1
|
Nuclear corepressor SMRT acts as a strong regulator of both β-oxidation and suppressor of fibrosis in the differentiation process of mouse skeletal muscle cells. PLoS One 2022; 17:e0277830. [PMID: 36454860 PMCID: PMC9714868 DOI: 10.1371/journal.pone.0277830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 11/03/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Silencing Mediator of Retinoid and Thyroid hormone receptors (SMRT; NCoR2) is a transcriptional corepressor (CoR) which has been recognized as an important player in the regulation of hepatic lipogenesis and in somatic development in mouse embryo. SMRT protein is also widely expressed in mouse connective tissues, for example adipocytes and muscle. We recently reported that mice with global deletion of SMRT develop significant obesity and muscle wasting which are independent from thyroid hormone (TH) signaling and thermogenesis. However, the tissue specific role of SMRT in skeletal muscle is still not clear. METHODS To clarify role of SMRT in muscle differentiation, we made myogenic C2C12 clones which lack SMRT protein (C2C12-SKO) by using CRISPR-Cas9. Wild-type C2C12 (C2C12-WT) and C2C12-SKO cells were cultured in differentiation medium, and the resulting gene and protein profiles were compared between the two cell lines both before and after differentiation. We also analyzed muscle tissues which were dissected from whole body SMRT knockout (KO) mice and their controls. RESULTS We found significant up-regulation of muscle specific β-oxidation markers; Peroxisome proliferator-activated receptor δ (PPARδ) and PPARγ coactivator-1α (PGC-1α) in the C2C12-SKO cells, suggesting that the cells had a similar gene profile to what is found in exercised rodent skeletal muscle. On the other hand, confocal microscopic analysis showed the significant loss of myotubes in C2C12-SKO cells similar to the morphology found in immature myoblasts. Proteomics analysis also confirmed that the C2C12-SKO cells had higher expression of markers of fibrosis (ex. Collagen1A1; COL1A1 and Fibroblast growth factor-2; FGF-2), indicating the up-regulation of Transforming growth factor-β (TGF-β) receptor signaling. Consistent with this, treatment with a specific TGF-β receptor inhibitor ameliorated both the defects in myotube differentiation and fibrosis. CONCLUSION Taken together, we demonstrate that SMRT functions as a pivotal transcriptional mediator for both β-oxidation and the prevention for the fibrosis via TGF-β receptor signaling in the differentiation of C2C12 myoblasts. In contrast to the results from C2C12 cells, SMRT does not appear to play a role in adult skeletal muscle of whole body SMRT KO mice. Thus, SMRT plays a significant role in the differentiation of myoblasts.
Collapse
|
2
|
Ehyai S, Miyake T, Williams D, Vinayak J, Bayfield MA, McDermott JC. FMRP recruitment of β-catenin to the translation pre-initiation complex represses translation. EMBO Rep 2018; 19:embr.201745536. [PMID: 30361391 DOI: 10.15252/embr.201745536] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 09/12/2018] [Accepted: 09/19/2018] [Indexed: 01/07/2023] Open
Abstract
Canonical Wnt/β-catenin signaling is an essential regulator of various cellular functions throughout development and adulthood. Aberrant Wnt/β-catenin signaling also contributes to various pathologies including cancer, necessitating an understanding of cell context-dependent mechanisms regulating this pathway. Since protein-protein interactions underpin β-catenin function and localization, we sought to identify novel β-catenin interacting partners by affinity purification coupled with tandem mass spectrometry in vascular smooth muscle cells (VSMCs), where β-catenin is involved in both physiological and pathological control of cell proliferation. Here, we report novel components of the VSMC β-catenin interactome. Bioinformatic analysis of the protein networks implies potentially novel functions for β-catenin, particularly in mRNA translation, and we confirm a direct interaction between β-catenin and the fragile X mental retardation protein (FMRP). Biochemical studies reveal a basal recruitment of β-catenin to the messenger ribonucleoprotein and translational pre-initiation complex, fulfilling a translational repressor function. Wnt stimulation antagonizes this function, in part, by sequestering β-catenin away from the pre-initiation complex. In conclusion, we present evidence that β-catenin fulfills a previously unrecognized function in translational repression.
Collapse
Affiliation(s)
- Saviz Ehyai
- Department of Biology, York University, Toronto, ON, Canada.,Muscle Health Research Centre (MHRC), York University, Toronto, ON, Canada.,Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada
| | - Tetsuaki Miyake
- Department of Biology, York University, Toronto, ON, Canada.,Muscle Health Research Centre (MHRC), York University, Toronto, ON, Canada.,Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada
| | - Declan Williams
- Department of Chemistry, York University, Toronto, ON, Canada.,Centre for Research in Mass Spectrometry (CRMS), York University, Toronto, ON, Canada
| | - Jyotsna Vinayak
- Department of Biology, York University, Toronto, ON, Canada.,Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada
| | - Mark A Bayfield
- Department of Biology, York University, Toronto, ON, Canada.,Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada
| | - John C McDermott
- Department of Biology, York University, Toronto, ON, Canada .,Muscle Health Research Centre (MHRC), York University, Toronto, ON, Canada.,Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada.,Department of Chemistry, York University, Toronto, ON, Canada.,Centre for Research in Mass Spectrometry (CRMS), York University, Toronto, ON, Canada
| |
Collapse
|
3
|
Ally A, Balasundaram M, Carlsen R, Chuah E, Clarke A, Dhalla N, Holt RA, Jones SJ, Lee D, Ma Y, Marra MA, Mayo M, Moore RA, Mungall AJ, Schein JE, Sipahimalani P, Tam A, Thiessen N, Cheung D, Wong T, Brooks D, Robertson AG, Bowlby R, Mungall K, Sadeghi S, Xi L, Covington K, Shinbrot E, Wheeler DA, Gibbs RA, Donehower LA, Wang L, Bowen J, Gastier-Foster JM, Gerken M, Helsel C, Leraas KM, Lichtenberg TM, Ramirez NC, Wise L, Zmuda E, Gabriel SB, Meyerson M, Cibulskis C, Murray BA, Shih J, Beroukhim R, Cherniack AD, Schumacher SE, Saksena G, Pedamallu CS, Chin L, Getz G, Noble M, Zhang H, Heiman D, Cho J, Gehlenborg N, Saksena G, Voet D, Lin P, Frazer S, Defreitas T, Meier S, Lawrence M, Kim J, Creighton CJ, Muzny D, Doddapaneni H, Hu J, Wang M, Morton D, Korchina V, Han Y, Dinh H, Lewis L, Bellair M, Liu X, Santibanez J, Glenn R, Lee S, Hale W, Parker JS, Wilkerson MD, Hayes DN, Reynolds SM, Shmulevich I, Zhang W, Liu Y, Iype L, Makhlouf H, Torbenson MS, Kakar S, Yeh MM, Jain D, Kleiner DE, Jain D, Dhanasekaran R, El-Serag HB, Yim SY, Weinstein JN, Mishra L, Zhang J, Akbani R, Ling S, Ju Z, Su X, Hegde AM, Mills GB, Lu Y, Chen J, Lee JS, Sohn BH, Shim JJ, Tong P, Aburatani H, Yamamoto S, Tatsuno K, Li W, Xia Z, Stransky N, Seiser E, Innocenti F, Gao J, Kundra R, Zhang H, Heins Z, Ochoa A, Sander C, Ladanyi M, Shen R, Arora A, Sanchez-Vega F, Schultz N, Kasaian K, Radenbaugh A, Bissig KD, Moore DD, Totoki Y, Nakamura H, Shibata T, Yau C, Graim K, Stuart J, Haussler D, Slagle BL, Ojesina AI, Katsonis P, Koire A, Lichtarge O, Hsu TK, Ferguson ML, Demchok JA, Felau I, Sheth M, Tarnuzzer R, Wang Z, Yang L, Zenklusen JC, Zhang J, Hutter CM, Sofia HJ, Verhaak RG, Zheng S, Lang F, Chudamani S, Liu J, Lolla L, Wu Y, Naresh R, Pihl T, Sun C, Wan Y, Benz C, Perou AH, Thorne LB, Boice L, Huang M, Rathmell WK, Noushmehr H, Saggioro FP, Tirapelli DPDC, Junior CGC, Mente ED, Silva ODC, Trevisan FA, Kang KJ, Ahn KS, Giama NH, Moser CD, Giordano TJ, Vinco M, Welling TH, Crain D, Curley E, Gardner J, Mallery D, Morris S, Paulauskis J, Penny R, Shelton C, Shelton T, Kelley R, Park JW, Chandan VS, Roberts LR, Bathe OF, Hagedorn CH, Auman JT, O'Brien DR, Kocher JPA, Jones CD, Mieczkowski PA, Perou CM, Skelly T, Tan D, Veluvolu U, Balu S, Bodenheimer T, Hoyle AP, Jefferys SR, Meng S, Mose LE, Shi Y, Simons JV, Soloway MG, Roach J, Hoadley KA, Baylin SB, Shen H, Hinoue T, Bootwalla MS, Van Den Berg DJ, Weisenberger DJ, Lai PH, Holbrook A, Berrios M, Laird PW. Comprehensive and Integrative Genomic Characterization of Hepatocellular Carcinoma. Cell 2017; 169:1327-1341.e23. [PMID: 28622513 PMCID: PMC5680778 DOI: 10.1016/j.cell.2017.05.046] [Citation(s) in RCA: 1634] [Impact Index Per Article: 204.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 04/02/2017] [Accepted: 05/26/2017] [Indexed: 12/12/2022]
Abstract
Liver cancer has the second highest worldwide cancer mortality rate and has limited therapeutic options. We analyzed 363 hepatocellular carcinoma (HCC) cases by whole-exome sequencing and DNA copy number analyses, and we analyzed 196 HCC cases by DNA methylation, RNA, miRNA, and proteomic expression also. DNA sequencing and mutation analysis identified significantly mutated genes, including LZTR1, EEF1A1, SF3B1, and SMARCA4. Significant alterations by mutation or downregulation by hypermethylation in genes likely to result in HCC metabolic reprogramming (ALB, APOB, and CPS1) were observed. Integrative molecular HCC subtyping incorporating unsupervised clustering of five data platforms identified three subtypes, one of which was associated with poorer prognosis in three HCC cohorts. Integrated analyses enabled development of a p53 target gene expression signature correlating with poor survival. Potential therapeutic targets for which inhibitors exist include WNT signaling, MDM4, MET, VEGFA, MCL1, IDH1, TERT, and immune checkpoint proteins CTLA-4, PD-1, and PD-L1.
Collapse
|
4
|
Ziv E, Yarmohammadi H, Boas FE, Petre EN, Brown KT, Solomon SB, Solit D, Reidy D, Erinjeri JP. Gene Signature Associated with Upregulation of the Wnt/β-Catenin Signaling Pathway Predicts Tumor Response to Transarterial Embolization. J Vasc Interv Radiol 2017; 28:349-355.e1. [PMID: 28126478 DOI: 10.1016/j.jvir.2016.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 11/05/2016] [Accepted: 11/05/2016] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To identify gene mutations in tumors undergoing transarterial embolization and explore the relationship between gene mutations and tumor response to embolization. MATERIALS AND METHODS This was a retrospective review that included 17 patients with primary or metastatic liver tumors treated with embolization and had specimens analyzed for a 341-gene panel next-generation sequence assay. Pathologic conditions included hepatocellular, carcinoid, pancreatic neuroendocrine, melanoma, medullary thyroid, and liver acinar-cell carcinoma. Disease, procedure data, and tumor response data were collected. Dimensionality reduction was performed by using principal component analysis. A linear support vector machine was used to learn a prediction rule and identify the genes most predictive of objective tumor response (partial or complete) per modified Response Evaluation Criteria In Solid Tumors. Cross-validation was used to test the prediction on the holdout set. Permutation testing was used to determine statistical significance of prediction accuracy. Recursive feature elimination was used to identify the most predictive genes. RESULTS At 4 months after embolization, 9 tumors showed a response and 8 did not. Using the top two principal components, prediction accuracy of the gene mutation signature was 70% (±11%), which was statistically significant (P < .05). The most predictive genes were CTNNB1, MEN1, and NCOR1: three genes associated with the Wnt/β-catenin and hypoxia signaling pathways. CONCLUSIONS This study identifies gene mutations in tumors treated with transarterial embolization. A gene-mutation signature obtained from the mutation data suggests that upregulation of the Wnt/β-catenin signaling pathway may be associated with sensitivity to embolization.
Collapse
Affiliation(s)
- Etay Ziv
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, Howard-118, 1275 York Ave., New York, NY10065.
| | - Hooman Yarmohammadi
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, Howard-118, 1275 York Ave., New York, NY10065
| | - F Edward Boas
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, Howard-118, 1275 York Ave., New York, NY10065
| | - Elena Nadia Petre
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, Howard-118, 1275 York Ave., New York, NY10065
| | - Karen T Brown
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, Howard-118, 1275 York Ave., New York, NY10065
| | - Stephen B Solomon
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, Howard-118, 1275 York Ave., New York, NY10065
| | - David Solit
- Human Oncology and Pathogenesis Program, Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Howard-118, 1275 York Ave., New York, NY10065
| | - Diane Reidy
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Howard-118, 1275 York Ave., New York, NY10065
| | - Joseph P Erinjeri
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, Howard-118, 1275 York Ave., New York, NY10065
| |
Collapse
|
5
|
Qu C, He D, Lu X, Dong L, Zhu Y, Zhao Q, Jiang X, Chang P, Jiang X, Wang L, Zhang Y, Bi L, He J, Peng Y, Su J, Zhang H, Huang H, Li Y, Zhou S, Qu Y, Zhao Y, Zhang Z. Salt-inducible Kinase (SIK1) regulates HCC progression and WNT/β-catenin activation. J Hepatol 2016; 64:1076-1089. [PMID: 26778753 DOI: 10.1016/j.jhep.2016.01.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS In this study, we investigated the role of salt-inducible kinase 1 (SIK1) and its possible mechanisms in human hepatocellular carcinoma (HCC). METHODS Immunoprecipitation, immunohistochemistry, luciferase reporter, Chromatin immunoprecipitation, in vitro kinase assays and a mouse model were used to examine the role of SIK1 on the β-catenin signaling pathway. RESULTS SIK1 was significantly downregulated in HCC compared with normal controls. Its introduction in HCC cells markedly suppresses epithelial-to-mesenchymal transition (EMT), tumor growth and lung metastasis in xenograft tumor models. The effect of SIK1 on tumor development occurs at least partially through regulation of β-catenin, as evidenced by the fact that SIK1 overexpression leads to repression of β-catenin transcriptional activity, while SIK1 depletion has the opposite effect. Mechanistically, SIK1 phosphorylates the silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) at threonine (T)1391, which promotes the association of nuclear receptor corepressor (NCoR)/SMRT with transducin-beta-like protein 1 (TBL1)/transducing-beta-like 1 X-linked receptor 1 (TBLR1) and disrupts the binding of β-catenin to the TBL1/TBLR1 complex, thereby inactivating the Wnt/β-catenin pathway. However, SMRT-T1391A reverses the phenotype of SIK1 and promotes β-catenin transactivation. Twist1 is identified as a critical factor downstream of SIK1/β-catenin axis, and Twist1 knockdown (Twist1(KD)) reverses SIK1(KD)-mediated changes, whereas SIK1(KD)/Twist1(KD) double knockdown cells were less efficient in establishing tumor growth and metastasis than SIK1(KD) cells. The promoter activity of SIK1 were negatively regulated by Twist1, indicating that a double-negative feedback loop exists. Importantly, levels of SIK1 inversely correlate with Twist1 expression in human HCC specimens. CONCLUSIONS Our findings highlight the critical roles of SIK1 and its targets in the regulation of HCC development and provides potential new candidates for HCC therapy.
Collapse
Affiliation(s)
- Chao Qu
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
| | - De He
- Department of General Surgery, Affiliated Baoan Hospital of Southern Medical University, Shenzhen, China
| | - Xiaoling Lu
- National Center for International Research of Biological Targeting Diagnosis and Therapy(Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research)Guangxi Medical University, Nanning, Guangxi, China
| | - Lihua Dong
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
| | - Yuekun Zhu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qin Zhao
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
| | - Pengyu Chang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
| | - Xinping Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
| | - Lizhe Wang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
| | - Yuyu Zhang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
| | - Lirong Bi
- Department of Pathology, The First Hospital of Jilin University, Changchun, China
| | - Jian He
- National Center for International Research of Biological Targeting Diagnosis and Therapy(Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research)Guangxi Medical University, Nanning, Guangxi, China
| | - Yi Peng
- National Center for International Research of Biological Targeting Diagnosis and Therapy(Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research)Guangxi Medical University, Nanning, Guangxi, China
| | - Jing Su
- National Center for International Research of Biological Targeting Diagnosis and Therapy(Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research)Guangxi Medical University, Nanning, Guangxi, China
| | - Heng Zhang
- Department of Medicine, College of Clinical Science, Three Gorges University, Yichang, Hubei, China
| | - He Huang
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Yan Li
- National Center for International Research of Biological Targeting Diagnosis and Therapy(Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research)Guangxi Medical University, Nanning, Guangxi, China
| | - Sufang Zhou
- National Center for International Research of Biological Targeting Diagnosis and Therapy(Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research)Guangxi Medical University, Nanning, Guangxi, China
| | - Yaqin Qu
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China.
| | - Yongxiang Zhao
- National Center for International Research of Biological Targeting Diagnosis and Therapy(Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research)Guangxi Medical University, Nanning, Guangxi, China.
| | - Zhiyong Zhang
- National Center for International Research of Biological Targeting Diagnosis and Therapy(Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research)Guangxi Medical University, Nanning, Guangxi, China; Department of Surgery, Robert-Wood-Johnson Medical School University Hospital, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
6
|
Trans-ancestry mutational landscape of hepatocellular carcinoma genomes. Nat Genet 2014; 46:1267-73. [PMID: 25362482 DOI: 10.1038/ng.3126] [Citation(s) in RCA: 594] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 10/03/2014] [Indexed: 11/08/2022]
Abstract
Diverse epidemiological factors are associated with hepatocellular carcinoma (HCC) prevalence in different populations. However, the global landscape of the genetic changes in HCC genomes underpinning different epidemiological and ancestral backgrounds still remains uncharted. Here a collection of data from 503 liver cancer genomes from different populations uncovered 30 candidate driver genes and 11 core pathway modules. Furthermore, a collaboration of two large-scale cancer genome projects comparatively analyzed the trans-ancestry substitution signatures in 608 liver cancer cases and identified unique mutational signatures that predominantly contribute to Asian cases. This work elucidates previously unexplored ancestry-associated mutational processes in HCC development. A combination of hotspot TERT promoter mutation, TERT focal amplification and viral genome integration occurs in more than 68% of cases, implicating TERT as a central and ancestry-independent node of hepatocarcinogenesis. Newly identified alterations in genes encoding metabolic enzymes, chromatin remodelers and a high proportion of mTOR pathway activations offer potential therapeutic and diagnostic opportunities.
Collapse
|
7
|
Grivas PD, Papavassiliou AG. Transcriptional corepressors in cancer: emerging targets for therapeutic intervention. Cancer 2012; 119:1120-8. [PMID: 23224952 DOI: 10.1002/cncr.27908] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 10/02/2012] [Accepted: 10/31/2012] [Indexed: 01/08/2023]
Abstract
The normal cell transcriptional process entails a high degree of combinatorial effects and time-dependent "flexibility" to translate cellular signaling into differential gene expression levels. Transcriptional corepressors can function as histone-modifying enzymes to regulate epigenetic events, modulate chromatin structure, and hence control transcriptional activity. Various corepressor complexes have been described; qualitative and quantitative alterations of corepressors can crucially influence the transcriptional output of both normal and malignant cells. Because these molecules can exert epigenetic control of tumorigenic signaling pathways, they can be considered potential regulators of cancer cell-related phenomena. Alterations of the expression level and/or function of transcriptional corepressors have been reported in a wide range of human cancers; thus, corepressors may present rational therapeutic targets as well as potential biomarkers of response to selective therapeutic interventions. Deeper insights into the context-specific and time-specific physical connections among transcription factors, coregulators, and gene regulatory elements, as well as epigenetic modifications, and their interactions, can enhance the capacity to interfere with small molecules that may restore the normal transcriptome/interactome in a cancer cell. There are several conceivable mechanisms of corepressor targeting in cancer that create enthusiasm. However, design, discovery, and testing of such innovative treatment approaches require extensive elaboration before they can achieve practical implementation in the clinic.
Collapse
Affiliation(s)
- Petros D Grivas
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
8
|
Guo L, Chen C, Liang Q, Karim MZ, Gorska MM, Alam R. Nuclear translocation of MEK1 triggers a complex T cell response through the corepressor silencing mediator of retinoid and thyroid hormone receptor. THE JOURNAL OF IMMUNOLOGY 2012; 190:159-67. [PMID: 23225884 DOI: 10.4049/jimmunol.1201657] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
MEK1 phosphorylates ERK1/2 and regulates T cell generation, differentiation, and function. MEK1 has recently been shown to translocate to the nucleus. Its nuclear function is largely unknown. By studying human CD4 T cells, we demonstrate that a low level of MEK1 is present in the nucleus of CD4 T cells under basal conditions. T cell activation further increases the nuclear translocation of MEK1. MEK1 interacts with the nuclear receptor corepressor silencing mediator of retinoid and thyroid hormone receptor (SMRT). MEK1 reduces the nuclear level of SMRT in an activation-dependent manner. MEK1 is recruited to the promoter of c-Fos upon TCR stimulation. Conversely, SMRT is bound to the c-Fos promoter under basal conditions and is removed upon TCR stimulation. We examined the role of SMRT in regulation of T cell function. Small interfering RNA-mediated knockdown of SMRT results in a biphasic effect on cytokine production. The production of the cytokines IL-2, IL-4, IL-10, and IFN-γ increases in the early phase (8 h) and then decreases in the late phase (48 h). The late-phase decrease is associated with inhibition of T cell proliferation. The late-phase inhibition of T cell activation is, in part, mediated by IL-10 that is produced in the early phase and, in part, by β-catenin signaling. Thus, we have identified a novel nuclear function of MEK1. MEK1 triggers a complex pattern of early T cell activation, followed by a late inhibition through its interaction with SMRT. This biphasic dual effect most likely reflects a homeostatic regulation of T cell function by MEK1.
Collapse
Affiliation(s)
- Lei Guo
- Division of Allergy and Immunology, Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| | | | | | | | | | | |
Collapse
|
9
|
The many faces and functions of β-catenin. EMBO J 2012; 31:2714-36. [PMID: 22617422 DOI: 10.1038/emboj.2012.150] [Citation(s) in RCA: 1220] [Impact Index Per Article: 93.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 04/30/2012] [Indexed: 02/07/2023] Open
Abstract
β-Catenin (Armadillo in Drosophila) is a multitasking and evolutionary conserved molecule that in metazoans exerts a crucial role in a multitude of developmental and homeostatic processes. More specifically, β-catenin is an integral structural component of cadherin-based adherens junctions, and the key nuclear effector of canonical Wnt signalling in the nucleus. Imbalance in the structural and signalling properties of β-catenin often results in disease and deregulated growth connected to cancer and metastasis. Intense research into the life of β-catenin has revealed a complex picture. Here, we try to capture the state of the art: we try to summarize and make some sense of the processes that regulate β-catenin, as well as the plethora of β-catenin binding partners. One focus will be the interaction of β-catenin with different transcription factors and the potential implications of these interactions for direct cross-talk between β-catenin and non-Wnt signalling pathways.
Collapse
|
10
|
Dela Cruz F, Terry M, Matushansky I. A transgenic, mesodermal specific, Dkk1 mouse model recapitulates a spectrum of human congenital limb reduction defects. Differentiation 2012; 83:220-30. [PMID: 22406973 DOI: 10.1016/j.diff.2012.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 12/14/2011] [Accepted: 01/17/2012] [Indexed: 01/02/2023]
Abstract
Congenital limb reduction defects occurring in isolation of other developmental abnormalities continue to be an important medical problem in which little progress has been made. Herein we generated transgenic mice expressing Dkk1 in an appendicular mesodermal pattern. Prx1-Dkk1 mice recapitulate a full spectrum of human congenital limb reduction defects, without other developmental issues, and have normal life-spans. Importantly, a close examination of the inheritance pattern suggests that there is a significant degree of incomplete penetrance as progeny of phenotypically positive or phenotypically negative, but genotypically positive Prx1-Dkk1 mice, consistently give rise to both phenotypically positive mice and phenotypically normal-appearing mice. Thus, this heterogeneous phenotype is reproducible with each generation regardless of the phenotype of the parents. We further go on to identify that mesenchymal stem cells from Prx1-Dkk1 mice have limited proliferative ability, but normal differentiation potential, which may explain the mechanism for the limb reduction defects observed. We believe Prx1-Dkk1 mice may prove useful in the future to study the mechanisms underlying the development of congenital limb reduction defects.
Collapse
Affiliation(s)
- Filemon Dela Cruz
- Division of Pediatric Oncology, Department of Pediatrics, Columbia University Medical Center, 161 Fort Washington Ave, IP-7, New York, NY 10032, USA
| | | | | |
Collapse
|
11
|
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
|
12
|
Carter WB, Tourtelot JB, Savell JG, Lilienfeld H. New Treatments and Shifting Paradigms in Differentiated Thyroid Cancer Management. Cancer Control 2011; 18:96-103. [DOI: 10.1177/107327481101800204] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- W. Bradford Carter
- Endocrine Tumor Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - John B. Tourtelot
- Endocrine Tumor Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jason G. Savell
- Anatomic Pathology Program at the H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Howard Lilienfeld
- Endocrine Tumor Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| |
Collapse
|
13
|
Battaglia S, Maguire O, Campbell MJ. Transcription factor co-repressors in cancer biology: roles and targeting. Int J Cancer 2010; 126:2511-9. [PMID: 20091860 DOI: 10.1002/ijc.25181] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Normal transcription displays a high degree of flexibility over the choice, timing and magnitude of mRNA expression levels that tend to oscillate and cycle. These processes allow for combinatorial actions, feedback control and fine-tuning. A central role has emerged for the transcriptional co-repressor proteins such as NCOR1, NCOR2/SMRT, CoREST and CTBPs, to control the actions of many transcriptional factors, in large part, by recruitment and activation of a range of chromatin remodeling enzymes. Thus, co-repressors and chromatin remodeling factors are recruited to transcription factors at specific promoter/enhancer regions and execute changes in the chromatin structure. The specificity of this recruitment is controlled in a spatial-temporal manner. By playing a central role in transcriptional control, as they move and target transcription factors, co-repressors act as a key driver in the epigenetic economy of the nucleus. Co-repressor functions are selectively distorted in malignancy, by both loss and gain of function and contribute to the generation of transcriptional rigidity. Features of transcriptional rigidity apparent in cancer cells include the distorted signaling of nuclear receptors and the WNTs/beta-catenin axis. Understanding and predicting the consequences of altered co-repressor expression patterns in cancer cells has diagnostic and prognostic significance, and also have the capacity to be targeted through selective epigenetic therapies.
Collapse
Affiliation(s)
- Sebastiano Battaglia
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | | |
Collapse
|
14
|
Beildeck ME, Islam M, Shah S, Welsh J, Byers SW. Control of TCF-4 expression by VDR and vitamin D in the mouse mammary gland and colorectal cancer cell lines. PLoS One 2009; 4:e7872. [PMID: 19924301 PMCID: PMC2774944 DOI: 10.1371/journal.pone.0007872] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Accepted: 10/14/2009] [Indexed: 11/18/2022] Open
Abstract
Background The vitamin D receptor (VDR) pathway is important in the prevention and potentially in the treatment of many cancers. One important mechanism of VDR action is related to its interaction with the Wnt/β-catenin pathway. Agonist-bound VDR inhibits the oncogenic Wnt/β-catenin/TCF pathway by interacting directly with β-catenin and in some cells by increasing cadherin expression which, in turn, recruits β-catenin to the membrane. Here we identify TCF-4, a transcriptional regulator and β-catenin binding partner as an indirect target of the VDR pathway. Methodology/Principal Findings In this work, we show that TCF-4 (gene name TCF7L2) is decreased in the mammary gland of the VDR knockout mouse as compared to the wild-type mouse. Furthermore, we show 1,25(OH)2D3 increases TCF-4 at the RNA and protein levels in several human colorectal cancer cell lines, the effect of which is completely dependent on the VDR. In silico analysis of the human and mouse TCF7L2 promoters identified several putative VDR binding elements. Although TCF7L2 promoter reporters responded to exogenous VDR, and 1,25(OH)2D3, mutation analysis and chromatin immunoprecipitation assays, showed that the increase in TCF7L2 did not require recruitment of the VDR to the identified elements and indicates that the regulation by VDR is indirect. This is further confirmed by the requirement of de novo protein synthesis for this up-regulation. Conclusions/Significance Although it is generally assumed that binding of β-catenin to members of the TCF/LEF family is cancer-promoting, recent studies have indicated that TCF-4 functions instead as a transcriptional repressor that restricts breast and colorectal cancer cell growth. Consequently, we conclude that the 1,25(OH)2D3/VDR-mediated increase in TCF-4 may have a protective role in colon cancer as well as diabetes and Crohn's disease.
Collapse
Affiliation(s)
- Marcy E. Beildeck
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University School of Medicine, Washington, D. C., United States of America
| | - Md Islam
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University School of Medicine, Washington, D. C., United States of America
| | - Salimuddin Shah
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University School of Medicine, Washington, D. C., United States of America
| | - JoEllen Welsh
- GenNYsis Center for Excellence in Cancer Genomics, SUNY at Albany, Rensselaer, New York, United States of America
| | - Stephen W. Byers
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University School of Medicine, Washington, D. C., United States of America
- * E-mail:
| |
Collapse
|
15
|
Iioka H, Doerner SK, Tamai K. Kaiso is a bimodal modulator for Wnt/beta-catenin signaling. FEBS Lett 2009; 583:627-32. [PMID: 19166851 DOI: 10.1016/j.febslet.2009.01.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 01/12/2009] [Accepted: 01/12/2009] [Indexed: 11/16/2022]
Abstract
The Wnt family of secreted ligands plays critical roles during embryonic development and tumorigenesis. Here we show that Kaiso, a dual specific DNA-binding protein, functions as a bimodal regulator of canonical Wnt signaling. Loss-of-function analysis of Kaiso abrogated Wnt-mediated reporter activity and axis duplication, whereas gain-of-function analysis of Kaiso dose-dependently resulted in synergistic and suppressive effects. Our analyses further suggest Kaiso can regulate TCF/LEF1-activity for these effects via modulating HDAC1 and beta-catenin-complex formation. Our studies together provide insights into why Kaiso null mice display resistance to intestinal tumors when crossed onto an Apc(Min/+) background.
Collapse
Affiliation(s)
- Hidekazu Iioka
- BRB 723, Department of Genetics, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
16
|
Thorne JL, Campbell MJ, Turner BM. Transcription factors, chromatin and cancer. Int J Biochem Cell Biol 2008; 41:164-75. [PMID: 18804550 DOI: 10.1016/j.biocel.2008.08.029] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2008] [Revised: 08/15/2008] [Accepted: 08/18/2008] [Indexed: 01/26/2023]
Abstract
Transcription factors, chromatin and chromatin-modifying enzymes are key components in a complex network through which the genome interacts with its environment. For many transcription factors, binding motifs are found adjacent to the promoter regions of a large proportion of genes, requiring mechanisms that confer binding specificity in any given cell type. These include association of the factor with other proteins and packaging of DNA, as chromatin, at the binding sequence so as to inhibit or facilitate binding. Recent evidence suggests that specific post-translational modifications of the histones packaging promoter DNA can help guide transcription factors to selected sites. The enzymes that put such modifications in place are dependent on metabolic components (e.g. acetyl CoA, S-adenosyl methionine) and susceptible to inhibition or activation by environmental factors. Local patterns of histone modification can be altered or maintained through direct interaction between the transcription factor and histone modifying enzymes. The functional consequences of transcription factor binding are also dependent on protein modifying enzymes, particularly those that alter lysine methylation at selected residues. Remarkably, the role of these enzymes is not limited to promoter-proximal events, but can be linked to changes in the intranuclear location of target genes. In this review we describe results that begin to define how transcription factors, chromatin and environmental variables interact and how these interactions are subverted in cancer. We focus on the nuclear receptor family of transcription factors, where binding of ligands such as steroid hormones and dietary derived factors provides an extra level of environmental input.
Collapse
Affiliation(s)
- James L Thorne
- University of Birmingham Medical School, Edgbaston, Birmingham, B15 2TT, UK
| | | | | |
Collapse
|