1
|
Özden-Yılmaz G, Savas B, Bursalı A, Eray A, Arıbaş A, Senturk S, Karaca E, Karakülah G, Erkek-Ozhan S. Differential Occupancy and Regulatory Interactions of KDM6A in Bladder Cell Lines. Cells 2023; 12:cells12060836. [PMID: 36980177 PMCID: PMC10047809 DOI: 10.3390/cells12060836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/16/2023] [Accepted: 03/01/2023] [Indexed: 03/30/2023] Open
Abstract
Epigenetic deregulation is a critical theme which needs further investigation in bladder cancer research. One of the most highly mutated genes in bladder cancer is KDM6A, which functions as an H3K27 demethylase and is one of the MLL3/4 complexes. To decipher the role of KDM6A in normal versus tumor settings, we identified the genomic landscape of KDM6A in normal, immortalized, and cancerous bladder cells. Our results showed differential KDM6A occupancy in the genes involved in cell differentiation, chromatin organization, and Notch signaling depending on the cell type and the mutation status of KDM6A. Transcription factor motif analysis revealed HES1 to be enriched at KDM6A peaks identified in the T24 bladder cancer cell line; moreover, it has a truncating mutation in KDM6A and lacks a demethylase domain. Our co-immunoprecipitation experiments revealed TLE co-repressors and HES1 as potential truncated and wild-type KDM6A interactors. With the aid of structural modeling, we explored how truncated KDM6A could interact with TLE and HES1, as well as RUNX and HHEX transcription factors. These structures provide a solid means of studying the functions of KDM6A independently of its demethylase activity. Collectively, our work provides important contributions to the understanding of KDM6A malfunction in bladder cancer.
Collapse
Affiliation(s)
| | - Busra Savas
- Izmir Biomedicine and Genome Center, Inciralti, 35340 Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Inciralti, 35340 Izmir, Turkey
| | - Ahmet Bursalı
- Izmir Biomedicine and Genome Center, Inciralti, 35340 Izmir, Turkey
| | - Aleyna Eray
- Izmir Biomedicine and Genome Center, Inciralti, 35340 Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Inciralti, 35340 Izmir, Turkey
| | - Alirıza Arıbaş
- Izmir Biomedicine and Genome Center, Inciralti, 35340 Izmir, Turkey
| | - Serif Senturk
- Izmir Biomedicine and Genome Center, Inciralti, 35340 Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Inciralti, 35340 Izmir, Turkey
| | - Ezgi Karaca
- Izmir Biomedicine and Genome Center, Inciralti, 35340 Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Inciralti, 35340 Izmir, Turkey
| | - Gökhan Karakülah
- Izmir Biomedicine and Genome Center, Inciralti, 35340 Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Inciralti, 35340 Izmir, Turkey
| | | |
Collapse
|
2
|
Haws W, England S, Grieb G, Susana G, Hernandez S, Mirer H, Lewis K. Analyses of binding partners and functional domains for the developmentally essential protein Hmx3a/HMX3. Sci Rep 2023; 13:1151. [PMID: 36670152 PMCID: PMC9859826 DOI: 10.1038/s41598-023-27878-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 01/09/2023] [Indexed: 01/22/2023] Open
Abstract
HMX3 is a homeodomain protein with essential roles in CNS and ear development. Homeodomains are DNA-binding domains and hence homeodomain-containing proteins are usually assumed to be transcription factors. However, intriguingly, our recent data suggest that zebrafish Hmx3a may not require its homeodomain to function, raising the important question of what molecular interactions mediate its effects. To investigate this, we performed a yeast two-hybrid screen and identified 539 potential binding partners of mouse HMX3. Using co-immunoprecipitation, we tested whether a prioritized subset of these interactions are conserved in zebrafish and found that Tle3b, Azin1b, Prmt2, Hmgb1a, and Hmgn3 bind Hmx3a. Next, we tested whether these proteins bind the products of four distinct hmx3a mutant alleles that all lack the homeodomain. Embryos homozygous for two of these alleles develop abnormally and die, whereas zebrafish homozygous for the other two alleles are viable. We found that all four mutations abrogate binding to Prmt2 and Tle3b, whereas Azin1b binding was preserved in all cases. Interestingly, Hmgb1a and Hmgn3 had more affinity for products of the viable mutant alleles. These data shed light on how HMX3/Hmx3a might function at a molecular level and identify new targets for future study in these vital developmental processes.
Collapse
Affiliation(s)
- William Haws
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - Samantha England
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - Ginny Grieb
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - Gabriela Susana
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - Sophie Hernandez
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - Hunter Mirer
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - Katharine Lewis
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA.
| |
Collapse
|
3
|
Shirakawa T, Toyono T, Inoue A, Matsubara T, Kawamoto T, Kokabu S. Factors Regulating or Regulated by Myogenic Regulatory Factors in Skeletal Muscle Stem Cells. Cells 2022; 11:cells11091493. [PMID: 35563799 PMCID: PMC9104119 DOI: 10.3390/cells11091493] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 11/23/2022] Open
Abstract
MyoD, Myf5, myogenin, and MRF4 (also known as Myf6 or herculin) are myogenic regulatory factors (MRFs). MRFs are regarded as master transcription factors that are upregulated during myogenesis and influence stem cells to differentiate into myogenic lineage cells. In this review, we summarize MRFs, their regulatory factors, such as TLE3, NF-κB, and MRF target genes, including non-myogenic genes such as taste receptors. Understanding the function of MRFs and the physiology or pathology of satellite cells will contribute to the development of cell therapy and drug discovery for muscle-related diseases.
Collapse
Affiliation(s)
- Tomohiko Shirakawa
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan; (T.S.); (A.I.); (T.K.)
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan;
| | - Takashi Toyono
- Division of Anatomy, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Japan;
| | - Asako Inoue
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan; (T.S.); (A.I.); (T.K.)
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan;
| | - Takuma Matsubara
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan;
| | - Tatsuo Kawamoto
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan; (T.S.); (A.I.); (T.K.)
| | - Shoichiro Kokabu
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan;
- Correspondence: ; Tel.: +81-93-582-1131; Fax: +81-93-285-6000
| |
Collapse
|
4
|
Kornspan D, Smith Y, Nechushtan H. Differential functions of TLE1 and TLE3 depending on a specific phosphorylation site. Biochem Biophys Res Commun 2021; 545:164-170. [PMID: 33571907 DOI: 10.1016/j.bbrc.2021.01.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 01/19/2021] [Indexed: 10/22/2022]
Abstract
Mammalian Transducin-like enhancer of split (TLE) confer global repression of numerous target genes in conjunction with a myriad of DNA-binding repressors. These factors have a major role in the regulation of multiple signal transduction pathways. Evidence have been obtained regarding the possible role of some of these proteins in cancer. TLE3 was suggested as a marker for increased chemosensitivity from pathological studies. Here we demonstrate, using the TCGA data base, differences in expression of this gene compared to TLE1 in several cancers. In-vitro transduction of a retrovirus encoding TLE3 to A549 lung cancer cells increased paclitaxel effectivity while TLE1 introduction to these cells decreased it. While TLE1 and TLE3 share ∼80% amino acid identity, we show that mutating or reconstituting an amino-terminal phosphorylation site, which is present only in TLE1 but absent from TLE3, and is evolutionary conserved, converts the activity of TLE1 to that of TLE3 like and vice versa. We repeated these results in an adipocytes differentiation system. Our results reveal how a single phosphorylation site can confer distinct qualitative or quantitative activities on highly homologous transcriptional regulators.
Collapse
Affiliation(s)
- David Kornspan
- Department of Oncology, Hadassah Hebrew University Medical Center, POB 12000, Jerusalem, 9112001, Israel; Department of Bacteriology, Kimron Veterinary Institute, POB 12, Bet Dagan, 50250, Israel.
| | - Yoav Smith
- Genomic Data Analysis Unit, The Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, Jerusalem, 9112001, Israel.
| | - Hovav Nechushtan
- Department of Oncology, Hadassah Hebrew University Medical Center, POB 12000, Jerusalem, 9112001, Israel.
| |
Collapse
|
5
|
Anthony CC, Robbins DJ, Ahmed Y, Lee E. Nuclear Regulation of Wnt/β-Catenin Signaling: It's a Complex Situation. Genes (Basel) 2020; 11:genes11080886. [PMID: 32759724 PMCID: PMC7465203 DOI: 10.3390/genes11080886] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/29/2020] [Accepted: 07/31/2020] [Indexed: 12/26/2022] Open
Abstract
Wnt signaling is an evolutionarily conserved metazoan cell communication pathway required for proper animal development. Of the myriad of signaling events that have been ascribed to cellular activation by Wnt ligands, the canonical Wnt/β-catenin pathway has been the most studied and best understood. Misregulation of Wnt/β-catenin signaling has been implicated in developmental defects in the embryo and major diseases in the adult. Despite the latter, no drugs that inhibit the Wnt/β-catenin pathway have been approved by the FDA. In this review, we explore the least understood step in the Wnt/β-catenin pathway-nuclear regulation of Wnt target gene transcription. We initially describe our current understanding of the importation of β-catenin into the nucleus. We then focus on the mechanism of action of the major nuclear proteins implicated in driving gene transcription. Finally, we explore the concept of a nuclear Wnt enhanceosome and propose a modified model that describes the necessary components for the transcription of Wnt target genes.
Collapse
Affiliation(s)
- Christin C. Anthony
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA;
| | - David J. Robbins
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Yashi Ahmed
- Department of Molecular and Systems Biology and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755, USA;
| | - Ethan Lee
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA;
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence:
| |
Collapse
|
6
|
Harvey S, Kumari P, Lapin D, Griebel T, Hickman R, Guo W, Zhang R, Parker JE, Beynon J, Denby K, Steinbrenner J. Downy Mildew effector HaRxL21 interacts with the transcriptional repressor TOPLESS to promote pathogen susceptibility. PLoS Pathog 2020; 16:e1008835. [PMID: 32785253 PMCID: PMC7446885 DOI: 10.1371/journal.ppat.1008835] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/24/2020] [Accepted: 07/24/2020] [Indexed: 01/22/2023] Open
Abstract
Hyaloperonospora arabidopsidis (Hpa) is an oomycete pathogen causing Arabidopsis downy mildew. Effector proteins secreted from the pathogen into the plant play key roles in promoting infection by suppressing plant immunity and manipulating the host to the pathogen's advantage. One class of oomycete effectors share a conserved 'RxLR' motif critical for their translocation into the host cell. Here we characterize the interaction between an RxLR effector, HaRxL21 (RxL21), and the Arabidopsis transcriptional co-repressor Topless (TPL). We establish that RxL21 and TPL interact via an EAR motif at the C-terminus of the effector, mimicking the host plant mechanism for recruiting TPL to sites of transcriptional repression. We show that this motif, and hence interaction with TPL, is necessary for the virulence function of the effector. Furthermore, we provide evidence that RxL21 uses the interaction with TPL, and its close relative TPL-related 1, to repress plant immunity and enhance host susceptibility to both biotrophic and necrotrophic pathogens.
Collapse
Affiliation(s)
- Sarah Harvey
- Department of Biology, University of York, York, United Kingdom
| | - Priyanka Kumari
- Institut für Phytopathologie, Universität Gießen, Gießen, Germany
| | - Dmitry Lapin
- Max Planck Institute for Plant Breeding Research, Carl-von-Linné-Weg, Cologne, Germany
- Cluster of Excellence in Plant Sciences (CEPLAS), Cologne, Germany
| | - Thomas Griebel
- Max Planck Institute for Plant Breeding Research, Carl-von-Linné-Weg, Cologne, Germany
- Dahlem Center of Plant Sciences, Plant Physiology, Freie Universität Berlin, Berlin, Germany
| | - Richard Hickman
- Department of Biology, University of York, York, United Kingdom
| | - Wenbin Guo
- The James Hutton Institute, Invergowrie, Dundee, Scotland United Kingdom
| | - Runxuan Zhang
- The James Hutton Institute, Invergowrie, Dundee, Scotland United Kingdom
| | - Jane E. Parker
- Max Planck Institute for Plant Breeding Research, Carl-von-Linné-Weg, Cologne, Germany
- Cluster of Excellence in Plant Sciences (CEPLAS), Cologne, Germany
| | - Jim Beynon
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Katherine Denby
- Department of Biology, University of York, York, United Kingdom
| | | |
Collapse
|
7
|
Ogawa M, Yaginuma T, Nakatomi C, Nakajima T, Tada-Shigeyama Y, Addison WN, Urata M, Matsubara T, Watanabe K, Matsuo K, Sato T, Honda H, Hikiji H, Watanabe S, Kokabu S. Transducin-like enhancer of split 3 regulates proliferation of melanoma cells via histone deacetylase activity. Oncotarget 2019; 10:404-414. [PMID: 30719233 PMCID: PMC6349449 DOI: 10.18632/oncotarget.26552] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 12/20/2018] [Indexed: 11/25/2022] Open
Abstract
Melanoma, one of the most aggressive neoplasms, is characterized by rapid cell proliferation. Transducin-like Enhancer of Split (TLE) is an important regulator of cell proliferation via Histone deacetylase (HDAC) recruitment. Given that HDAC activity is associated with melanoma progression, we examined the relationship between TLE3, a TLE family member, and melanoma. TLE3 expression was increased during the progression of human patient melanoma (p < 0.05). Overexpression of Tle3 in B16 murine melanoma cells led to an increase in cell proliferation (p < 0.01) as well as the number of cyclinD1-positive cells. in vivo injection of mice with B16 cells overexpressing Tle3 resulted in larger tumor formation than in mice injected with control cells (p < 0.05). In contrast, siRNA-mediated knockdown of Tle3 in B16 cells or TLE3 in HMV-II human melanoma cells decreased proliferation (p < 0.01). Treatment of B16 cells with trichostatin A (2.5 μM), a class I and II HDAC inhibitor, prevented the effect s of Tle3 on proliferation. In conclusion, these data indicate that Tle3 is required, at least in part, for proliferation in the B16 mouse melanoma model.
Collapse
Affiliation(s)
- Masahiro Ogawa
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka, Japan.,Division of Dental Anesthesiology, Department of Science of Physical Functions, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Tatsuki Yaginuma
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Chihiro Nakatomi
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Tsuyoshi Nakajima
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Yukiyo Tada-Shigeyama
- Division of Dental Anesthesiology, Department of Science of Physical Functions, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - William N Addison
- Research Unit, Shriners Hospitals for Children-Canada, Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Mariko Urata
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Takuma Matsubara
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Koji Watanabe
- Division of Developmental Stomatognathic Function Science, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Kou Matsuo
- Division of Oral Pathology, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Tsuyoshi Sato
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Saitama Medical University, Moroyama-machi, Iruma-gun, Saitama, Japan
| | - Hiromi Honda
- School of Oral Health Sciences, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Hisako Hikiji
- School of Oral Health Sciences, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Seiji Watanabe
- Division of Dental Anesthesiology, Department of Science of Physical Functions, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Shoichiro Kokabu
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| |
Collapse
|
8
|
Sena E, Rocques N, Borday C, Amin HSM, Parain K, Sitbon D, Chesneau A, Durand BC. Barhl2 maintains T-cell factors as repressors, and thereby switches off the Wnt/β-Catenin response driving Spemann organizer formation. Development 2019; 146:dev.173112. [DOI: 10.1242/dev.173112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 04/11/2019] [Indexed: 12/19/2022]
Abstract
A hallmark of Wnt/β-Catenin signaling is the extreme diversity of its transcriptional response, which varies depending on cell and developmental context. What controls this diversity is poorly understood. In all cases, the switch from transcriptional repression to activation depends on a nuclear increase in β-Catenin, which detaches the transcription factor T-cell Factor-7 like 1 (Tcf7l1) bound to Groucho (Gro) transcriptional co-repressors from its DNA binding sites and transiently converts Tcf7/Lymphoid enhancer binding factor 1 (Lef1) into a transcriptional activator. One of the earliest and evolutionarily conserved functions of Wnt/β-Catenin signaling is the induction of the blastopore lip organizer. Here, we demonstrate that the evolutionarily conserved BarH-like homeobox-2 (Barhl2) protein stabilizes the Tcf7l1-Gro complex and maintains repressed expression of Tcf target genes by a mechanism that depends on histone deacetylase 1 (Hdac-1) activity. In this way, Barhl2 switches off the Wnt/β-Catenin-dependent early transcriptional response, thereby limiting the formation of the organizer in time and/or space. This study reveals a novel nuclear inhibitory mechanism of Wnt/Tcf signaling that switches off organizer fate determination.
Collapse
Affiliation(s)
- Elena Sena
- Institut Curie, Research Division, PSL Research University, Université Paris Sud, CNRS UMR 3347, INSERM U1021, Centre Universitaire, Bâtiment 110 F-91405 Orsay Cedex
| | - Nathalie Rocques
- Institut Curie, Research Division, PSL Research University, Université Paris Sud, CNRS UMR 3347, INSERM U1021, Centre Universitaire, Bâtiment 110 F-91405 Orsay Cedex
| | - Caroline Borday
- Institut Curie, Research Division, PSL Research University, Université Paris Sud, CNRS UMR 3347, INSERM U1021, Centre Universitaire, Bâtiment 110 F-91405 Orsay Cedex
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, 91405 Orsay, France
| | - Harem Sabr Muhamad Amin
- Ecole Normale Supérieure, Institut de Biologie de l'ENS, IBENS, S1.7 CNRS 8197, INSERM U1024 46 rue d'Ulm 75005, Paris F-75005, France
| | - Karine Parain
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, 91405 Orsay, France
| | - David Sitbon
- Institut Curie, PSL Research University, CNRS, UMR3664, Equipe Labellisée Ligue contre le Cancer, Paris, France
| | - Albert Chesneau
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, 91405 Orsay, France
| | - Béatrice C. Durand
- Institut Curie, Research Division, PSL Research University, Université Paris Sud, CNRS UMR 3347, INSERM U1021, Centre Universitaire, Bâtiment 110 F-91405 Orsay Cedex
- Ecole Normale Supérieure, Institut de Biologie de l'ENS, IBENS, S1.7 CNRS 8197, INSERM U1024 46 rue d'Ulm 75005, Paris F-75005, France
| |
Collapse
|
9
|
Doenz G, Dorn S, Aghaallaei N, Bajoghli B, Riegel E, Aigner M, Bock H, Werner B, Lindhorst T, Czerny T. The function of tcf3 in medaka embryos: efficient knockdown with pePNAs. BMC Biotechnol 2018; 18:1. [PMID: 29316906 PMCID: PMC5759164 DOI: 10.1186/s12896-017-0411-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 12/11/2017] [Indexed: 12/16/2022] Open
Abstract
Background The application of antisense molecules, such as morpholino oligonucleotides, is an efficient method of gene inactivation in vivo. We recently introduced phosphonic ester modified peptide nucleic acids (PNA) for in vivo loss-of-function experiments in medaka embryos. Here we tested novel modifications of the PNA backbone to knockdown the medaka tcf3 gene. Results A single tcf3 gene exists in the medaka genome and its inactivation strongly affected eye development of the embryos, leading to size reduction and anophthalmia in severe cases. The function of Tcf3 strongly depends on co-repressor interactions. We found interactions with Groucho/Tle proteins to be most important for eye development. Using a dominant negative approach for combined inactivation of all groucho/tle genes also resulted in eye phenotypes, as did interference with three individual tle genes. Conclusions Our results show that side chain modified PNAs come close to the knockdown efficiency of morpholino oligonucleotides in vivo. A single medaka tcf3 gene combines the function of the two zebrafish paralogs hdl and tcf3b. In combination with Groucho/Tle corepressor proteins Tcf3 acts in anterior development and is critical for eye formation. Electronic supplementary material The online version of this article (10.1186/s12896-017-0411-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gerlinde Doenz
- Department for Applied Life Sciences, University of Applied Sciences, FH Campus Wien, Helmut-Qualtinger-Gasse 2, A-1030, Vienna, Austria
| | - Sebastian Dorn
- Department for Applied Life Sciences, University of Applied Sciences, FH Campus Wien, Helmut-Qualtinger-Gasse 2, A-1030, Vienna, Austria
| | - Narges Aghaallaei
- Centre for Organismal Studies (COS), University of Heidelberg, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany.,Department of Hematology, Oncology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Otfried-Mueller-Strasse 10, 72076, Tübingen, Germany
| | - Baubak Bajoghli
- Department of Hematology, Oncology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Otfried-Mueller-Strasse 10, 72076, Tübingen, Germany
| | - Elisabeth Riegel
- Department for Applied Life Sciences, University of Applied Sciences, FH Campus Wien, Helmut-Qualtinger-Gasse 2, A-1030, Vienna, Austria
| | | | - Holger Bock
- CAST Gründungszentrum GmbH, Wilhelm-Greil-Straße 15, A-6020, Innsbruck, Austria
| | - Birgit Werner
- UGISense AG, c/o Nordwind Capital GmbH, Residenzstrasse 18, 80333, München, Germany
| | - Thomas Lindhorst
- UGISense AG, c/o Nordwind Capital GmbH, Residenzstrasse 18, 80333, München, Germany
| | - Thomas Czerny
- Department for Applied Life Sciences, University of Applied Sciences, FH Campus Wien, Helmut-Qualtinger-Gasse 2, A-1030, Vienna, Austria.
| |
Collapse
|
10
|
Ramakrishnan AB, Sinha A, Fan VB, Cadigan KM. The Wnt Transcriptional Switch: TLE Removal or Inactivation? Bioessays 2017; 40. [PMID: 29250807 DOI: 10.1002/bies.201700162] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/12/2017] [Indexed: 01/06/2023]
Abstract
Many targets of the Wnt/β-catenin signaling pathway are regulated by TCF transcription factors, which play important roles in animal development, stem cell biology, and oncogenesis. TCFs can regulate Wnt targets through a "transcriptional switch," repressing gene expression in unstimulated cells and promoting transcription upon Wnt signaling. However, it is not clear whether this switch mechanism is a general feature of Wnt gene regulation or limited to a subset of Wnt targets. Co-repressors of the TLE family are known to contribute to the repression of Wnt targets in the absence of signaling, but how they are inactivated or displaced by Wnt signaling is poorly understood. In this mini-review, we discuss several recent reports that address the prevalence and molecular mechanisms of the Wnt transcription switch, including the finding of Wnt-dependent ubiquitination/inactivation of TLEs. Together, these findings highlight the growing complexity of the regulation of gene expression by the Wnt pathway.
Collapse
Affiliation(s)
| | - Abhishek Sinha
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109-1048
| | - Vinson B Fan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109-1048
| | - Ken M Cadigan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109-1048
| |
Collapse
|
11
|
Kokabu S, Nakatomi C, Matsubara T, Ono Y, Addison WN, Lowery JW, Urata M, Hudnall AM, Hitomi S, Nakatomi M, Sato T, Osawa K, Yoda T, Rosen V, Jimi E. The transcriptional co-repressor TLE3 regulates myogenic differentiation by repressing the activity of the MyoD transcription factor. J Biol Chem 2017; 292:12885-12894. [PMID: 28607151 DOI: 10.1074/jbc.m116.774570] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 06/08/2017] [Indexed: 11/06/2022] Open
Abstract
Satellite cells are skeletal muscle stem cells that provide myonuclei for postnatal muscle growth, maintenance, and repair/regeneration in adults. Normally, satellite cells are mitotically quiescent, but they are activated in response to muscle injury, in which case they proliferate extensively and exhibit up-regulated expression of the transcription factor MyoD, a master regulator of myogenesis. MyoD forms a heterodimer with E proteins through their basic helix-loop-helix domain, binds to E boxes in the genome and thereby activates transcription at muscle-specific promoters. The central role of MyoD in muscle differentiation has increased interest in finding potential MyoD regulators. Here we identified transducin-like enhancer of split (TLE3), one of the Groucho/TLE family members, as a regulator of MyoD function during myogenesis. TLE3 was expressed in activated and proliferative satellite cells in which increased TLE3 levels suppressed myogenic differentiation, and, conversely, reduced TLE3 levels promoted myogenesis with a concomitant increase in proliferation. We found that, via its glutamine- and serine/proline-rich domains, TLE3 interferes with MyoD function by disrupting the association between the basic helix-loop-helix domain of MyoD and E proteins. Our findings indicate that TLE3 participates in skeletal muscle homeostasis by dampening satellite cell differentiation via repression of MyoD transcriptional activity.
Collapse
Affiliation(s)
- Shoichiro Kokabu
- Divisions of Molecular Signaling and Biochemistry, Kyushu Dental University, Kitakyushu 803-8580, Japan; Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Saitama Medical University, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan; Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts 02115.
| | - Chihiro Nakatomi
- Divisions of Molecular Signaling and Biochemistry, Kyushu Dental University, Kitakyushu 803-8580, Japan
| | - Takuma Matsubara
- Divisions of Molecular Signaling and Biochemistry, Kyushu Dental University, Kitakyushu 803-8580, Japan
| | - Yusuke Ono
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8102, Japan
| | - William N Addison
- Research Unit, Department of Human Genetics, Shriners Hospitals for Children, McGill University, Montreal, Quebec H4A 0A9, Canada
| | - Jonathan W Lowery
- Division of Biomedical Science, College of Osteopathic Medicine, Marian University, Indianapolis, Indiana 46222
| | - Mariko Urata
- Divisions of Molecular Signaling and Biochemistry, Kyushu Dental University, Kitakyushu 803-8580, Japan
| | - Aaron M Hudnall
- Division of Biomedical Science, College of Osteopathic Medicine, Marian University, Indianapolis, Indiana 46222
| | - Suzuro Hitomi
- Division of Physiology, Kyushu Dental University, Kitakyushu 803-8580, Japan
| | - Mitsushiro Nakatomi
- Division of Anatomy, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Japan
| | - Tsuyoshi Sato
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Saitama Medical University, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan
| | - Kenji Osawa
- Division of Oral Medicine, Department of Science of Physical Functions, Kyushu Dental University, Kitakyushu 803-8580, Japan
| | - Tetsuya Yoda
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Saitama Medical University, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts 02115
| | - Eijiro Jimi
- Divisions of Molecular Signaling and Biochemistry, Kyushu Dental University, Kitakyushu 803-8580, Japan; Oral Health Brain Health Total Health, Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
12
|
Chambers M, Turki-Judeh W, Kim MW, Chen K, Gallaher SD, Courey AJ. Mechanisms of Groucho-mediated repression revealed by genome-wide analysis of Groucho binding and activity. BMC Genomics 2017; 18:215. [PMID: 28245789 PMCID: PMC5331681 DOI: 10.1186/s12864-017-3589-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 02/13/2017] [Indexed: 12/24/2022] Open
Abstract
Background The transcriptional corepressor Groucho (Gro) is required for the function of many developmentally regulated DNA binding repressors, thus helping to define the gene expression profile of each cell during development. The ability of Gro to repress transcription at a distance together with its ability to oligomerize and bind to histones has led to the suggestion that Gro may spread along chromatin. However, much is unknown about the mechanism of Gro-mediated repression and about the dynamics of Gro targeting. Results Our chromatin immunoprecipitation sequencing analysis of temporally staged Drosophila embryos shows that Gro binds in a highly dynamic manner primarily to clusters of discrete (<1 kb) segments. Consistent with the idea that Gro may facilitate communication between silencers and promoters, Gro binding is enriched at both cis-regulatory modules, as well as within the promotors of potential target genes. While this Gro-recruitment is required for repression, our data show that it is not sufficient for repression. Integration of Gro binding data with transcriptomic analysis suggests that, contrary to what has been observed for another Gro family member, Drosophila Gro is probably a dedicated repressor. This analysis also allows us to define a set of high confidence Gro repression targets. Using publically available data regarding the physical and genetic interactions between these targets, we are able to place them in the regulatory network controlling development. Through analysis of chromatin associated pre-mRNA levels at these targets, we find that genes regulated by Gro in the embryo are enriched for characteristics of promoter proximal paused RNA polymerase II. Conclusions Our findings are inconsistent with a one-dimensional spreading model for long-range repression and suggest that Gro-mediated repression must be regulated at a post-recruitment step. They also show that Gro is likely a dedicated repressor that sits at a prominent highly interconnected regulatory hub in the developmental network. Furthermore, our findings suggest a role for RNA polymerase II pausing in Gro-mediated repression. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3589-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michael Chambers
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, 90095, USA
| | - Wiam Turki-Judeh
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, 90095, USA.,Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - Min Woo Kim
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, 90095, USA
| | - Kenny Chen
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, 90095, USA
| | - Sean D Gallaher
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, 90095, USA.,Department of Energy, Institute of Genomics and Proteomics, University of California, Los Angeles, CA, 90095, USA
| | - Albert J Courey
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, 90095, USA. .,Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
13
|
Cell Fate and Differentiation of Bone Marrow Mesenchymal Stem Cells. Stem Cells Int 2016; 2016:3753581. [PMID: 27298623 PMCID: PMC4889852 DOI: 10.1155/2016/3753581] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/05/2016] [Indexed: 01/18/2023] Open
Abstract
Osteoblasts and bone marrow adipocytes originate from bone marrow mesenchymal stem cells (BMMSCs) and there appears to be a reciprocal relationship between adipogenesis and osteoblastogenesis. Alterations in the balance between adipogenesis and osteoblastogenesis in BMMSCs wherein adipogenesis is increased relative to osteoblastogenesis are associated with decreased bone quality and quantity. Several proteins have been reported to regulate this reciprocal relationship but the exact nature of the signals regulating the balance between osteoblast and adipocyte formation within the bone marrow space remains to be determined. In this review, we focus on the role of Transducin-Like Enhancer of Split 3 (TLE3), which was recently reported to regulate the balance between osteoblast and adipocyte formation from BMMSCs. We also discuss evidence implicating canonical Wnt signalling, which plays important roles in both adipogenesis and osteoblastogenesis, in regulating TLE3 expression. Currently, there is demand for new effective therapies that target the stimulation of osteoblast differentiation to enhance bone formation. We speculate that reducing TLE3 expression or activity in BMMSCs could be a useful approach towards increasing osteoblast numbers and reducing adipogenesis in the bone marrow environment.
Collapse
|
14
|
Tle1 tumor suppressor negatively regulates inflammation in vivo and modulates NF-κB inflammatory pathway. Proc Natl Acad Sci U S A 2016; 113:1871-6. [PMID: 26831087 DOI: 10.1073/pnas.1511380113] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Tle1 (transducin-like enhancer of split 1) is a corepressor that interacts with a variety of DNA-binding transcription factors and has been implicated in many cellular functions; however, physiological studies are limited. Tle1-deficient (Tle1(Δ/Δ)) mice, although grossly normal at birth, exhibit skin defects, lung hypoplasia, severe runting, poor body condition, and early mortality. Tle1(Δ/Δ) mice display a chronic inflammatory phenotype with increased expression of inflammatory cytokines and chemokines in the skin, lung, and intestine and increased circulatory IL-6 and G-CSF, along with a hematopoietic shift toward granulocyte macrophage progenitor and myeloid cells. Tle1(Δ/Δ) macrophages produce increased inflammatory cytokines in response to Toll-like receptor (TLR) agonists and lipopolysaccharides (LPS), and Tle1(Δ/Δ) mice display an enhanced inflammatory response to ear skin 12-O-tetradecanoylphorbol-13-acetate treatment. Loss of Tle1 not only results in increased phosphorylation and activation of proinflammatory NF-κB but also results in decreased Hes1 (hairy and enhancer of split-1), a negative regulator of inflammation in macrophages. Furthermore, Tle1(Δ/Δ) mice exhibit accelerated growth of B6-F10 melanoma xenografts. Our work provides the first in vivo evidence, to our knowledge, that TLE1 is a major counterregulator of inflammation with potential roles in a variety of inflammatory diseases and in cancer progression.
Collapse
|
15
|
Kwong PN, Chambers M, Vashisht AA, Turki-Judeh W, Yau TY, Wohlschlegel JA, Courey AJ. The Central Region of the Drosophila Co-repressor Groucho as a Regulatory Hub. J Biol Chem 2015; 290:30119-30. [PMID: 26483546 DOI: 10.1074/jbc.m115.681171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Indexed: 12/23/2022] Open
Abstract
Groucho (Gro) is a Drosophila co-repressor that regulates the expression of a large number of genes, many of which are involved in developmental control. Previous studies have shown that its central region is essential for function even though its three domains are poorly conserved and intrinsically disordered. Using these disordered domains as affinity reagents, we have now identified multiple embryonic Gro-interacting proteins. The interactors include protein complexes involved in chromosome organization, mRNA processing, and signaling. Further investigation of the interacting proteins using a reporter assay showed that many of them modulate Gro-mediated repression either positively or negatively. The positive regulators include components of the spliceosomal subcomplex U1 small nuclear ribonucleoprotein (U1 snRNP). A co-immunoprecipitation experiment confirms this finding and suggests that a sizable fraction of nuclear U1 snRNP is associated with Gro. The use of RNA-seq to analyze the gene expression profile of cells subjected to knockdown of Gro or snRNP-U1-C (a component of U1 snRNP) showed a significant overlap between genes regulated by these two factors. Furthermore, comparison of our RNA-seq data with Gro and RNA polymerase II ChIP data led to a number of insights, including the finding that Gro-repressed genes are enriched for promoter-proximal RNA polymerase II. We conclude that the Gro central domains mediate multiple interactions required for repression, thus functioning as a regulatory hub. Furthermore, interactions with the spliceosome may contribute to repression by Gro.
Collapse
Affiliation(s)
- Pak N Kwong
- From the Departments of Chemistry and Biochemistry and
| | | | | | - Wiam Turki-Judeh
- From the Departments of Chemistry and Biochemistry and Molecular Biology Institute, UCLA, Los Angeles, California 90095
| | - Tak Yu Yau
- From the Departments of Chemistry and Biochemistry and
| | - James A Wohlschlegel
- Biological Chemistry and Molecular Biology Institute, UCLA, Los Angeles, California 90095
| | - Albert J Courey
- From the Departments of Chemistry and Biochemistry and Molecular Biology Institute, UCLA, Los Angeles, California 90095
| |
Collapse
|
16
|
Fiedler M, Graeb M, Mieszczanek J, Rutherford TJ, Johnson CM, Bienz M. An ancient Pygo-dependent Wnt enhanceosome integrated by Chip/LDB-SSDP. eLife 2015; 4:e09073. [PMID: 26312500 PMCID: PMC4571689 DOI: 10.7554/elife.09073] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/26/2015] [Indexed: 12/15/2022] Open
Abstract
TCF/LEF factors are ancient context-dependent enhancer-binding proteins that are activated by β-catenin following Wnt signaling. They control embryonic development and adult stem cell compartments, and their dysregulation often causes cancer. β-catenin-dependent transcription relies on the NPF motif of Pygo proteins. Here, we use a proteomics approach to discover the Chip/LDB-SSDP (ChiLS) complex as the ligand specifically binding to NPF. ChiLS also recognizes NPF motifs in other nuclear factors including Runt/RUNX2 and Drosophila ARID1, and binds to Groucho/TLE. Studies of Wnt-responsive dTCF enhancers in the Drosophila embryonic midgut indicate how these factors interact to form the Wnt enhanceosome, primed for Wnt responses by Pygo. Together with previous evidence, our study indicates that ChiLS confers context-dependence on TCF/LEF by integrating multiple inputs from lineage and signal-responsive factors, including enhanceosome switch-off by Notch. Its pivotal function in embryos and stem cells explain why its integrity is crucial in the avoidance of cancer.
Collapse
Affiliation(s)
- Marc Fiedler
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Michael Graeb
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Juliusz Mieszczanek
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Trevor J Rutherford
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Christopher M Johnson
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Mariann Bienz
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
17
|
Kaul A, Schuster E, Jennings BH. The Groucho co-repressor is primarily recruited to local target sites in active chromatin to attenuate transcription. PLoS Genet 2014; 10:e1004595. [PMID: 25165826 PMCID: PMC4148212 DOI: 10.1371/journal.pgen.1004595] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 07/03/2014] [Indexed: 12/25/2022] Open
Abstract
Gene expression is regulated by the complex interaction between transcriptional activators and repressors, which function in part by recruiting histone-modifying enzymes to control accessibility of DNA to RNA polymerase. The evolutionarily conserved family of Groucho/Transducin-Like Enhancer of split (Gro/TLE) proteins act as co-repressors for numerous transcription factors. Gro/TLE proteins act in several key pathways during development (including Notch and Wnt signaling), and are implicated in the pathogenesis of several human cancers. Gro/TLE proteins form oligomers and it has been proposed that their ability to exert long-range repression on target genes involves oligomerization over broad regions of chromatin. However, analysis of an endogenous gro mutation in Drosophila revealed that oligomerization of Gro is not always obligatory for repression in vivo. We have used chromatin immunoprecipitation followed by DNA sequencing (ChIP-seq) to profile Gro recruitment in two Drosophila cell lines. We find that Gro predominantly binds at discrete peaks (<1 kilobase). We also demonstrate that blocking Gro oligomerization does not reduce peak width as would be expected if Gro oligomerization induced spreading along the chromatin from the site of recruitment. Gro recruitment is enriched in “active” chromatin containing developmentally regulated genes. However, Gro binding is associated with local regions containing hypoacetylated histones H3 and H4, which is indicative of chromatin that is not fully open for efficient transcription. We also find that peaks of Gro binding frequently overlap the transcription start sites of expressed genes that exhibit strong RNA polymerase pausing and that depletion of Gro leads to release of polymerase pausing and increased transcription at a bona fide target gene. Our results demonstrate that Gro is recruited to local sites by transcription factors to attenuate rather than silence gene expression by promoting histone deacetylation and polymerase pausing. Repression by transcription factors plays a central role in gene regulation. The Groucho/Transducin-Like Enhancer of split (Gro/TLE) family of co-repressors interacts with many different transcription factors and has many essential roles during animal development. Groucho/TLE proteins form oligomers that are necessary for target gene repression in some contexts. We have profiled the genome-wide recruitment of the founding member of this family, Groucho (from Drosophila) to gain insight into how and where it binds with respect to target genes and to identify factors associated with its binding. We find that Groucho binds in discrete peaks, frequently at transcription start sites, and that blocking Groucho from forming oligomers does not significantly change the pattern of Groucho recruitment. Although Groucho acts as a repressor, Groucho binding is enriched in chromatin that is permissive for transcription, and we find that it acts to attenuate rather than completely silence target gene expression. Thus, Groucho does not act as an “on/off” switch on target gene expression, but rather as a “mute” button.
Collapse
Affiliation(s)
- Aamna Kaul
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Eugene Schuster
- Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Barbara H. Jennings
- UCL Cancer Institute, University College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
18
|
Wheat JC, Krause DS, Shin TH, Chen X, Wang J, Ding D, Yamin R, Sweetser DA. The corepressor Tle4 is a novel regulator of murine hematopoiesis and bone development. PLoS One 2014; 9:e105557. [PMID: 25153823 PMCID: PMC4143290 DOI: 10.1371/journal.pone.0105557] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 07/23/2014] [Indexed: 11/18/2022] Open
Abstract
Hematopoiesis is a complex process that relies on various cell types, signaling pathways, transcription factors and a specific niche. The integration of these various components is of critical importance to normal blood development, as deregulation of these may lead to bone marrow failure or malignancy. Tle4, a transcriptional corepressor, acts as a tumor suppressor gene in a subset of acute myeloid leukemia, yet little is known about its function in normal and malignant hematopoiesis or in mammalian development. We report here that Tle4 knockout mice are runted and die at around four weeks with defects in bone development and BM aplasia. By two weeks of age, Tle4 knockout mice exhibit leukocytopenia, B cell lymphopenia, and significant reductions in hematopoietic stem and progenitor cells. Tle4 deficient hematopoietic stem cells are intrinsically defective in B lymphopoiesis and exhaust upon stress, such as serial transplantation. In the absence of Tle4 there is a profound decrease in bone mineralization. In addition, Tle4 knockout stromal cells are defective at maintaining wild-type hematopoietic stem cell function in vitro. In summary, we illustrate a novel and essential role for Tle4 in the extrinsic and intrinsic regulation of hematopoiesis and in bone development.
Collapse
Affiliation(s)
- Justin C. Wheat
- Department of Pediatrics, Divisions of Pediatric Hematology/Oncology and Medical Genetics, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Daniela S. Krause
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Thomas H. Shin
- Department of Pediatrics, Divisions of Pediatric Hematology/Oncology and Medical Genetics, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Molecular and Translational Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Xi Chen
- Department of Pediatrics, Divisions of Pediatric Hematology/Oncology and Medical Genetics, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Jianfeng Wang
- Department of Pediatrics, Divisions of Pediatric Hematology/Oncology and Medical Genetics, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Dacheng Ding
- Department of Pediatrics, Divisions of Pediatric Hematology/Oncology and Medical Genetics, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Rae’e Yamin
- Department of Pediatrics, Divisions of Pediatric Hematology/Oncology and Medical Genetics, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - David A. Sweetser
- Department of Pediatrics, Divisions of Pediatric Hematology/Oncology and Medical Genetics, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
19
|
Chodaparambil JV, Pate KT, Hepler MRD, Tsai BP, Muthurajan UM, Luger K, Waterman ML, Weis WI. Molecular functions of the TLE tetramerization domain in Wnt target gene repression. EMBO J 2014; 33:719-31. [PMID: 24596249 DOI: 10.1002/embj.201387188] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Wnt signaling activates target genes by promoting association of the co-activator β-catenin with TCF/LEF transcription factors. In the absence of β-catenin, target genes are silenced by TCF-mediated recruitment of TLE/Groucho proteins, but the molecular basis for TLE/TCF-dependent repression is unclear. We describe the unusual three-dimensional structure of the N-terminal Q domain of TLE1 that mediates tetramerization and binds to TCFs. We find that differences in repression potential of TCF/LEFs correlates with their affinities for TLE-Q, rather than direct competition between β-catenin and TLE for TCFs as part of an activation-repression switch. Structure-based mutation of the TLE tetramer interface shows that dimers cannot mediate repression, even though they bind to TCFs with the same affinity as tetramers. Furthermore, the TLE Q tetramer, not the dimer, binds to chromatin, specifically to K20 methylated histone H4 tails, suggesting that the TCF/TLE tetramer complex promotes structural transitions of chromatin to mediate repression.
Collapse
Affiliation(s)
- Jayanth V Chodaparambil
- Departments of Structural Biology and Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Toffolo E, Rusconi F, Paganini L, Tortorici M, Pilotto S, Heise C, Verpelli C, Tedeschi G, Maffioli E, Sala C, Mattevi A, Battaglioli E. Phosphorylation of neuronal Lysine-Specific Demethylase 1LSD1/KDM1A impairs transcriptional repression by regulating interaction with CoREST and histone deacetylases HDAC1/2. J Neurochem 2013; 128:603-16. [PMID: 24111946 DOI: 10.1111/jnc.12457] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 09/10/2013] [Accepted: 09/16/2013] [Indexed: 01/31/2023]
Abstract
Epigenetic mechanisms play important roles in brain development, orchestrating proliferation, differentiation, and morphogenesis. Lysine-Specific Demethylase 1 (LSD1 also known as KDM1A and AOF2) is a histone modifier involved in transcriptional repression, forming a stable core complex with the corepressors corepressor of REST (CoREST) and histone deacetylases (HDAC1/2). Importantly, in the mammalian CNS, neuronal LSD1-8a, an alternative splicing isoform of LSD1 including the mini-exon E8a, sets alongside LSD1 and is capable of enhancing neurite growth and morphogenesis. Here, we describe that the morphogenic properties of neuronal LSD1-8a require switching off repressive activity and this negative modulation is mediated in vivo by phosphorylation of the Thr369b residue coded by exon E8a. Three-dimensional crystal structure analysis using a phospho-mimetic mutant (Thr369bAsp), indicate that phosphorylation affects the residues surrounding the exon E8a-coded amino acids, causing a local conformational change. We suggest that phosphorylation, without affecting demethylase activity, causes in neurons CoREST and HDAC1/2 corepressors detachment from LSD1-8a and impairs neuronal LSD1-8a repressive activity. In neurons, Thr369b phosphorylation is required for morphogenic activity, converting neuronal LSD1-8a in a dominant-negative isoform, challenging LSD1-mediated transcriptional repression on target genes.
Collapse
Affiliation(s)
- Emanuela Toffolo
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Sugiyama Y, Ikeshita N, Shibahara H, Yamamoto D, Kawagishi M, Iguchi G, Iida K, Takahashi Y, Kaji H, Chihara K, Okimura Y. A PROP1-binding factor, AES cloned by yeast two-hybrid assay represses PROP1-induced Pit-1 gene expression. Mol Cell Endocrinol 2013; 376:93-8. [PMID: 23732115 DOI: 10.1016/j.mce.2013.05.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Revised: 04/22/2013] [Accepted: 05/28/2013] [Indexed: 10/26/2022]
Abstract
PROP1 mutation causes combined pituitary hormone deficiency (CPHD). Several mutations are located in a transactivation domain (TAD) of Prop1, and the loss of TAD binding to cofactors is likely the cause of CPHD. PROP1 cofactors have not yet been identified. In the present study, we aimed to identify the PROP1-interacting proteins from the human brain cDNA library. Using a yeast two-hybrid assay, we cloned nine candidate proteins that may bind to PROP1. Of those nine candidates, amino-terminal enhancer of split (AES) was the most abundant, and we analyzed the AES function. AES dose-dependently decreased the PROP1-induced Pit-1 reporter gene expression. An immunoprecipitation assay revealed the relationship between AES and PROP1. In a mammalian two-hybrid assay, a leucine zipper-like motif of the AES Q domain was identified as a region that interacted with TAD. These results indicated that AES was a corepressor of PROP1.
Collapse
Affiliation(s)
- Yuka Sugiyama
- Department of Biophysics, Kobe University Graduate School of Health Science, 7-10-2, Tomogaoka, Suma-ku, Kobe 654-0142, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
GRG5/AES interacts with T-cell factor 4 (TCF4) and downregulates Wnt signaling in human cells and zebrafish embryos. PLoS One 2013; 8:e67694. [PMID: 23840876 PMCID: PMC3698143 DOI: 10.1371/journal.pone.0067694] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 05/22/2013] [Indexed: 12/27/2022] Open
Abstract
Transcriptional control by TCF/LEF proteins is crucial in key developmental processes such as embryo polarity, tissue architecture and cell fate determination. TCFs associate with β-catenin to activate transcription in the presence of Wnt signaling, but in its absence act as repressors together with Groucho-family proteins (GRGs). TCF4 is critical in vertebrate intestinal epithelium, where TCF4-β-catenin complexes are necessary for the maintenance of a proliferative compartment, and their abnormal formation initiates tumorigenesis. However, the extent of TCF4-GRG complexes' roles in development and the mechanisms by which they repress transcription are not completely understood. Here we characterize the interaction between TCF4 and GRG5/AES, a Groucho family member whose functional relationship with TCFs has been controversial. We map the core GRG interaction region in TCF4 to a 111-amino acid fragment and show that, in contrast to other GRGs, GRG5/AES-binding specifically depends on a 4-amino acid motif (LVPQ) present only in TCF3 and some TCF4 isoforms. We further demonstrate that GRG5/AES represses Wnt-mediated transcription both in human cells and zebrafish embryos. Importantly, we provide the first evidence of an inherent repressive function of GRG5/AES in dorsal-ventral patterning during early zebrafish embryogenesis. These results improve our understanding of TCF-GRG interactions, have significant implications for models of transcriptional repression by TCF-GRG complexes, and lay the groundwork for in depth direct assessment of the potential role of Groucho-family proteins in both normal and abnormal development.
Collapse
|
23
|
Matsumura H, Kusaka N, Nakamura T, Tanaka N, Sagegami K, Uegaki K, Inoue T, Mukai Y. Crystal structure of the N-terminal domain of the yeast general corepressor Tup1p and its functional implications. J Biol Chem 2012; 287:26528-38. [PMID: 22707714 DOI: 10.1074/jbc.m112.369652] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The yeast Cyc8p-Tup1p protein complex is a general transcriptional corepressor of genes involved in many different physiological processes. Herein, we present the crystal structure of the Tup1p N-terminal domain (residues 1-92), essential for Tup1p self-assembly and interaction with Cyc8p. This domain tetramerizes to form a novel antiparallel four-helix bundle. Coiled coil interactions near the helical ends hold each dimer together, whereas interdimeric association involves only two sets of two residues located toward the chain centers. A mutagenesis study confirmed that the nonpolar residues responsible for the association of the protomers as dimers are also required for transcriptional repression. An additional structural study demonstrated that the domain containing an Leu(62) → Arg mutation that had been shown not to bind Cyc8p exhibits an altered structure, distinct from the wild type. This altered structure explains why the mutant cannot bind Cyc8p. The data presented herein highlight the importance of the architecture of the Tup1p N-terminal domain for self-association.
Collapse
Affiliation(s)
- Hiroyoshi Matsumura
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Functional features of RUNX1 mutants in acute transformation of chronic myeloid leukemia and their contribution to inducing murine full-blown leukemia. Blood 2012; 119:2873-82. [PMID: 22318203 DOI: 10.1182/blood-2011-08-370981] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The BCR-ABL fusion protein generated by t(9;22)(q34;q11) in chronic myeloid leukemia (CML) plays an essential role in the pathogenesis of the myeloproliferative disorder status at the chronic phase of the disease, but progression from the chronic phase to blast crisis (BC) is believed to require additional mutations. To explore the underlying mechanisms for BC, which is characterized by a blockage of blood cell differentiation, we screened several genes crucial to hematopoiesis and identified 10 types of mutations in RUNX1 among 11 of 85 (12.9%) patients with acute transformation of CML. Most of the mutations occurred in the runt homology domain, including H78Q, W79C, R139G, D171G, R174Q, L71fs-ter94, and V91fs-ter94. Further studies indicated that RUNX1 mutants not only exhibited decreased transactivation activity but also had an inhibitory effect on the WT RUNX1. To investigate the leukemogenic effect of mutated RUNX1, H78Q and V91fs-ter94 were transduced into 32D cells or BCR-ABL-harboring murine cells, respectively. Consistent with the myeloblastic features of advanced CML patients with RUNX1 mutations, H78Q and V91fs-ter94 disturbed myeloid differentiation and induced a BC or accelerated phase-like phenotype in mice. These results suggest that RUNX1 abnormalities may promote acute myeloid leukemic transformation in a subset of CML patients.
Collapse
|
25
|
Turki-Judeh W, Courey AJ. The unconserved groucho central region is essential for viability and modulates target gene specificity. PLoS One 2012; 7:e30610. [PMID: 22319573 PMCID: PMC3272004 DOI: 10.1371/journal.pone.0030610] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 12/26/2011] [Indexed: 12/31/2022] Open
Abstract
Groucho (Gro) is a Drosophila corepressor required by numerous DNA-binding repressors, many of which are distributed in gradients and provide positional information during development. Gro contains well-conserved domains at its N- and C-termini, and a poorly conserved central region that includes the GP, CcN, and SP domains. All lethal point mutations in gro map to the conserved regions, leading to speculation that the unconserved central domains are dispensable. However, our sequence analysis suggests that the central domains are disordered leading us to suspect that the lack of lethal mutations in this region reflects a lack of order rather than an absence of essential functions. In support of this conclusion, genomic rescue experiments with Gro deletion variants demonstrate that the GP and CcN domains are required for viability. Misexpression assays using these same deletion variants show that the SP domain prevents unrestrained and promiscuous repression by Gro, while the GP and CcN domains are indispensable for repression. Deletion of the GP domain leads to loss of nuclear import, while deletion of the CcN domain leads to complete loss of repression. Changes in Gro activity levels reset the threshold concentrations at which graded repressors silence target gene expression. We conclude that co-regulators such as Gro are not simply permissive components of the repression machinery, but cooperate with graded DNA-binding factors in setting borders of gene expression. We suspect that disorder in the Gro central domains may provide the flexibility that allows this region to mediate multiple interactions required for repression.
Collapse
Affiliation(s)
- Wiam Turki-Judeh
- Department of Chemistry and Biochemistry and Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Albert J. Courey
- Department of Chemistry and Biochemistry and Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
26
|
Abstract
Wnts are conserved, secreted signaling proteins that can influence cell behavior by stabilizing β-catenin. Accumulated β-catenin enters the nucleus, where it physically associates with T-cell factor (TCF) family members to regulate target gene expression in many developmental and adult tissues. Recruitment of β-catenin to Wnt response element (WRE) chromatin converts TCFs from transcriptional repressors to activators. This review will outline the complex interplay between factors contributing to TCF repression and coactivators working with β-catenin to regulate Wnt targets. In addition, three variations of the standard transcriptional switch model will be discussed. One is the Wnt/β-catenin symmetry pathway in Caenorhabditis elegans, where Wnt-mediated nuclear efflux of TCF is crucial for activation of targets. Another occurs in vertebrates, where distinct TCF family members are associated with repression and activation, and recent evidence suggests that Wnt signaling facilitates a "TCF exchange" on WRE chromatin. Finally, a "reverse switch" mechanism for target genes that are directly repressed by Wnt/β-catenin signaling occurs in Drosophila cells. The diversity of TCF regulatory mechanisms may help to explain how a small group of transcription factors can function in so many different contexts to regulate target gene expression.
Collapse
Affiliation(s)
- Ken M Cadigan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
27
|
Abstract
Drosophila Groucho (Gro) is the founding member of a family of metazoan corepressors. Gro mediates repression through interactions with a myriad of DNA-binding repressor proteins to direct the silencing of genes involved in many developmental processes, including neurogenesis and patterning of the main body axis, as well as receptor tyrosine kinase/Ras/MAPK, Notch, Wingless (Wg)/Wnt, and Decapentaplegic (Dpp) signaling. Gro mediates repression by multiple molecular mechanisms, depending on the regulatory context. Because Gro is a broadly expressed nuclear factor, whereas its repressor partners display restricted temporal and spatial distribution, it was presumed that this corepressor played permissive rather than instructive roles in development. However, a wide range of studies demonstrates that this is not the case. Gro can sense and integrate many cellular inputs to modulate the expression of variety of genes, making it a versatile corepressor with crucial instructive roles in development and signaling.
Collapse
Affiliation(s)
- Wiam Turki-Judeh
- Department of Chemistry & Biochemistry and Molecular Biology Institute, University of California, Los Angeles, California, USA
| | | |
Collapse
|
28
|
Abstract
The human Transducin-like Enhancer of Split (TLE) and mouse homologue, Groucho gene-related protein (GRG), represent a family of conserved non-DNA binding transcriptional modulatory proteins divided into two subgroups based upon size. The long TLE/GRGs consist of four pentadomain proteins that are dedicated co-repressors for multiple transcription factors (TF). The second TLE/GRG subgroup is composed of the Amino-terminal Enhancer of Split (AES) in humans and its mouse homolog GRG5 (AES/GRG5). In contrast to the dedicated co-repressor function of long TLE/GRGs, AES/GRG5 can both positively or negatively modulate various TF as well as non-TF proteins in a long TLE/GRG-dependent or -independent manner. Therefore, AES/GRG5 is a functionally dynamic protein that is not exclusively defined by its role as a long TLE/GRG antagonist. AES/GRG5 may function in various developmental and pathological processes but the functional characteristics of endogenous AES/GRG5 in a physiologically relevant context remains to be determined. Developmental Dynamics 239:2795–2805, 2010. © 2010 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Brandon Beagle
- Department of Anesthesiology, University of Rochester, Rochester, New York 14642, USA
| | | |
Collapse
|
29
|
Wang S, Du J, Tang H, Ding X, Zha M, Xu Z. Expression, purification, crystallization, and preliminary X-ray diffraction analysis of the human TLE1 Q domain. Acta Biochim Biophys Sin (Shanghai) 2011; 43:149-53. [PMID: 21183761 DOI: 10.1093/abbs/gmq116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Human transducin-like enhancer of split 1 (TLE1) plays crucial roles in a number of developmental processes and is involved in pathogenesis of malignancy tumors. The N-terminal glutamine-rich domain (Q domain) of TLE1 mediates its tetramerization and interactions with different DNA-binding transcription factors to regulate Notch and Wnt signaling pathways. To better understand the molecular mechanism of TLE1's functions in these pathways, we cloned, purified, and crystallized the TLE1 Q domain (TLE1-Q). The crystals belong to space group C222(1), with the complete diffraction data of the native and Se-Met TLE1-Q collected to 3.5 and 4.1 Å resolutions, respectively. The phasing-solving and model building are in progress.
Collapse
Affiliation(s)
- Su Wang
- Department of Thoracic and Cardiovascular Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
30
|
Varlakhanova N, Hahm JB, Privalsky ML. Regulation of SMRT corepressor dimerization and composition by MAP kinase phosphorylation. Mol Cell Endocrinol 2011; 332:180-8. [PMID: 20965228 PMCID: PMC3011023 DOI: 10.1016/j.mce.2010.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Revised: 09/30/2010] [Accepted: 10/12/2010] [Indexed: 10/18/2022]
Abstract
The SMRT (Silencing Mediator of Retinoid and Thyroid hormone receptors) corepressor mediates gene repression by nuclear receptors and other transcriptional factors. The SMRT protein serves as a key nucleating core that organizes the assembly of a larger corepressor complex. We report here that SMRT interacts with itself to form a protein dimer, and that Erk2, a mitogen-activated protein (MAP) kinase, disrupts this SMRT self-dimerization in vitro and in vivo. Notably Erk2 phosphorylation also results in a re-organization of the overall corepressor complex, characterized by a reduced sedimentation coefficient, partial release of HDAC3, TBL-1, and TBLR-1, and inhibition of transcriptional repression. We propose that SMRT dimers form the central platform on which additional corepressor components assemble, and that kinase signaling modifies the architecture, composition, and function of this complex. These observations contribute to our understanding of how the SMRT corepressor complex assembles and is regulated during cell proliferation and differentiation.
Collapse
Affiliation(s)
- Natalia Varlakhanova
- Department of Microbiology, College of Biological Sciences, University of California at Davis, United States
| | | | | |
Collapse
|
31
|
Payankaulam S, Li LM, Arnosti DN. Transcriptional repression: conserved and evolved features. Curr Biol 2011; 20:R764-71. [PMID: 20833321 DOI: 10.1016/j.cub.2010.06.037] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The regulation of gene expression by transcriptional repression is an ancient and conserved mechanism that manifests itself in diverse ways. Here we summarize conserved pathways for transcriptional repression prevalent throughout all forms of life, as well as indirect mechanisms that appear to have originated in eukaryotes, consistent with the unique chromatin environment of eukaryotic genes. The direct interactions between transcriptional repressors and the core transcriptional machinery in bacteria and archaea are sufficient to generate a sophisticated suite of mechanisms that provide flexible control. These direct interactions contrast with the activity of corepressors, which provide an additional regulatory control in eukaryotes. Their modulation of chromatin structure represents an indirect pathway to downregulate transcription, and their diversity and modulation provide additional complexity suited to the requirements of elaborate eukaryotic repression patterns. New findings indicate that corepressors are not necessarily restricted to generating a single stereotypic output, but can rather exhibit diverse functional responses depending on the context in which they are recruited, providing a hitherto unsuspected additional source of diversity in transcriptional control. Mechanisms within eukaryotes appear to be highly conserved, with novel aspects chiefly represented by addition of lineage-specific corepressor scaffolds that provide additional opportunities for recruiting the same core machinery.
Collapse
Affiliation(s)
- Sandhya Payankaulam
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48910, USA
| | | | | |
Collapse
|
32
|
Wang S, Xu Z, Tang H, Wei L, Zhao X. [Prokaryotic expression and purification of human TLE1 N-terminal Q domain fragment and production of its polyclonal antibody]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2010; 13:1004-8. [PMID: 21081038 PMCID: PMC6000491 DOI: 10.3779/j.issn.1009-3419.2010.11.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
背景与目的 TLE1是参与Wnt、Notch以及EGFR信号通路调控的一种重要蛋白。TLE1 N端Q结构域片段通过介导自身寡聚化及与LEF1结合发挥调控作用。本实验旨在构建人TLE1 N端Q区基因在大肠杆菌的融合表达载体,制备并纯化人TLE1 N端Q结构域蛋白片段,制备TLE1 Q结构域多克隆抗体。 方法 以人肺腺癌cDNA库为模板聚合酶链反应(polymerase chain reaction, PCR)特异性扩增TLE1-Q(1-136)基因序列,与pGEX-4T-1质粒连接后转化感受态大肠杆菌E.coli。以异丙基-β-D-硫代半乳糖苷(isopropyl-β-D thiogalactoside, IPTG)诱导表达产生融合蛋白GSTTLE1-Q(1-136)。经亲和层析,Thrombin酶切,FPLC纯化,SDS-PAGE鉴定目的蛋白TLE1-Q(1-136)。免疫家兔,制备多克隆抗体。 结果 测序证实重组表达质粒中的人TLE1 N端Q结构域基因序列正确,成功构建表达型重组质粒pGEX-4T1-TLE1-Q。重组质粒转化大肠杆菌C+后,经诱导,重组蛋白GST-TLE1-Q(1-136)得到表达。SDS-PAGE鉴定示纯化蛋白为目的蛋白人TLE1 N端Q结构域片段TLE1-Q(1-136)。免疫家兔后收获抗血清,ELISA显示抗体效价为1:20 000,具有高度特异性。免疫印迹结果显示,制备的多抗可与TLE1-Q(1-136)蛋白特异性结合。 结论 成功构建了人TLE1 N端Q结构域重组融合蛋白表达质粒pGEX-4T1-TLE1-Q,表达纯化了稳定可溶TLE1 N端Q结构域蛋白TLE1-Q(1-136),制备了人TLE1 N端Q结构域蛋白片段多克隆抗体,为进一步研究TLE1在肺癌生成中的作用奠定了基础。
Collapse
Affiliation(s)
- Su Wang
- Department of Thoracic Surgery, Changzheng Hospital, the Second Military Medical University, Shanghai 200003, China
| | | | | | | | | |
Collapse
|
33
|
Differential modulation of TCF/LEF-1 activity by the soluble LRP6-ICD. PLoS One 2010; 5:e11821. [PMID: 20676368 PMCID: PMC2911377 DOI: 10.1371/journal.pone.0011821] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Accepted: 07/05/2010] [Indexed: 12/28/2022] Open
Abstract
The canonical Wnt/β-catenin (Wnt) pathway is a master transcriptional regulatory signaling pathway that controls numerous biological processes including proliferation and differentiation. As such, transcriptional activity of the Wnt pathway is tightly regulated and/or modulated by numerous proteins at the level of the membrane, cytosol and/or nucleus. In the nucleus, transcription of Wnt target genes by TCF/LEF-1 is repressed by the long Groucho/TLE co-repressor family. However, a truncated member of the Groucho/TLE family, amino terminal enhancer of Split (AES) can positively modulate TCF/LEF-1 activity by antagonizing long Groucho/TLE members in a dominant negative manner. We have previously shown the soluble intracellular domain of the LRP6 receptor, a receptor required for activation of the Wnt pathway, can positively regulate transcriptional activity within the Wnt pathway. In the current study, we show the soluble LRP6 intracellular domain (LRP6-ICD) can also translocate to the nucleus in CHO and HEK 293T cells and in contrast to cytosolic LRP6-ICD; nuclear LRP6-ICD represses TCF/LEF-1 activity. In agreement with previous reports, we show AES enhances TCF/LEF-1 mediated reporter transcription and further we demonstrate that AES activity is spatially regulated in HEK 293T cells. LRP6-ICD interacts with AES exclusively in the nucleus and represses AES mediated TCF/LEF-1 reporter transcription. These results suggest that LRP6-ICD can differentially modulate Wnt pathway transcriptional activity depending upon its subcellular localization and differential protein-protein interactions.
Collapse
|
34
|
Flowers EB, Poole RJ, Tursun B, Bashllari E, Pe'er I, Hobert O. The Groucho ortholog UNC-37 interacts with the short Groucho-like protein LSY-22 to control developmental decisions in C. elegans. Development 2010; 137:1799-805. [PMID: 20431118 DOI: 10.1242/dev.046219] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Transcriptional co-repressors of the Groucho/TLE family are important regulators of development in many species. A subset of Groucho/TLE family members that lack the C-terminal WD40 domains have been proposed to act as dominant-negative regulators of Groucho/TLE proteins, yet such a role has not been conclusively proven. Through a mutant screen for genes controlling a left/right asymmetric cell fate decision in the nervous system of the nematode C. elegans, we have retrieved loss-of-function alleles in two distinct loci that display identical phenotypes in neuronal fate specification and in other developmental contexts. Using the novel technology of whole-genome sequencing, we find that these loci encode the C. elegans ortholog of Groucho, UNC-37, and, surprisingly, a short Groucho-like protein, LSY-22, that is similar to truncated Groucho proteins in other species. Besides their phenotypic similarities, unc-37 and lsy-22 show genetic interactions and UNC-37 and LSY-22 proteins also physically bind to each other in vivo. Our findings suggest that rather than acting as negative regulators of Groucho, small Groucho-like proteins may promote Groucho function. We propose that Groucho-mediated gene regulatory events involve heteromeric complexes of distinct Groucho-like proteins.
Collapse
Affiliation(s)
- Eileen B Flowers
- Department of Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute, Columbia University Medical Center, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | |
Collapse
|
35
|
Structural and functional analysis of amino-terminal enhancer of split in androgen-receptor-driven transcription. Biochem J 2010; 427:499-511. [PMID: 20163360 DOI: 10.1042/bj20091631] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We previously demonstrated that the Groucho protein AES (amino-terminal enhancer of split) functions as a co-repressor of the AR (androgen receptor). It physically interacts with the N-terminal domain of AR and inhibits AR-driven transcription, but the molecular mechanism of its action remained unclear. In the present paper we report that the AES protein contains one inhibitory domain, and one positive and one negative regulatory domain. The negative regulatory domain inhibits AES dimerization and AES-mediated inhibition of AR-driven transcription through an interaction with the inhibitory domain. The positive regulatory domain blocked this interaction and relieved the inhibitory effect. In addition, we discovered mechanisms by which AES regulates AR transcriptional activity, which included disruption of the interaction between the AR N-terminal and C-terminal domains, and inhibition of AR-DNA interaction. Although AES broadly inhibited the activity of androgen-dependent luciferase reporters in a transient transfection assay, it selectively regulated the expression of endogenous androgen-dependent genes in prostate cancer cells.
Collapse
|
36
|
Winkler CJ, Ponce A, Courey AJ. Groucho-mediated repression may result from a histone deacetylase-dependent increase in nucleosome density. PLoS One 2010; 5:e10166. [PMID: 20405012 PMCID: PMC2854148 DOI: 10.1371/journal.pone.0010166] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 03/21/2010] [Indexed: 01/19/2023] Open
Abstract
Groucho (Gro) is a Drosophila melanogaster transcriptional corepressor that directly interacts with the histone deacetylase Rpd3. Although previous studies suggest that this interaction is required for repression of Gro-responsive reporters in cultured cells, the in vivo significance of this interaction and the mechanism by which it leads to repression remain largely unexplored. In this study, we show that Gro is partially dependent on Rpd3 for repression, supporting the idea that Rpd3-mediated repression is one mode of Gro-mediated repression. We demonstrate that Gro colocalizes with Rpd3 to the chromatin of a target gene and that this is accompanied by the deacetylation of specific lysines within the N-terminal tails of histones H3 and H4. Gro overexpression leads to wing patterning defects and ectopic repression in the wing disc of transcription directed by the vestigial quadrant enhancer. These effects are reversed by the histone deacetylase inhibitors TSA and HC-Toxin and by the reduction of Rpd3 gene dosage. Furthermore, repression of the vestigial quadrant enhancer is accompanied by a Gro-mediated increase in nucleosome density, an effect that is reversed by histone deacetylase inhibitors. We propose a model in which Gro-mediated histone deacetylation results in increased nucleosome density leading to transcriptional repression.
Collapse
Affiliation(s)
- Clint J. Winkler
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Alberto Ponce
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Albert J. Courey
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
37
|
Boeke J, Regnard C, Cai W, Johansen J, Johansen KM, Becker PB, Imhof A. Phosphorylation of SU(VAR)3-9 by the chromosomal kinase JIL-1. PLoS One 2010; 5:e10042. [PMID: 20386606 PMCID: PMC2850320 DOI: 10.1371/journal.pone.0010042] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 03/17/2010] [Indexed: 11/23/2022] Open
Abstract
The histone methyltransferase SU(VAR)3–9 plays an important role in the formation of heterochromatin within the eukaryotic nucleus. Several studies have shown that the formation of condensed chromatin is highly regulated during development, suggesting that SU(VAR)3–9's activity is regulated as well. However, no mechanism by which this may be achieved has been reported so far. As we and others had shown previously that the N-terminus of SU(VAR)3–9 plays an important role for its activity, we purified interaction partners from Drosophila embryo nuclear extract using as bait a GST fusion protein containing the SU(VAR)3–9 N-terminus. Among several other proteins known to bind Su(VAR)3–9 we isolated the chromosomal kinase JIL-1 as a strong interactor. We show that SU(VAR)3–9 is a substrate for JIL-1 in vitro as well as in vivo and map the site of phosphorylation. These findings may provide a molecular explanation for the observed genetic interaction between SU(VAR)3–9 and JIL-1.
Collapse
Affiliation(s)
- Joern Boeke
- Adolf-Butenandt Institute and Munich Center of Integrated Protein Science (CIPS), Ludwig Maximilians University of Munich, Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
38
|
Alternative splicing of the histone demethylase LSD1/KDM1 contributes to the modulation of neurite morphogenesis in the mammalian nervous system. J Neurosci 2010; 30:2521-32. [PMID: 20164337 DOI: 10.1523/jneurosci.5500-09.2010] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
A variety of chromatin remodeling complexes are thought to orchestrate transcriptional programs that lead neuronal precursors from earliest commitment to terminal differentiation. Here we show that mammalian neurons have a specialized chromatin remodeling enzyme arising from a neurospecific splice variant of LSD1/KDM1, histone lysine specific demethylase 1, whose demethylase activity on Lys4 of histone H3 has been related to gene repression. We found that alternative splicing of LSD1 transcript generates four full-length isoforms from combinatorial retention of two identified exons: the 4 aa exon E8a is internal to the amine oxidase domain, and its inclusion is restricted to the nervous system. Remarkably, the expression of LSD1 splice variants is dynamically regulated throughout cortical development, particularly during perinatal stages, with a progressive increase of LSD1 neurospecific isoforms over the ubiquitous ones. Notably, the same LSD1 splice dynamics can be fairly recapitulated in cultured cortical neurons. Functionally, LSD1 isoforms display in vitro a comparable demethylase activity, yet the inclusion of the sole exon E8a reduces LSD1 repressor activity on a reporter gene. Additional distinction among isoforms is supported by the knockdown of neurospecific variants in cortical neurons resulting in the inhibition of neurite maturation, whereas overexpression of the same variants enhances it. Instead, perturbation of LSD1 isoforms that are devoid of the neurospecific exon elicits no morphogenic effect. Collectively, results demonstrate that the arousal of neuronal LSD1 isoforms pacemakes early neurite morphogenesis, conferring a neurospecific function to LSD1 epigenetic activity.
Collapse
|
39
|
Arce L, Pate KT, Waterman ML. Groucho binds two conserved regions of LEF-1 for HDAC-dependent repression. BMC Cancer 2009; 9:159. [PMID: 19460168 PMCID: PMC2701438 DOI: 10.1186/1471-2407-9-159] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Accepted: 05/21/2009] [Indexed: 11/28/2022] Open
Abstract
Background Drosophila Groucho and its human Transducin-like-Enhancer of Split orthologs (TLEs) function as transcription co-repressors within the context of Wnt signaling, a pathway with strong links to cancer. The current model for how Groucho/TLE's modify Wnt signaling is by direct competition with β-catenin for LEF/TCF binding. The molecular events involved in this competitive interaction are not defined and the actions of Groucho/TLEs within the context of Wnt-linked cancer are unknown. Methods We used in vitro protein interaction assays with the LEF/TCF family member LEF-1, and in vivo assays with Wnt reporter plasmids to define Groucho/TLE interaction and repressor function. Results Mapping studies reveal that Groucho/TLE binds two regions in LEF-1. The primary site of recognition is a 20 amino acid region in the Context Dependent Regulatory domain. An auxiliary site is in the High Mobility Group DNA binding domain. Mutation of an eight amino acid sequence within the primary region (RFSHHMIP) results in a loss of Groucho action in a transient reporter assay. Drosophila Groucho, human TLE-1, and a truncated human TLE isoform Amino-enhancer-of-split (AES), work equivalently to repress LEF-1•β-catenin transcription in transient reporter assays, and these actions are sensitive to the HDAC inhibitor Trichostatin A. A survey of Groucho/TLE action in a panel of six colon cancer cell lines with elevated β-catenin shows that Groucho is not able to repress transcription in a subset of these cell lines. Conclusion Our data shows that Groucho/TLE repression requires two sites of interaction in LEF-1 and that a central, conserved amino acid sequence within the primary region (F S/T/P/xx y I/L/V) is critical. Our data also reveals that AES opposes LEF-1 transcription activation and that both Groucho and AES repression require histone deacetylase activity suggesting multiple steps in Groucho competition with β-catenin. The variable ability of Groucho/TLE to oppose Wnt signaling in colon cancer cells suggests there may be defects in one or more of these steps.
Collapse
Affiliation(s)
- Laura Arce
- Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, CA 92697-4025, USA.
| | | | | |
Collapse
|
40
|
Abstract
The PRH (proline-rich homeodomain) [also known as Hex (haematopoietically expressed homeobox)] protein is a transcription factor that functions as an important regulator of vertebrate development and many other processes in the adult including haematopoiesis. The Groucho/TLE (transducin-like enhancer) family of co-repressor proteins also regulate development and modulate the activity of many DNA-binding transcription factors during a range of diverse cellular processes including haematopoiesis. We have shown previously that PRH is a repressor of transcription in haematopoietic cells and that an Eh-1 (Engrailed homology) motif present within the N-terminal transcription repression domain of PRH mediates binding to Groucho/TLE proteins and enables co-repression. In the present study we demonstrate that PRH regulates the nuclear retention of TLE proteins during cellular fractionation. We show that transcriptional repression and the nuclear retention of TLE proteins requires PRH to bind to both TLE and DNA. In addition, we characterize a trans-dominant-negative PRH protein that inhibits wild-type PRH activity by sequestering TLE proteins to specific subnuclear domains. These results demonstrate that transcriptional repression by PRH is dependent on TLE availability and suggest that subnuclear localization of TLE plays an important role in transcriptional repression by PRH.
Collapse
|
41
|
Zhang X, Chen HM, Jaramillo E, Wang L, D'Mello SR. Histone deacetylase-related protein inhibits AES-mediated neuronal cell death by direct interaction. J Neurosci Res 2008; 86:2423-31. [PMID: 18438919 DOI: 10.1002/jnr.21680] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Histone deacetylase-related protein (HDRP), an alternatively spliced and truncated form of histone deacetylase-9 that lacks a C-terminal catalytic domain, protects neurons from death. In an effort to understand the mechanism by which HDRP mediates its neuroprotective effect, we screened for proteins in the brain that interact with HDRP by using a yeast two-hybrid assay. One of the HDRP-interacting proteins identified in this screen was amino enhancer of split (AES), a 197-amino acid protein belonging to the Groucho family. Interaction between HDRP and AES was verified by in vitro binding assays, coimmunoprecipitation, and colocalization studies. To investigate the significance of the HDRP-AES association to the regulation of neuronal survival, we used cultured cerebellar granule neurons, which undergo apoptosis when treated with low potassium (LK) medium. We found that in contrast to HDRP, whose expression is markedly reduced by LK treatment, AES expression was not appreciably altered. Forced expression of AES in healthy neurons results in cell death, an action that is blocked by the coexpression of HDRP. AES is a truncated version of larger Groucho-related proteins, one of which is transducin-like enhancer of split (TLE)-1. We found that the expression of TLE1 is reduced in LK-treated neurons and the forced expression of TLE1 blocks LK-induced neuronal death as well as death induced by AES. Our results show that AES has apoptotic activity in neurons and suggest that neuroprotection by HDRP is mediated by the inhibition of this activity through direct interaction.
Collapse
Affiliation(s)
- Xiaoguang Zhang
- Department of Molecular and Cell Biology, University of Texas at Dallas, Richardson, Texas 75080, USA
| | | | | | | | | |
Collapse
|
42
|
Cinnamon E, Paroush Z. Context-dependent regulation of Groucho/TLE-mediated repression. Curr Opin Genet Dev 2008; 18:435-40. [PMID: 18721877 DOI: 10.1016/j.gde.2008.07.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2008] [Revised: 07/14/2008] [Accepted: 07/23/2008] [Indexed: 01/15/2023]
Abstract
Groucho/TLE proteins are global corepressors that are recruited to target promoters by different families of DNA-binding repressors. As these corepressors are widely expressed, the long-standing view had been that Groucho/TLE-mediated repression is regulated solely by the spatial and temporal distribution of partner repressors. It has recently emerged, however, that Groucho/TLE repressor activity is itself regulated, in a signal induced, context-dependent manner. Here we review the essential roles played by Groucho/TLE factors in different cell-signalling processes that illustrate different modes for regulating Groucho/TLE-mediated repression: (i) via the expression of partner repressors; (ii) by competition with coactivators and (iii) through post-translational modifications of Groucho/TLE. We also discuss how the intrinsic properties of repressors can result in differential responses to Groucho/TLE regulation.
Collapse
Affiliation(s)
- Einat Cinnamon
- Department of Biochemistry, Faculty of Medicine, The Hebrew University, PO Box 12272, Jerusalem 91120, Israel.
| | | |
Collapse
|
43
|
Ratnaparkhi GS, Duong HA, Courey AJ. Dorsal interacting protein 3 potentiates activation by Drosophila Rel homology domain proteins. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2008; 32:1290-1300. [PMID: 18538389 PMCID: PMC2603422 DOI: 10.1016/j.dci.2008.04.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Revised: 04/11/2008] [Accepted: 04/12/2008] [Indexed: 05/26/2023]
Abstract
Dorsal interacting protein 3 (Dip3) contains a MADF DNA-binding domain and a BESS protein interaction domain. The Dip3 BESS domain was previously shown to bind to the Dorsal Rel homology domain. We show here that Dip3 also binds to the Relish Rel homology domain and enhances Rel family transcription factor function in both dorsoventral patterning and the immune response. While Dip3 is not essential, Dip3 mutations enhance the embryonic patterning defects that result from dorsal haplo-insufficiency, indicating that Dip3 may render dorsoventral patterning more robust. Dip3 is also required for optimal resistance to immune challenge since Dip3 mutant adults and larvae infected with bacteria have shortened lifetimes relative to infected wild-type flies. Furthermore, the mutant larvae exhibit significantly reduced expression of antimicrobial defense genes. Chromatin immunoprecipitation experiments in S2 cells indicate the presence of Dip3 at the promoters of these genes, and this binding requires the presence of Rel proteins at these promoters.
Collapse
Affiliation(s)
- Girish S Ratnaparkhi
- Department of Chemistry and Biochemistry, University of California at Los Angeles, Los Angeles, CA, USA
| | | | | |
Collapse
|
44
|
An J, Tsunekawa K, Feng GG, Li C, Huang L, Ito Y, Sugiyama S, Kurokawa T, Koide T, Nonami T, Ishikawa N. Roles of naofen, a novel WD-repeat-2 protein, in the CCl4-treated livers--a possible relationship to cell proliferation. Eur J Pharmacol 2008; 587:285-90. [PMID: 18472094 DOI: 10.1016/j.ejphar.2008.04.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Revised: 03/21/2008] [Accepted: 04/02/2008] [Indexed: 11/29/2022]
Abstract
Naofen (GenBank accession no. EF613262), a newly found intracellular protein in the WD-repeat-2 protein family, has been cloned as an anti-verotoxin II antibody immunoreactive substance, and the nucleotide- and amino acid-sequences have been clarified. The present study was undertaken to evaluate the roles of naofen especially in carbon tetrachloride (CCl4-induced cirrhosis model of rats, also in partial hepatectomy. Naofen mRNA expressions were observed from the early phases of cirrhosis development and during regenerative phases after partial hepatectomy, more remarkable in the former. Naofen immunoreactive fragments located in the vascular endothelial cells and peri-vascular spaces in normal livers especially in Glisson's areas, being strongly stained in the connective tissues 8 weeks after starting CCl4-injections, besides in the cytoplasm of hepatocytes in pseudo-lobules. In contrast, partial hepatectomy caused a small increase of naofen expressions in the whole hepatocytes, and significantly in the endothelial cells of portal veins and hepatic arterioles. Furthermore, in parallel to the degree of naofen mRNA and protein expressions, the rates of double-nuclei cells to total hepatocytes in the Glisson's areas increased in both cirrhosis and partial hepatectomy, suggesting a relationship between naofen expression and mitosis. In in-vitro studies with cell lines, vascular endothelial growth factor, a cell proliferation stimulant, increased the naofen mRNA expressions in HepG(2) cell lines, whereas paclitaxel, a cytotoxic anti-cancer drug, diminished them in NRK52E, both concentration-dependently. These results indicated that naofen immunoreactive fragments play an important role in the cell proliferation, relevant for analyzing the regenerative phases during cirrhosis developments and after partial hepatectomy.
Collapse
Affiliation(s)
- Jun An
- Department of Pharmacology, Aichi Medical University School of Medicine, Nagakute, Aichi pref., 480-1195, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The Groucho family of co-repressor proteins are essential for development and may also have a role in some human cancers.
Collapse
Affiliation(s)
- Barbara H Jennings
- Developmental Genetics Laboratory, Cancer Research UK, Lincoln's Inn Fields, London, WC2A 3PX, UK
| | | |
Collapse
|
46
|
Choi HK, Choi KC, Kang HB, Kim HC, Lee YH, Haam S, Park HG, Yoon HG. Function of multiple Lis-Homology domain/WD-40 repeat-containing proteins in feed-forward transcriptional repression by silencing mediator for retinoic and thyroid receptor/nuclear receptor corepressor complexes. Mol Endocrinol 2008; 22:1093-104. [PMID: 18202150 DOI: 10.1210/me.2007-0396] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Lis-homology (LisH) motifs are involved in protein dimerization, and the discovery of the conserved N-terminal LisH domain in transducin beta-like protein 1 and its receptor (TBL1 and TBLR1) led us to examine the role of this domain in transcriptional repression. Here we show that multiple beta-transducin (WD-40) repeat-containing proteins interact to form oligomers in solution and that oligomerization depends on the presence of the LisH domain in each protein. Repression of transcription, as assayed using Gal4 fusion proteins, also depended on the presence of the LisH domain, suggesting that oligomerization is a prerequisite for efficient transcriptional repression. Furthermore, we show that the LisH domain is responsible for the binding to the hypoacetylated histone H4 tail and for stable chromatin targeting by the nuclear receptor corepressor complex. Mutations in conserved residues in the LisH motif of TBL1 and TBLR1 block histone binding, oligomerization, and transcriptional repression, supporting the functional importance of the LisH motif in transcriptional repression. Our results indicate that another WD-40 protein, TBL3, also preferentially binds to the N-terminal domain of TBL1 and TBLR1, and forms oligomers with other WD-40 proteins. Finally, we observed that the WD-40 proteins RbAp46 and RbAp48 of the sin3A corepressor complex failed to dimerize. We also found the specific interaction UbcH/E2 with TBL1, but not RbAp46/48. Altogether, our results thus indicate that the presence of multiple LisH/WD-40 repeat containing proteins is exclusive to nuclear receptor corepressor/ silencing mediator for retinoic and thyroid receptor complexes compared with other class 1 histone deacetylase-containing corepessor complexes.
Collapse
Affiliation(s)
- Hyo-Kyoung Choi
- Department of Biochemistry and Molecular Biology, Center for Chronic Metabolic Disease Research, Yonsei University College of Medicine, Seoul 120-752, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Repression by Groucho/TLE/Grg proteins: genomic site recruitment generates compacted chromatin in vitro and impairs activator binding in vivo. Mol Cell 2008; 28:291-303. [PMID: 17964267 DOI: 10.1016/j.molcel.2007.10.002] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2007] [Revised: 07/13/2007] [Accepted: 08/12/2007] [Indexed: 11/21/2022]
Abstract
Groucho-related (Gro/TLE/Grg) corepressors meditate embryonic segmentation, dorsal-ventral patterning, neurogenesis, and Notch and Wnt signaling. Although Gro/TLE/Grgs disrupt activator complexes and recruit histone deacetylases (HDAC), activator complexes can be disrupted in various ways, HDAC recruitment does not account for full corepressor activity, and a direct role for Gro/TLE/Grg binding and altering chromatin structure has not been explored. Using diverse chromatin substrates in vitro, we show that Grg3 creates higher-order, condensed complexes of polynucleosome arrays. Surprisingly, such complexes are in an open, exposed configuration. We find that chromatin binding enables Grg3 recruitment by a transcription factor and the creation of a closed, poorly accessible domain spanning three to four nucleosomes. Targeted recruitment of Grg3 blankets a similar-sized region in vivo, impairing activator recruitment and repressing transcription. These activities of a Groucho protein represent a newly discovered mechanism which differs from that of other classes of corepressors.
Collapse
|
48
|
Differential in vivo requirements for oligomerization during Groucho-mediated repression. EMBO Rep 2007; 9:76-83. [PMID: 18034187 DOI: 10.1038/sj.embor.7401122] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Revised: 10/17/2007] [Accepted: 10/19/2007] [Indexed: 11/09/2022] Open
Abstract
The Groucho (Gro)/transducin-like enhancer of split family of transcriptional corepressors are implicated in many signalling pathways that are important in development and disease, including those mediated by Notch, Wnt and Hedgehog. Here, we describe a genetic screen in Drosophila that yielded 50 new gro alleles, including the first protein-null allele, and has two mutations in the conserved Q oligomerization domain that have been proposed to have an essential role in corepressor activity. One of these latter mutations, encoding an amino-terminal protein truncation that lacks part of the Q domain, abolishes oligomerization in vitro and renders the protein unstable in vivo. Nevertheless, the mutation is not a null: maternal mutant embryos have intermediate segmentation phenotypes and relatively normal terminal patterning suggesting that the mutant protein retains partial corepressor activity. Our results show that homo-oligomerization of Gro is not obligatory for its action in vivo, and that Gro represses transcription through more than one molecular mechanism.
Collapse
|
49
|
Heimbucher T, Murko C, Bajoghli B, Aghaallaei N, Huber A, Stebegg R, Eberhard D, Fink M, Simeone A, Czerny T. Gbx2 and Otx2 interact with the WD40 domain of Groucho/Tle corepressors. Mol Cell Biol 2006; 27:340-51. [PMID: 17060451 PMCID: PMC1800652 DOI: 10.1128/mcb.00811-06] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
One of the earliest organizational decisions in the development of the vertebrate brain is the division of the neural plate into Otx2-positive anterior and Gbx2-positive posterior territories. At the junction of these two expression domains, a local signaling center is formed, known as the midbrain-hindbrain boundary (MHB). This tissue coordinates or "organizes" the development of neighboring brain structures, such as the midbrain and cerebellum. Correct positioning of the MHB is thought to depend on mutual repression involving these two homeobox genes. Using a cell culture colocalization assay and coimmunoprecipitation experiments, we show that engrailed homology region 1 (eh1)-like motifs of both transcription factors physically interact with the WD40 domain of Groucho/Tle corepressor proteins. In addition, heat shock-induced expression of wild-type and mutant Otx2 and Gbx2 in medaka embryos demonstrates that Groucho is required for the repression of Otx2 by Gbx2. On the other hand, the repressive functions of Otx2 on Gbx2 do not appear to be dependent on corepressor interaction. Interestingly, the association of Groucho with Otx2 is also required for the repression of Fgf8 in the MHB. Therefore Groucho/Tle family members appear to regulate key aspects in the MHB development of the vertebrate brain.
Collapse
Affiliation(s)
- Thomas Heimbucher
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Veterinärplatz 1, A-1210 Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Jennings BH, Pickles LM, Wainwright SM, Roe SM, Pearl LH, Ish-Horowicz D. Molecular recognition of transcriptional repressor motifs by the WD domain of the Groucho/TLE corepressor. Mol Cell 2006; 22:645-55. [PMID: 16762837 DOI: 10.1016/j.molcel.2006.04.024] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2006] [Revised: 03/20/2006] [Accepted: 04/24/2006] [Indexed: 11/18/2022]
Abstract
The Groucho (Gro)/TLE/Grg family of corepressors operates in many signaling pathways (including Notch and Wnt). Gro/TLE proteins recognize a wide range of transcriptional repressors by binding to divergent short peptide sequences, including a C-terminal WRPW/Y motif (Hairy/Hes/Runx) and internal eh1 motifs (FxIxxIL; Engrailed/Goosecoid/Pax/Nkx). Here, we identify several missense mutations in Drosophila Gro, which demonstrate peptide binding to the central pore of the WD (WD40) beta propeller domain in vitro and in vivo. We define these interactions at the molecular level with crystal structures of the WD domain of human TLE1 bound to either WRPW or eh1 peptides. The two distinct peptide motifs adopt markedly different bound conformations but occupy overlapping sites across the central pore of the beta propeller. Our structural and functional analysis explains the rigid conservation of the WRPW motif, the sequence flexibility of eh1 motifs, and other aspects of repressor recognition by Gro in vivo.
Collapse
Affiliation(s)
- Barbara H Jennings
- Developmental Genetics Laboratory, Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 3PX, United Kingdom
| | | | | | | | | | | |
Collapse
|