locus Control Region (LCR) and regulatory elements in or near the gene. We previously showed that LCR activity requires specific β-globin intron elements to consistently activate transgene expression in mice. These important intronic elements fail to transmit through lentivirus vectors designed for gene therapy of Sickle Cell Anemia. In this study, we identify intron modifications that reveal functional cooperation between the β-globin intronic enhancer and an intronic Oct-1 site. LCR activity in transgenic mice is also potentiated by an intronically located Igμ 3′MAR element. During induction of erythroid gene expression, the modified intron directs relocalization of the transgene away from the nuclear periphery towards more central neighbourhoods, and this movement mimics relocalization by the endogenous β-globin locus. Lentivirus vectors with the modified intron produce high titer virus stocks that express the transgene to therapeutic levels in erythroid cells. These findings have implications for understanding the mechanism of LCR activity, and for designing safe and effective lentivirus vectors for gene therapy.
Collapse
|
285
|
Tan JAT, Sun Y, Song J, Chen Y, Krontiris TG, Durrin LK. SUMO conjugation to the matrix attachment region-binding protein, special AT-rich sequence-binding protein-1 (SATB1), targets SATB1 to promyelocytic nuclear bodies where it undergoes caspase cleavage. J Biol Chem 2008; 283:18124-34. [PMID: 18408014 DOI: 10.1074/jbc.m800512200] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
SATB1 (special AT-rich sequence-binding protein-1) provides a key link between DNA loop organization, chromatin modification/remodeling, and association of transcription factors at matrix attachment regions (MARs). To investigate the role of SATB1 in cellular events, we performed a yeast two-hybrid screen that identified SUMO-1, Ubc9, and protein inhibitor of activated STAT (PIAS) family members as SATB1 interaction partners. These proteins, working in concert, enhanced SUMO conjugation to lysine-744 of SATB1. Overexpression of SUMO or PIAS in Jurkat cells, which express high levels of endogenous SATB1, exhibited enhanced caspase cleavage of this MAR-associating protein. Sumoylation-deficient SATB1 (SATB1(K744R)) failed to display the characteristic caspase cleavage pattern; however, fusion of SUMO in-frame to SATB1(K744R) restored cleavage. A SUMO-independent interaction of inactive caspase-6 and SATB1 was noted. A subset of total cellular SATB1 localized into promyelocytic leukemia nuclear bodies where enhanced SATB1 cleavage was detected subsequent to caspase activation. These results reveal a novel sumoylation-directed caspase cleavage of this key regulatory molecule. The role of regulated proteolysis of SATB1 may be to control transcription in immune cells during normal cell functions or to assist in efficient and rapid clearance of nonfunctional or potentially damaging immune cells.
Collapse
Affiliation(s)
- Joseph-Anthony T Tan
- Division of Molecular Medicine, Division of Immunology, and Graduate School of Biological Sciences, Beckman Research Institute of the City of Hope Medical Center, Duarte, CA 91010, USA
| | | | | | | | | | | |
Collapse
|
286
|
Mateos-Langerak J, Goetze S, Leonhardt H, Cremer T, van Driel R, Lanctôt C. Nuclear architecture: Is it important for genome function and can we prove it? J Cell Biochem 2008; 102:1067-75. [PMID: 17786936 DOI: 10.1002/jcb.21521] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Gene regulation in higher eukaryotes has been shown to involve regulatory sites, such as promoters and enhancers which act at the level of individual genes, and mechanisms which control the functional state of gene clusters. A fundamental question is whether additional levels of genome control exist. Nuclear organization and large-scale chromatin structure may constitute such a level and play an important role in the cell-type specific orchestration of the expression of thousands of genes in eukaryotic cells. Numerous observations indicate a tight correlation between genome activity and nuclear and large-scale chromatin structure. However, causal relationships are rare. Here we explore how these might be uncovered.
Collapse
Affiliation(s)
- Julio Mateos-Langerak
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
287
|
SATB1 reprogrammes gene expression to promote breast tumour growth and metastasis. Nature 2008; 452:187-93. [PMID: 18337816 DOI: 10.1038/nature06781] [Citation(s) in RCA: 390] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2007] [Accepted: 01/22/2008] [Indexed: 12/13/2022]
|
288
|
Chromatin remodelling at promoters suppresses antisense transcription. Nature 2008; 450:1031-5. [PMID: 18075583 DOI: 10.1038/nature06391] [Citation(s) in RCA: 323] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2007] [Accepted: 10/18/2007] [Indexed: 01/14/2023]
Abstract
Chromatin allows the eukaryotic cell to package its DNA efficiently. To understand how chromatin structure is controlled across the Saccharomyces cerevisiae genome, we have investigated the role of the ATP-dependent chromatin remodelling complex Isw2 in positioning nucleosomes. We find that Isw2 functions adjacent to promoter regions where it repositions nucleosomes at the interface between genic and intergenic sequences. Nucleosome repositioning by Isw2 is directional and results in increased nucleosome occupancy of the intergenic region. Loss of Isw2 activity leads to inappropriate transcription, resulting in the generation of both coding and noncoding transcripts. Here we show that Isw2 repositions nucleosomes to enforce directionality on transcription by preventing transcription initiation from cryptic sites. Our analyses reveal how chromatin is organized on a global scale and advance our understanding of how transcription is regulated.
Collapse
|
289
|
Purbey PK, Singh S, Kumar PP, Mehta S, Ganesh KN, Mitra D, Galande S. PDZ domain-mediated dimerization and homeodomain-directed specificity are required for high-affinity DNA binding by SATB1. Nucleic Acids Res 2008; 36:2107-22. [PMID: 18187506 PMCID: PMC2367706 DOI: 10.1093/nar/gkm1151] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
To better understand DNA recognition and transcription activity by SATB1, the T-lineage-enriched chromatin organizer and transcription factor, we have determined its optimal DNA-binding sequence by random oligonucleotide selection. The consensus SATB1-binding sequence (CSBS) comprises a palindromic sequence in which two identical AT-rich half-sites are arranged as inverted repeats flanking a central cytosine or guanine. Strikingly, the CSBS half-site is identical to the conserved element ‘TAATA’ bound by the known homeodomains (HDs). Furthermore, we show that the high-affinity binding of SATB1 to DNA is dimerization-dependent and the HD also binds in similar fashion. Binding studies using HD-lacking SATB1 and binding target with increased spacer between the two half-sites led us to propose a model for SATB1–DNA complex in which the HDs bind in an antiparallel fashion to the palindromic consensus element via minor groove, bridged by the PDZ-like dimerization domain. CSBS-driven in vivo reporter analysis indicated that SATB1 acts as a repressor upon binding to the CSBS and most of its derivatives and the extent of repression is proportional to SATB1's binding affinity to these sequences. These studies provide mechanistic insights into the mode of DNA binding and its effect on the regulation of transcription by SATB1.
Collapse
|
290
|
Cam HP, Noma KI, Ebina H, Levin HL, Grewal SIS. Host genome surveillance for retrotransposons by transposon-derived proteins. Nature 2007; 451:431-6. [PMID: 18094683 DOI: 10.1038/nature06499] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Accepted: 11/22/2007] [Indexed: 12/23/2022]
Abstract
Transposable elements and their remnants constitute a substantial fraction of eukaryotic genomes. Host genomes have evolved defence mechanisms, including chromatin modifications and RNA interference, to regulate transposable elements. Here we describe a genome surveillance mechanism for retrotransposons by transposase-derived centromeric protein CENP-B homologues of the fission yeast Schizosaccharomyces pombe. CENP-B homologues of S. pombe localize at and recruit histone deacetylases to silence Tf2 retrotransposons. CENP-Bs also repress solo long terminal repeats (LTRs) and LTR-associated genes. Tf2 elements are clustered into 'Tf' bodies, the organization of which depends on CENP-Bs that display discrete nuclear structures. Furthermore, CENP-Bs prevent an 'extinct' Tf1 retrotransposon from re-entering the host genome by blocking its recombination with extant Tf2, and silence and immobilize a Tf1 integrant that becomes sequestered into Tf bodies. Our results reveal a probable ancient retrotransposon surveillance pathway important for host genome integrity, and highlight potential conflicts between DNA transposons and retrotransposons, major transposable elements believed to have greatly moulded the evolution of genomes.
Collapse
Affiliation(s)
- Hugh P Cam
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
291
|
Galande S, Purbey PK, Notani D, Kumar PP. The third dimension of gene regulation: organization of dynamic chromatin loopscape by SATB1. Curr Opin Genet Dev 2007; 17:408-14. [PMID: 17913490 DOI: 10.1016/j.gde.2007.08.003] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2007] [Revised: 07/27/2007] [Accepted: 08/17/2007] [Indexed: 02/07/2023]
Abstract
Compartmentalized distribution of functional components is a hallmark of the eukaryotic nucleus. Technological advances in recent years have provided unprecedented insights into the role of chromatin organization and interactions of various structural-functional components toward gene regulation. SATB1, the global chromatin organizer and transcription factor, has emerged as a key factor integrating higher-order chromatin architecture with gene regulation. Studies in recent years have unraveled the role of SATB1 in organization of chromatin 'loopscape' and its dynamic nature in response to physiological stimuli. SATB1 organizes the MHC class-I locus into distinct chromatin loops by tethering MARs to nuclear matrix at fixed distances. Silencing of SATB1 mimics the effects of IFNgamma treatment on chromatin loop architecture of the MHC class-I locus and altered expression of genes within the locus. At genome-wide level, SATB1 seems to play a role in organization of the transcriptionally poised chromatin.
Collapse
Affiliation(s)
- Sanjeev Galande
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| | | | | | | |
Collapse
|
292
|
Steidl U, Steidl C, Ebralidze A, Chapuy B, Han HJ, Will B, Rosenbauer F, Becker A, Wagner K, Koschmieder S, Kobayashi S, Costa DB, Schulz T, O’Brien KB, Verhaak RG, Delwel R, Haase D, Trümper L, Krauter J, Kohwi-Shigematsu T, Griesinger F, Tenen DG. A distal single nucleotide polymorphism alters long-range regulation of the PU.1 gene in acute myeloid leukemia. J Clin Invest 2007; 117:2611-20. [PMID: 17694175 PMCID: PMC1937499 DOI: 10.1172/jci30525] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2006] [Accepted: 05/14/2007] [Indexed: 01/20/2023] Open
Abstract
Targeted disruption of a highly conserved distal enhancer reduces expression of the PU.1 transcription factor by 80% and leads to acute myeloid leukemia (AML) with frequent cytogenetic aberrations in mice. Here we identify a SNP within this element in humans that is more frequent in AML with a complex karyotype, leads to decreased enhancer activity, and reduces PU.1 expression in myeloid progenitors in a development-dependent manner. This SNP inhibits binding of the chromatin-remodeling transcriptional regulator special AT-rich sequence binding protein 1 (SATB1). Overexpression of SATB1 increased PU.1 expression, and siRNA inhibition of SATB1 downregulated PU.1 expression. Targeted disruption of the distal enhancer led to a loss of regulation of PU.1 by SATB1. Interestingly, disruption of SATB1 in mice led to a selective decrease of PU.1 RNA in specific progenitor types (granulocyte-macrophage and megakaryocyte-erythrocyte progenitors) and a similar effect was observed in AML samples harboring this SNP. Thus we have identified a SNP within a distal enhancer that is associated with a subtype of leukemia and exerts a deleterious effect through remote transcriptional dysregulation in specific progenitor subtypes.
Collapse
MESH Headings
- Animals
- Base Sequence
- Cell Adhesion Molecules, Neuronal/genetics
- Cell Adhesion Molecules, Neuronal/metabolism
- Cell Line, Tumor
- Down-Regulation
- Gene Expression Regulation, Neoplastic
- Genome, Human/genetics
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Mice
- Mice, Knockout
- Molecular Sequence Data
- Polymorphism, Single Nucleotide/genetics
- Proto-Oncogene Proteins/deficiency
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Receptors, Lymphocyte Homing/genetics
- Receptors, Lymphocyte Homing/metabolism
- Stem Cells/metabolism
- Trans-Activators/deficiency
- Trans-Activators/genetics
- Trans-Activators/metabolism
Collapse
Affiliation(s)
- Ulrich Steidl
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Christian Steidl
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Alexander Ebralidze
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Björn Chapuy
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Hye-Jung Han
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Britta Will
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Frank Rosenbauer
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Annegret Becker
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Katharina Wagner
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Steffen Koschmieder
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Susumu Kobayashi
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Daniel B. Costa
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Thomas Schulz
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Karen B. O’Brien
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Roel G.W. Verhaak
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ruud Delwel
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Detlef Haase
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Lorenz Trümper
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jürgen Krauter
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Terumi Kohwi-Shigematsu
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Frank Griesinger
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Daniel G. Tenen
- Harvard Institutes of Medicine, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
Department of Hematology and Oncology, Georg-August University of Göttingen, Goettingen, Germany.
Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
Department of Cell Biology, University of Freiburg, Freiburg, Germany.
Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Hannover, Germany.
Department of Medicine, Hematology and Oncology, University Hospital Münster, Muenster, Germany.
Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
293
|
Abstract
T lymphocyte development is directed by a gene-expression program that occurs in the complex nucleoprotein environment of chromatin. This review examines basic principles of chromatin regulation and evaluates ongoing progress toward understanding how the chromatin template is manipulated to control gene expression and gene recombination in developing thymocytes. Special attention is devoted to the loci encoding T cell receptors alpha and beta, T cell coreceptors CD4 and CD8, and the enzyme terminal deoxynucleotidyl transferase. The properties of SATB1, a notable organizer of thymocyte chromatin, are also addressed.
Collapse
Affiliation(s)
- Michael S Krangel
- Department of Immunology, Duke University Medical Center, Durham, North Carolina 27710 USA.
| |
Collapse
|
294
|
Lee J, Klase Z, Gao X, Caldwell JS, Stinski MF, Kashanchi F, Chao SH. Cellular homeoproteins, SATB1 and CDP, bind to the unique region between the human cytomegalovirus UL127 and major immediate-early genes. Virology 2007; 366:117-25. [PMID: 17512569 DOI: 10.1016/j.virol.2007.04.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Revised: 03/19/2007] [Accepted: 04/20/2007] [Indexed: 01/19/2023]
Abstract
An AT-rich region of the human cytomegalovirus (CMV) genome between the UL127 open reading frame and the major immediate-early (MIE) enhancer is referred to as the unique region (UR). It has been shown that the UR represses activation of transcription from the UL127 promoter and functions as a boundary between the divergent UL127 and MIE genes during human CMV infection [Angulo, A., Kerry, D., Huang, H., Borst, E.M., Razinsky, A., Wu, J., Hobom, U., Messerle, M., Ghazal, P., 2000. Identification of a boundary domain adjacent to the potent human cytomegalovirus enhancer that represses transcription of the divergent UL127 promoter. J. Virol. 74 (6), 2826-2839; Lundquist, C.A., Meier, J.L., Stinski, M.F., 1999. A strong negative transcriptional regulatory region between the human cytomegalovirus UL127 gene and the major immediate-early enhancer. J. Virol. 73 (11), 9039-9052]. A putative forkhead box-like (FOX-like) site, AAATCAATATT, was identified in the UR and found to play a key role in repression of the UL127 promoter in recombinant virus-infected cells [Lashmit, P.E., Lundquist, C.A., Meier, J.L., Stinski, M.F., 2004. Cellular repressor inhibits human cytomegalovirus transcription from the UL127 promoter. J. Virol. 78 (10), 5113-5123]. However, the cellular factors which associate with the UR and FOX-like region remain to be determined. We reported previously that pancreatic-duodenal homeobox factor-1 (PDX1) bound to a 45-bp element located within the UR [Chao, S.H., Harada, J.N., Hyndman, F., Gao, X., Nelson, C.G., Chanda, S.K., Caldwell, J.S., 2004. PDX1, a Cellular Homeoprotein, Binds to and Regulates the Activity of Human Cytomegalovirus Immediate Early Promoter. J. Biol. Chem. 279 (16), 16111-16120]. Here we demonstrate that two additional cellular homeoproteins, special AT-rich sequence binding protein 1 (SATB1) and CCAAT displacement protein (CDP), bind to the human CMV UR in vitro and in vivo. Furthermore, CDP is identified as a FOX-like binding protein and a repressor of the UL127 promoter, while SATB1 has no effect on UL127 expression. Since CDP is known as a transcription repressor and a nuclear matrix-associated region binding protein, CDP may have a role in the regulation of human CMV transcription.
Collapse
Affiliation(s)
- Jialing Lee
- Expression Engineering Group, Bioprocessing Technology Institute, 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | | | | | | | | | | | | |
Collapse
|
295
|
Schüle B, Li HH, Fisch-Kohl C, Purmann C, Francke U. DLX5 and DLX6 expression is biallelic and not modulated by MeCP2 deficiency. Am J Hum Genet 2007; 81:492-506. [PMID: 17701895 PMCID: PMC1950824 DOI: 10.1086/520063] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2007] [Accepted: 05/21/2007] [Indexed: 11/03/2022] Open
Abstract
Mutations in MECP2 and Mecp2 (encoding methyl-CpG binding protein 2 [MeCP2]) cause distinct neurological phenotypes in humans and mice, respectively, but the molecular pathology is unclear. Recent literature claimed that the developmental homeobox gene DLX5 is imprinted and that its imprinting status is modulated by MeCP2, leading to biallelic expression in Rett syndrome and twofold overexpression of Dlx5 and Dlx6 in Mecp2-null mice. The conclusion that DLX5 is a direct target of MeCP2 has implications for research on the molecular bases of Rett syndrome, autism, and genomic imprinting. Attempting to replicate the reported data, we evaluated allele-specific expression of DLX5 and DLX6 in mouse x human somatic cell hybrids, lymphoblastoid cell lines, and frontal cortex from controls and individuals with MECP2 mutations. We identified novel single-nucleotide polymorphisms in DLX5 and DLX6, enabling the first imprinting studies of DLX6. We found that DLX5 and DLX6 are biallelically expressed in somatic cell hybrids and in human cell lines and brain, with no differences between affected and control samples. We also determined expression levels of Dlx5 and Dlx6 in forebrain from seven male Mecp2-mutant mice and eight wild-type littermates by real-time quantitative reverse-transcriptase polymerase chain reaction assays. Expression of Dlx5 and Dlx6, as well as of the imprinted gene Peg3, in mouse forebrain was highly variable, with no consistent differences between Mecp2-null mutants and controls. We conclude that DLX5 and DLX6 are not imprinted in humans and are not likely to be direct targets of MeCP2 modulation. In contrast, the imprinting status of PEG3 and PEG10 is maintained in MeCP2-deficient tissues. Our results confirm that MeCP2 plays no role in the maintenance of genomic imprinting and add PEG3 and PEG10 to the list of studied imprinted genes.
Collapse
Affiliation(s)
- Birgitt Schüle
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
296
|
Christova R, Jones T, Wu PJ, Bolzer A, Costa-Pereira AP, Watling D, Kerr IM, Sheer D. P-STAT1 mediates higher-order chromatin remodelling of the human MHC in response to IFNgamma. J Cell Sci 2007; 120:3262-70. [PMID: 17726060 DOI: 10.1242/jcs.012328] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Transcriptional activation of the major histocompatibility complex (MHC) by IFNgamma is a key step in cell-mediated immunity. At an early stage of IFNgamma induction, chromatin carrying the entire MHC locus loops out from the chromosome 6 territory. We show here that JAK/STAT signalling triggers this higher-order chromatin remodelling and the entire MHC locus becomes decondensed prior to transcriptional activation of the classical HLA class II genes. A single point mutation of STAT1 that prevents phosphorylation is sufficient to abolish chromatin remodelling, thus establishing a direct link between the JAK/STAT signalling pathway and human chromatin architecture. The onset of chromatin remodelling corresponds with the binding of activated STAT1 and the chromatin remodelling enzyme BRG1 at specific sites within the MHC, and is followed by RNA-polymerase recruitment and histone hyperacetylation. We propose that the higher-order chromatin remodelling of the MHC locus is an essential step to generate a transcriptionally permissive chromatin environment for subsequent activation of classical HLA genes.
Collapse
Affiliation(s)
- Rossitza Christova
- Human Cytogenetics Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, WC2A 3PX, UK
| | | | | | | | | | | | | | | |
Collapse
|
297
|
Girod PA, Nguyen DQ, Calabrese D, Puttini S, Grandjean M, Martinet D, Regamey A, Saugy D, Beckmann JS, Bucher P, Mermod N. Genome-wide prediction of matrix attachment regions that increase gene expression in mammalian cells. Nat Methods 2007; 4:747-53. [PMID: 17676049 DOI: 10.1038/nmeth1076] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2007] [Accepted: 07/02/2007] [Indexed: 01/13/2023]
Abstract
Gene transfer in eukaryotic cells and organisms suffers from epigenetic effects that result in low or unstable transgene expression and high clonal variability. Use of epigenetic regulators such as matrix attachment regions (MARs) is a promising approach to alleviate such unwanted effects. Dissection of a known MAR allowed the identification of sequence motifs that mediate elevated transgene expression. Bioinformatics analysis implied that these motifs adopt a curved DNA structure that positions nucleosomes and binds specific transcription factors. From these observations, we computed putative MARs from the human genome. Cloning of several predicted MARs indicated that they are much more potent than the previously known element, boosting the expression of recombinant proteins from cultured cells as well as mediating high and sustained expression in mice. Thus we computationally identified potent epigenetic regulators, opening new strategies toward high and stable transgene expression for research, therapeutic production or gene-based therapies.
Collapse
Affiliation(s)
- Pierre-Alain Girod
- Institute of Biotechnology, University of Lausanne, and Center for Biotechnology of the University of Lausanne and Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
298
|
Yamasaki K, Akiba T, Yamasaki T, Harata K. Structural basis for recognition of the matrix attachment region of DNA by transcription factor SATB1. Nucleic Acids Res 2007; 35:5073-84. [PMID: 17652321 PMCID: PMC1976457 DOI: 10.1093/nar/gkm504] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Special AT-rich sequence binding protein 1 (SATB1) regulates gene expression essential in immune T-cell maturation and switching of fetal globin species, by binding to matrix attachment regions (MARs) of DNA and inducing a local chromatin remodeling. Previously we have revealed a five-helix structure of the N-terminal CUT domain, which is essentially the folded region in the MAR-binding domain, of human SATB1 by NMR. Here we determined crystal structure of the complex of the CUT domain and a MAR DNA, in which the third helix of the CUT domain deeply enters the major groove of DNA in the B-form. Bases of 5'-CTAATA-3' sequence are contacted by this helix, through direct and water-mediated hydrogen bonds and apolar and van der Waals contacts. Mutations at conserved base-contacting residues, Gln402 and Gly403, reduced the DNA-binding activity, which confirmed the importance of the observed interactions involving these residues. A significant number of equivalent contacts are observed also for typically four-helix POU-specific domains of POU-homologous proteins, indicating that these domains share a common framework of the DNA-binding mode, recognizing partially similar DNA sequences.
Collapse
Affiliation(s)
- Kazuhiko Yamasaki
- Age Dimension Research Center, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan.
| | | | | | | |
Collapse
|
299
|
Abstract
An important tenet of evolutionary developmental biology ("evo devo") is that adaptive mutations affecting morphology are more likely to occur in the cis-regulatory regions than in the protein-coding regions of genes. This argument rests on two claims: (1) the modular nature of cis-regulatory elements largely frees them from deleterious pleiotropic effects, and (2) a growing body of empirical evidence appears to support the predominant role of gene regulatory change in adaptation, especially morphological adaptation. Here we discuss and critique these assertions. We first show that there is no theoretical or empirical basis for the evo devo contention that adaptations involving morphology evolve by genetic mechanisms different from those involving physiology and other traits. In addition, some forms of protein evolution can avoid the negative consequences of pleiotropy, most notably via gene duplication. In light of evo devo claims, we then examine the substantial data on the genetic basis of adaptation from both genome-wide surveys and single-locus studies. Genomic studies lend little support to the cis-regulatory theory: many of these have detected adaptation in protein-coding regions, including transcription factors, whereas few have examined regulatory regions. Turning to single-locus studies, we note that the most widely cited examples of adaptive cis-regulatory mutations focus on trait loss rather than gain, and none have yet pinpointed an evolved regulatory site. In contrast, there are many studies that have both identified structural mutations and functionally verified their contribution to adaptation and speciation. Neither the theoretical arguments nor the data from nature, then, support the claim for a predominance of cis-regulatory mutations in evolution. Although this claim may be true, it is at best premature. Adaptation and speciation probably proceed through a combination of cis-regulatory and structural mutations, with a substantial contribution of the latter.
Collapse
Affiliation(s)
- Hopi E Hoekstra
- Department of Organismic and Evolutionary Biology and the Museum of Comparative Zoology, Harvard University, 26 Oxford Street, Cambridge, Massachusetts 02138, USA.
| | | |
Collapse
|
300
|
Jordan C, Li HH, Kwan HC, Francke U. Cerebellar gene expression profiles of mouse models for Rett syndrome reveal novel MeCP2 targets. BMC MEDICAL GENETICS 2007; 8:36. [PMID: 17584923 PMCID: PMC1931432 DOI: 10.1186/1471-2350-8-36] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2007] [Accepted: 06/20/2007] [Indexed: 01/01/2023]
Abstract
Background MeCP2, methyl-CpG-binding protein 2, binds to methylated cytosines at CpG dinucleotides, as well as to unmethylated DNA, and affects chromatin condensation. MECP2 mutations in females lead to Rett syndrome, a neurological disorder characterized by developmental stagnation and regression, loss of purposeful hand movements and speech, stereotypic hand movements, deceleration of brain growth, autonomic dysfunction and seizures. Most mutations occur de novo during spermatogenesis. Located at Xq28, MECP2 is subject to X inactivation, and affected females are mosaic. Rare hemizygous males suffer from a severe congenital encephalopathy. Methods To identify the pathways mis-regulated by MeCP2 deficiency, microarray-based global gene expression studies were carried out in cerebellum of Mecp2 mutant mice. We compared transcript levels in mutant/wildtype male sibs of two different MeCP2-deficient mouse models at 2, 4 and 8 weeks of age. Increased transcript levels were evaluated by real-time quantitative RT-PCR. Chromatin immunoprecipitation assays were used to document in vivo MeCP2 binding to promoter regions of candidate target genes. Results Of several hundred genes with altered expression levels in the mutants, twice as many were increased than decreased, and only 27 were differentially expressed at more than one time point. The number of misregulated genes was 30% lower in mice with the exon 3 deletion (Mecp2tm1.1Jae) than in mice with the larger deletion (Mecp2tm1.1Bird). Between the mutants, few genes overlapped at each time point. Real-time quantitative RT-PCR assays validated increased transcript levels for four genes: Irak1, interleukin-1 receptor-associated kinase 1; Fxyd1, phospholemman, associated with Na, K-ATPase;Reln, encoding an extracellular signaling molecule essential for neuronal lamination and synaptic plasticity; and Gtl2/Meg3, an imprinted maternally expressed non-translated RNA that serves as a host gene for C/D box snoRNAs and microRNAs. Chromatin immunoprecipitation assays documented in vivo MeCP2 binding to promoter regions of Fxyd1, Reln, and Gtl2. Conclusion Transcriptional profiling of cerebellum failed to detect significant global changes in Mecp2-mutant mice. Increased transcript levels of Irak1, Fxyd1, Reln, and Gtl2 may contribute to the neuronal dysfunction in MeCP2-deficient mice and individuals with Rett syndrome. Our data provide testable hypotheses for future studies of the regulatory or signaling pathways that these genes act on.
Collapse
Affiliation(s)
- ChaRandle Jordan
- Department of Genetics, Stanford University School of Medicine, Stanford CA 94305-5323, USA
| | - Hong Hua Li
- Department of Genetics, Stanford University School of Medicine, Stanford CA 94305-5323, USA
| | - Helen C Kwan
- Department of Genetics, Stanford University School of Medicine, Stanford CA 94305-5323, USA
| | - Uta Francke
- Department of Genetics, Stanford University School of Medicine, Stanford CA 94305-5323, USA
| |
Collapse
|