1
|
Rodriguez-Blanco J, Salvador AD, Suter RK, Swiderska-Syn M, Palomo-Caturla I, Kliebe V, Shahani P, Peterson K, Turos-Cabal M, Vieira ME, Wynn DT, Howell AJ, Yang F, Ban Y, McCrea HJ, Zindy F, Danis E, Vibhakar R, Jermakowicz A, Martin V, Coss CC, Harris BT, de Cubas A, Chen XS, Barnoud T, Roussel MF, Ayad NG, Robbins DJ. Triptolide and its prodrug Minnelide target high-risk MYC-amplified medulloblastoma in preclinical models. J Clin Invest 2024; 134:e171136. [PMID: 38885332 PMCID: PMC11290968 DOI: 10.1172/jci171136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Most children with medulloblastoma (MB) achieve remission, but some face very aggressive metastatic tumors. Their dismal outcome highlights the critical need to advance therapeutic approaches that benefit such high-risk patients. Minnelide, a clinically relevant analog of the natural product triptolide, has oncostatic activity in both preclinical and early clinical settings. Despite its efficacy and tolerable toxicity, this compound has not been evaluated in MB. Utilizing a bioinformatic data set that integrates cellular drug response data with gene expression, we predicted that Group 3 (G3) MB, which has a poor 5-year survival, would be sensitive to triptolide/Minnelide. We subsequently showed that both triptolide and Minnelide attenuate the viability of G3 MB cells ex vivo. Transcriptomic analyses identified MYC signaling, a pathologically relevant driver of G3 MB, as a downstream target of this class of drugs. We validated this MYC dependency in G3 MB cells and showed that triptolide exerts its efficacy by reducing both MYC transcription and MYC protein stability. Importantly, Minnelide acted on MYC to reduce tumor growth and leptomeningeal spread, which resulted in improved survival of G3 MB animal models. Moreover, Minnelide improved the efficacy of adjuvant chemotherapy, further highlighting its potential for the treatment of MYC-driven G3 MB.
Collapse
Affiliation(s)
- Jezabel Rodriguez-Blanco
- Darby Children’s Research Institute, Department of Pediatrics, and
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | - Robert K. Suter
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
| | | | | | - Valentin Kliebe
- Darby Children’s Research Institute, Department of Pediatrics, and
| | - Pritika Shahani
- Darby Children’s Research Institute, Department of Pediatrics, and
| | - Kendell Peterson
- Darby Children’s Research Institute, Department of Pediatrics, and
| | | | - Megan E. Vieira
- Darby Children’s Research Institute, Department of Pediatrics, and
| | - Daniel T. Wynn
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
| | - Ashley J. Howell
- Darby Children’s Research Institute, Department of Pediatrics, and
| | - Fan Yang
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
| | - Yuguang Ban
- Department of Public Health Sciences, and
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Heather J. McCrea
- Departments of Neurological Surgery and Pediatrics, University of Miami, Jackson Health System, Miller School of Medicine, Miami, Florida, USA
| | - Frederique Zindy
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Etienne Danis
- University of Colorado Cancer Center
- Department of Biomedical Informatics, and
| | - Rajeev Vibhakar
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Anna Jermakowicz
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
| | - Vanesa Martin
- Department of Morphology and Cell Biology, University of Oviedo, Oviedo, Asturias, Spain
| | | | - Brent T. Harris
- Departments of Neurology and Pathology, Georgetown University Medical Center, Washington DC, USA
| | - Aguirre de Cubas
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Microbiology and Immunology, and
| | - X. Steven Chen
- Department of Public Health Sciences, and
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Thibaut Barnoud
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Martine F. Roussel
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Nagi G. Ayad
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
| | - David J. Robbins
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
| |
Collapse
|
2
|
Copeland CA, Olenchock BA, Ziehr D, McGarrity S, Leahy K, Young JD, Loscalzo J, Oldham WM. MYC overrides HIF-1α to regulate proliferating primary cell metabolism in hypoxia. eLife 2023; 12:e82597. [PMID: 37428010 DOI: 10.7554/elife.82597] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 06/27/2023] [Indexed: 07/11/2023] Open
Abstract
Hypoxia requires metabolic adaptations to sustain energetically demanding cellular activities. While the metabolic consequences of hypoxia have been studied extensively in cancer cell models, comparatively little is known about how primary cell metabolism responds to hypoxia. Thus, we developed metabolic flux models for human lung fibroblast and pulmonary artery smooth muscle cells proliferating in hypoxia. Unexpectedly, we found that hypoxia decreased glycolysis despite activation of hypoxia-inducible factor 1α (HIF-1α) and increased glycolytic enzyme expression. While HIF-1α activation in normoxia by prolyl hydroxylase (PHD) inhibition did increase glycolysis, hypoxia blocked this effect. Multi-omic profiling revealed distinct molecular responses to hypoxia and PHD inhibition, and suggested a critical role for MYC in modulating HIF-1α responses to hypoxia. Consistent with this hypothesis, MYC knockdown in hypoxia increased glycolysis and MYC over-expression in normoxia decreased glycolysis stimulated by PHD inhibition. These data suggest that MYC signaling in hypoxia uncouples an increase in HIF-dependent glycolytic gene transcription from glycolytic flux.
Collapse
Affiliation(s)
- Courtney A Copeland
- Department of Medicine, Brigham and Women's Hospital, Boston, United States
- Department of Medicine, Harvard Medical School, Boston, United States
| | - Benjamin A Olenchock
- Department of Medicine, Brigham and Women's Hospital, Boston, United States
- Department of Medicine, Harvard Medical School, Boston, United States
| | - David Ziehr
- Department of Medicine, Brigham and Women's Hospital, Boston, United States
- Department of Medicine, Harvard Medical School, Boston, United States
- Department of Medicine, Massachusetts General Hospital, Boston, United States
| | - Sarah McGarrity
- Department of Medicine, Brigham and Women's Hospital, Boston, United States
- Department of Medicine, Harvard Medical School, Boston, United States
- Center for Systems Biology, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Kevin Leahy
- Department of Medicine, Brigham and Women's Hospital, Boston, United States
- Department of Medicine, Harvard Medical School, Boston, United States
| | - Jamey D Young
- Departments of Chemical & Biomolecular Engineering and Molecular Physiology & Biophysics, Vanderbilt University, Nashville, United States
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Boston, United States
- Department of Medicine, Harvard Medical School, Boston, United States
| | - William M Oldham
- Department of Medicine, Brigham and Women's Hospital, Boston, United States
- Department of Medicine, Harvard Medical School, Boston, United States
| |
Collapse
|
3
|
Gonzalez-Pecchi V, Kwan AK, Doyle S, Ivanov AA, Du Y, Fu H. NSD3S stabilizes MYC through hindering its interaction with FBXW7. J Mol Cell Biol 2021; 12:438-447. [PMID: 31638140 PMCID: PMC7333476 DOI: 10.1093/jmcb/mjz098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 06/08/2019] [Accepted: 09/15/2019] [Indexed: 01/16/2023] Open
Abstract
The MYC transcription factor plays a key role in cell growth control. Enhanced MYC protein stability has been found to promote tumorigenesis. Thus, understanding how MYC stability is controlled may have significant implications for revealing MYC-driven growth regulatory mechanisms in physiological and pathological processes. Our previous work identified the histone lysine methyltransferase nuclear receptor binding SET domain protein 3 (NSD3) as a MYC modulator. NSD3S, a noncatalytic isoform of NSD3 with oncogenic activity, appears to bind, stabilize, and activate the transcriptional activity of MYC. However, the mechanism by which NSD3S stabilizes MYC remains to be elucidated. To uncover the nature of the interaction and the underlying mechanism of MYC regulation by NSD3S, we characterized the binding interface between both proteins by narrowing the interface to a 15-amino acid region in NSD3S that is partially required for MYC regulation. Mechanistically, NSD3S binds to MYC and reduces the association of F-box and WD repeat domain containing 7 (FBXW7) with MYC, which results in suppression of FBXW7-mediated proteasomal degradation of MYC and an increase in MYC protein half-life. These results support a critical role for NSD3S in the regulation of MYC function and provide a novel mechanism for NSD3S oncogenic function through inhibition of FBXW7-mediated degradation of MYC.
Collapse
Affiliation(s)
- Valentina Gonzalez-Pecchi
- Graduate Program in Cancer Biology, Emory University, Atlanta, GA, USA.,Department of Pharmacology and Chemical Biology, Emory Chemical Biology Discovery Center, Emory University, Atlanta, GA, USA
| | - Albert K Kwan
- Department of Pharmacology and Chemical Biology, Emory Chemical Biology Discovery Center, Emory University, Atlanta, GA, USA
| | - Sean Doyle
- Department of Pharmacology and Chemical Biology, Emory Chemical Biology Discovery Center, Emory University, Atlanta, GA, USA
| | - Andrey A Ivanov
- Department of Pharmacology and Chemical Biology, Emory Chemical Biology Discovery Center, Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Hematology & Medical Oncology, Emory University, Atlanta, GA, USA
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology, Emory Chemical Biology Discovery Center, Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, Emory Chemical Biology Discovery Center, Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Hematology & Medical Oncology, Emory University, Atlanta, GA, USA
| |
Collapse
|
4
|
Sun XX, Li Y, Sears RC, Dai MS. Targeting the MYC Ubiquitination-Proteasome Degradation Pathway for Cancer Therapy. Front Oncol 2021; 11:679445. [PMID: 34178666 PMCID: PMC8226175 DOI: 10.3389/fonc.2021.679445] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/24/2021] [Indexed: 12/23/2022] Open
Abstract
Deregulated MYC overexpression and activation contributes to tumor growth and progression. Given the short half-life and unstable nature of the MYC protein, it is not surprising that the oncoprotein is highly regulated via diverse posttranslational mechanisms. Among them, ubiquitination dynamically controls the levels and activity of MYC during normal cell growth and homeostasis, whereas the disturbance of the ubiquitination/deubiquitination balance enables unwanted MYC stabilization and activation. In addition, MYC is also regulated by SUMOylation which crosstalks with the ubiquitination pathway and controls MYC protein stability and activity. In this mini-review, we will summarize current updates regarding MYC ubiquitination and provide perspectives about these MYC regulators as potential therapeutic targets in cancer.
Collapse
Affiliation(s)
- Xiao-Xin Sun
- Department of Molecular & Medical Genetics, School of Medicine and the OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Yanping Li
- Department of Molecular & Medical Genetics, School of Medicine and the OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Rosalie C Sears
- Department of Molecular & Medical Genetics, School of Medicine and the OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Mu-Shui Dai
- Department of Molecular & Medical Genetics, School of Medicine and the OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
5
|
Qiu Z, Fa P, Liu T, Prasad CB, Ma S, Hong Z, Chan ER, Wang H, Li Z, He K, Wang QE, Williams TM, Yan C, Sizemore ST, Narla G, Zhang J. A Genome-Wide Pooled shRNA Screen Identifies PPP2R2A as a Predictive Biomarker for the Response to ATR and CHK1 Inhibitors. Cancer Res 2020; 80:3305-3318. [PMID: 32522823 PMCID: PMC7518641 DOI: 10.1158/0008-5472.can-20-0057] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/17/2020] [Accepted: 06/04/2020] [Indexed: 01/18/2023]
Abstract
There is currently a lack of precise predictive biomarkers for patient selection in clinical trials of inhibitors targeting replication stress (RS) response proteins ATR and CHK1. The objective of this study was to identify novel predictive biomarkers for the response to these agents in treating non-small cell lung cancer (NSCLC). A genome-wide loss-of-function screen revealed that tumor suppressor PPP2R2A, a B regulatory subunit of protein phosphatase 2 (PP2A), determines sensitivity to CHK1 inhibition. A synthetic lethal interaction between PPP2R2A deficiency and ATR or CHK1 inhibition was observed in NSCLC in vitro and in vivo and was independent of p53 status. ATR and CHK1 inhibition resulted in significantly increased levels of RS and altered replication dynamics, particularly in PPP2R2A-deficient NSCLC cells. Mechanistically, PPP2R2A negatively regulated translation of oncogene c-Myc protein. c-Myc activity was required for PPP2R2A deficiency-induced alterations of replication initiation/RS and sensitivity to ATR/CHK1 inhibitors. We conclude that PPP2R2A deficiency elevates RS by upregulating c-Myc activity, rendering cells reliant on the ATR/CHK1 axis for survival. Our studies show a novel synthetic lethal interaction and identify PPP2R2A as a potential new predictive biomarker for patient stratification in the clinical use of ATR and CHK1 inhibitors. SIGNIFICANCE: This study reveals new approaches to specifically target PPP2R2A-deficient lung cancer cells and provides a novel biomarker that will significantly improve treatment outcome with ATR and CHK1 inhibitors.
Collapse
MESH Headings
- Animals
- Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors
- Biomarkers, Tumor/deficiency
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Non-Small-Cell Lung/chemistry
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Cell Line, Tumor
- Checkpoint Kinase 1/antagonists & inhibitors
- DNA Damage
- DNA Replication
- Drug Resistance, Neoplasm
- Female
- Gene Knockdown Techniques
- Genes, p53
- Genome-Wide Association Study
- Heterografts
- Humans
- Lung Neoplasms/chemistry
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Male
- Mice
- Mice, Nude
- Protein Phosphatase 2/deficiency
- Protein Phosphatase 2/genetics
- Protein Phosphatase 2/metabolism
- Proto-Oncogene Proteins c-myc/metabolism
- RNA, Small Interfering
Collapse
Affiliation(s)
- Zhaojun Qiu
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Pengyan Fa
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Tao Liu
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Chandra B Prasad
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Shanhuai Ma
- University of Rochester, Rochester, New York
| | - Zhipeng Hong
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Ernest R Chan
- Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Zaibo Li
- Department of Pathology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Kai He
- Department of Internal Medicine, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Qi-En Wang
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Terence M Williams
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Chunhong Yan
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Steven T Sizemore
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Goutham Narla
- Department of Medicine, University of Michigan, Ann Arbor, Michigan
| | - Junran Zhang
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio.
| |
Collapse
|
6
|
Abstract
MYC is a master transcriptional regulator that controls almost all cellular processes. Over the last several decades, researchers have strived to define the context-dependent transcriptional gene programs that are controlled by MYC, as well as the mechanisms that regulate MYC function, in an effort to better understand the contribution of this oncoprotein to cancer progression. There are a wealth of data indicating that deregulation of MYC activity occurs in a large number of cancers and significantly contributes to disease progression, metastatic potential, and therapeutic resistance. Although the therapeutic targeting of MYC in cancer is highly desirable, there remain substantial structural and functional challenges that have impeded direct MYC-targeted drug development and efficacy. While efforts to drug the ‘undruggable’ may seem futile given these challenges and considering the broad reach of MYC, significant strides have been made to identify points of regulation that can be exploited for therapeutic purposes. These include targeting the deregulation of MYC transcription in cancer through small-molecule inhibitors that induce epigenetic silencing or that regulate the G-quadruplex structures within the MYC promoter. Alternatively, compounds that disrupt the DNA-binding activities of MYC have been the long-standing focus of many research groups, since this method would prevent downstream MYC oncogenic activities regardless of upstream alterations. Finally, proteins involved in the post-translational regulation of MYC have been identified as important surrogate targets to reduce MYC activity downstream of aberrant cell stimulatory signals. Given the complex regulation of the MYC signaling pathway, a combination of these approaches may provide the most durable response, but this has yet to be shown. Here, we provide a comprehensive overview of the different therapeutic strategies being employed to target oncogenic MYC function, with a focus on post-translational mechanisms.
Collapse
|
7
|
Chen Y, Sun XX, Sears RC, Dai MS. Writing and erasing MYC ubiquitination and SUMOylation. Genes Dis 2019; 6:359-371. [PMID: 31832515 PMCID: PMC6889025 DOI: 10.1016/j.gendis.2019.05.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/23/2019] [Accepted: 05/29/2019] [Indexed: 12/22/2022] Open
Abstract
The transcription factor c-MYC (MYC thereafter) controls diverse transcription programs and plays a key role in the development of many human cancers. Cells develop multiple mechanisms to ensure that MYC levels and activity are precisely controlled in normal physiological context. As a short half-lived protein, MYC protein levels are tightly regulated by the ubiquitin proteasome system. Over a dozen of ubiquitin ligases have been found to ubiquitinate MYC whereas a number of deubiquitinating enzymes counteract this process. Recent studies show that SUMOylation and deSUMOylation can also regulate MYC protein stability and activity. Interestingly, evidence suggests an intriguing crosstalk between MYC ubiquitination and SUMOylation. Deregulation of the MYC ubiquitination-SUMOylation regulatory network may contribute to tumorigenesis. This review is intended to provide the current understanding of the complex regulation of the MYC biology by dynamic ubiquitination and SUMOylation and their crosstalk.
Collapse
Affiliation(s)
- Yingxiao Chen
- Departments of Molecular & Medical Genetics, School of Medicine, OHSU Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Xiao-Xin Sun
- Departments of Molecular & Medical Genetics, School of Medicine, OHSU Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Rosalie C Sears
- Departments of Molecular & Medical Genetics, School of Medicine, OHSU Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Mu-Shui Dai
- Departments of Molecular & Medical Genetics, School of Medicine, OHSU Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
8
|
Swan AH, Gruscheski L, Boland LA, Brand T. The Popeye domain containing gene family encoding a family of cAMP-effector proteins with important functions in striated muscle and beyond. J Muscle Res Cell Motil 2019; 40:169-183. [PMID: 31197601 PMCID: PMC6726836 DOI: 10.1007/s10974-019-09523-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022]
Abstract
The Popeye domain containing (POPDC) gene family encodes a novel class of membrane-bound cyclic AMP effector proteins. POPDC proteins are abundantly expressed in cardiac and skeletal muscle. Consistent with its predominant expression in striated muscle, Popdc1 and Popdc2 null mutants in mouse and zebrafish develop cardiac arrhythmia and muscular dystrophy. Likewise, mutations in POPDC genes in patients have been associated with cardiac arrhythmia and muscular dystrophy phenotypes. A membrane trafficking function has been identified in this context. POPDC proteins have also been linked to tumour formation. Here, POPDC1 plays a role as a tumour suppressor by limiting c-Myc and WNT signalling. Currently, a common functional link between POPDC's role in striated muscle and as a tumour suppressor is lacking. We also discuss several alternative working models to better understand POPDC protein function.
Collapse
Affiliation(s)
- Alexander H Swan
- National Heart and Lung Institute, Imperial College London, 4th Floor ICTEM Building, Du Cane Road, London, W12 0NN, UK
- Institute of Chemical Biology, Imperial College London, London, UK
| | - Lena Gruscheski
- National Heart and Lung Institute, Imperial College London, 4th Floor ICTEM Building, Du Cane Road, London, W12 0NN, UK
| | - Lauren A Boland
- National Heart and Lung Institute, Imperial College London, 4th Floor ICTEM Building, Du Cane Road, London, W12 0NN, UK
| | - Thomas Brand
- National Heart and Lung Institute, Imperial College London, 4th Floor ICTEM Building, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
9
|
Chakravorty D, Banerjee K, Mapder T, Saha S. In silico modeling of phosphorylation dependent and independent c-Myc degradation. BMC Bioinformatics 2019; 20:230. [PMID: 31068129 PMCID: PMC6505206 DOI: 10.1186/s12859-019-2846-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 04/24/2019] [Indexed: 12/24/2022] Open
Abstract
Background c-Myc plays an important role in cell proliferation, cell growth and in differentiation, making it a key regulator for carcinogenesis and pluripotency. Tight control of c-myc turnover is required by ubiquitin-mediated degradation. This is achieved in the system by two F-box proteins Skp2 and FBXW7. Results Dynamic modelling technique was used to build two exclusive models for phosphorylation dependent degradation of Myc by FBXW7 (Model 1) and phosphorylation independent degradation by Skp2 (Model 2). Sensitivity analysis performed on these two models revealed that these models were corroborating experimental studies. It was also seen that Model 1 was more robust and perhaps more efficient in degrading c-Myc. These results questioned the existence of the two models in the system and to answer the question a combined model was hypothesised which had a decision making switch. The combined model had both Skp2 and FBXW7 mediated degradation where again the latter played a more important role. This model was able to achieve the lowest levels of ubiquitylated Myc and therefore functioned most efficiently in degradation of Myc. Conclusion In this report, c-Myc degradation by two F-box proteins was mathematically evaluated based on the importance of c-Myc turnover. The study was performed in a homeostatic system and therefore, prompts the exploration of c-Myc degradation in cancer state and in pluripotent state. Electronic supplementary material The online version of this article (10.1186/s12859-019-2846-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | - Tarunendu Mapder
- ARC CoE for Mathematical and Statistical Frontiers, School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia.
| | - Sudipto Saha
- Bioinformatics Centre, Bose Institute, Kolkata, India.
| |
Collapse
|
10
|
Wu L, Yi B, Wei S, Rao D, He Y, Naik G, Bae S, Liu XM, Yang WH, Sonpavde G, Liu R, Wang L. Loss of FOXP3 and TSC1 Accelerates Prostate Cancer Progression through Synergistic Transcriptional and Posttranslational Regulation of c-MYC. Cancer Res 2019; 79:1413-1425. [PMID: 30733194 DOI: 10.1158/0008-5472.can-18-2049] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 11/07/2018] [Accepted: 02/04/2019] [Indexed: 01/05/2023]
Abstract
Although c-MYC and mTOR are frequently activated proteins in prostate cancer, any interaction between the two is largely untested. Here, we characterize the functional cross-talk between FOXP3-c-MYC and TSC1-mTOR signaling during tumor progression. Deletion of Tsc1 in mouse embryonic fibroblasts (MEF) decreased phosphorylation of c-MYC at threonine 58 (pT58) and increased phosphorylation at serine 62 (pS62), an observation validated in prostate cancer cells. Conversely, inhibition of mTOR increased pT58 but decreased pS62. Loss of both FOXP3 and TSC1 in prostate cancer cells synergistically enhanced c-MYC expression via regulation of c-Myc transcription and protein phosphorylation. This crosstalk between FOXP3 and TSC1 appeared to be mediated by both the mTOR-4EBP1-c-MYC and FOXP3-c-MYC pathways. In mice, Tsc1 and Foxp3 double deletions in the prostate led to prostate carcinomas at an early age; this did not occur in these mice with an added c-Myc deletion. In addition, we observed synergistic antitumor effects of cotreating mice with inhibitors of mTOR and c-MYC in prostate cancer cells and in Foxp3 and Tsc1 double-mutant mice. In human prostate cancer, loss of nuclear FOXP3 is often accompanied by low expression of TSC1. Because loss of FOXP3 transcriptionally induces c-Myc expression and loss of TSC1 activates mTOR signaling, these data suggest cross-talk between FOXP3-c-MYC and TSC1-mTOR signaling that converges on c-MYC to regulate tumor progression. Coadministration of c-MYC and mTOR inhibitors may overcome the resistance to mTOR inhibition commonly observed in prostate cancer cells. SIGNIFICANCE: These results establish the principle of a synergistic action of TSC1 and FOXP3 during prostate cancer progression and provide new therapeutic targets for patients who have prostate cancer with two signaling defects.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/7/1413/F1.large.jpg.
Collapse
Affiliation(s)
- Lianpin Wu
- Institute of Translational Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Baozhu Yi
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Shi Wei
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Dapeng Rao
- Department of Urology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Youhua He
- Department of Urology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gurudatta Naik
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sejong Bae
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Xiaoguang M Liu
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wei-Hsiung Yang
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia
| | | | - Runhua Liu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama.
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lizhong Wang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama.
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
11
|
Su Y, Pelz C, Huang T, Torkenczy K, Wang X, Cherry A, Daniel CJ, Liang J, Nan X, Dai MS, Adey A, Impey S, Sears RC. Post-translational modification localizes MYC to the nuclear pore basket to regulate a subset of target genes involved in cellular responses to environmental signals. Genes Dev 2018; 32:1398-1419. [PMID: 30366908 PMCID: PMC6217735 DOI: 10.1101/gad.314377.118] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 09/04/2018] [Indexed: 12/14/2022]
Abstract
In this study, Su et al. investigate how post-translational modifications of Myc that affect stability and oncogenic activity regulate its function. They show that Ser62 phosphorylation and PIN1-mediated isomerization of MYC dynamically regulate the spatial distribution of MYC in the nucleus, promoting its association with the inner basket of the nuclear pore in response to proliferative signals, where it recruits the histone acetyltransferase GCN5 to bind and regulate local gene acetylation and expression, thus providing new insights into how post-translational modification of MYC controls its spatial activity. The transcription factor MYC (also c-Myc) induces histone modification, chromatin remodeling, and the release of paused RNA polymerase to broadly regulate transcription. MYC is subject to a series of post-translational modifications that affect its stability and oncogenic activity, but how these control MYC's function on the genome is largely unknown. Recent work demonstrates an intimate connection between nuclear compartmentalization and gene regulation. Here, we report that Ser62 phosphorylation and PIN1-mediated isomerization of MYC dynamically regulate the spatial distribution of MYC in the nucleus, promoting its association with the inner basket of the nuclear pore in response to proliferative signals, where it recruits the histone acetyltransferase GCN5 to bind and regulate local gene acetylation and expression. We demonstrate that PIN1-mediated localization of MYC to the nuclear pore regulates MYC target genes responsive to mitogen stimulation that are involved in proliferation and migration pathways. These changes are also present at the chromatin level, with an increase in open regulatory elements in response to stimulation that is PIN1-dependent and associated with MYC chromatin binding. Taken together, our study indicates that post-translational modification of MYC controls its spatial activity to optimally regulate gene expression in response to extrinsic signals in normal and diseased states.
Collapse
Affiliation(s)
- Yulong Su
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Oregon 97239, USA
| | - Carl Pelz
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Oregon 97239, USA.,Oregon Stem Cell Center, Oregon Health and Science University, Oregon 97239, USA
| | - Tao Huang
- Department of Biomedical Engineering, Oregon Health and Science University, Oregon 97239, USA
| | - Kristof Torkenczy
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Oregon 97239, USA
| | - Xiaoyan Wang
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Oregon 97239, USA
| | - Allison Cherry
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Oregon 97239, USA
| | - Colin J Daniel
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Oregon 97239, USA
| | - Juan Liang
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Oregon 97239, USA
| | - Xiaolin Nan
- Department of Biomedical Engineering, Oregon Health and Science University, Oregon 97239, USA
| | - Mu-Shui Dai
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Oregon 97239, USA
| | - Andrew Adey
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Oregon 97239, USA
| | - Soren Impey
- Oregon Stem Cell Center, Oregon Health and Science University, Oregon 97239, USA
| | - Rosalie C Sears
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Oregon 97239, USA
| |
Collapse
|
12
|
Macek P, Cliff MJ, Embrey KJ, Holdgate GA, Nissink JWM, Panova S, Waltho JP, Davies RA. Myc phosphorylation in its basic helix-loop-helix region destabilizes transient α-helical structures, disrupting Max and DNA binding. J Biol Chem 2018; 293:9301-9310. [PMID: 29695509 DOI: 10.1074/jbc.ra118.002709] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/16/2018] [Indexed: 01/01/2023] Open
Abstract
Myelocytomatosis proto-oncogene transcription factor (Myc) is an intrinsically disordered protein with critical roles in cellular homeostasis and neoplastic transformation. It is tightly regulated in the cell, with Myc phosphorylation playing a major role. In addition to the well-described tandem phosphorylation of Thr-52 and Ser-62 in the Myc transactivation domain linked to its degradation, P21 (RAC1)-activated kinase 2 (PAK2)-mediated phosphorylation of serine and threonine residues in the C-terminal basic helix-loop-helix leucine zipper (bHLH-LZ) region regulates Myc transcriptional activity. Here we report that PAK2 preferentially phosphorylates Myc twice, at Thr-358 and Ser-373, with only a minor fraction being modified at the previously identified Thr-400 site. For transcriptional activity, Myc binds E-box DNA elements, requiring its heterodimerization with Myc-associated factor X (Max) via the bHLH-LZ regions. Using isothermal calorimetry (ITC), we found that Myc phosphorylation destabilizes this ternary protein-DNA complex by decreasing Myc's affinity for Max by 2 orders of magnitude, suggesting a major effect of phosphorylation on this complex. Phosphomimetic substitutions revealed that Ser-373 dominates the effect on Myc-Max heterodimerization. Moreover, a T400D substitution disrupted Myc's affinity for Max. ITC, NMR, and CD analyses of several Myc variants suggested that the effect of phosphorylation on the Myc-Max interaction is caused by secondary structure disruption during heterodimerization rather than by a change in the structurally disordered state of Myc or by phosphorylation-induced electrostatic repulsion in the heterodimer. Our findings provide critical insights into the effects of PAK2-catalyzed phosphorylation of Myc on its interactions with Max and DNA.
Collapse
Affiliation(s)
- Pavel Macek
- From AstraZeneca, IMED Discovery Sciences, Alderley Park SK10 4TG, United Kingdom,
| | - Matthew J Cliff
- the Manchester Institute of Biotechnology and School of Chemistry, University of Manchester, Manchester M1 7DN, United Kingdom
| | - Kevin J Embrey
- AstraZeneca, IMED Discovery Sciences, Cambridge CB4 0WG, United Kingdom
| | | | | | - Stanislava Panova
- the Manchester Institute of Biotechnology and School of Chemistry, University of Manchester, Manchester M1 7DN, United Kingdom
| | - Jonathan P Waltho
- the Manchester Institute of Biotechnology and School of Chemistry, University of Manchester, Manchester M1 7DN, United Kingdom
| | - Rick A Davies
- From AstraZeneca, IMED Discovery Sciences, Alderley Park SK10 4TG, United Kingdom,
| |
Collapse
|
13
|
A Proteomics Analysis Reveals 9 Up-Regulated Proteins Associated with Altered Cell Signaling in Colon Cancer Patients. Protein J 2017; 36:513-522. [PMID: 29128960 DOI: 10.1007/s10930-017-9746-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Colorectal cancer is the second most common cancer in women and third most common cancer in men. Cell signaling alterations in colon cancer, especially in aggressive metastatic tumors, require further investigations. The present study aims to compare the expression pattern of proteins associated with cell signaling in paired tumor and non-tumor samples of patients with colon cancer, as well as to define the cluster of proteins to differentiate patients with non-metastatic (Dukes' grade B) and metastatic (Dukes' grade C&D) colon cancer. Frozen tumor and non-tumor samples were collected after tumor resection from 19 patients with colon cancer. The Panorama™ Antibody Microarray-Cell Signaling kits were used for the analyses. The expression ratios of paired tumor/non-tumor samples were calculated for the each protein. We employed R packages 'samr', 'gplots', 'supclust' (pelora, wilma algorithms), 'glmnet' for the differential expression analysis, supervised clustering and penalized logistic regression. Significance analysis of microarrays revealed 9 significantly up-regulated proteins, including protein kinase C gamma, c-Myc, MDM2, pan cytokeratin, and 1 significantly down-regulated protein (GAP1) in tumoral mucosa. Pan-cytokeratin and APP were up-regulated in tumor versus non-tumor tissue, and were selected in the predictive cluster to discriminate colon cancer type. Higher levels of S-100b and phospho-Tau-pSer199/202 were confirmed as the predictors of non-metastatic colon cancer by all employed regression/clustering methods. Deregulated proteins in colon cancer are involved in oncogenic signal transduction, cell cycle control, and regulation of cytoskeleton/transport. Further studies are needed to validate potential protein markers of colon cancer development and metastatic progression.
Collapse
|
14
|
Farrell AS, Joly MM, Allen-Petersen BL, Worth PJ, Lanciault C, Sauer D, Link J, Pelz C, Heiser LM, Morton JP, Muthalagu N, Hoffman MT, Manning SL, Pratt ED, Kendsersky ND, Egbukichi N, Amery TS, Thoma MC, Jenny ZP, Rhim AD, Murphy DJ, Sansom OJ, Crawford HC, Sheppard BC, Sears RC. MYC regulates ductal-neuroendocrine lineage plasticity in pancreatic ductal adenocarcinoma associated with poor outcome and chemoresistance. Nat Commun 2017; 8:1728. [PMID: 29170413 PMCID: PMC5701042 DOI: 10.1038/s41467-017-01967-6] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 10/26/2017] [Indexed: 01/06/2023] Open
Abstract
Intratumoral phenotypic heterogeneity has been described in many tumor types, where it can contribute to drug resistance and disease recurrence. We analyzed ductal and neuroendocrine markers in pancreatic ductal adenocarcinoma, revealing heterogeneous expression of the neuroendocrine marker Synaptophysin within ductal lesions. Higher percentages of Cytokeratin-Synaptophysin dual positive tumor cells correlate with shortened disease-free survival. We observe similar lineage marker heterogeneity in mouse models of pancreatic ductal adenocarcinoma, where lineage tracing indicates that Cytokeratin-Synaptophysin dual positive cells arise from the exocrine compartment. Mechanistically, MYC binding is enriched at neuroendocrine genes in mouse tumor cells and loss of MYC reduces ductal-neuroendocrine lineage heterogeneity, while deregulated MYC expression in KRAS mutant mice increases this phenotype. Neuroendocrine marker expression is associated with chemoresistance and reducing MYC levels decreases gemcitabine-induced neuroendocrine marker expression and increases chemosensitivity. Altogether, we demonstrate that MYC facilitates ductal-neuroendocrine lineage plasticity in pancreatic ductal adenocarcinoma, contributing to poor survival and chemoresistance.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Carcinoma, Neuroendocrine/drug therapy
- Carcinoma, Neuroendocrine/metabolism
- Carcinoma, Neuroendocrine/pathology
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cell Differentiation
- Cell Line, Tumor
- Cell Lineage
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/therapeutic use
- Drug Resistance, Neoplasm
- Female
- Heterografts
- Humans
- Keratins/metabolism
- Male
- Mice
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasm Transplantation
- Neuroendocrine Cells/metabolism
- Neuroendocrine Cells/pathology
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Prognosis
- Proto-Oncogene Proteins c-myc/metabolism
- Synaptophysin/metabolism
- Gemcitabine
Collapse
Affiliation(s)
- Amy S Farrell
- Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Meghan Morrison Joly
- Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Brittany L Allen-Petersen
- Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Patrick J Worth
- Department of Surgery, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Christian Lanciault
- Department of Pathology, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA
| | - David Sauer
- Department of Pathology, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Jason Link
- Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, 3181 S.W Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Carl Pelz
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, 3181 S.W Sam Jackson Park Road, Portland, OR, 97239, USA
- Computational Biology, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Laura M Heiser
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Jennifer P Morton
- Cancer Research UK, Beatson Institute, Switchback Road, Bearsden, Glasgow, G61 1BD, UK
| | - Nathiya Muthalagu
- Cancer Research UK, Beatson Institute, Switchback Road, Bearsden, Glasgow, G61 1BD, UK
| | - Megan T Hoffman
- Department of Molecular and Integrative Physiology, University of Michigan, 7744 MS II, 1137 E. Catherine St., Ann Arbor, MI, 48109, USA
| | - Sara L Manning
- Department of Gastroenterology, Hepatology and Nutrition and Zayed Center for Pancreatic Cancer Research, University of Texas M.D. Anderson Cancer Center, Unit 1466, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Erica D Pratt
- Department of Gastroenterology, Hepatology and Nutrition and Zayed Center for Pancreatic Cancer Research, University of Texas M.D. Anderson Cancer Center, Unit 1466, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Nicholas D Kendsersky
- Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Nkolika Egbukichi
- Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Taylor S Amery
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, 3181 S.W Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Mary C Thoma
- Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Zina P Jenny
- Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Andrew D Rhim
- Department of Gastroenterology, Hepatology and Nutrition and Zayed Center for Pancreatic Cancer Research, University of Texas M.D. Anderson Cancer Center, Unit 1466, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Daniel J Murphy
- Cancer Research UK, Beatson Institute, Switchback Road, Bearsden, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, University Avenue, Glasgow, G12 8QQ, UK
| | - Owen J Sansom
- Cancer Research UK, Beatson Institute, Switchback Road, Bearsden, Glasgow, G61 1BD, UK
| | - Howard C Crawford
- Department of Molecular and Integrative Physiology, University of Michigan, 7744 MS II, 1137 E. Catherine St., Ann Arbor, MI, 48109, USA
| | - Brett C Sheppard
- Department of Surgery, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, 3181 S.W Sam Jackson Park Road, Portland, OR, 97239, USA
- Knight Cancer Institute, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Rosalie C Sears
- Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA.
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, 3181 S.W Sam Jackson Park Road, Portland, OR, 97239, USA.
- Knight Cancer Institute, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA.
| |
Collapse
|
15
|
Brägelmann J, Böhm S, Guthrie MR, Mollaoglu G, Oliver TG, Sos ML. Family matters: How MYC family oncogenes impact small cell lung cancer. Cell Cycle 2017; 16:1489-1498. [PMID: 28737478 DOI: 10.1080/15384101.2017.1339849] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Small cell lung cancer (SCLC) is one of the most deadly cancers and currently lacks effective targeted treatment options. Recent advances in the molecular characterization of SCLC has provided novel insight into the biology of this disease and raises hope for a paradigm shift in the treatment of SCLC. We and others have identified activation of MYC as a driver of susceptibility to Aurora kinase inhibition in SCLC cells and tumors that translates into a therapeutic option for the targeted treatment of MYC-driven SCLC. While MYC shares major features with its paralogs MYCN and MYCL, the sensitivity to Aurora kinase inhibitors is unique for MYC-driven SCLC. In this review, we will compare the distinct molecular features of the 3 MYC family members and address the potential implications for targeted therapy of SCLC.
Collapse
Affiliation(s)
- Johannes Brägelmann
- a Molecular Pathology, Institute of Pathology, University of Cologne , Cologne , Germany.,b Department of Translational Genomics , Medical Faculty, University of Cologne , Cologne , Germany
| | - Stefanie Böhm
- a Molecular Pathology, Institute of Pathology, University of Cologne , Cologne , Germany.,b Department of Translational Genomics , Medical Faculty, University of Cologne , Cologne , Germany
| | - Matthew R Guthrie
- c Department of Oncological Sciences , University of Utah, Huntsman Cancer Institute , Salt Lake City , UT , USA
| | - Gurkan Mollaoglu
- c Department of Oncological Sciences , University of Utah, Huntsman Cancer Institute , Salt Lake City , UT , USA
| | - Trudy G Oliver
- c Department of Oncological Sciences , University of Utah, Huntsman Cancer Institute , Salt Lake City , UT , USA
| | - Martin L Sos
- a Molecular Pathology, Institute of Pathology, University of Cologne , Cologne , Germany.,b Department of Translational Genomics , Medical Faculty, University of Cologne , Cologne , Germany.,d Center for Molecular Medicine Cologne , University of Cologne , Cologne , Germany
| |
Collapse
|
16
|
MYC Modulation around the CDK2/p27/SKP2 Axis. Genes (Basel) 2017; 8:genes8070174. [PMID: 28665315 PMCID: PMC5541307 DOI: 10.3390/genes8070174] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 06/23/2017] [Accepted: 06/24/2017] [Indexed: 12/20/2022] Open
Abstract
MYC is a pleiotropic transcription factor that controls a number of fundamental cellular processes required for the proliferation and survival of normal and malignant cells, including the cell cycle. MYC interacts with several central cell cycle regulators that control the balance between cell cycle progression and temporary or permanent cell cycle arrest (cellular senescence). Among these are the cyclin E/A/cyclin-dependent kinase 2 (CDK2) complexes, the CDK inhibitor p27KIP1 (p27) and the E3 ubiquitin ligase component S-phase kinase-associated protein 2 (SKP2), which control each other by forming a triangular network. MYC is engaged in bidirectional crosstalk with each of these players; while MYC regulates their expression and/or activity, these factors in turn modulate MYC through protein interactions and post-translational modifications including phosphorylation and ubiquitylation, impacting on MYC's transcriptional output on genes involved in cell cycle progression and senescence. Here we elaborate on these network interactions with MYC and their impact on transcription, cell cycle, replication and stress signaling, and on the role of other players interconnected to this network, such as CDK1, the retinoblastoma protein (pRB), protein phosphatase 2A (PP2A), the F-box proteins FBXW7 and FBXO28, the RAS oncoprotein and the ubiquitin/proteasome system. Finally, we describe how the MYC/CDK2/p27/SKP2 axis impacts on tumor development and discuss possible ways to interfere therapeutically with this system to improve cancer treatment.
Collapse
|
17
|
Kalkat M, De Melo J, Hickman KA, Lourenco C, Redel C, Resetca D, Tamachi A, Tu WB, Penn LZ. MYC Deregulation in Primary Human Cancers. Genes (Basel) 2017; 8:genes8060151. [PMID: 28587062 PMCID: PMC5485515 DOI: 10.3390/genes8060151] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022] Open
Abstract
MYC regulates a complex biological program by transcriptionally activating and repressing its numerous target genes. As such, MYC is a master regulator of many processes, including cell cycle entry, ribosome biogenesis, and metabolism. In cancer, the activity of the MYC transcriptional network is frequently deregulated, contributing to the initiation and maintenance of disease. Deregulation often leads to constitutive overexpression of MYC, which can be achieved through gross genetic abnormalities, including copy number alterations, chromosomal translocations, increased enhancer activity, or through aberrant signal transduction leading to increased MYC transcription or increased MYC mRNA and protein stability. Herein, we summarize the frequency and modes of MYC deregulation and describe both well-established and more recent findings in a variety of cancer types. Notably, these studies have highlighted that with an increased appreciation for the basic mechanisms deregulating MYC in cancer, new therapeutic vulnerabilities can be discovered and potentially exploited for the inhibition of this potent oncogene in cancer.
Collapse
Affiliation(s)
- Manpreet Kalkat
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - Jason De Melo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - Katherine Ashley Hickman
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada.
| | - Corey Lourenco
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - Cornelia Redel
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - Diana Resetca
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - Aaliya Tamachi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - William B Tu
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - Linda Z Penn
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| |
Collapse
|
18
|
Zaytseva O, Quinn LM. Controlling the Master: Chromatin Dynamics at the MYC Promoter Integrate Developmental Signaling. Genes (Basel) 2017; 8:genes8040118. [PMID: 28398229 PMCID: PMC5406865 DOI: 10.3390/genes8040118] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/15/2017] [Accepted: 04/07/2017] [Indexed: 02/06/2023] Open
Abstract
The transcription factor and cell growth regulator MYC is potently oncogenic and estimated to contribute to most cancers. Decades of attempts to therapeutically target MYC directly have not resulted in feasible clinical applications, and efforts have moved toward indirectly targeting MYC expression, function and/or activity to treat MYC-driven cancer. A multitude of developmental and growth signaling pathways converge on the MYC promoter to modulate transcription through their downstream effectors. Critically, even small increases in MYC abundance (<2 fold) are sufficient to drive overproliferation; however, the details of how oncogenic/growth signaling networks regulate MYC at the level of transcription remain nebulous even during normal development. It is therefore essential to first decipher mechanisms of growth signal-stimulated MYC transcription using in vivo models, with intact signaling environments, to determine exactly how these networks are dysregulated in human cancer. This in turn will provide new modalities and approaches to treat MYC-driven malignancy. Drosophila genetic studies have shed much light on how complex networks signal to transcription factors and enhancers to orchestrate Drosophila MYC (dMYC) transcription, and thus growth and patterning of complex multicellular tissue and organs. This review will discuss the many pathways implicated in patterning MYC transcription during development and the molecular events at the MYC promoter that link signaling to expression. Attention will also be drawn to parallels between mammalian and fly regulation of MYC at the level of transcription.
Collapse
Affiliation(s)
- Olga Zaytseva
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600, Australia.
- School of Biomedical Sciences, University of Melbourne, Parkville 3010, Australia.
| | - Leonie M Quinn
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600, Australia.
- School of Biomedical Sciences, University of Melbourne, Parkville 3010, Australia.
| |
Collapse
|
19
|
Santha S, Davaakhuu G, Basu A, Ke R, Das S, Rana A, Rana B. Modulation of glycogen synthase kinase-3β following TRAIL combinatorial treatment in cancer cells. Oncotarget 2016; 7:66892-66905. [PMID: 27602497 PMCID: PMC5341845 DOI: 10.18632/oncotarget.11834] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 08/29/2016] [Indexed: 12/12/2022] Open
Abstract
Glycogen Synthase Kinase-3β (GSK3β) is a serine/threonine kinase, known to regulate various cellular processes including proliferation, differentiation, survival, apoptosis as well as TRAIL-resistance. Thus pathways that can modulate GSK3β axis are important targets for cancer drug development. Our earlier studies have shown that combinatorial treatment with Troglitazone (TZD) and TRAIL can induce apoptosis in TRAIL-resistant cancer cells. The current studies were undertaken to investigate whether GSK3β pathway was modulated during this apoptosis. Our results indicated an increase in inhibitory GSK3βSer9 phosphorylation during apoptosis, mediated via AKT. At a later time, however, TZD alone and TRAIL-TZD combination produced a dramatic reduction of GSK3β expression, which was abolished by cycloheximide. Luciferase assays with GSK3β-luc promoter reporter showed that TZD can effectively antagonize GSK3β promoter activity. Since TZD is a ligand for transcription factor PPARγ and can activate AMPK, we determined their roles on antagonism of GSK3β. Knockdown of PPARγ was unable to restore GSK3β expression or antagonize GSK3βSer9 phosphorylation. Although pretreatment with Compound C (pharmacological inhibitor of AMPK) partially rescued GSK3β expression, knockdown of AMPKα1 or α2 alone or in combination were ineffective. These studies suggested a novel PPARγ-AMPK-independent mechanism of targeting GSK3β by TZD, elucidation of which might provide newer insights to improve our understanding of TRAIL-resistance.
Collapse
Affiliation(s)
- Sreevidya Santha
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Gantulga Davaakhuu
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Aninda Basu
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Rong Ke
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Subhasis Das
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA.,University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA.,Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Basabi Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA.,University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA.,Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
20
|
Gao R, Wang L, Cai H, Zhu J, Yu L. E3 Ubiquitin Ligase RLIM Negatively Regulates c-Myc Transcriptional Activity and Restrains Cell Proliferation. PLoS One 2016; 11:e0164086. [PMID: 27684546 PMCID: PMC5042457 DOI: 10.1371/journal.pone.0164086] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 09/19/2016] [Indexed: 11/19/2022] Open
Abstract
RNF12/RLIM is a RING domain-containing E3 ubiquitin ligase whose function has only begun to be elucidated recently. Although RLIM was reported to play important roles in some biological processes such as imprinted X-chromosome inactivation and regulation of TGF-β pathway etc., other functions of RLIM are largely unknown. Here, we identified RLIM as a novel E3 ubiquitin ligase for c-Myc, one of the most frequently deregulated oncoproteins in human cancers. RLIM associates with c-Myc in vivo and in vitro independently of the E3 ligase activity of RLIM. Moreover, RLIM promotes the polyubiquitination of c-Myc protein independently of Ser62 and Thr58 phosphorylation of c-Myc. However, RLIM-mediated ubiquitination does not affect c-Myc stability. Instead, RLIM inhibits the transcriptional activity of c-Myc through which RLIM restrains cell proliferation. Our results suggest that RLIM may function as a tumor suppressor by controlling the activity of c-Myc oncoprotein.
Collapse
Affiliation(s)
- Rui Gao
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, P.R. China
- * E-mail:
| | - Lan Wang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, P.R. China
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine Ministry of Education, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Hao Cai
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Jingjing Zhu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Long Yu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, P.R. China
| |
Collapse
|
21
|
Abstract
Ubiquitination plays a key and complex role in the regulation of c-Myc stability, transactivation, and oncogenic activity. c-Myc is ubiquitinated by a number of ubiquitin ligases (E3s), such as SCF(Fbw7) and SCF(Skp2). Depending on the E3s, ubiquitination can either positively or negatively regulate c-Myc levels and activity. Meanwhile, c-Myc ubiquitination can be reversed by deubiquitination. An early study showed that USP28 deubiquitinates c-Myc via interacting with Fbw7α whereas a recent study reveals that USP37 deubiquitinates c-Myc independently of Fbw7 and c-Myc phosphorylation. Consequently, both USP28 and USP37 stabilize c-Myc and enhance its activity. We recently found the nucleolar USP36 as a novel c-Myc deubiquitinase that controls the end-point of c-Myc degradation pathway in the nucleolus. Here we briefly review the current understanding of ubiquitination and deubiquitination regulation of c-Myc and further discuss the USP36-c-Myc regulatory pathway.
Collapse
Affiliation(s)
- Xiao-Xin Sun
- a Departments of Molecular & Medical Genetics ; School of Medicine and the OHSU Knight Cancer Institute; Oregon Health & Science University ; Portland , OR USA
| | - Rosalie C Sears
- a Departments of Molecular & Medical Genetics ; School of Medicine and the OHSU Knight Cancer Institute; Oregon Health & Science University ; Portland , OR USA
| | - Mu-Shui Dai
- a Departments of Molecular & Medical Genetics ; School of Medicine and the OHSU Knight Cancer Institute; Oregon Health & Science University ; Portland , OR USA
| |
Collapse
|
22
|
The ubiquitin ligase Huwe1 regulates the maintenance and lymphoid commitment of hematopoietic stem cells. Nat Immunol 2016; 17:1312-1321. [PMID: 27668798 PMCID: PMC5117833 DOI: 10.1038/ni.3559] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 08/17/2016] [Indexed: 02/07/2023]
Abstract
Hematopoietic stem cells (HSCs) are dormant in the bone marrow and can be activated in response to diverse stresses to replenish all blood cell types. Here we identify the ubiquitin ligase Huwe1 as a crucial regulator of HSC functions via its post-translational control of N-myc. We found Huwe1 to be essential for HSC self-renewal, quiescence and lymphoid fate specification. Using a novel fluorescent fusion allele (MycnM), we observed that N-myc expression was restricted to the most immature, multipotent stem and progenitor populations. N-myc was upregulated in response to stress or upon loss of Huwe1, leading to increased proliferation and stem cell exhaustion. Mycn depletion reversed most of these phenotypes in vivo, suggesting that the attenuation of N-myc by Huwe1 is essential to reestablish homeostasis following stress.
Collapse
|
23
|
González-Prieto R, Cuijpers SA, Kumar R, Hendriks IA, Vertegaal AC. c-Myc is targeted to the proteasome for degradation in a SUMOylation-dependent manner, regulated by PIAS1, SENP7 and RNF4. Cell Cycle 2016; 14:1859-72. [PMID: 25895136 DOI: 10.1080/15384101.2015.1040965] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
c-Myc is the most frequently overexpressed oncogene in tumors, including breast cancer, colon cancer and lung cancer. Post-translational modifications comprising phosphorylation, acetylation and ubiquitylation regulate the activity of c-Myc. Recently, it was shown that c-Myc-driven tumors are strongly dependent on the SUMO pathway. Currently, the relevant SUMO target proteins in this pathway are unknown. Here we show that c-Myc is a target protein for SUMOylation, and that SUMOylated c-Myc is subsequently ubiquitylated and degraded by the proteasome. SUMO chains appeared to be dispensable for this process, polymerization-deficient SUMO mutants supported proteolysis of SUMOylated c-Myc. These results indicate that multiple SUMO monomers conjugated to c-Myc could be sufficient to direct SUMOylated c-Myc to the ubiquitin-proteasome pathway. Knocking down the SUMO-targeted ubiquitin ligase RNF4 enhanced the levels of SUMOylated c-Myc, indicating that RNF4 could recognize a multi-SUMOylated protein as a substrate in addition to poly-SUMOylated proteins. Knocking down the SUMO E3 ligase PIAS1 resulted in reduced c-Myc SUMOylation and increased c-Myc transcriptional activity, indicating that PIAS1 mediates c-Myc SUMOylation. Increased SUMOylation of c-Myc was noted upon knockdown of the SUMO protease SENP7, indicating that it also could regulate a multi-SUMOylated protein in addition to poly-SUMOylated proteins. C-Myc lacks KxE-type SUMOylation consensus motifs. We used mass spectrometry to identify 10 SUMO acceptor lysines: K52, K148, K157, K317, K323, K326, K389, K392, K398 and K430. Intriguingly, mutating all 10 SUMO acceptor lysines did not reduce c-Myc SUMOylation, suggesting that SUMO acceptor lysines in c-Myc act promiscuously. Our results provide novel insight into the complexity of c-Myc post-translational regulation.
Collapse
Affiliation(s)
- Román González-Prieto
- a Department of Molecular Cell Biology; Leiden University Medical Center ; Leiden , The Netherlands
| | | | | | | | | |
Collapse
|
24
|
Bahram F, Hydbring P, Tronnersjö S, Zakaria SM, Frings O, Fahlén S, Nilsson H, Goodwin J, von der Lehr N, Su Y, Lüscher B, Castell A, Larsson LG. Interferon-γ-induced p27KIP1 binds to and targets MYC for proteasome-mediated degradation. Oncotarget 2016; 7:2837-54. [PMID: 26701207 PMCID: PMC4823075 DOI: 10.18632/oncotarget.6693] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/21/2015] [Indexed: 11/25/2022] Open
Abstract
The Myc oncoprotein is tightly regulated at multiple levels including ubiquitin-mediated protein turnover. We recently demonstrated that inhibition of Cdk2-mediated phosphorylation of Myc at Ser-62 pharmacologically or through interferon (IFN)-γ-induced expression of p27(Kip1) (p27) repressed Myc's activity to suppress cellular senescence and differentiation. In this study we identified an additional activity of p27 to interfere with Myc independent of Ser-62 phosphorylation. p27 is required and sufficient for IFN-γ-induced turnover of Myc. p27 interacted with Myc in the nucleus involving the C-termini of the two proteins, including Myc box 4 of Myc. The C-terminus but not the Cdk2 binding fragment of p27 was sufficient for inducing Myc degradation. Protein expression data of The Cancer Genome Atlas breast invasive carcinoma set revealed significantly lower Myc protein levels in tumors with highly expressed p27 lacking phosphorylation at Thr-157--a marker for active p27 localized in the nucleus. Further, these conditions correlated with favorable tumor stage and patient outcome. This novel regulation of Myc by IFN-γ/p27(KIP1) potentially offers new possibilities for therapeutic intervention in tumors with deregulated Myc.
Collapse
Affiliation(s)
- Fuad Bahram
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
- Department of Plant Biology and Forest Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
- Moreinx AB, Uppsala, Sweden
| | - Per Hydbring
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Susanna Tronnersjö
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
- GE Healthcare, Uppsala, Sweden
| | - Siti Mariam Zakaria
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Oliver Frings
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Sara Fahlén
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
- Department of Neuroscience, Swedish Medical Nanoscience Center, Karolinska Institutet, Stockholm, Sweden
| | - Helén Nilsson
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Pathology, Lund University, Lund, Sweden
| | - Jacob Goodwin
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Natalie von der Lehr
- Department of Plant Biology and Forest Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
- NatScience, Uppsala, Sweden
| | - Yingtao Su
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
- Anxun International Co., Limited, Hong Kong, China
| | - Bernhard Lüscher
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Alina Castell
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Lars-Gunnar Larsson
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
25
|
Myant K, Qiao X, Halonen T, Come C, Laine A, Janghorban M, Partanen JI, Cassidy J, Ogg EL, Cammareri P, Laiterä T, Okkeri J, Klefström J, Sears RC, Sansom OJ, Westermarck J. Serine 62-Phosphorylated MYC Associates with Nuclear Lamins and Its Regulation by CIP2A Is Essential for Regenerative Proliferation. Cell Rep 2015; 12:1019-31. [PMID: 26235622 PMCID: PMC4535171 DOI: 10.1016/j.celrep.2015.07.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 04/24/2015] [Accepted: 07/01/2015] [Indexed: 02/07/2023] Open
Abstract
An understanding of the mechanisms determining MYC's transcriptional and proliferation-promoting activities in vivo could facilitate approaches for MYC targeting. However, post-translational mechanisms that control MYC function in vivo are poorly understood. Here, we demonstrate that MYC phosphorylation at serine 62 enhances MYC accumulation on Lamin A/C-associated nuclear structures and that the protein phosphatase 2A (PP2A) inhibitor protein CIP2A is required for this process. CIP2A is also critical for serum-induced MYC phosphorylation and for MYC-elicited proliferation induction in vitro. Complementary transgenic approaches and an intestinal regeneration model further demonstrated the in vivo importance of CIP2A and serine 62 phosphorylation for MYC activity upon DNA damage. However, targeting of CIP2A did not influence the normal function of intestinal crypt cells. These data underline the importance of nuclear organization in the regulation of MYC phosphorylation, leading to an in vivo demonstration of a strategy for inhibiting MYC activity without detrimental physiological effects.
Collapse
Affiliation(s)
- Kevin Myant
- The Beatson Institute for Cancer Research, Glasgow G61 1BD, UK
| | - Xi Qiao
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Department of Pathology, University of Turku, 20520 Turku, Finland
| | - Tuuli Halonen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Christophe Come
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Anni Laine
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Mahnaz Janghorban
- Department of Molecular and Medical Genetics and Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Johanna I Partanen
- Research Programs Unit, Translational Cancer Biology and Institute of Biomedicine, University of Helsinki, 00014 Helsinki, Finland
| | - John Cassidy
- The Beatson Institute for Cancer Research, Glasgow G61 1BD, UK
| | - Erinn-Lee Ogg
- The Beatson Institute for Cancer Research, Glasgow G61 1BD, UK
| | | | - Tiina Laiterä
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Juha Okkeri
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Juha Klefström
- Research Programs Unit, Translational Cancer Biology and Institute of Biomedicine, University of Helsinki, 00014 Helsinki, Finland
| | - Rosalie C Sears
- Department of Molecular and Medical Genetics and Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Owen J Sansom
- The Beatson Institute for Cancer Research, Glasgow G61 1BD, UK.
| | - Jukka Westermarck
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Department of Pathology, University of Turku, 20520 Turku, Finland.
| |
Collapse
|
26
|
Koh CM, Khattar E, Leow SC, Liu CY, Muller J, Ang WX, Li Y, Franzoso G, Li S, Guccione E, Tergaonkar V. Telomerase regulates MYC-driven oncogenesis independent of its reverse transcriptase activity. J Clin Invest 2015; 125:2109-22. [PMID: 25893605 PMCID: PMC4463203 DOI: 10.1172/jci79134] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 03/12/2015] [Indexed: 12/25/2022] Open
Abstract
Constitutively active MYC and reactivated telomerase often coexist in cancers. While reactivation of telomerase is thought to be essential for replicative immortality, MYC, in conjunction with cofactors, confers several growth advantages to cancer cells. It is known that the reactivation of TERT, the catalytic subunit of telomerase, is limiting for reconstituting telomerase activity in tumors. However, while reactivation of TERT has been functionally linked to the acquisition of several "hallmarks of cancer" in tumors, the molecular mechanisms by which this occurs and whether these mechanisms are distinct from the role of telomerase on telomeres is not clear. Here, we demonstrated that first-generation TERT-null mice, unlike Terc-null mice, show delayed onset of MYC-induced lymphomagenesis. We further determined that TERT is a regulator of MYC stability in cancer. TERT stabilized MYC levels on chromatin, contributing to either activation or repression of its target genes. TERT regulated MYC ubiquitination and proteasomal degradation, and this effect of TERT was independent of its reverse transcriptase activity and role in telomere elongation. Based on these data, we conclude that reactivation of TERT, a direct transcriptional MYC target in tumors, provides a feed-forward mechanism to potentiate MYC-dependent oncogenesis.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cell Transformation, Neoplastic/genetics
- Enzyme Activation
- Feedback, Physiological
- Gene Expression Regulation, Neoplastic/genetics
- Genes, myc
- Glycogen Synthase Kinase 3/physiology
- Glycogen Synthase Kinase 3 beta
- Heterografts
- Humans
- Lymphoma, Non-Hodgkin/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Neoplasm Proteins/physiology
- Neoplasm Transplantation
- Phosphorylation
- Promoter Regions, Genetic
- Protein Processing, Post-Translational
- Protein Stability
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- Proto-Oncogene Proteins c-myc/physiology
- RNA/genetics
- RNA/physiology
- RNA Interference
- Telomerase/deficiency
- Telomerase/genetics
- Telomerase/physiology
- Telomere Homeostasis/genetics
- Time Factors
- Transcription, Genetic
- Ubiquitination
Collapse
Affiliation(s)
- Cheryl M. Koh
- Division of Cancer Genetics and Therapeutics, Laboratory of Methyltransferases in Development and Disease, and
| | - Ekta Khattar
- Division of Cancer Genetics and Therapeutics, Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Shi Chi Leow
- Division of Cancer Genetics and Therapeutics, Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Chia Yi Liu
- Division of Cancer Genetics and Therapeutics, Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Julius Muller
- Division of Cancer Genetics and Therapeutics, Laboratory of Methyltransferases in Development and Disease, and
| | - Wei Xia Ang
- Division of Cancer Genetics and Therapeutics, Laboratory of Methyltransferases in Development and Disease, and
| | - Yinghui Li
- Division of Cancer Genetics and Therapeutics, Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Guido Franzoso
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Shang Li
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore
- Department of Physiology and
| | - Ernesto Guccione
- Division of Cancer Genetics and Therapeutics, Laboratory of Methyltransferases in Development and Disease, and
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Vinay Tergaonkar
- Division of Cancer Genetics and Therapeutics, Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
27
|
The nucleolar ubiquitin-specific protease USP36 deubiquitinates and stabilizes c-Myc. Proc Natl Acad Sci U S A 2015; 112:3734-9. [PMID: 25775507 DOI: 10.1073/pnas.1411713112] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
c-Myc protein stability and activity are tightly regulated by the ubiquitin-proteasome system. Aberrant stabilization of c-Myc contributes to many human cancers. c-Myc is ubiquitinated by SCF(Fbw7) (a SKP1-cullin-1-F-box complex that contains the F-box and WD repeat domain-containing 7, Fbw7, as the F-box protein) and several other ubiquitin ligases, whereas it is deubiquitinated and stabilized by ubiquitin-specific protease (USP) 28. The bulk of c-Myc degradation appears to occur in the nucleolus. However, whether c-Myc is regulated by deubiquitination in the nucleolus is not known. Here, we report that the nucleolar deubiquitinating enzyme USP36 is a novel c-Myc deubiquitinase. USP36 interacts with and deubiquitinates c-Myc in cells and in vitro, leading to the stabilization of c-Myc. This USP36 regulation of c-Myc occurs in the nucleolus. Interestingly, USP36 interacts with the nucleolar Fbw7γ but not the nucleoplasmic Fbw7α. However, it abolished c-Myc degradation mediated both by Fbw7γ and by Fbw7α. Consistently, knockdown of USP36 reduces the levels of c-Myc and suppresses cell proliferation. We further show that USP36 itself is a c-Myc target gene, suggesting that USP36 and c-Myc form a positive feedback regulatory loop. High expression levels of USP36 are found in a subset of human breast and lung cancers. Altogether, these results identified USP36 as a crucial and bono fide deubiquitinating enzyme controlling c-Myc's nucleolar degradation pathway.
Collapse
|
28
|
Hill R, Kalathur RKR, Callejas S, Colaço L, Brandão R, Serelde B, Cebriá A, Blanco-Aparicio C, Pastor J, Futschik M, Dopazo A, Link W. A novel phosphatidylinositol 3-kinase (PI3K) inhibitor directs a potent FOXO-dependent, p53-independent cell cycle arrest phenotype characterized by the differential induction of a subset of FOXO-regulated genes. Breast Cancer Res 2014; 16:482. [PMID: 25488803 PMCID: PMC4303209 DOI: 10.1186/s13058-014-0482-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 11/14/2014] [Indexed: 02/01/2023] Open
Abstract
Introduction The activation of the phosphoinositide 3-kinase (PI3K)/AKT signalling pathway is one the most frequent genetic events in breast cancer, consequently the development of PI3K inhibitors has attracted much attention. Here we evaluate the effect of PI3K inhibition on global gene expression in breast cancer cells. Methods We used a range of methodologies that include in silico compound analysis, in vitro kinase assays, cell invasion assays, proliferation assays, genome-wide transcription studies (Agilent Technologies full genome arrays), gene set enrichment analysis, quantitative real-time PCR, immunoblotting in addition to chromatin immunoprecipitation. Results We defined the physico-chemical and the biological properties of ETP-45658, a novel potent PI3K inhibitor. We demonstrated that ETP-45658 potently inhibited cell proliferation within a broad range of human cancer cells, most potently suppressing the growth of breast cancer cells via inhibiting cell cycle. We show that this response is Forkhead box O (FOXO) protein dependent and p53 independent. Our genome-wide microarray analysis revealed that the cell cycle was the most affected biological process after exposure to ETP-45658 (or our control PI3K inhibitor PI-103), that despite the multiple transcription factors that are regulated by the PI3K/AKT signalling cascade, only the binding sites for FOXO transcription factors were significantly enriched and only a subset of all FOXO-dependent genes were induced. This disparity in gene transcription was not due to differential FOXO promoter recruitment. Conclusions The constitutive activation of PI3Ks and thus the exclusion of FOXO transcription factors from the nucleus is a key feature of breast cancer. Our results presented here highlight that PI3K inhibition activates specific FOXO-dependent genes that mediate cell cycle arrest in breast cancer cells. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0482-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Richard Hill
- IBB-Institute for Biotechnology and Bioengineering, Centre for Molecular and Structural Biomedicine (CBME), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal. .,Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
| | - Ravi Kiran Reddy Kalathur
- IBB-Institute for Biotechnology and Bioengineering, Centre for Molecular and Structural Biomedicine (CBME), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
| | - Sergio Callejas
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle de Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| | - Laura Colaço
- IBB-Institute for Biotechnology and Bioengineering, Centre for Molecular and Structural Biomedicine (CBME), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
| | - Ricardo Brandão
- IBB-Institute for Biotechnology and Bioengineering, Centre for Molecular and Structural Biomedicine (CBME), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
| | - Beatriz Serelde
- Experimental Therapeutics Program, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| | - Antonio Cebriá
- Experimental Therapeutics Program, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| | - Carmen Blanco-Aparicio
- Experimental Therapeutics Program, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| | - Joaquín Pastor
- Experimental Therapeutics Program, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| | - Matthias Futschik
- IBB-Institute for Biotechnology and Bioengineering, Centre for Molecular and Structural Biomedicine (CBME), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
| | - Ana Dopazo
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle de Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| | - Wolfgang Link
- IBB-Institute for Biotechnology and Bioengineering, Centre for Molecular and Structural Biomedicine (CBME), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal. .,Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
| |
Collapse
|
29
|
Watnick RS, Rodriguez RK, Wang S, Blois AL, Rangarajan A, Ince T, Weinberg RA. Thrombospondin-1 repression is mediated via distinct mechanisms in fibroblasts and epithelial cells. Oncogene 2014; 34:2823-35. [PMID: 25109329 DOI: 10.1038/onc.2014.228] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 06/03/2014] [Accepted: 06/21/2014] [Indexed: 12/27/2022]
Abstract
Tumor-associated angiogenesis is postulated to be regulated by the balance between pro- and anti-angiogenic factors. We demonstrate here that the critical step in establishing the angiogenic capability of human tumor cells is the repression of a key secreted anti-angiogenic factor, thrombospondin-1 (Tsp-1). This repression is essential for tumor formation by mammary epithelial cells and kidney cells engineered to express SV40 early region proteins, hTERT, and H-RasV12. In transformed epithelial cells, a signaling pathway leading from Ras to Tsp-1 repression induces the sequential activation of PI3 kinase, Rho and ROCK, leading to activation of Myc through phosphorylation, thereby enabling Myc to repress Tsp-1 transcription. In transformed fibroblasts, however, the repression of Tsp-1 can be achieved by an alternative mechanism involving inactivation of both p53 and pRb. We thus describe novel mechanisms by which the activation of oncogenes in epithelial cells and the inactivation of tumor suppressors in fibroblasts permits angiogenesis and, in turn, tumor formation.
Collapse
Affiliation(s)
- R S Watnick
- 1] Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA [2] Department of Surgery, Harvard Medical School, Boston, MA, USA [3] Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - R K Rodriguez
- 1] Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA [2] Department of Surgery, Harvard Medical School, Boston, MA, USA [3] Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - S Wang
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA
| | - A L Blois
- 1] Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA [2] Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - A Rangarajan
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - T Ince
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - R A Weinberg
- 1] Whitehead Institute for Biomedical Research, Cambridge, MA, USA [2] Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
30
|
Hann SR. MYC cofactors: molecular switches controlling diverse biological outcomes. Cold Spring Harb Perspect Med 2014; 4:a014399. [PMID: 24939054 DOI: 10.1101/cshperspect.a014399] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The transcription factor MYC has fundamental roles in proliferation, apoptosis, tumorigenesis, and stem cell pluripotency. Over the last 30 years extensive information has been gathered on the numerous cofactors that interact with MYC and the target genes that are regulated by MYC as a means of understanding the molecular mechanisms controlling its diverse roles. Despite significant advances and perhaps because the amount of information learned about MYC is overwhelming, there has been little consensus on the molecular functions of MYC that mediate its critical biological roles. In this perspective, the major MYC cofactors that regulate the various transcriptional activities of MYC, including canonical and noncanonical transactivation and transcriptional repression, will be reviewed and a model of how these transcriptional mechanisms control MYC-mediated proliferation, apoptosis, and tumorigenesis will be presented. The basis of the model is that a variety of cofactors form dynamic MYC transcriptional complexes that can switch the molecular and biological functions of MYC to yield a diverse range of outcomes in a cell-type- and context-dependent fashion.
Collapse
Affiliation(s)
- Stephen R Hann
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-2175
| |
Collapse
|
31
|
Su WM, Han GS, Carman GM. Cross-talk phosphorylations by protein kinase C and Pho85p-Pho80p protein kinase regulate Pah1p phosphatidate phosphatase abundance in Saccharomyces cerevisiae. J Biol Chem 2014; 289:18818-30. [PMID: 24876385 DOI: 10.1074/jbc.m114.581462] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Yeast Pah1p is the phosphatidate phosphatase that catalyzes the penultimate step in triacylglycerol synthesis and plays a role in the transcriptional regulation of phospholipid synthesis genes. The enzyme is multiply phosphorylated, some of which is mediated by Pho85p-Pho80p, Cdc28p-cyclin B, and protein kinase A. Here, we showed that Pah1p is a bona fide substrate of protein kinase C; the phosphorylation reaction was time- and dose-dependent and dependent on the concentrations of ATP (Km = 4.5 μm) and Pah1p (Km = 0.75 μm). The stoichiometry of the reaction was 0.8 mol of phosphate/mol of Pah1p. By combining mass spectrometry, truncation analysis, site-directed mutagenesis, and phosphopeptide mapping, we identified Ser-677, Ser-769, Ser-773, and Ser-788 as major sites of phosphorylation. Analysis of Pah1p phosphorylations by different protein kinases showed that prephosphorylation with protein kinase C reduces its subsequent phosphorylation with protein kinase A and vice versa. Prephosphorylation with Pho85p-Pho80p had an inhibitory effect on its subsequent phosphorylation with protein kinase C; however, prephosphorylation with protein kinase C had no effect on the subsequent phosphorylation with Pho85p-Pho80p. Unlike its phosphorylations by Pho85p-Pho80p and protein kinase A, which cause a significant reduction in phosphatidate phosphatase activity, the phosphorylation of Pah1p by protein kinase C had a small stimulatory effect on the enzyme activity. Analysis of phosphorylation-deficient forms of Pah1p indicated that protein kinase C does not have a major effect on its location or its function in triacylglycerol synthesis, but instead, the phosphorylation favors loss of Pah1p abundance when it is not phosphorylated with Pho85p-Pho80p.
Collapse
Affiliation(s)
- Wen-Min Su
- From the Department of Food Science, Rutgers Center for Lipid Research, and New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey 08901
| | - Gil-Soo Han
- From the Department of Food Science, Rutgers Center for Lipid Research, and New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey 08901
| | - George M Carman
- From the Department of Food Science, Rutgers Center for Lipid Research, and New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey 08901
| |
Collapse
|
32
|
Abstract
The MYC oncoprotein is an essential transcription factor that regulates the expression of many genes involved in cell growth, proliferation, and metabolic pathways. Thus, it is important to keep MYC activity in check in normal cells in order to avoid unwanted oncogenic changes. Normal cells have adapted several ways to control MYC levels, and these mechanisms can be disrupted in cancer cells. One of the major ways in which MYC levels are controlled in cells is through targeted degradation by the ubiquitin-proteasome system (UPS). Here, we discuss the role of the UPS in the regulation of MYC protein levels and review some of the many proteins that have been shown to regulate MYC protein stability. In addition, we discuss how this relates to MYC transcriptional activity, human cancers, and therapeutic targeting.
Collapse
Affiliation(s)
- Amy S Farrell
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon 97239
| | | |
Collapse
|
33
|
Abstract
Neuroblastoma, the most common extracranial solid tumor of childhood, is thought to originate from undifferentiated neural crest cells. Amplification of the MYC family member, MYCN, is found in ∼25% of cases and correlates with high-risk disease and poor prognosis. Currently, amplification of MYCN remains the best-characterized genetic marker of risk in neuroblastoma. This article reviews roles for MYCN in neuroblastoma and highlights recent identification of other driver mutations. Strategies to target MYCN at the level of protein stability and transcription are also reviewed.
Collapse
Affiliation(s)
- Miller Huang
- Departments of Neurology, Pediatrics, and Neurosurgery, University of California, San Francisco, California 94158-9001
| | | |
Collapse
|
34
|
Zhou X, Hao Q, Liao JM, Liao P, Lu H. Ribosomal protein S14 negatively regulates c-Myc activity. J Biol Chem 2013; 288:21793-801. [PMID: 23775087 PMCID: PMC3724636 DOI: 10.1074/jbc.m112.445122] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 05/16/2013] [Indexed: 12/13/2022] Open
Abstract
The ribosomal gene RPS14 is associated with the cancer-prone 5q-syndrome, which is caused by an interstitial deletion of the long arm of human chromosome 5. Previously, we found that ribosomal protein S14 (RPS14) binds to and inactivates MDM2, consequently leading to p53-dependent cell-cycle arrest and growth inhibition. However, it remains elusive whether RPS14 regulates cell proliferation in a p53-independent manner. Here, we show that RPS14 interacts with the Myc homology box II (MBII) and the C-terminal basic helix-loop-helix leucine zipper (bHLH-LZ) domains of the oncoprotein c-Myc. Further, RPS14 inhibited c-Myc transcriptional activity by preventing the recruitment of c-Myc and its cofactor, TRRAP, to the target gene promoters, as thus suppressing c-Myc-induced cell proliferation. Also, siRNA-mediated RPS14 depletion elevated c-Myc transcriptional activity determined by its target gene, Nucleolin, expression. Interestingly, RPS14 depletion also resulted in the induction of c-Myc mRNA and subsequent protein levels. Consistent with this, RPS14 promoted c-Myc mRNA turnover through an Argonaute 2 (Ago2)- and microRNA-mediated pathway. Taken together, our study demonstrates that RPS14 negates c-Myc functions by directly inhibiting its transcriptional activity and mediating its mRNA degradation via miRNA.
Collapse
Affiliation(s)
- Xiang Zhou
- From the Department of Biochemistry & Molecular Biology and Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - Qian Hao
- From the Department of Biochemistry & Molecular Biology and Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - Jun-ming Liao
- From the Department of Biochemistry & Molecular Biology and Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - Peng Liao
- From the Department of Biochemistry & Molecular Biology and Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - Hua Lu
- From the Department of Biochemistry & Molecular Biology and Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana 70112
| |
Collapse
|
35
|
Pin1 regulates the dynamics of c-Myc DNA binding to facilitate target gene regulation and oncogenesis. Mol Cell Biol 2013; 33:2930-49. [PMID: 23716601 DOI: 10.1128/mcb.01455-12] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The Myc oncoprotein is considered a master regulator of gene transcription by virtue of its ability to modulate the expression of a large percentage of all genes. However, mechanisms that direct Myc's recruitment to DNA and target gene selection to elicit specific cellular functions have not been well elucidated. Here, we report that the Pin1 prolyl isomerase enhances recruitment of serine 62-phosphorylated Myc and its coactivators to select promoters during gene activation, followed by promoting Myc's release associated with its degradation. This facilitates Myc's activation of genes involved in cell growth and metabolism, resulting in enhanced proproliferative activity, even while controlling Myc levels. In cancer cells with impaired Myc degradation, Pin1 still enhances Myc DNA binding, although it no longer facilitates Myc degradation. Thus, we find that Pin1 and Myc are cooverexpressed in cancer, and this drives a gene expression pattern that we show is enriched in poor-outcome breast cancer subtypes. This study provides new insight into mechanisms regulating Myc DNA binding and oncogenic activity, it reveals a novel role for Pin1 in the regulation of transcription factors, and it elucidates a mechanism that can contribute to oncogenic cooperation between Pin1 and Myc.
Collapse
|
36
|
Funakoshi-Tago M, Sumi K, Kasahara T, Tago K. Critical roles of Myc-ODC axis in the cellular transformation induced by myeloproliferative neoplasm-associated JAK2 V617F mutant. PLoS One 2013; 8:e52844. [PMID: 23300995 PMCID: PMC3536786 DOI: 10.1371/journal.pone.0052844] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 11/21/2012] [Indexed: 12/13/2022] Open
Abstract
The acquired mutation (V617F) of Janus kinase 2 (JAK2) is observed in the majority of patients with myeloproliferative neoplasms (MPNs). In the screening of genes whose expression was induced by JAK2 (V617F), we found the significant induction of c-Myc mRNA expression mediated by STAT5 activation. Interestingly, GSK-3β was inactivated in transformed Ba/F3 cells by JAK2 (V617F), and this enhanced the protein expression of c-Myc. The enforced expression of c-Myc accelerated cell proliferation but failed to inhibit apoptotic cell death caused by growth factor deprivation; however, the inhibition of GSK-3β completely inhibited the apoptosis of cells expressing c-Myc. Strikingly, c-Myc T58A mutant exhibited higher proliferative activity in a growth-factor-independent manner; however, this mutant failed to induce apoptosis. In addition, knockdown of c-Myc significantly inhibited the proliferation of transformed cells by JAK2 (V617F), suggesting that c-Myc plays an important role in oncogenic activity of JAK2 (V617F). Furthermore, JAK2 (V617F) induced the expression of a target gene of c-Myc, ornithine decarboxylase (ODC), known as the rate-limiting enzyme in polyamine biosynthesis. An ODC inhibitor, difluoromethylornithine (DFMO), prevented the proliferation of transformed cells by JAK2 (V617F). Importantly, administration of DFMO effectively delayed tumor formation in nude mice inoculated with transformed cells by JAK2 (V617F), resulting in prolonged survival; therefore, ODC expression through c-Myc is a critical step for JAK2 (V617F)-induced transformation and DFMO could be used as effective therapy for MPNs.
Collapse
Affiliation(s)
- Megumi Funakoshi-Tago
- Department of Biochemistry, Faculty of Pharmacology, Keio University, Tokyo, Japan
- * E-mail: (MF-T); (KT)
| | - Kazuya Sumi
- Department of Biochemistry, Faculty of Pharmacology, Keio University, Tokyo, Japan
| | - Tadashi Kasahara
- Department of Biochemistry, Faculty of Pharmacology, Keio University, Tokyo, Japan
| | - Kenji Tago
- Division of Structural Biochemistry, Department of Biochemistry, Jichi Medical University, Shimotsuke-shi, Japan
- * E-mail: (MF-T); (KT)
| |
Collapse
|
37
|
Chung EY, Psathas JN, Yu D, Li Y, Weiss MJ, Thomas-Tikhonenko A. CD19 is a major B cell receptor-independent activator of MYC-driven B-lymphomagenesis. J Clin Invest 2012; 122:2257-66. [PMID: 22546857 DOI: 10.1172/jci45851] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 03/27/2012] [Indexed: 12/13/2022] Open
Abstract
PAX5, a B cell-specific transcription factor, is overexpressed through chromosomal translocations in a subset of B cell lymphomas. Previously, we had shown that activation of immunoreceptor tyrosine-based activation motif (ITAM) proteins and B cell receptor (BCR) signaling by PAX5 contributes to B-lymphomagenesis. However, the effect of PAX5 on other oncogenic transcription factor-controlled pathways is unknown. Using a MYC-induced murine lymphoma model as well as MYC-transformed human B cell lines, we found that PAX5 controls c-MYC protein stability and steady-state levels. This promoter-independent, posttranslational mechanism of c-MYC regulation was independent of ITAM/BCR activity. Instead it was controlled by another PAX5 target, CD19, through the PI3K-AKT-GSK3β axis. Consequently, MYC levels in B cells from CD19-deficient mice were sharply reduced. Conversely, reexpression of CD19 in murine lymphomas with spontaneous silencing of PAX5 boosted MYC levels, expression of its key target genes, cell proliferation in vitro, and overall tumor growth in vivo. In human B-lymphomas, CD19 mRNA levels were found to correlate with those of MYC-activated genes. They also negatively correlated with the overall survival of patients with lymphoma in the same way that MYC levels do. Thus, CD19 is a major BCR-independent regulator of MYC-driven neoplastic growth in B cell neoplasms.
Collapse
Affiliation(s)
- Elaine Y Chung
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia 19104-4399, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
38
|
Taira N, Mimoto R, Kurata M, Yamaguchi T, Kitagawa M, Miki Y, Yoshida K. DYRK2 priming phosphorylation of c-Jun and c-Myc modulates cell cycle progression in human cancer cells. J Clin Invest 2012; 122:859-72. [PMID: 22307329 PMCID: PMC3287383 DOI: 10.1172/jci60818] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 12/21/2011] [Indexed: 02/06/2023] Open
Abstract
Dysregulation of the G(1)/S transition in the cell cycle contributes to tumor development. The oncogenic transcription factors c-Jun and c-Myc are indispensable regulators at this transition, and their aberrant expression is associated with many malignancies. Degradation of c-Jun/c-Myc is a critical process for the G(1)/S transition, which is initiated upon phosphorylation by glycogen synthase kinase 3 β (GSK3β). However, a specific kinase or kinases responsible for priming phosphorylation events that precede this GSK3β modification has not been definitively identified. Here, we found that the dual-specificity tyrosine phosphorylation-regulated kinase DYRK2 functions as a priming kinase of c-Jun and c-Myc. Knockdown of DYRK2 in human cancer cells shortened the G(1) phase and accelerated cell proliferation due to escape of c-Jun and c-Myc from ubiquitination-mediated degradation. In concert with these results, silencing DYRK2 increased cell proliferation in human cancer cells, and this promotion was completely impeded by codeprivation of c-Jun or c-Myc in vivo. We also found marked attenuation of DYRK2 expression in multiple human tumor samples. Downregulation of DYRK2 correlated with high levels of unphosphorylated c-Jun and c-Myc and, importantly, with invasiveness of human breast cancers. These results reveal that DYRK2 regulates tumor progression through modulation of c-Jun and c-Myc.
Collapse
Affiliation(s)
- Naoe Taira
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
Department of Comprehensive Pathology, Aging and Developmental Sciences, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.
Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Rei Mimoto
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
Department of Comprehensive Pathology, Aging and Developmental Sciences, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.
Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Morito Kurata
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
Department of Comprehensive Pathology, Aging and Developmental Sciences, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.
Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Tomoko Yamaguchi
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
Department of Comprehensive Pathology, Aging and Developmental Sciences, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.
Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Masanobu Kitagawa
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
Department of Comprehensive Pathology, Aging and Developmental Sciences, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.
Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshio Miki
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
Department of Comprehensive Pathology, Aging and Developmental Sciences, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.
Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Kiyotsugu Yoshida
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
Department of Comprehensive Pathology, Aging and Developmental Sciences, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.
Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
39
|
Abstract
The iconic history of the Myc oncoprotein encompasses 3 decades of intense scientific discovery. There is no question that Myc has been a pioneer, advancing insight into the molecular basis of cancer as well as functioning as a critical control center for several diverse biological processes and regulatory mechanisms. This narrative chronicles the journey and milestones that have defined the understanding of Myc, and it provides an opportunity to consider future directions in this challenging yet rewarding field.
Collapse
Affiliation(s)
- Amanda R Wasylishen
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
40
|
Kapeli K, Hurlin PJ. Differential regulation of N-Myc and c-Myc synthesis, degradation, and transcriptional activity by the Ras/mitogen-activated protein kinase pathway. J Biol Chem 2011; 286:38498-38508. [PMID: 21908617 DOI: 10.1074/jbc.m111.276675] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Myc transcription factors are important regulators of proliferation and can promote oncogenesis when deregulated. Deregulated Myc expression in cancers can result from MYC gene amplification and translocation but also from alterations in mitogenic signaling pathways that affect Myc levels through both transcriptional and post-transcription mechanisms. For example, mutations in Ras family GTPase proteins that cause their constitutive activation can increase cellular levels of c-Myc by interfering with its rapid proteasomal degradation. Although enhanced protein stability is generally thought to be applicable to other Myc family members, here we show that c-Myc and its paralog N-Myc respond to oncogenic H-Ras (H-Ras(G12V)) in very different ways. H-Ras(G12V) promotes accumulation of both c-Myc and N-Myc, but although c-Myc accumulation is achieved by enhanced protein stability, N-Myc accumulation is associated with an accelerated rate of translation that overcomes a surprising H-Ras(G12V)-mediated destabilization of N-Myc. We show that H-Ras(G12V)-mediated degradation of N-Myc functions independently of key phosphorylation sites in the highly conserved Myc homology box I region that controls c-Myc protein stability by oncogenic Ras. Finally, we found that N-Myc and c-Myc transcriptional activity is associated with their proteasomal degradation but that N-Myc may be uniquely dependent on Ras-stimulated proteolysis for target gene expression. Taken together, these studies provide mechanistic insight into how oncogenic Ras augments N-Myc levels in cells and suggest that enhanced N-Myc translation and degradation-coupled transactivation may contribute to oncogenesis.
Collapse
Affiliation(s)
- Katannya Kapeli
- Department of Cell and Developmental Biology, Oregon Health and Science University, Portland, Oregon 97239
| | - Peter J Hurlin
- Department of Cell and Developmental Biology, Oregon Health and Science University, Portland, Oregon 97239; Shriners Hospital for Children, Portland, Oregon 97239.
| |
Collapse
|
41
|
PP2A-B56α controls oncogene-induced senescence in normal and tumor human melanocytic cells. Oncogene 2011; 31:1484-92. [PMID: 21822300 PMCID: PMC3213274 DOI: 10.1038/onc.2011.339] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Oncoprotein C-MYC is overexpressed in human metastatic melanomas and melanoma-derived cells where it is required for suppression of oncogene-induced senescence (OIS). The genetic events that maintain high levels of C-MYC in melanoma cells and their role in OIS are unknown. Here, we report that C-MYC in cells from several randomly chosen melanoma lines was up-regulated at the protein level, and largely due to the increased protein stability. Of all known regulators of C-MYC stability, levels of B56α subunit of the PP2A tumor suppressor complex were substantially suppressed in all human melanoma cells compared to normal melanocytes. Accordingly, immuno-histochemical analysis revealed that the lowest and the highest amounts of PP2A-B56α were predominantly detected in metastatic melanoma tissues and in primary melanomas from patients with good clinical outcome, respectively. Importantly, PP2A-B56α overexpression suppressed C-MYC in melanoma cells and induced OIS, whereas depletion of PP2A-B56α in normal human melanocytes up-regulated C-MYC protein levels and suppressed BRAFV600E- and, less efficiently, NRASQ61R-induced senescence. Our data reveal a mechanism of C-MYC overexpression in melanoma cells and identify a functional role for PP2A-B56α in OIS of melanocytic cells.
Collapse
|
42
|
Paek H, Hwang JY, Zukin RS, Hébert JM. β-Catenin-dependent FGF signaling sustains cell survival in the anterior embryonic head by countering Smad4. Dev Cell 2011; 20:689-99. [PMID: 21571225 DOI: 10.1016/j.devcel.2011.04.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 03/02/2011] [Accepted: 04/26/2011] [Indexed: 12/22/2022]
Abstract
Growing evidence suggests that FGFs secreted from embryonic signaling centers are key mediators of cell survival. However, the mechanisms regulating FGF-dependent cell survival remain obscure. At the rostral end of the embryo, for example, ablation of FGF signaling leads to the rapid death of the precursor cells that form the anterior head, including the telencephalon. Here, we outline a core genetic circuit that regulates survival in the embryonic mouse head: WNT signaling through β-catenin directly maintains FGF expression and requires FGF function in vivo to oppose proapoptotic TGF-β signaling through SMAD4. Moreover, these antagonistic pathways converge on the transcriptional regulation of apoptosis, and genes such as Cdkn1a, suggesting a mechanism for how signaling centers in the embryonic head regulate cell survival.
Collapse
Affiliation(s)
- Hunki Paek
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | |
Collapse
|
43
|
Mayes PA, Dolloff NG, Daniel CJ, Liu JJ, Hart LS, Kuribayashi K, Allen JE, Jee DIH, Dorsey JF, Liu YY, Dicker DT, Brown JM, Furth EE, Klein PS, Sears RC, El-Deiry WS. Overcoming hypoxia-induced apoptotic resistance through combinatorial inhibition of GSK-3β and CDK1. Cancer Res 2011; 71:5265-75. [PMID: 21646472 DOI: 10.1158/0008-5472.can-11-1383] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor hypoxia is an inherent impediment to cancer treatment that is both clinically significant and problematic. In this study, we conducted a cell-based screen to identify small molecules that could reverse the apoptotic resistance of hypoxic cancer cells. Among the compounds, we identified were a structurally related group that sensitized hypoxic cancer cells to apoptosis by inhibiting the kinases GSK-3β and cyclin-dependent kinase (CDK) 1. Combinatorial inhibition of these proteins in hypoxic cancer cells and tumors increased levels of c-Myc and decreased expression of c-IAP2 and the central hypoxia response regulator hypoxia-inducible factor (HIF) 1α. In mice, these compounds augmented the hypoxic tumor cell death induced by cytotoxic chemotherapy, blocking angiogenesis and tumor growth. Taken together, our findings suggest that combinatorial inhibition of GSK-3β and CDK1 augment the apoptotic sensitivity of hypoxic tumors, and they offer preclinical validation of a novel and readily translatable strategy to improve cancer therapy.
Collapse
Affiliation(s)
- Patrick A Mayes
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Simpson DR, Yu M, Zheng S, Zhao Z, Muthuswamy SK, Tansey WP. Epithelial cell organization suppresses Myc function by attenuating Myc expression. Cancer Res 2011; 71:3822-30. [PMID: 21610111 DOI: 10.1158/0008-5472.can-10-3782] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
c-Myc is an oncogene transcription factor that causes cancer in many settings, including solid tumors that arise in the context of organized tissue structures. Given that disruption of tissue architecture frequently occurs in cancer, there is considerable interest in how cell organization impacts oncogene function. A previous report found that organization of mammary epithelial cells into defined 3-dimensional structures renders them insensitive to the effects of retrovirus-mediated overexpression of Myc, leading to the notion that organization tempers the sensitivity of individual cells to Myc activity. In this article, we report that epithelial cell organization does not profoundly alter Myc activity but, instead, suppresses Myc by modulating its expression. We show that the morphogenesis of mammary epithelial cells into organized acinar structures in vitro is accompanied by widespread changes in gene expression patterns, including a substantial decrease in the expression of Myc. Concomitant with the decrease in endogenous Myc expression, we observe a decrease in transcription from retroviral vectors during morphogenesis and find that Myc transgene expression in acini is much lower than in unorganized cells. This decrease in Myc transgene activity is responsible for the apparent recalcitrance of organized cells to ectopic Myc, as adenovirus-mediated expression of Myc in organized structures potently induces apoptosis. These observations reveal that organization does not alter the inherent response of epithelial cells to Myc and suggest that other tumor suppression mechanisms, apart from structure, antagonize Myc in the development of solid tumors.
Collapse
Affiliation(s)
- David R Simpson
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | | | | | | | |
Collapse
|
45
|
Role of helix-loop-helix proteins during differentiation of erythroid cells. Mol Cell Biol 2011; 31:1332-43. [PMID: 21282467 DOI: 10.1128/mcb.01186-10] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Helix-loop-helix (HLH) proteins play a profound role in the process of development and cellular differentiation. Among the HLH proteins expressed in differentiating erythroid cells are the ubiquitous proteins Myc, USF1, USF2, and TFII-I, as well as the hematopoiesis-specific transcription factor Tal1/SCL. All of these HLH proteins exhibit distinct functions during the differentiation of erythroid cells. For example, Myc stimulates the proliferation of erythroid progenitor cells, while the USF proteins and Tal1 regulate genes that specify the differentiated phenotype. This minireview summarizes the known activities of Myc, USF, TFII-I, and Tal11/SCL and discusses how they may function sequentially, cooperatively, or antagonistically in regulating expression programs during the differentiation of erythroid cells.
Collapse
|
46
|
Wang X, Cunningham M, Zhang X, Tokarz S, Laraway B, Troxell M, Sears RC. Phosphorylation regulates c-Myc's oncogenic activity in the mammary gland. Cancer Res 2011; 71:925-36. [PMID: 21266350 DOI: 10.1158/0008-5472.can-10-1032] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Expression of the c-Myc oncoprotein is affected by conserved threonine 58 (T58) and serine 62 (S62) phosphorylation sites that help to regulate c-Myc protein stability, and altered ratios of T58 and S62 phosphorylation have been observed in human cancer. Here, we report the development of 3 unique c-myc knock-in mice that conditionally express either c-Myc(WT) or the c-Myc(T58A) or c-Myc(S62A) phosphorylation mutant from the constitutively active ROSA26 locus in response to Cre recombinase to study the role of these phosphorylation sites in vivo. Using a mammary-specific Cre model, we found that expression of c-Myc(WT) resulted in increased mammary gland density, but normal morphology and no tumors at the level expressed from the ROSA promoter. In contrast, c-Myc(T58A) expression yielded enhanced mammary gland density, hyperplastic foci, cellular dysplasia, and mammary carcinoma, associated with increased genomic instability and suppressed apoptosis relative to c-Myc(WT). Alternatively, c-Myc(S62A) expression reduced mammary gland density relative to control glands, and this was associated with increased genomic instability and normal apoptotic function. Our results indicate that specific activities of c-Myc are differentially affected by T58 and S62 phosphorylation. This model provides a robust platform to interrogate the role that these phosphorylation sites play in c-Myc function during development and tumorigenesis.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Molecular and Medical Genetics Department, Oregon Health and Sciences University, Portland, Oregon, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Cao Z, Fan-Minogue H, Bellovin DI, Yevtodiyenko A, Arzeno J, Yang Q, Gambhir SS, Felsher DW. MYC phosphorylation, activation, and tumorigenic potential in hepatocellular carcinoma are regulated by HMG-CoA reductase. Cancer Res 2011; 71:2286-97. [PMID: 21262914 DOI: 10.1158/0008-5472.can-10-3367] [Citation(s) in RCA: 148] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
MYC is a potential target for many cancers but is not amenable to existing pharmacologic approaches. Inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMG-CoA reductase) by statins has shown potential efficacy against a number of cancers. Here, we show that inhibition of HMG-CoA reductase by atorvastatin (AT) blocks both MYC phosphorylation and activation, suppressing tumor initiation and growth in vivo in a transgenic model of MYC-induced hepatocellular carcinoma (HCC) as well as in human HCC-derived cell lines. To confirm specificity, we show that the antitumor effects of AT are blocked by cotreatment with the HMG-CoA reductase product mevalonate. Moreover, by using a novel molecular imaging sensor, we confirm that inhibition of HMG-CoA reductase blocks MYC phosphorylation in vivo. Importantly, the introduction of phosphorylation mutants of MYC at Ser62 or Thr58 into tumors blocks their sensitivity to inhibition of HMG-CoA reductase. Finally, we show that inhibition of HMG-CoA reductase suppresses MYC phosphorylation through Rac GTPase. Therefore, HMG-CoA reductase is a critical regulator of MYC phosphorylation, activation, and tumorigenic properties. The inhibition of HMG-CoA reductase may be a useful target for the treatment of MYC-associated HCC as well as other tumors.
Collapse
Affiliation(s)
- Zhongwei Cao
- Division of Medical Oncology, Department of Medicine and Pathology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California 94305, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Buraschi S, Pal N, Tyler-Rubinstein N, Owens RT, Neill T, Iozzo RV. Decorin antagonizes Met receptor activity and down-regulates {beta}-catenin and Myc levels. J Biol Chem 2010; 285:42075-85. [PMID: 20974860 PMCID: PMC3009933 DOI: 10.1074/jbc.m110.172841] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Revised: 10/13/2010] [Indexed: 12/18/2022] Open
Abstract
A theme emerging during the past few years is that members of the small leucine-rich proteoglycan gene family affect cell growth by interacting with multiple receptor tyrosine kinases (RTKs), mostly by a physical down-regulation of the receptors, thereby depriving tumor cells of pro-survival signals. Decorin binds and down-regulates several RTKs, including Met, the receptor for hepatocyte growth factor. Here we demonstrate that decorin blocks several biological activities mediated by the Met signaling axis, including cell scatter, evasion, and migration. These effects were mediated by a profound down-regulation of noncanonical β-catenin levels. In addition, Myc, a downstream target of β-catenin, was markedly down-regulated by decorin, whereas phosphorylation of Myc at threonine 58 was markedly induced. The latter is known to destabilize Myc and target it for proteasomal degradation. We also discovered that systemic delivery of decorin using three distinct tumor xenograft models caused down-regulation of Met and a concurrent suppression of β-catenin and Myc levels. We found that decorin protein core labeled with the near infrared dye IR800 specifically targeted the tumor cells expressing Met. Even 68-h post-injection, decorin was found to reside within the tumor xenografts with little or no binding to other tissues. Collectively, our results indicate a role for a secreted proteoglycan in suppressing the expression of key oncogenic factors required for tumor progression.
Collapse
Affiliation(s)
- Simone Buraschi
- From the Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107 and
| | - Nutan Pal
- From the Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107 and
| | - Nadia Tyler-Rubinstein
- From the Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107 and
| | | | - Thomas Neill
- From the Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107 and
| | - Renato V. Iozzo
- From the Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107 and
| |
Collapse
|
49
|
The ubiquitous nature of cancer: the role of the SCF(Fbw7) complex in development and transformation. Oncogene 2010; 29:4865-73. [PMID: 20543859 DOI: 10.1038/onc.2010.222] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The ubiquitin-proteasome system (UPS) is a multi-subunit pathway that allows for ubiquitin modification of proteins and leads to either degradation or other non-proteolytic processes such as trafficking or transcriptional activation. Given its role as a regulator of cellular homeostasis it is not surprising that members of the UPS are frequently aberrantly expressed in a number of disease states including cancer. This review will focus on one member of the UPS, the F-box protein, Fbw7 (also known as Sel-10, Ago, hCDC4) and mechanisms by which Fbw7 interacts with its substrates in the context of development and tumorigenesis will be discussed. In addition, antagonists of this pathway as well as current and future therapeutics for the UPS will be examined.
Collapse
|
50
|
Stem cell-specific activation of an ancestral myc protooncogene with conserved basic functions in the early metazoan Hydra. Proc Natl Acad Sci U S A 2010; 107:4051-6. [PMID: 20142507 DOI: 10.1073/pnas.0911060107] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The c-myc protooncogene encodes a transcription factor (Myc) with oncogenic potential. Myc and its dimerization partner Max are bHLH-Zip DNA binding proteins controlling fundamental cellular processes. Deregulation of c-myc leads to tumorigenesis and is a hallmark of many human cancers. We have identified and extensively characterized ancestral forms of myc and max genes from the early diploblastic cnidarian Hydra, the most primitive metazoan organism employed so far for the structural, functional, and evolutionary analysis of these genes. Hydra myc is specifically activated in all stem cells and nematoblast nests which represent the rapidly proliferating cell types of the interstitial stem cell system and in proliferating gland cells. In terminally differentiated nerve cells, nematocytes, or epithelial cells, myc expression is not detectable by in situ hybridization. Hydra max exhibits a similar expression pattern in interstitial cell clusters. The ancestral Hydra Myc and Max proteins display the principal design of their vertebrate derivatives, with the highest degree of sequence identities confined to the bHLH-Zip domains. Furthermore, the 314-amino acid Hydra Myc protein contains basic forms of the essential Myc boxes I through III. A recombinant Hydra Myc/Max complex binds to the consensus DNA sequence CACGTG with high affinity. Hybrid proteins composed of segments from the retroviral v-Myc oncoprotein and the Hydra Myc protein display oncogenic potential in cell transformation assays. Our results suggest that the principal functions of the Myc master regulator arose very early in metazoan evolution, allowing their dissection in a simple model organism showing regenerative ability but no senescence.
Collapse
|