201
|
Zhang M, Fang H, Zhou J, Herring BP. A novel role of Brg1 in the regulation of SRF/MRTFA-dependent smooth muscle-specific gene expression. J Biol Chem 2007; 282:25708-16. [PMID: 17599918 DOI: 10.1074/jbc.m701925200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Serum response factor (SRF) is a key regulator of smooth muscle differentiation, proliferation, and migration. Myocardin-related transcription factor A (MRTFA) is a co-activator of SRF that can induce expression of SRF-dependent, smooth muscle-specific genes and actin/Rho-dependent genes, but not MAPK-regulated growth response genes. How MRTFA and SRF discriminate between these sets of target genes is still unclear. We hypothesized that SWI/SNF ATP-dependent chromatin remodeling complexes, containing Brahma-related gene 1 (Brg1) or Brahma (Brm), may play a role in this process. Results from Western blotting and qRT-PCR analysis demonstrated that dominant negative Brg1 blocked the ability of MRTFA to induce expression of smooth muscle-specific genes, but not actin/Rho-dependent early response genes, in fibroblasts. In addition, dominant negative Brg1 attenuated expression of smooth muscle-specific genes in primary cultures of smooth muscle cells. MRTFA overexpression did not induce expression of smooth muscle-specific genes in SW13 cells, which lack endogenous Brg1 or Brm. Reintroduction of Brg1 or Brm into SW13 cells restored their responsiveness to MRTFA. Immunoprecipitation assays revealed that Brg1, SRF, and MRTFA form a complex in vivo, and Brg1 directly binds MRTFA, but not SRF, in vitro. Results from chromatin immunoprecipitation assays demonstrated that dominant negative Brg1 significantly attenuated the ability of MRTFA to increase SRF binding to the promoters of smooth muscle-specific genes, but not early response genes. Together these data suggest that Brg1/Brm containing SWI/SNF complexes play a critical role in regulating expression of SRF/MRTFA-dependent smooth muscle-specific genes but not SRF/MRTFA-dependent early response genes.
Collapse
Affiliation(s)
- Min Zhang
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202-5120, USA
| | | | | | | |
Collapse
|
202
|
Huang B, Ahn YT, McPherson L, Clayberger C, Krensky AM. Interaction of PRP4 with Kruppel-like factor 13 regulates CCL5 transcription. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:7081-7. [PMID: 17513757 PMCID: PMC2674583 DOI: 10.4049/jimmunol.178.11.7081] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Activation of resting T lymphocytes initiates differentiation into mature effector cells over 3-7 days. The chemokine CCL5 (RANTES) and its major transcriptional regulator, Krüppel-like factor 13 (KLF13), are expressed late (3-5 days) after activation in T lymphocytes. Using yeast two-hybrid screening of a human thymus cDNA library, PRP4, a serine/threonine protein kinase, was identified as a KLF13-binding protein. Specific interaction of KLF13 and PRP4 was confirmed by reciprocal coimmunoprecipitation. PRP4 is expressed in PHA-stimulated human T lymphocytes from days 1 and 7 with a peak at day 3. Using an in vitro kinase assay, it was found that PRP4 phosphorylates KLF13. Furthermore, although phosphorylation of KLF13 by PRP4 results in lower binding affinity to the A/B site of the CCL5 promoter, coexpression of PRP4 and KLF13 increases nuclear localization of KLF13 and CCL5 transcription. Finally, knock-down of PRP4 by small interfering RNA markedly decreases CCL5 expression in T lymphocytes. Thus, PRP4-mediated phosphorylation of KLF13 plays a role in the regulation of CCL5 expression in T lymphocytes.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/genetics
- Active Transport, Cell Nucleus/immunology
- Amino Acid Sequence
- Animals
- COS Cells
- Cell Cycle Proteins/biosynthesis
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/physiology
- Cells, Cultured
- Chemokine CCL5/biosynthesis
- Chemokine CCL5/genetics
- Chemokine CCL5/metabolism
- Chemokines, CC/biosynthesis
- Chemokines, CC/genetics
- Chemokines, CC/metabolism
- Chlorocebus aethiops
- Gene Expression Regulation/immunology
- Humans
- Kruppel-Like Transcription Factors/biosynthesis
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/physiology
- Mitogen-Activated Protein Kinases/metabolism
- Mitogen-Activated Protein Kinases/physiology
- Molecular Sequence Data
- Phosphorylation
- Protein Binding/genetics
- Protein Binding/immunology
- Protein Serine-Threonine Kinases/biosynthesis
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Repressor Proteins/biosynthesis
- Repressor Proteins/genetics
- Repressor Proteins/physiology
- Ribonucleoprotein, U4-U6 Small Nuclear/biosynthesis
- Ribonucleoprotein, U4-U6 Small Nuclear/genetics
- Ribonucleoprotein, U4-U6 Small Nuclear/metabolism
- Ribonucleoprotein, U4-U6 Small Nuclear/physiology
- T-Lymphocyte Subsets/enzymology
- T-Lymphocyte Subsets/immunology
- Thymus Gland/cytology
- Thymus Gland/enzymology
- Thymus Gland/immunology
- Transcription, Genetic
- Two-Hybrid System Techniques
Collapse
Affiliation(s)
- Boli Huang
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305
| | - Yong-Tae Ahn
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305
| | - Lisa McPherson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305
| | - Carol Clayberger
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305
| | - Alan M. Krensky
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
203
|
Gunawardena RW, Fox SR, Siddiqui H, Knudsen ES. SWI/SNF activity is required for the repression of deoxyribonucleotide triphosphate metabolic enzymes via the recruitment of mSin3B. J Biol Chem 2007; 282:20116-23. [PMID: 17510060 DOI: 10.1074/jbc.m701406200] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The SWI/SNF chromatin remodeling complex plays a critical role in the coordination of gene expression with physiological stimuli. The synthetic enzymes ribonucleotide reductase, dihydrofolate reductase, and thymidylate synthase are coordinately regulated to ensure appropriate deoxyribonucleotide triphosphate levels. Particularly, these enzymes are actively repressed as cells exit the cell cycle through the action of E2F transcription factors and the retinoblastoma tumor suppressor/p107/p130 family of pocket proteins. This process is found to be highly dependent on SWI/SNF activity as cells deficient in BRG-1 and Brm subunits fail to repress these genes with activation of pocket proteins, and this deficit in repression can be complemented, via the ectopic expression of BRG-1. The failure to repress transcription does not involve a blockade in the association of E2F or pocket proteins p107 and p130 with promoter elements. Rather, the deficit in repression is due to a failure to mediate histone deacetylation of ribonucleotide reductase, dihydrofolate reductase, and thymidylate synthase promoters in the absence of SWI/SNF activity. The basis for this is found to be a failure to recruit mSin3B and histone deacetylase proteins to promoters. Thus, the coordinate repression of deoxyribonucleotide triphosphate metabolic enzymes is dependent on the action of SWI/SNF in facilitating the assembly of repressor complexes at the promoter.
Collapse
Affiliation(s)
- Ranjaka W Gunawardena
- Department of Cell and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | | | | | | |
Collapse
|
204
|
Li J, Sutter C, Parker DS, Blauwkamp T, Fang M, Cadigan KM. CBP/p300 are bimodal regulators of Wnt signaling. EMBO J 2007; 26:2284-94. [PMID: 17410209 PMCID: PMC1864967 DOI: 10.1038/sj.emboj.7601667] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Accepted: 02/28/2007] [Indexed: 01/11/2023] Open
Abstract
Many Wnts influence cell behavior by a conserved signaling cascade that promotes the stabilization and nuclear accumulation of beta-catenin (beta-cat), which then associates with TCF family members to activate target genes. The histone acetyltransferase CREB binding protein (CBP) can bind to TCF and inhibit Wnt signaling in Drosophila. In contrast, studies in vertebrates indicate a positive role for CBP and the closely related protein p300 as beta-cat binding transcriptional co-activators. We address this discrepancy by demonstrating that in addition to its negative role, CBP has an essential positive role in Wnt signaling in flies. CBP binds directly to the C-terminus of Armadillo (Arm, the fly beta-cat) and is recruited to a Wnt-regulated enhancer (WRE) in a Wnt- and Arm-dependent manner. In a human colorectal cancer cell line, we show that CBP and p300 can inhibit Wnt signaling and demonstrate that human p300 can bind directly to TCF4 in vitro. Our results argue that CBP/p300 has an evolutionarily conserved role as a buffer regulating TCF-beta-cat/Arm binding. Subsequent to this interaction, it also has an essential role in mediating the transactivation activity of beta-cat/Arm.
Collapse
Affiliation(s)
- Jiong Li
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Chris Sutter
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - David S Parker
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Timothy Blauwkamp
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Ming Fang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Ken M Cadigan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, 830 North University Avenue, Ann Arbor, MI 48109-1048, USA. Tel.: +1 734 936-3246; Fax: +1 734 647 0884; E-mail:
| |
Collapse
|
205
|
de la Roche M, Bienz M. Wingless-independent association of Pygopus with dTCF target genes. Curr Biol 2007; 17:556-61. [PMID: 17320388 DOI: 10.1016/j.cub.2007.01.063] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2006] [Revised: 01/25/2007] [Accepted: 01/25/2007] [Indexed: 11/16/2022]
Abstract
The Wnt signaling pathway controls numerous cell fates during animal development. Its inappropriate activity can lead to cancer in many human tissues. A key effector of the canonical Wnt pathway is beta-catenin (or Drosophila Armadillo), a highly unstable phosphorylated protein that shuttles rapidly between nucleus and cytoplasm. Wnt signaling inhibits its phosphorylation and degradation; this allows it to associate with TCF/LEF factors bound to Wnt target genes and to stimulate their transcription by recruiting chromatin modifying and remodeling factors. The transcriptional activity of Armadillo/beta-catenin also depends on Pygopus (Pygo), a nuclear protein with which it associates through the Legless/BCL9 adaptor. It has been proposed that Pygo associates with TCF target genes during Wnt signaling through Armadillo and Legless to recruit a transcriptional coactivator through its Nbox motif. Here, we report that Pygo is associated constitutively with dTCF target genes in Drosophila salivary glands and tissue-culture cells. Our evidence indicates that this association depends on dTCF and on the Nbox motif of Pygo, but not on Legless. We thus propose an alternative model according to which Pygo functions at the onset of Wnt signaling, or at low signaling levels, to capture Armadillo at dTCF target genes, thus enabling the interaction between Armadillo and dTCF and, consequently, the Armadillo-mediated recruitment of transcriptional coactivators.
Collapse
Affiliation(s)
- Marc de la Roche
- MRC Laboratory of Molecular Biology, Hills Road, Cambridge, United Kingdom
| | | |
Collapse
|
206
|
Chen YH, Yang CK, Xia M, Ou CY, Stallcup MR. Role of GAC63 in transcriptional activation mediated by beta-catenin. Nucleic Acids Res 2007; 35:2084-92. [PMID: 17344318 PMCID: PMC1874623 DOI: 10.1093/nar/gkm095] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2006] [Revised: 01/03/2007] [Accepted: 02/02/2007] [Indexed: 01/04/2023] Open
Abstract
Beta-catenin is a key mediator in the canonical Wnt signaling pathway, which plays important roles in multiple developmental processes. Inappropriate activation of this pathway leads to developmental defects and development of certain cancers. Upon Wnt signaling, beta-catenin binds TCF/LEF transcription factors. The TCF/LEF-beta-catenin complex then recruits a variety of transcriptional coactivators to the promoter/enhancer region of Wnt-responsive genes and activates target gene transcription. In this article, we demonstrate that GRIP1-associated coactivator 63 (GAC63), a recently identified nuclear receptor (NR) coactivator, interacts with beta-catenin. The N-terminus of GAC63 is the binding site for beta-catenin, whereas a C-terminal fragment of beta-catenin including armadillo repeats 10-12 binds to GAC63. Over-expression of GAC63 enhanced the transcriptional activity of beta-catenin, and also greatly enhanced TCF/LEF-regulated reporter gene activity in a beta-catenin-dependent manner. Endogenous GAC63 was recruited to TCF/LEF-responsive enhancer elements when beta-catenin levels were induced by LiCl. In addition, reduction of endogenous GAC63 level by small interfering RNA (siRNA) inhibited TCF/LEF-mediated gene transcription. Our findings reveal a new function of GAC63 in transcriptional activation of Wnt-responsive genes.
Collapse
Affiliation(s)
- Yong-Heng Chen
- Department of Pathology, Department of Biochemistry and Molecular Biology, and Division of Gastroenterology and Liver Diseases, University of Southern California, Los Angeles, California 90089, USA
| | - Catherine K. Yang
- Department of Pathology, Department of Biochemistry and Molecular Biology, and Division of Gastroenterology and Liver Diseases, University of Southern California, Los Angeles, California 90089, USA
| | - Meng Xia
- Department of Pathology, Department of Biochemistry and Molecular Biology, and Division of Gastroenterology and Liver Diseases, University of Southern California, Los Angeles, California 90089, USA
| | - Chen-Yin Ou
- Department of Pathology, Department of Biochemistry and Molecular Biology, and Division of Gastroenterology and Liver Diseases, University of Southern California, Los Angeles, California 90089, USA
| | - Michael R. Stallcup
- Department of Pathology, Department of Biochemistry and Molecular Biology, and Division of Gastroenterology and Liver Diseases, University of Southern California, Los Angeles, California 90089, USA
| |
Collapse
|
207
|
Feng Y, Bommer GT, Zhai Y, Akyol A, Hinoi T, Winer I, Lin HV, Cadigan KM, Cho KR, Fearon ER. Drosophila split ends homologue SHARP functions as a positive regulator of Wnt/beta-catenin/T-cell factor signaling in neoplastic transformation. Cancer Res 2007; 67:482-91. [PMID: 17234755 DOI: 10.1158/0008-5472.can-06-2314] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Wnt ligands have pleiotropic and context-specific roles in embryogenesis and adult tissues. Among other effects, certain Wnts stabilize the beta-catenin protein, leading to the ability of beta-catenin to activate T-cell factor (TCF)-mediated transcription. Mutations resulting in constitutive beta-catenin stabilization underlie development of several human cancers. Genetic studies in Drosophila highlighted the split ends (spen) gene as a positive regulator of Wnt-dependent signaling. We have assessed the role of SHARP, a human homologue of spen, in Wnt/beta-catenin/TCF function in mammalian cells. We found that SHARP gene and protein expression is elevated in human colon and ovarian endometrioid adenocarcinomas and mouse colon adenomas and carcinomas carrying gene defects leading to beta-catenin dysregulation. When ectopically expressed, the silencing mediator for retinoid and thyroid receptors/histone deacetylase 1-associated repressor protein (SHARP) protein potently enhanced beta-catenin/TCF transcription of a model reporter gene and cellular target genes. Inhibition of endogenous SHARP function via RNA inhibitory (RNAi) approaches antagonized beta-catenin/TCF-mediated activation of target genes. The effect of SHARP on beta-catenin/TCF-regulated genes was mediated via a functional interaction between SHARP and TCF. beta-Catenin-dependent neoplastic transformation of RK3E cells was enhanced by ectopic expression of SHARP, and RNAi-mediated inhibition of endogenous SHARP in colon cancer cells inhibited their transformed growth. In toto, our findings implicate SHARP as an important positive regulator of Wnt signaling in cancers with beta-catenin dysregulation.
Collapse
Affiliation(s)
- Ying Feng
- Department of Internal Medicine, The Cancer Center, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
208
|
Fillingham JS, Garg J, Tsao N, Vythilingum N, Nishikawa T, Pearlman RE. Molecular genetic analysis of an SNF2/brahma-related gene in Tetrahymena thermophila suggests roles in growth and nuclear development. EUKARYOTIC CELL 2007; 5:1347-59. [PMID: 16896218 PMCID: PMC1539136 DOI: 10.1128/ec.00149-06] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We used a reverse genetic approach to identify three members of the SNF2 superfamily of chromatin remodeling genes in the ciliated protozoan Tetrahymena thermophila in order to investigate possible functions of ATP-dependent chromatin remodeling factors in growth and nuclear development. Comparative sequence analysis of the gene product of the Tetrahymena brahma-related gene (TtBRG1) indicates it is a member of the SNF2/BRM subgroup of the SNF2 superfamily. Northern analysis suggests that TtBRG1 has roles in growth and nuclear development in Tetrahymena. Indirect immunofluorescence analysis during nuclear development indicates that TtBrg1p localizes to both the parental and developing macronucleus of Tetrahymena during the time period corresponding to genome rearrangements. We generated germ line knockout heterokaryons for TtBRG1 and demonstrated that expression of the gene is required to complete nuclear development of Tetrahymena. In addition, the formation of distinct Pdd1p-containing structures is disturbed during the late stages of conjugation in TtBRG1 germ line knockout heterokaryons. We discuss these results in light of possible roles of SNF2-related proteins in growth and nuclear development of Tetrahymena.
Collapse
Affiliation(s)
- Jeffrey S Fillingham
- Department of Biology, York University, 4700 Keele St, Toronto, Ontario, Canada M3J 1P3
| | | | | | | | | | | |
Collapse
|
209
|
Eroglu B, Wang G, Tu N, Sun X, Mivechi NF. Critical role of Brg1 member of the SWI/SNF chromatin remodeling complex during neurogenesis and neural crest induction in zebrafish. Dev Dyn 2007; 235:2722-35. [PMID: 16894598 DOI: 10.1002/dvdy.20911] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Brg1 is a member of the SWI/SNF chromatin-remodeling complex, and in some organisms Brg1 has been shown to interact with beta-catenin and positively control the TCF/LEF transcription factor that is located downstream of the Wnt signal transduction pathway. During development, TCF/LEF activity is critical during neurogenesis and head induction. In zebrafish, Brg1-deficient embryos exhibit retinal cell differentiation and eye defects; however, the role of Brg1 in neurogenesis and neural crest cell induction remains elusive. We used zebrafish deficient in Brg1 (yng) or Brg1 specific-morpholino oligonucleotide-mediated knockdown to analyze the embryonic requirements of Brg1. Our results indicate that reduction in Brg1 expression leads to the expansion of the forebrain-specific transcription factor, six3, and marked reduction in expression of the mid/hind-brain boundary and hind-brain genes, engrailed2 and krox20, respectively. At 12 hpf, the expression of neural crest specifiers are severely affected in Brg1-morpholino-injected embryos. These results suggest that Brg1 is involved in neural crest induction, which is critical for the development of neurons, glia, pigment cells, and craniofacial structures. Brg1 is a maternal factor, and brg1-deficient embryos bearing the yng mutation derived from heterozygote intercrosses exhibit lesser effects on neural crest-specific gene expression, but show defects in neurogenesis and neural crest cell differentiation. This is exhibited by the aberrant brain patterning, a reduction in the sensory neurons, and craniofacial defects. These results further elucidate the critical role for Brg1 in neurogenesis, neural crest induction, and differentiation.
Collapse
Affiliation(s)
- Binnur Eroglu
- Center for Molecular Chaperone/Radiobiology and Cancer Virology, Medical College of Georgia, Augusta, Georgia, USA
| | | | | | | | | |
Collapse
|
210
|
Abstract
Aberrant activation of the Wnt pathway is implicated in driving the formation of various human cancers, particularly those of the digestive tract. Inhibition of aberrant Wnt pathway activity in cancer cell lines efficiently blocks their growth, highlighting the great potential of therapeutics designed to achieve this in cancer patients. Here we provide an overview of the promise and pitfalls of current drug development strategies striving to inhibit the Wnt pathway and present new opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Nick Barker
- Hubrecht laboratory, Netherlands Institute for Developmental Biology, Uppsalalaan 8 3584CT, Utrecht, The Netherlands.
| | | |
Collapse
|
211
|
Parker DS, Blauwkamp T, Cadigan KM. Wnt/β‐catenin‐mediated transcriptional regulation. WNT SIGNALING IN EMBRYONIC DEVELOPMENT 2007. [DOI: 10.1016/s1574-3349(06)17001-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
212
|
Toualbi K, Güller MC, Mauriz JL, Labalette C, Buendia MA, Mauviel A, Bernuau D. Physical and functional cooperation between AP-1 and beta-catenin for the regulation of TCF-dependent genes. Oncogene 2006; 26:3492-502. [PMID: 17146436 DOI: 10.1038/sj.onc.1210133] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Stabilization of cytoplasmic beta-catenin is a hallmark of a variety of cancers. The stabilized beta-catenin is able to translocate to the nucleus, where it acts as a transcriptional activator of T-cell factor (TCF)-regulated genes. beta-Catenin may cross-talk with many signalling cascades to activate target genes. Whether beta-catenin cooperates with AP-1, another transcriptional complex activated during tumorigenesis is not fully clarified. We show that beta-catenin co-immunoprecipitates with c-Jun and c-Fos. GST pull-down experiments indicate a physical association of the armadillo repeat domain of beta-catenin with the DNA-binding domain of c-Jun and of the C-terminal domain of beta-catenin with the N-terminal domain of c-Fos. Promoter studies indicate that overexpression of AP-1 activates the transcription of two beta-catenin target genes, cyclin D1 and c-myc, by a mechanism independent of the AP-1 site, and fully dependent on the TCF-binding site. We further demonstrate that AP-1/beta-catenin synergism is involved during serum-induced cyclin D1 transcriptional activation. We identify a TCF-binding site on the cyclin D1 promoter which binds in vivo a complex induced by serum, containing beta-catenin, TCF4, c-Fos, c-Jun, JunB and JunD. This novel mechanism of interaction between two signalling cascades might contribute to the potentiation of malignancy.
Collapse
|
213
|
Ahn YT, Huang B, McPherson L, Clayberger C, Krensky AM. Dynamic interplay of transcriptional machinery and chromatin regulates "late" expression of the chemokine RANTES in T lymphocytes. Mol Cell Biol 2006; 27:253-66. [PMID: 17074812 PMCID: PMC1800668 DOI: 10.1128/mcb.01071-06] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The chemokine RANTES (regulated upon activation normal T cell expressed and secreted) is expressed "late" (3 to 5 days) after activation in T lymphocytes. In order to understand the molecular events that accompany changes in gene expression, a detailed analysis of the interplay between transcriptional machinery and chromatin on the RANTES promoter over time was undertaken. Krüppel-like factor 13 (KLF13), a sequence-specific DNA binding transcription factor, orchestrates the induction of RANTES expression in T lymphocytes by ordered recruitment of effector molecules, including Nemo-like kinase, p300/cyclic AMP response element binding protein (CBP), p300/CBP-associated factor, and Brahma-related gene 1, that initiate sequential changes in phosphorylation and acetylation of histones and ATP-dependent chromatin remodeling near the TATA box of the RANTES promoter. These events recruit RNA polymerase II to the RANTES promoter and are responsible for late expression of RANTES in T lymphocytes. Therefore, KLF13 is a key regulator of late RANTES expression in T lymphocytes.
Collapse
Affiliation(s)
- Yong-Tae Ahn
- Division of Immunology and Transplantation Biology, Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305-5164, USA
| | | | | | | | | |
Collapse
|
214
|
Hoffmans R, Basler K. BCL9-2 binds Arm/beta-catenin in a Tyr142-independent manner and requires Pygopus for its function in Wg/Wnt signaling. Mech Dev 2006; 124:59-67. [PMID: 17113272 DOI: 10.1016/j.mod.2006.09.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Revised: 09/24/2006] [Accepted: 09/26/2006] [Indexed: 11/24/2022]
Abstract
The Wingless (Wg)/Wnt signal transduction pathway controls fundamental processes during animal development. Deregulation of the Wg/Wnt pathway has been causally linked to several forms of cancer, most notably to colorectal cancer. In response to Wg/Wnt signaling, Armadillo/beta-catenin associates in the nucleus with DNA bound TCF and several co-factors, among them Legless/BCL9, which provides a link to Pygopus. Recently, the second vertebrate homologue of Legless, BCL9-2 (or B9L), was characterized and proposed to mediate Wnt signaling in a Pygopus-independent manner, by binding to a Tyrosine-142-phosphorylated form of beta-catenin. Here we examine the role of Tyrosine-142 phosphorylation in several assays and find that it is neither important for the recruitment of BCL9-2, nor for the transcriptional activity of beta-catenin in cultured mammalian cells, nor in Drosophila for Wg signaling activity in vivo. Furthermore, we demonstrate that BCL9-2 can functionally replace Lgs both in cultured cells as well as in vivo and that this rescue activity depends on the ability of BCL9-2 to bind Pygo. Our results do not show a significant functional difference between BCL9-2 and BCL9 but rather suggest that the two proteins represent evolutionary duplicates of Legless, which have acquired distinct expression patterns while acting in a largely redundant manner.
Collapse
Affiliation(s)
- Raymond Hoffmans
- Institut für Molekularbiologie, Universität Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | | |
Collapse
|
215
|
Lee YH, Stallcup MR. Interplay of Fli-I and FLAP1 for regulation of beta-catenin dependent transcription. Nucleic Acids Res 2006; 34:5052-9. [PMID: 16990252 PMCID: PMC1636430 DOI: 10.1093/nar/gkl652] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
β-catenin mediates Wnt/wingless signaling and transcriptional activation by lymphocyte enhancer binding factor 1/T cell factor (LEF1/TCF) proteins with the assistance of multiple coregulators, including positive cofactors like p300/CBP and negative cofactors like HDACs. We previously demonstrated that a developmentally essential protein, Flightless-I (Fli-I), serves as a coactivator for nuclear receptor-mediated transcription. To further understand the action mechanism of Fli-I, we investigated the functional roles of Fli-I and Fli-I leucine rich repeat associated protein 1 (FLAP1) in transcriptional activation by β-catenin and LEF1/TCF. β-catenin-dependent transcription was activated by exogenous FLAP1 but inhibited by Fli-I. Reduction of endogenous FLAP1 levels compromised transcriptional activation by LEF1/TCF, β-catenin and the p160 coactivator GRIP1. FLAP1 interacted directly with β-catenin, GRIP1 and p300 and enhanced their activity. Furthermore, FLAP1 was strongly synergistic with p300 in supporting transcriptional activation by β-catenin and LEF1/TCF, but Fli-I disrupted the synergy of FLAP1 with p300 and β-catenin. Thus the opposing effects of Fli-I and FLAP1 may be a key regulatory mechanism for β-catenin and LEF1/TCF-mediated transcription and thus for Wnt signaling, and some mutations of Fli-I may result in developmental defects, such as the flightless phenotype of Drosophila, by causing dysregulation of the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
| | - Michael R. Stallcup
- To whom correspondence should be addressed. Tel: +1 323 442 1145; Fax: +1 323 442 1224;
| |
Collapse
|
216
|
Abstract
The Wingless/Wnt pathway controls cell fates during animal development and regulates tissue homeostasis as well as stem cell number and differentiation in epithelia. Deregulation of Wnt signaling has been associated with cancer in humans. In the nucleus, the Wingless/Wnt signal is transmitted via the key effector protein Armadillo/beta-catenin. The recent identification and functional analysis of novel Armadillo/beta-catenin interaction partners provide new and exciting insights into the highly complex mechanism of Wingless/Wnt target gene activation.
Collapse
Affiliation(s)
- Reto Städeli
- These authors contributed equally. Institut für Molekularbiologie, Universität Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | | | | |
Collapse
|
217
|
Yang CK, Kim JH, Stallcup MR. Role of the N-terminal activation domain of the coiled-coil coactivator in mediating transcriptional activation by beta-catenin. Mol Endocrinol 2006; 20:3251-62. [PMID: 16931570 PMCID: PMC1770943 DOI: 10.1210/me.2006-0200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The coiled-coil coactivator (CoCoA) is involved in transcriptional activation of target genes by nuclear receptors and the xenobiotic aryl hydrocarbon receptor, as well as target genes of the Wnt signaling pathway, which is mediated by the lymphocyte enhancer factor (LEF)/T cell factor transcription factors and the coactivator beta-catenin. The recruitment of CoCoA by nuclear receptors is accomplished by the interaction of the central coiled-coiled domain of CoCoA with p160 coactivators; the C-terminal activation domain (AD) of CoCoA is used for downstream signaling, whereas the function of the N-terminal region is undefined. Here we report that the N terminus of CoCoA contains another AD, which is necessary and sufficient for synergistic activation of LEF1-mediated transcription by CoCoA and beta-catenin. The N-terminal AD contains a p300 binding motif, which is important for synergistic cooperation of CoCoA and p300 as coactivators for LEF1 and beta-catenin. p300 contributes to the function of the CoCoA N-terminal AD primarily through its histone acetyltransferase activity. Moreover, in cultured cells, endogenous p300 is recruited to the promoter of an integrated reporter gene by the N terminus of CoCoA. Thus, the coactivator function of CoCoA for nuclear receptors and LEF1/beta-catenin involves differential utilization of two different CoCoA ADs.
Collapse
Affiliation(s)
| | | | - Michael R. Stallcup
- Corresponding Author to whom reprint requests should be addressed: Michael R. Stallcup, Department of Biochemistry and Molecular Biology, University of Southern California, 1333 San Pablo Street, MCA 51A, Los Angeles, CA 90089-9151. Phone: 323-442-1289; Fax: 323-442-1224. E-mail:
| |
Collapse
|
218
|
Li TWH, Ting JHT, Yokoyama NN, Bernstein A, van de Wetering M, Waterman ML. Wnt activation and alternative promoter repression of LEF1 in colon cancer. Mol Cell Biol 2006; 26:5284-99. [PMID: 16809766 PMCID: PMC1592719 DOI: 10.1128/mcb.00105-06] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Alternative promoters within the LEF1 locus produce polypeptides of opposing biological activities. Promoter 1 produces full-length LEF-1 protein, which recruits beta-catenin to Wnt target genes. Promoter 2 produces a truncated form that cannot interact with beta-catenin and instead suppresses Wnt regulation of target genes. Here we show that promoter 1 is aberrantly activated in colon cancers because it is a direct target of the Wnt pathway. T-cell factor (TCF)-beta-catenin complexes bind to Wnt response elements in exon 1 and dynamically regulate chromatin acetylation and promoter 1 activity. Promoter 2 is delimited to the intron 2/exon 3 boundary and, like promoter 1, is also directly regulated by TCF-beta-catenin complexes. Promoter 2 is nevertheless silent in colon cancer because an upstream repressor selectively targets the basal promoter leading to destabilized TCF-beta-catenin binding. We conclude that the biological outcome of aberrant LEF1 activation in colon cancer is directed by differential promoter activation and repression.
Collapse
Affiliation(s)
- Tony W-H Li
- Department of Microbiology and Molecular Genetics, Rm. B240, Medical Sciences I, University of California, Irvine, Irvine, CA 92697-4025, USA.
| | | | | | | | | | | |
Collapse
|
219
|
Rossi A, Mukerjee R, Ferrante P, Khalili K, Amini S, Sawaya BE. Human immunodeficiency virus type 1 Tat prevents dephosphorylation of Sp1 by TCF-4 in astrocytes. J Gen Virol 2006; 87:1613-1623. [PMID: 16690926 DOI: 10.1099/vir.0.81691-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Previous examination of the effect of TCF-4 on transcription of the human immunodeficiency virus type 1 (HIV-1) promoter in human astrocytic cells found that TCF-4 affects the HIV-1 promoter through the GC-rich domain (nt -80 to nt -68). Here, the physical interaction and a functional consequence of TCF4-Sp1 contact were characterized. It was shown that expression of TCF-4 in U-87 MG (human astrocytic) cells decreased basal and Sp1-mediated transcription of the HIV-1 promoter. Results from a GST pull-down assay, as well as combined immunoprecipitation and Western blot analysis of protein extracts from U-87 MG cells, revealed an interaction of Sp1 with TCF-4. Using in vitro protein chromatography, the region of Sp1 that contacts TCF-4 was mapped to aa 266-350. It was also found that, in cell-free extracts, TCF-4 prevented dsDNA-dependent protein kinase (DNA-PK)-mediated Sp1 phosphorylation. Surprisingly, TCF-4 failed to decrease Sp1-mediated transcription of the HIV-1 long terminal repeat (LTR) and Sp1 phosphorylation in cells expressing HIV-1 Tat. Results from immunoprecipitation/Western blotting demonstrated that TCF-4 lost its ability to interact with Sp1, but not with Tat, in Tat-transfected cells. Taken together, these findings suggest that activity at the HIV-1 promoter is influenced by phosphorylation of Sp1, which is affected by Tat and DNA-PK. Interactions among TCF-4, Sp1 and/or Tat may determine the level of viral gene transcription in human astrocytic cells.
Collapse
Affiliation(s)
- Andrea Rossi
- Laboratory of Biology, Don C. Gnocchi Foundation, IRCCS, 20148 Milan, Italy
- Department of Neuroscience & Center for Neurovirology, Temple University School of Medicine, 1900 N 12th Street (015-96), Philadelphia, PA 19122, USA
| | - Ruma Mukerjee
- Department of Neuroscience & Center for Neurovirology, Temple University School of Medicine, 1900 N 12th Street (015-96), Philadelphia, PA 19122, USA
| | - Pasquale Ferrante
- Laboratory of Biology, Don C. Gnocchi Foundation, IRCCS, 20148 Milan, Italy
| | - Kamel Khalili
- Department of Neuroscience & Center for Neurovirology, Temple University School of Medicine, 1900 N 12th Street (015-96), Philadelphia, PA 19122, USA
| | - Shohreh Amini
- Department of Biology, Temple University School of Medicine, 1900 N 12th Street (015-96), Philadelphia, PA 19122, USA
- Department of Neuroscience & Center for Neurovirology, Temple University School of Medicine, 1900 N 12th Street (015-96), Philadelphia, PA 19122, USA
| | - Bassel E Sawaya
- Department of Neuroscience & Center for Neurovirology, Temple University School of Medicine, 1900 N 12th Street (015-96), Philadelphia, PA 19122, USA
| |
Collapse
|
220
|
Gunduz E, Gunduz M, Nagatsuka H, Beder L, Demircan K, Tamamura R, Hatipoglu OF, Mahmut N, Katase N, Naomoto Y, Nagai N. Epigenetic alterations of BRG1 leads to cancer development through its nuclear-cytoplasmic shuttling abnormalities. Med Hypotheses 2006; 67:1313-6. [PMID: 16824695 DOI: 10.1016/j.mehy.2006.05.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2006] [Accepted: 05/15/2006] [Indexed: 11/30/2022]
Abstract
SWI/SNF is a multiprotein chromatin remodeling complex important for gene regulation. BRG1 and its close relative BRM, have ATPase activity necessary for transcriptional regulation by conformational change of nucleosomes. Due to this role on gene expression, several members of SWI/SNF complex including BRG1 and BRM function as a tumor suppressor or negative regulator of cellular proliferation. On the other hand, the shuttling of proteins between nucleus and cytoplasm is strongly involved in the regulation of cell cycle and proliferation. Many of tumor suppressor gene (TSG)s including p53, BRCA1, ING1 play some of their functions through nucleocytoplasmic shuttling. Abnormalities related with this process abrogate the subcellular localization of the TSGs and lead to cancer development. We recently demonstrated BRG1 as a TSG in oral cancer. Our analysis also revealed an interesting finding that one of the splicing forms of BRG1 is selectively lost in cancer tissue as compared to normal counterparts. Our further analysis revealed a putative nuclear retention signal domain for this splicing form. In this article, we speculate the possible mechanism for the inactivation of BRG1 gene in oral cancer through an abnormality in its subcellular localization.
Collapse
Affiliation(s)
- Esra Gunduz
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikatacho, Okayamashi 700-8558, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
221
|
Olson LE, Tollkuhn J, Scafoglio C, Krones A, Zhang J, Ohgi KA, Wu W, Taketo MM, Kemler R, Grosschedl R, Rose D, Li X, Rosenfeld MG. Homeodomain-mediated beta-catenin-dependent switching events dictate cell-lineage determination. Cell 2006; 125:593-605. [PMID: 16678101 DOI: 10.1016/j.cell.2006.02.046] [Citation(s) in RCA: 217] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2005] [Revised: 12/13/2005] [Accepted: 02/09/2006] [Indexed: 11/24/2022]
Abstract
While the biological roles of canonical Wnt/beta-catenin signaling in development and disease are well documented, understanding the molecular logic underlying the functionally distinct nuclear transcriptional programs mediating the diverse functions of beta-catenin remains a major challenge. Here, we report an unexpected strategy for beta-catenin-dependent regulation of cell-lineage determination based on interactions between beta-catenin and a specific homeodomain factor, Prop1, rather than Lef/Tcfs. beta-catenin acts as a binary switch to simultaneously activate expression of the critical lineage-determining transcription factor, Pit1, and to repress the gene encoding the lineage-inhibiting transcription factor, Hesx1, acting via TLE/Reptin/HDAC1 corepressor complexes. The strategy of functionally distinct actions of a homeodomain factor in response to Wnt signaling is suggested to be prototypic of a widely used mechanism for generating diverse cell types from pluripotent precursor cells in response to common signaling pathways during organogenesis.
Collapse
Affiliation(s)
- Lorin E Olson
- Howard Hughes Medical Institute, Department and School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
222
|
Zhang W, Chen X, Kato Y, Evans PM, Yuan S, Yang J, Rychahou PG, Yang VW, He X, Evers BM, Liu C. Novel cross talk of Kruppel-like factor 4 and beta-catenin regulates normal intestinal homeostasis and tumor repression. Mol Cell Biol 2006; 26:2055-64. [PMID: 16507986 PMCID: PMC1430292 DOI: 10.1128/mcb.26.6.2055-2064.2006] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Epithelial cells of the intestinal mucosa undergo a continual process of proliferation, differentiation, and apoptosis which is regulated by multiple signaling pathways. The Wnt/beta-catenin pathway plays a critical role in this process. Mutations in the Wnt pathway, however, are associated with colorectal cancers. Krüppel-like factor 4 (KLF4) is an epithelial transcriptional factor that is down-regulated in many colorectal cancers. Here, we show that KLF4 interacts with beta-catenin and represses beta-catenin-mediated gene expression. Moreover, KLF4 inhibits the axis formation of Xenopus embryos and inhibits xenograft tumor growth in athymic nude mice. Our findings suggest that the cross talk of KLF4 and beta-catenin plays a critical role in homeostasis of the normal intestine as well as in tumorigenesis of colorectal cancers.
Collapse
Affiliation(s)
- Wen Zhang
- Sealy Center for Cancer Cell Biology, University of Texas Medical Branch, Galveston, TX 77555-1048, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
223
|
Takayama S, Rogatsky I, Schwarcz LE, Darimont BD. The Glucocorticoid Receptor Represses Cyclin D1 by Targeting the Tcf-β-Catenin Complex. J Biol Chem 2006; 281:17856-63. [PMID: 16644723 DOI: 10.1074/jbc.m602290200] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The ability of glucocorticoids (GCs) to regulate cell proliferation plays an important role in their therapeutic use. The canonical Wnt pathway, which promotes the proliferation of many cancers and differentiated tissues, is an emerging target for the actions of GCs, albeit existing links between these signaling pathways are indirect. By screening known Wnt target genes for their ability to respond differently to GCs in cells whose proliferation is either positively or negatively regulated by GCs, we identified c-myc, c-jun, and cyclin D1, which encode rate-limiting factors for G(1) progression of the cell cycle. Here we show that in U2OS/GR cells, which are growth-arrested by GCs, the glucocorticoid receptor (GR) represses cyclin D1 via Tcf-beta-catenin, the transcriptional effector of the canonical Wnt pathway. We demonstrate that GR can bind beta-catenin in vitro, suggesting that GC and Wnt signaling pathways are linked directly through their effectors. Down-regulation of beta-catenin by RNA interference impeded the expression of cyclin D1 but not of c-myc or c-jun and had no significant effect on the proliferation of U2OS/GR cells. Although these results revealed that beta-catenin and cyclin D1 are not essential for the regulation of U2OS/GR cell proliferation, considering the importance of the Wnt pathway for proliferation and differentiation of other cells, the repression of Tcf-beta-catenin activity by GR could open new possibilities for tissue-selective GC therapies.
Collapse
Affiliation(s)
- Sachiko Takayama
- Institute of Molecular Biology and Department of Chemistry, University of Oregon, Eugene, OR 97403-1229, USA
| | | | | | | |
Collapse
|
224
|
Abstract
The Wnt signaling pathway controls cell proliferation and body patterning throughout development. A surprising number of cytoplasmic Wnt regulators (e.g., beta-catenin, Bcl-9/Lgs, APC, Axin) also appear, often transiently, in the nucleus. beta-Catenin is an integral component of E-cadherin complexes at intercellular adherens junctions, but also recruits chromatin remodeling complexes to activate transcription in the nucleus. The APC tumor suppressor is a part of the cytoplasmic beta-catenin destruction complex, yet also counteracts beta-catenin transactivation and histone H3K4 methylation at Wnt target genes. Furthermore, APC coordinates the cyclic exchange of Wnt coregulator complexes at the DNA. These opposing roles of APC and beta-catenin enable a rapid coordination of gene expression and cytoskeletal organization throughout the cell in response to signaling.
Collapse
Affiliation(s)
- Karl Willert
- Department of Molecular and Cellular Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | | |
Collapse
|
225
|
Baba Y, Garrett KP, Kincade PW. Constitutively active beta-catenin confers multilineage differentiation potential on lymphoid and myeloid progenitors. Immunity 2006; 23:599-609. [PMID: 16356858 PMCID: PMC1850237 DOI: 10.1016/j.immuni.2005.10.009] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2005] [Revised: 09/29/2005] [Accepted: 10/19/2005] [Indexed: 01/13/2023]
Abstract
Beta-catenin-mediated Wnt signaling may contribute to the self-renewal of hematopoietic stem cells and proliferation in some malignancies. We now show that expression of constitutively active beta-catenin in normal lymphoid or myeloid progenitors generated uncommitted cells with multilineage differentiation potential. Inappropriate gene expression occurred in cells destined to produce either cell type and caused corresponding changes in their characteristics. For example, forced activation of beta-catenin quickly increased C/EBPalpha while reducing EBF and Pax-5 in lymphoid progenitors that then generated myeloid cells. Inversely, EBF dramatically increased in transduced myeloid progenitors and lymphocytes were produced. The results indicate that ectopic activation of beta-catenin destabilizes lineage fate decisions and confers some, but not all, stem cell properties on committed progenitors.
Collapse
Affiliation(s)
- Yoshihiro Baba
- Immunobiology and Cancer Program, Oklahoma Medical Research Foundation, 825 NE 13(th) Street, Oklahoma City, Oklahoma 73104, USA
| | | | | |
Collapse
|
226
|
Fang M, Li J, Blauwkamp T, Bhambhani C, Campbell N, Cadigan KM. C-terminal-binding protein directly activates and represses Wnt transcriptional targets in Drosophila. EMBO J 2006; 25:2735-45. [PMID: 16710294 PMCID: PMC1500853 DOI: 10.1038/sj.emboj.7601153] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2005] [Accepted: 04/27/2006] [Indexed: 01/27/2023] Open
Abstract
Regulation of Wnt transcriptional targets is thought to occur by a transcriptional switch. In the absence of Wnt signaling, sequence-specific DNA-binding proteins of the TCF family repress Wnt target genes. Upon Wnt stimulation, stabilized beta-catenin binds to TCFs, converting them into transcriptional activators. C-terminal-binding protein (CtBP) is a transcriptional corepressor that has been reported to inhibit Wnt signaling by binding to TCFs or by preventing beta-catenin from binding to TCF. Here, we show that CtBP is also required for the activation of some Wnt targets in Drosophila. CtBP is recruited to Wnt-regulated enhancers in a Wnt-dependent manner, where it augments Armadillo (the fly beta-catenin) transcriptional activation. We also found that CtBP is required for repression of a subset of Wnt targets in the absence of Wnt stimulation, but in a manner distinct from previously reported mechanisms. CtBP binds to Wnt-regulated enhancers in a TCF-independent manner and represses target genes in parallel with TCF. Our data indicate dual roles for CtBP as a gene-specific activator and repressor of Wnt target gene transcription.
Collapse
Affiliation(s)
- Ming Fang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Jiong Li
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Timothy Blauwkamp
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Chandan Bhambhani
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Nathan Campbell
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Ken M Cadigan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, 830 North University Avenue, Ann Arbor, MI 48109-1048, USA. Tel.: +1 734 936 3246; Fax: +1 734 647 0884; E-mail:
| |
Collapse
|
227
|
Andreu P, Colnot S, Godard C, Laurent-Puig P, Lamarque D, Kahn A, Perret C, Romagnolo B. Identification of the IFITM family as a new molecular marker in human colorectal tumors. Cancer Res 2006; 66:1949-55. [PMID: 16488993 DOI: 10.1158/0008-5472.can-05-2731] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We analyzed the expression profiles of intestinal adenomas from a new murine familial adenomatous polyposis model (Apc(delta14/+)) using suppression subtractive hybridization to identify novel diagnostic markers of colorectal carcinogenesis. We identified 18 candidate genes having increased expression levels in the adenoma. Subsequent Northern blotting, real-time reverse transcription-PCR, and in situ hybridization analysis confirmed their induction in beta-catenin-activated epithelial cells of murine adenomas. We showed that most of the genes also have altered expression levels in human colonic adenomas and carcinomas. We focused on the IFITM genes that encode IFN-inducible transmembrane proteins. Serial analyses of gene expression levels revealed high levels of expression in early and late intestinal neoplasm in both mice and humans. Using a conditional mouse model of Apc inactivation and a human colon carcinoma cell line, we showed that IFITM gene expression is rapidly induced after activation of the beta-catenin signaling. Using a large-scale analysis of human tumors, we showed that IFITM gene expression is significantly up-regulated specifically in colorectal tumors and thus may be a useful diagnostic tool in these tumors.
Collapse
Affiliation(s)
- Pauline Andreu
- Institut Cochin, INSERM U567, Centre National de la Recherche Scientifique UMR8104, Université Paris V, France
| | | | | | | | | | | | | | | |
Collapse
|
228
|
Tréand C, du Chéné I, Brès V, Kiernan R, Benarous R, Benkirane M, Emiliani S. Requirement for SWI/SNF chromatin-remodeling complex in Tat-mediated activation of the HIV-1 promoter. EMBO J 2006; 25:1690-9. [PMID: 16601680 PMCID: PMC1440843 DOI: 10.1038/sj.emboj.7601074] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2005] [Accepted: 03/08/2006] [Indexed: 11/09/2022] Open
Abstract
Activation of the human immunodeficiency virus type-1 (HIV-1) promoter in infected cells requires the sequential recruitment of several cellular factors to facilitate the formation of a processive elongation complex. The nucleosomal reorganization of the HIV-1 long terminal repeat (LTR) observed upon Tat stimulation suggests that chromatin-remodeling complexes could play a role during this process. Here, we reported that Tat interacts directly with Brm, a DNA-dependent ATPase subunit of the SWI/SNF chromatin-remodeling complex, to activate the HIV-1 LTR. Inhibition of Brm via small interfering RNAs impaired Tat-mediated transactivation of an integrated HIV-1 promoter. Furthermore, Brm is recruited in vivo to the HIV-1 LTR in a Tat-dependent manner. Interestingly, we found that Tat/Brm interaction is regulated by Tat lysine 50 acetylation. These data show the requirement of Tat-mediated recruitment of SWI/SNF chromatin-remodeling complex to HIV-1 promoter in the activation of the LTR.
Collapse
Affiliation(s)
- Céline Tréand
- Institut Cochin, Département de Génétique et Développement, Paris, France
- Inserm, U567, Paris, France
- CNRS, UMR 8104, Paris, France
- Université Paris 5, Faculté de Médecine René Descartes, UM 3, Paris, France
| | - Isaure du Chéné
- Institut de Génétique Humaine, CNRSUPR1142, Montpellier, France
| | - Vanessa Brès
- Institut de Génétique Humaine, CNRSUPR1142, Montpellier, France
| | | | - Richard Benarous
- Institut Cochin, Département de Génétique et Développement, Paris, France
- Inserm, U567, Paris, France
- CNRS, UMR 8104, Paris, France
- Université Paris 5, Faculté de Médecine René Descartes, UM 3, Paris, France
| | | | - Stéphane Emiliani
- Institut Cochin, Département de Génétique et Développement, Paris, France
- Inserm, U567, Paris, France
- CNRS, UMR 8104, Paris, France
- Université Paris 5, Faculté de Médecine René Descartes, UM 3, Paris, France
- Institut Cochin, Département Maladies Infectieuses, 27 rue du faubourg Saint Jacques, Gustave Roussy, 75014 Paris, France. Tel.: +33 1 40 51 65 76; Fax: +33 1 40 51 65 70; E-mail:
| |
Collapse
|
229
|
Mosimann C, Hausmann G, Basler K. Parafibromin/Hyrax Activates Wnt/Wg Target Gene Transcription by Direct Association with β-catenin/Armadillo. Cell 2006; 125:327-41. [PMID: 16630820 DOI: 10.1016/j.cell.2006.01.053] [Citation(s) in RCA: 241] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Revised: 11/10/2005] [Accepted: 01/24/2006] [Indexed: 12/21/2022]
Abstract
The Wnt pathway controls cell fates, tissue homeostasis, and cancer. Its activation entails the association of beta-catenin with nuclear TCF/LEF proteins and results in transcriptional activation of target genes. The mechanism by which nuclear beta-catenin controls transcription is largely unknown. Here we genetically identify a novel Wnt/Wg pathway component that mediates the transcriptional outputs of beta-catenin/Armadillo. We show that Drosophila Hyrax and its human ortholog, Parafibromin, components of the Polymerase-Associated Factor 1 (PAF1) complex, are required for nuclear transduction of the Wnt/Wg signal and bind directly to the C-terminal region of beta-catenin/Armadillo. Moreover, we find that the transactivation potential of Parafibromin/Hyrax depends on the recruitment of Pygopus to beta-catenin/Armadillo. Our results assign to the tumor suppressor Parafibromin an unexpected role in Wnt signaling and provide a molecular mechanism for Wnt target gene control, in which the nuclear Wnt signaling complex directly engages the PAF1 complex, thereby controlling transcriptional initiation and elongation by RNA Polymerase II.
Collapse
Affiliation(s)
- Christian Mosimann
- National Research Center Frontiers in Genetics, Institut für Molekularbiologie, Universität Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | | | | |
Collapse
|
230
|
Abstract
Signal transduction within the canonical Wnt/beta-catenin pathway drives development and carcinogenesis through programmed or unprogrammed changes in gene transcription. Although the upstream events linked to signal-induced activation of beta-catenin in the cytoplasm have been deciphered in considerable detail, much less is known regarding the mechanism by which beta-catenin stimulates target gene transcription in the nucleus. Here, we show that beta-catenin physically and functionally targets the MED12 subunit in Mediator to activate transcription. The beta-catenin transactivation domain bound directly to isolated MED12 and intact Mediator both in vitro and in vivo, and Mediator was recruited to Wnt-responsive genes in a beta-catenin-dependent manner. Disruption of the beta-catenin/MED12 interaction through dominant-negative interference- or RNA interference-mediated MED12 suppression inhibited beta-catenin transactivation in response to Wnt signaling. This study thus identifies the MED12 interface within Mediator as a new component and a potential therapeutic target in the Wnt/beta-catenin pathway.
Collapse
Affiliation(s)
- Seokjoong Kim
- Department of Molecular Medicine and the Institute of Biotechnology, University of Texas Health Science Center, San Antonio, Texas 78245-3207, USA
| | | | | | | |
Collapse
|
231
|
|
232
|
Sierra J, Yoshida T, Joazeiro CA, Jones KA. The APC tumor suppressor counteracts beta-catenin activation and H3K4 methylation at Wnt target genes. Genes Dev 2006; 20:586-600. [PMID: 16510874 PMCID: PMC1410807 DOI: 10.1101/gad.1385806] [Citation(s) in RCA: 321] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The APC tumor suppressor controls the stability and nuclear export of beta-catenin (beta-cat), a transcriptional coactivator of LEF-1/TCF HMG proteins in the Wnt/Wg signaling pathway. We show here that beta-cat and APC have opposing actions at Wnt target genes in vivo. The beta-cat C-terminal activation domain associates with TRRAP/TIP60 and mixed-lineage-leukemia (MLL1/MLL2) SET1-type chromatin-modifying complexes in vitro, and we show that beta-cat promotes H3K4 trimethylation at the c-Myc gene in vivo. H3K4 trimethylation in vivo requires prior ubiquitination of H2B, and we find that ubiquitin is necessary for transcription initiation on chromatin but not nonchromatin templates in vitro. Chromatin immunoprecipitation experiments reveal that beta-cat recruits Pygopus, Bcl-9/Legless, and MLL/SET1-type complexes to the c-Myc enhancer together with the negative Wnt regulators, APC, and betaTrCP. Interestingly, APC-mediated repression of c-Myc transcription in HT29-APC colorectal cancer cells is initiated by the transient binding of APC, betaTrCP, and the CtBP corepressor to the c-Myc enhancer, followed by stable binding of the TLE-1 and HDAC1 corepressors. Moreover, nuclear CtBP physically associates with full-length APC, but not with mutant SW480 or HT29 APC proteins. We conclude that, in addition to regulating the stability of beta-cat, APC facilitates CtBP-mediated repression of Wnt target genes in normal, but not in colorectal cancer cells.
Collapse
Affiliation(s)
- Jose Sierra
- Regulatory Biology Laboratory, The Salk Insitute for Biological Studies, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
233
|
Weerkamp F, Baert MRM, Naber BAE, Koster EEL, de Haas EFE, Atkuri KR, van Dongen JJM, Herzenberg LA, Staal FJT. Wnt signaling in the thymus is regulated by differential expression of intracellular signaling molecules. Proc Natl Acad Sci U S A 2006; 103:3322-6. [PMID: 16492759 PMCID: PMC1413930 DOI: 10.1073/pnas.0511299103] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Wnt signaling is essential for T cell development in the thymus, but the stages in which it occurs and the molecular mechanisms underlying Wnt responsiveness have remained elusive. Here we examined Wnt signaling activity in both human and murine thymocyte populations by determining beta-catenin levels, Tcf-reporter activation and expression of Wnt-target genes. We demonstrate that Wnt signaling occurs in all thymocyte subsets, including the more mature populations, but most prominently in the double negative (DN) subsets. This differential sensitivity to Wnt signaling was not caused by differences in the presence of Wnts or Wnt receptors, as these appeared to be expressed at comparable levels in all thymocyte subsets. Rather, it can be explained by high expression of activating signaling molecules in DN cells, e.g., beta-catenin, plakoglobin, and long forms of Tcf-1, and by low levels of inhibitory molecules. By blocking Wnt signaling from the earliest stage onwards using overexpression of Dickkopf, we show that inhibition of the canonical Wnt pathway blocks development at the most immature DN1 stage. Thus, responsiveness to developmental signals can be regulated by differential expression of intracellular mediators rather than by abundance of receptors or ligands.
Collapse
Affiliation(s)
- Floor Weerkamp
- *Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam NL-3015 GE, The Netherlands; and
| | - Miranda R. M. Baert
- *Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam NL-3015 GE, The Netherlands; and
| | - Brigitta A. E. Naber
- *Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam NL-3015 GE, The Netherlands; and
| | - Esther E. L. Koster
- *Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam NL-3015 GE, The Netherlands; and
| | - Edwin F. E. de Haas
- *Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam NL-3015 GE, The Netherlands; and
| | - Kondala R. Atkuri
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305
| | - Jacques J. M. van Dongen
- *Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam NL-3015 GE, The Netherlands; and
| | | | - Frank J. T. Staal
- *Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam NL-3015 GE, The Netherlands; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
234
|
Yang CK, Kim JH, Li H, Stallcup MR. Differential use of functional domains by coiled-coil coactivator in its synergistic coactivator function with beta-catenin or GRIP1. J Biol Chem 2006; 281:3389-97. [PMID: 16344550 PMCID: PMC1626527 DOI: 10.1074/jbc.m510403200] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
beta-Catenin, a pivotal component of the Wnt-signaling pathway, binds to and serves as a transcriptional coactivator for the T-cell factor/lymphoid enhancer factor (TCF/LEF) family of transcriptional activator proteins and for the androgen receptor (AR), a nuclear receptor. Three components of the p160 nuclear receptor coactivator complex, including CARM1, p300/CBP, and GRIP1 (one of the p160 coactivators), bind to and cooperate with beta-catenin to enhance transcriptional activation by TCF/LEF and AR. Here we report that another component of the p160 nuclear receptor coactivator complex, the coiled-coil coactivator (CoCoA), directly binds to and cooperates synergistically with beta-catenin as a coactivator for AR and TCF/LEF. CoCoA uses different domains to bind GRIP1 and beta-catenin, and it uses different domains to transmit the activating signal to the transcription machinery, depending on whether it is bound to GRIP1 or beta-catenin. CoCoA associated specifically with the promoters of transiently transfected and endogenous target genes of TCF/LEF, and reduction of the endogenous CoCoA level decreased the ability of TCF/LEF and beta-catenin to activate transcription of transient and endogenous target genes. Thus, CoCoA uses different combinations of functional domains to serve as a physiologically relevant component of the Wnt/beta-catenin signaling pathway and the androgen signaling pathway.
Collapse
Affiliation(s)
| | - Jeong Hoon Kim
- Pathology, University of Southern California, Los Angeles, California 90089
| | - Hongwei Li
- Pathology, University of Southern California, Los Angeles, California 90089
| | - Michael R. Stallcup
- Departments of Biochemistry and Molecular Biology and of
- Pathology, University of Southern California, Los Angeles, California 90089
| |
Collapse
|
235
|
Pretto D, Barco R, Rivera J, Neel N, Gustavson MD, Eid JE. The synovial sarcoma translocation protein SYT-SSX2 recruits β-catenin to the nucleus and associates with it in an active complex. Oncogene 2006; 25:3661-9. [PMID: 16462762 DOI: 10.1038/sj.onc.1209413] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Localization of beta-catenin in the cell is a key determinant in its decision to function as a critical mediator of cell adhesion at the surface or a transcription activator in the nucleus. SYT-SSX2 is the fusion product of the chromosomal translocation, t(X;18)(p11.2;q11.2), which occurs in synovial sarcoma, a soft tissue tumor. SYT-SSX2 is known to associate with chromatin remodeling complexes and is proposed to be involved in controlling gene expression. We report that SYT-SSX2 plays a direct role in beta-catenin regulation. When expressed in mammalian cells, SYT-SSX2-induced beta-catenin recruitment to the nucleus. Interestingly, known target genes of canonical Wnt were not activated as a result of SYT-SSX2 expression, nor was the nuclear localization of beta-catenin due to one of the signaling pathways normally implicated in this event. beta-Catenin accumulation in the nucleus led to the formation of a transcriptionally active nuclear complex that contained SYT-SSX2 and beta-catenin. More importantly, depletion of SYT-SSX2 in primary synovial sarcoma cells resulted in loss of nuclear beta-catenin signal and a significant decrease in its signaling activity. These results unravel a novel pathway in the control of beta-catenin cellular transport and strongly suggest that SYT-SSX2 contributes to tumor development, in part through beta-catenin signaling.
Collapse
Affiliation(s)
- D Pretto
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | |
Collapse
|
236
|
Abstract
The lymph nodes (LNs) harbor a cryptic T-lymphopoietic pathway that is dramatically amplified by oncostatin M (OM). OM-transgenic mice generate massive amounts of T lymphocytes in the absence of Lin(-)c-Kit(hi)IL-7Ralpha- lymphoid progenitors and of reticular epithelial cells. Extrathymic T cells that develop along the OM-dependent LN pathway originate from Lin(-)c-Kit(lo)IL-7Ralpha+ lymphoid progenitors and are different from classic T cells in terms of turnover kinetics and function. Positive selection does not obey the same rules in the thymus and the LNs, where positive selection of developing T cells is supported primarily by epithelial and hematopoietic cells, respectively. Extrathymic T cells undergo enhanced homeostatic proliferation and thereby acquire some properties of memory T cells. Following antigen encounter, extrathymic T-cells initiate proliferation and cytokine secretion more readily than classic T cells, but their accumulation is limited by an exquisite susceptibility to apoptosis. Studies on in vitro and in vivo extrathymic T-cell development have yielded novel insights into the essence of a primary T-lymphoid organ. Furthermore, comparison of the thymic and OM-dependent extrathymic pathways shows how the division of labor between primary and secondary lymphoid organs influences the repertoire and homeostasis of T lymphocytes.
Collapse
Affiliation(s)
- Marie-Eve Blais
- Institute of Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, Canada
| | | | | |
Collapse
|
237
|
Chai J, Charboneau AL, Betz BL, Weissman BE. Loss of the hSNF5 gene concomitantly inactivates p21CIP/WAF1 and p16INK4a activity associated with replicative senescence in A204 rhabdoid tumor cells. Cancer Res 2006; 65:10192-8. [PMID: 16288006 DOI: 10.1158/0008-5472.can-05-1896] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
hSNF5, the smallest member of the SWI/SNF chromatin remodeling complex, is lost in most malignant rhabdoid tumors (MRT). In MRT cell lines, reexpression of hSNF5 induces G1 cell cycle arrest, elevated p16INK4a, and activated replicative senescence markers, such as beta-galactosidase (beta-Gal) and plasminogen activator inhibitor-1. To compare the replicative senescence caused by hSNF5 in A204 cells to normal cellular senescence, we examined the activation of both p16INK4a and p21CIP/WAF1. Analogous to normal cellular senescence, both p16INK4a and p21CIP/WAF1 were up-regulated following hSNF5 restoration. Furthermore, we found that hSNF5 bound the p16INK4a and p21CIP/WAF1 promoters, suggesting that it directly regulates transcription of these genes. Using p16INK4a RNA interference, we showed its requirement for the replicative senescence caused by hSNF5 but not the growth arrest. Instead, p21CIP/WAF1 remained activated by hSNF5 in the absence of high p16INK4a expression, apparently causing the growth arrest in A204. Interestingly, we also found that, in the absence of p16INK4a, reexpression of hSNF5 also increased protein levels of a second cyclin-dependent kinase (CDK) inhibitor, p18INK4c. However, our data show that lack of hSNF5 does not abrogate cellular responsiveness to DNA damage or growth-inhibitory factors. In summary, our studies suggest that hSNF5 loss may influence the regulation of multiple CDK inhibitors involved in replicative senescence.
Collapse
Affiliation(s)
- Jingjing Chai
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina 27599-7295, USA
| | | | | | | |
Collapse
|
238
|
Standley HJ, Destrée O, Kofron M, Wylie C, Heasman J. Maternal XTcf1 and XTcf4 have distinct roles in regulating Wnt target genes. Dev Biol 2006; 289:318-28. [PMID: 16325796 DOI: 10.1016/j.ydbio.2005.10.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2005] [Revised: 09/23/2005] [Accepted: 10/04/2005] [Indexed: 11/21/2022]
Abstract
Wnt signaling pathways have essential roles in developing embryos and adult tissue, and alterations in their function are implicated in many disease processes including cancers. The major nuclear transducers of Wnt signals are the Tcf/LEF family of transcription factors, which have binding sites for both the transcriptional co-repressor groucho, and the co-activator beta-catenin. The early Xenopus embryo expresses three maternally inherited Tcf/LEF mRNAs, and their relative roles in regulating the expression of Wnt target genes are not understood. We have addressed this by using antisense oligonucleotides to deplete maternal XTcf1 and XTcf4 mRNAs in oocytes. We find that XTcf1 represses expression of Wnt target genes ventrally and laterally, and activates their expression dorsally. Double depletions of XTcf1 and XTcf3 suggest that they act cooperatively to repress Wnt target genes ventrally. In contrast, XTcf4 has no repressive role but is required to activate expression of Xnr3 and chordin in organizer cells at the gastrula stage. This work provides evidence for distinct roles for XTcfs in regulating Wnt target gene expression.
Collapse
Affiliation(s)
- Henrietta J Standley
- Division of Developmental Biology ML7007, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, OH 45229-3039, USA
| | | | | | | | | |
Collapse
|
239
|
Brembeck FH, Rosário M, Birchmeier W. Balancing cell adhesion and Wnt signaling, the key role of beta-catenin. Curr Opin Genet Dev 2005; 16:51-9. [PMID: 16377174 DOI: 10.1016/j.gde.2005.12.007] [Citation(s) in RCA: 523] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2005] [Accepted: 12/08/2005] [Indexed: 01/06/2023]
Abstract
Controlled regulation of cell proliferation and differentiation is essential for embryonic development and requires the coordinated regulation of cell-cell adhesion and gene transcription. The armadillo repeat protein beta-catenin is an important integrator of both processes. Beta-catenin acts in the Wnt signaling pathway, activating the transcription of crucial target genes responsible for cellular proliferation and differentiation. Beta-catenin also controls E-cadherin-mediated cell adhesion at the plasma membrane and mediates the interplay of adherens junction molecules with the actin cytoskeleton. Both functions of beta-catenin are de-regulated in human malignancies, thereby leading both to the loss of cell-cell adhesion and to the increased transcription of Wnt target genes.
Collapse
Affiliation(s)
- Felix H Brembeck
- Max Delbrueck Centre for Molecular Medicine, Robert-Roessle-Strasse 10, D-13125 Berlin, Germany
| | | | | |
Collapse
|
240
|
Rosson GB, Bartlett C, Reed W, Weissman BE. BRG1 loss in MiaPaCa2 cells induces an altered cellular morphology and disruption in the organization of the actin cytoskeleton. J Cell Physiol 2005; 205:286-94. [PMID: 15887247 DOI: 10.1002/jcp.20397] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BRG1 and Brahma are critical and mutually exclusive subunits of the multi-constituent SWI/SNF chromatin remodeling complexes. These complexes play a key role in transcriptional regulation by dynamically altering chromatin architecture. Although the two proteins are very similar in structure, murine models demonstrate a clear dichotomy in BRG1/BRM function as heterozygous loss of BRG1 results in tumor development whereas homozygous loss of BRM does not. BRG1 and/or BRM protein is absent or disrupted in approximately 17% of all human adenocarcinomas. Concomitant loss is frequent in non-small cell lung carcinomas and incurs a negative prognosis. The mechanism(s) whereby loss of BRG1 (but apparently not BRM) may contribute to tumor development and/or progression is/are ill defined. In this study, we employ MiaPaCa2, a human pancreatic adenocarcinoma cell line that lacks BRM but retains BRG1 expression to evaluate the impact of BRG1 and BRM individually on growth and tumorigenicity. We show that the MiaPaca2 cell line can apparently tolerate only very low levels of BRM after restoration of stable expression. Reduction of expression of BRG1 via shRNAi in stable clones of MiaPaCa2 results in a marked change in morphology and alterations in actin cytoskeletal organization but does not appear to exert a significant effect on in vitro growth of the cell line. Our results implicate a role for the SWI/SNF complex in the regulation of cellular differentiation.
Collapse
Affiliation(s)
- Gary B Rosson
- Department of Pathology and Laboratory Medicine, University of North Carolina, Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina 27599, USA
| | | | | | | |
Collapse
|
241
|
Behrens J. The role of the Wnt signalling pathway in colorectal tumorigenesis. Biochem Soc Trans 2005; 33:672-5. [PMID: 16042571 DOI: 10.1042/bst0330672] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Colorectal cancer (CRC) is the second largest cause of cancer-related deaths in Western countries. CRC arises from the colorectal epithelium as a result of the accumulation of genetic alterations in defined oncogenes and tumour suppressor genes. Mutations in the tumour suppressor APC (adenomatous polyposis coli) genes occur early in the development of CRC and lead to the stabilization of the Wnt pathway component beta-catenin and to the constitutive activation of Wnt signalling. Stabilizing mutations of beta-catenin can also lead to its accumulation, qualifying beta-catenin as a proto-oncogene. Here I will summarize the biochemical interactions occurring in Wnt signalling and describe how alterations in Wnt pathway components lead to CRC.
Collapse
Affiliation(s)
- J Behrens
- Nikolaus-Fiebiger-Center for Molecular Medicine, University Erlangen-Nürnberg, Glueckstr. 6, D-91054 Erlangen, Germany.
| |
Collapse
|
242
|
Hendriksen J, Fagotto F, van der Velde H, van Schie M, Noordermeer J, Fornerod M. RanBP3 enhances nuclear export of active (beta)-catenin independently of CRM1. J Cell Biol 2005; 171:785-97. [PMID: 16314428 PMCID: PMC2171279 DOI: 10.1083/jcb.200502141] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2005] [Accepted: 10/31/2005] [Indexed: 01/12/2023] Open
Abstract
beta-Catenin is the nuclear effector of the Wnt signaling cascade. The mechanism by which nuclear activity of beta-catenin is regulated is not well defined. Therefore, we used the nuclear marker RanGTP to screen for novel nuclear beta-catenin binding proteins. We identified a cofactor of chromosome region maintenance 1 (CRM1)-mediated nuclear export, Ran binding protein 3 (RanBP3), as a novel beta-catenin-interacting protein that binds directly to beta-catenin in a RanGTP-stimulated manner. RanBP3 inhibits beta-catenin-mediated transcriptional activation in both Wnt1- and beta-catenin-stimulated human cells. In Xenopus laevis embryos, RanBP3 interferes with beta-catenin-induced dorsoventral axis formation. Furthermore, RanBP3 depletion stimulates the Wnt pathway in both human cells and Drosophila melanogaster embryos. In human cells, this is accompanied by an increase of dephosphorylated beta-catenin in the nucleus. Conversely, overexpression of RanBP3 leads to a shift of active beta-catenin toward the cytoplasm. Modulation of beta-catenin activity and localization by RanBP3 is independent of adenomatous polyposis coli protein and CRM1. We conclude that RanBP3 is a direct export enhancer for beta-catenin, independent of its role as a CRM1-associated nuclear export cofactor.
Collapse
Affiliation(s)
- Jolita Hendriksen
- Department of Tumor Biology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
243
|
van Es JH, Clevers H. Notch and Wnt inhibitors as potential new drugs for intestinal neoplastic disease. Trends Mol Med 2005; 11:496-502. [PMID: 16214417 DOI: 10.1016/j.molmed.2005.09.008] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2005] [Revised: 09/02/2005] [Accepted: 09/22/2005] [Indexed: 01/22/2023]
Abstract
Colorectal cancer is a major cause of death in the western world. Recent advances in treatment comprise variations on the classical themes of surgical resection combined with chemotherapy using cytotoxic drugs and radiation therapy. Because this therapy is only moderately successful, novel approaches to the treatment of colorectal cancer are required. Our rapidly increasing knowledge of molecular signalling pathways that are deregulated in colorectal cancer might provide a platform from which to develop new rational cancer therapies. Here, we give an update on the roles of the Wnt and Notch signalling pathways in the self renewal of the intestinal epithelium and the consequences of Wnt deregulation in colorectal cancer. We focus on the potential of recently identified small-molecule inhibitors of the Wnt pathway and gamma-secretase inhibitors of the Notch pathway as novel colon cancer therapeutics.
Collapse
Affiliation(s)
- Johan H van Es
- Hubrecht Laboratory, Netherlands Institute for Developmental Biology, Uppsalalaan 8, 3584CT Utrecht, The Netherlands
| | | |
Collapse
|
244
|
Pedersen AH, Heller RS. A possible role for the canonical Wnt pathway in endocrine cell development in chicks. Biochem Biophys Res Commun 2005; 333:961-8. [PMID: 15970275 DOI: 10.1016/j.bbrc.2005.05.189] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2005] [Accepted: 05/23/2005] [Indexed: 12/27/2022]
Abstract
Wnt signalling is involved in many developmental processes such as proliferation, differentiation, cell fate decisions, and morphogenesis. However, little is known about Wnt signalling during pancreas development. Multiple Wnt ligands and Frizzled receptors are expressed in the embryonic mouse pancreas, the surrounding mesenchyme, and have also been detected in the chicken endoderm during development. The aim of this study was to investigate the role of canonical Wnt signalling on endocrine cell development by use of the in ovo electroporation of the chicken endoderm. Overexpression with a constitutive active form of beta-catenin in combination with Ngn3 resulted in reduced numbers of glucagon cells. dnLEF-1 or naked-1 did not alter endocrine cell differentiation when co-expressed with Ngn3, but dnLEF-1 appeared to have some potential for inhibiting delamination of Ngn3 cells. In addition, neuronal beta-III-tubulin, which had previously been considered a specific marker for neuronal cells, was observed in the pancreas and was upregulated in the electroporated Ngn3 cells and thus may be a new endocrine marker in the chicken.
Collapse
Affiliation(s)
- Anna Hauntoft Pedersen
- Department of Developmental Biology, Hagedorn Research Institute, Niels Steensensvej 6, DK2820 Gentofte, Denmark
| | | |
Collapse
|
245
|
Brabletz T, Hlubek F, Spaderna S, Schmalhofer O, Hiendlmeyer E, Jung A, Kirchner T. Invasion and metastasis in colorectal cancer: epithelial-mesenchymal transition, mesenchymal-epithelial transition, stem cells and beta-catenin. Cells Tissues Organs 2005; 179:56-65. [PMID: 15942193 DOI: 10.1159/000084509] [Citation(s) in RCA: 419] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Invasion by colorectal carcinomas is characterized by an epithelial-mesenchymal transition (EMT)-like dedifferentiation of the tumor cells. However, a redifferentiation towards an epithelial phenotype, resembling a mesenchymal-epithelial transition, is detectable in metastases. This indicates that malignant progression is based on dynamic processes, which cannot be explained solely by irreversible genetic alterations, but must be additionally regulated by the tumor environment. The main oncoprotein in colorectal cancer is the Wnt pathway effector beta-catenin, which is overexpressed due to mutations in the APC tumor suppressor in most cases. EMT of the tumor cells is associated with a nuclear accumulation of the transcriptional activator beta-catenin, which is reversed in metastases. Nuclear beta-catenin is involved in two fundamental processes in embryonic development: EMT and stem cell formation. Accumulating data demonstrate that aberrant nuclear expression of beta-catenin can also confer these two abilities to tumor cells, thereby driving malignant tumor progression.
Collapse
Affiliation(s)
- Thomas Brabletz
- Department of Pathology, University of Erlangen-Nurnberg, Erlangen, Germany.
| | | | | | | | | | | | | |
Collapse
|
246
|
Abstract
Wnt signaling elicits changes in gene expression and cell physiology through beta-catenin and LEF1/TCF proteins. The signal transduction pathway regulates many cellular and developmental processes, including cell proliferation, cell fate decisions and differentiation. In cells that have been stimulated by a Wnt protein, cytoplasmic beta-catenin is stabilized and transferred to the nucleus, where it interacts with the nuclear mediators of Wnt signaling, the LEF1/TCF proteins, to elicit a transcriptional response. Loss-of-function and gain-of-function experiments in the mouse have provided insight into the role of this signaling pathway in lymphopoiesis. The self-renewal and maintenance of hematopoietic stem cells is regulated by Wnt signals. Differentiation of T cells and natural killer cells is blocked in the absence of LEF1/TCF proteins, and pro-B cell proliferation is regulated by Wnt signaling.
Collapse
Affiliation(s)
- A Timm
- Gene Center and Institute of Biochemistry, University of Munich, Germany
| | | |
Collapse
|
247
|
Städeli R, Basler K. Dissecting nuclear Wingless signalling: recruitment of the transcriptional co-activator Pygopus by a chain of adaptor proteins. Mech Dev 2005; 122:1171-82. [PMID: 16169192 DOI: 10.1016/j.mod.2005.07.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2005] [Revised: 07/22/2005] [Accepted: 07/25/2005] [Indexed: 10/25/2022]
Abstract
Members of the Wingless (Wg)/Wnt family of secreted glycoproteins control cell fate during embryonic development and adult homeostasis. Wnt signals regulate the expression of target genes by activating a conserved signal transduction pathway. Upon receptor activation, the signal is transmitted intracellularly by stabilization of Armadillo (Arm)/beta-catenin. Arm/beta-catenin translocates to the nucleus, interacts with DNA-binding factors of the Pangolin (Pan)/TCF/LEF class and activates transcription of target genes in cooperation with the recently identified proteins Legless/BCL9 (Lgs) and Pygopus (Pygo). Here, we analyse the mode of action of Pan, Arm, Lgs, and Pygo in Drosophila cultured cells. We provide evidence that together these four proteins form a 'chain of adaptors' linking the NH2-terminal homology domain (NHD) of Pygo to the DNA-binding domain of Pan. We show that the NHD has potent transcriptional activation capacity, which differs from that of acidic activator domains and depends on a conserved NPF tripeptide. A single point mutation within this NPF motif abolishes the transcriptional activity of the Pygo NHD in vitro and strongly reduces Wg signalling in vivo. Together, our results suggest that the transcriptional output of Wg pathway activity largely relies on a 'chain of adaptors' design to direct the Pygo NHD to Wg target promoters in an Arm-dependent manner.
Collapse
Affiliation(s)
- Reto Städeli
- Institut für Molekularbiologie, Universität Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | | |
Collapse
|
248
|
Klebes A, Sustar A, Kechris K, Li H, Schubiger G, Kornberg TB. Regulation of cellular plasticity inDrosophilaimaginal disc cells by the Polycomb group, trithorax group andlamagenes. Development 2005; 132:3753-65. [PMID: 16077094 DOI: 10.1242/dev.01927] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Drosophila imaginal disc cells can switch fates by transdetermining from one determined state to another. We analyzed the expression profiles of cells induced by ectopic Wingless expression to transdetermine from leg to wing by dissecting transdetermined cells and hybridizing probes generated by linear RNA amplification to DNA microarrays. Changes in expression levels implicated a number of genes: lamina ancestor, CG12534 (a gene orthologous to mouse augmenter of liver regeneration), Notch pathway members, and the Polycomb and trithorax groups of chromatin regulators. Functional tests revealed that transdetermination was significantly affected in mutants for lama and seven different PcG and trxG genes. These results validate our methods for expression profiling as a way to analyze developmental programs, and show that modifications to chromatin structure are key to changes in cell fate. Our findings are likely to be relevant to the mechanisms that lead to disease when homologs of Wingless are expressed at abnormal levels and to the manifestation of pluripotency of stem cells.
Collapse
Affiliation(s)
- Ansgar Klebes
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143, USA
| | | | | | | | | | | |
Collapse
|
249
|
Jung HC, Kim K. Identification of MYCBP as a beta-catenin/LEF-1 target using DNA microarray analysis. Life Sci 2005; 77:1249-62. [PMID: 15979100 DOI: 10.1016/j.lfs.2005.02.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2004] [Accepted: 02/22/2005] [Indexed: 02/07/2023]
Abstract
Abnormal activation of the beta-catenin signaling pathway can cause various types of cancer. Activation of Wnt pathway leads to stabilization of the beta-catenin protein, which results in its translocation to the nucleus and the formation of complexes with lymphoid enhancer factor-1 (LEF-1) and other T-cell factor (TCF) family of transcription factors to affect the transcription of target genes. However, the entrapment pattern of beta-catenin in the nucleus of normal epithelial cells differs from that in colon carcinoma cells. Normal epithelial cells may have different binding partners of beta-catenin and LEF-1 compared to tumor cells, which may result in differential expression of target genes. To investigate LEF-1-induced gene expression profiles, we used DNA microarrays to search the alterations of gene expression in normal epithelia versus cancer cells. Here, we reported 10 potential targets genes of beta-catenin/LEF-1. We showed that the expression of c-myc binding protein (MYCBP) in colon carcinoma cells was consistently upregulated by overexpressed LEF-1, which is confirmed by microarray data, RT-PCR and luciferase assay. We suggest that the MYCBP gene can be a direct target of beta-catenin/LEF-1 pathway through its LEF-1 binding site(s) in the MYCBP promoter, and that MYCBP up-regulation in colon carcinoma cell may play a co-activator role of c-MYC.
Collapse
Affiliation(s)
- Ho-Chul Jung
- College of Pharmacy, Chonnam National University, Gwangju, Korea
| | | |
Collapse
|
250
|
Beiter K, Hiendlmeyer E, Brabletz T, Hlubek F, Haynl A, Knoll C, Kirchner T, Jung A. β-Catenin regulates the expression of tenascin-C in human colorectal tumors. Oncogene 2005; 24:8200-4. [PMID: 16091738 DOI: 10.1038/sj.onc.1208960] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Tenascin-C (TN-C) is a component of the extracellular matrix (ECM). It is expressed during development and re-expressed in many types of cancers, where it is involved in the modulation of adhesion and proliferation. TN-C expression is especially high at sites of epithelial mesenchymal transition (EMT), which are found frequently at the invasion front of well-differentiated human colorectal adenocarcinomas. Tumor cells in this compartment are characterized by a strong nuclear expression of the oncogenic transcription factor beta-catenin. Here, we demonstrate that TN-C is a beta-catenin target gene in human colorectal tumors. Thus, by far the most common mutations in colorectal tumors, found in the Wnt-signaling pathway and leading to the stabilizing of beta-catenin, might influence invasion by altering adhesive properties and EMT of tumor cells.
Collapse
Affiliation(s)
- Katharina Beiter
- Pathologisch-Anatomisches Institut, Friedrich-Alexander-Universität Erlangen-Nürnberg, Krankenhausstr. 8-10, D-91054 Erlangen, Germany
| | | | | | | | | | | | | | | |
Collapse
|