1
|
Montoya-Novoa I, Gardeazábal-Torbado JL, Alegre-Martí A, Fuentes-Prior P, Estébanez-Perpiñá E. Androgen receptor post-translational modifications and their implications for pathology. Biochem Soc Trans 2024; 52:1673-1694. [PMID: 38958586 DOI: 10.1042/bst20231082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 07/04/2024]
Abstract
A major mechanism to modulate the biological activities of the androgen receptor (AR) involves a growing number of post-translational modifications (PTMs). In this review we summarise the current knowledge on the structural and functional impact of PTMs that affect this major transcription factor. Next, we discuss the cross-talk between these different PTMs and the presence of clusters of modified residues in the AR protein. Finally, we discuss the implications of these covalent modifications for the aetiology of diseases such as spinal and bulbar muscular atrophy (Kennedy's disease) and prostate cancer, and the perspectives for pharmacological intervention.
Collapse
Affiliation(s)
- Inés Montoya-Novoa
- Structural Biology of Nuclear Receptors, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona (UB), 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona (UB), 08028 Barcelona, Spain
| | - José Luis Gardeazábal-Torbado
- Structural Biology of Nuclear Receptors, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona (UB), 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona (UB), 08028 Barcelona, Spain
| | - Andrea Alegre-Martí
- Structural Biology of Nuclear Receptors, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona (UB), 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona (UB), 08028 Barcelona, Spain
| | - Pablo Fuentes-Prior
- Structural Biology of Nuclear Receptors, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona (UB), 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona (UB), 08028 Barcelona, Spain
| | - Eva Estébanez-Perpiñá
- Structural Biology of Nuclear Receptors, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona (UB), 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona (UB), 08028 Barcelona, Spain
| |
Collapse
|
2
|
Pornour M, Jeon HY, Ryu H, Khadka S, Xu R, Chen H, Hussain A, Lam HM, Zhuang Z, Oo HZ, Gleave M, Dong X, Wang Q, Barbieri C, Qi J. USP11 promotes prostate cancer progression by up-regulating AR and c-Myc activity. Proc Natl Acad Sci U S A 2024; 121:e2403331121. [PMID: 39052835 PMCID: PMC11295044 DOI: 10.1073/pnas.2403331121] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024] Open
Abstract
Androgen receptor (AR) is a main driver for castration-resistant prostate cancer (CRPC). c-Myc is an oncogene underlying prostate tumorigenesis. Here, we find that the deubiquitinase USP11 targets both AR and c-Myc in prostate cancer (PCa). USP11 expression was up-regulated in metastatic PCa and CRPC. USP11 knockdown (KD) significantly inhibited PCa cell growth. Our RNA-seq studies revealed AR and c-Myc as the top transcription factors altered after USP11 KD. ChIP-seq analysis showed that either USP11 KD or replacement of endogenous USP11 with a catalytic-inactive USP11 mutant significantly decreased chromatin binding by AR and c-Myc. We find that USP11 employs two mechanisms to up-regulate AR and c-Myc levels: namely, deubiquitination of AR and c-Myc proteins to increase their stability and deubiquitination of H2A-K119Ub, a repressive histone mark, on promoters of AR and c-Myc genes to increase their transcription. AR and c-Myc reexpression in USP11-KD PCa cells partly rescued cell growth defects. Thus, our studies reveal a tumor-promoting role for USP11 in aggressive PCa through upregulation of AR and c-Myc activities and support USP11 as a potential target against PCa.
Collapse
Affiliation(s)
- Majid Pornour
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD21201
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD21201
| | - Hee-Young Jeon
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD21201
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD21201
| | - Hyunju Ryu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD21201
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD21201
| | - Sudeep Khadka
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD21201
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD21201
| | - Rui Xu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD21201
- Department of Marine Biotechnology, Institute of Marine and Environmental Technology, University of Maryland, Baltimore, MD21202
| | - Hegang Chen
- Department of Epidemiology and Public Health, University of Maryland, Baltimore, MD21201
| | - Arif Hussain
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD21201
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD21201
- Baltimore Veterans Affairs Medical Center, Baltimore, MD21201
| | - Hung-Ming Lam
- Department of Urology, University of Washington, Seattle, WA98195
| | - Zhihao Zhuang
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE19716
| | - Htoo Zarni Oo
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BCV6H 3Z6, Canada
| | - Martin Gleave
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BCV6H 3Z6, Canada
| | - Xuesen Dong
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BCV6H 3Z6, Canada
| | - Qianben Wang
- Department of Pathology and Duke Cancer Institute, Duke University School of Medicine, Durham, NC27710
| | - Christopher Barbieri
- Department of Urology, Weill Cornell Medical College, Cornell University, New York, NY10065
| | - Jianfei Qi
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD21201
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD21201
| |
Collapse
|
3
|
Migale R, Neumann M, Mitter R, Rafiee MR, Wood S, Olsen J, Lovell-Badge R. FOXL2 interaction with different binding partners regulates the dynamics of ovarian development. SCIENCE ADVANCES 2024; 10:eadl0788. [PMID: 38517962 PMCID: PMC10959415 DOI: 10.1126/sciadv.adl0788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 02/16/2024] [Indexed: 03/24/2024]
Abstract
The transcription factor FOXL2 is required in ovarian somatic cells for female fertility. Differential timing of Foxl2 deletion, in embryonic versus adult mouse ovary, leads to distinctive outcomes, suggesting different roles across development. Here, we comprehensively investigated FOXL2's role through a multi-omics approach to characterize gene expression dynamics and chromatin accessibility changes, coupled with genome-wide identification of FOXL2 targets and on-chromatin interacting partners in somatic cells across ovarian development. We found that FOXL2 regulates more targets postnatally, through interaction with factors regulating primordial follicle formation and steroidogenesis. Deletion of one interactor, ubiquitin-specific protease 7 (Usp7), results in impairment of somatic cell differentiation, germ cell nest breakdown, and ovarian development, leading to sterility. Our datasets constitute a comprehensive resource for exploration of the molecular mechanisms of ovarian development and causes of female infertility.
Collapse
Affiliation(s)
- Roberta Migale
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| | - Michelle Neumann
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| | - Richard Mitter
- Bioinformatics core, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Mahmoud-Reza Rafiee
- RNA Networks Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Sophie Wood
- Genetic Modification Service, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Jessica Olsen
- Genetic Modification Service, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Robin Lovell-Badge
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| |
Collapse
|
4
|
Lumahan LEV, Arif M, Whitener AE, Yi P. Regulating Androgen Receptor Function in Prostate Cancer: Exploring the Diversity of Post-Translational Modifications. Cells 2024; 13:191. [PMID: 38275816 PMCID: PMC10814774 DOI: 10.3390/cells13020191] [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: 12/01/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/27/2024] Open
Abstract
Androgen receptor (AR) transcriptional activity significantly influences prostate cancer (PCa) progression. In addition to ligand stimulation, AR transcriptional activity is also influenced by a variety of post-translational modifications (PTMs). A number of oncogenes and tumor suppressors have been observed leveraging PTMs to influence AR activity. Subjectively targeting these post-translational modifiers based on their impact on PCa cell proliferation is a rapidly developing area of research. This review elucidates the modifiers, contextualizes the effects of these PTMs on AR activity, and connects these cellular interactions to the progression of PCa.
Collapse
Affiliation(s)
- Lance Edward V. Lumahan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77204, USA
| | - Mazia Arif
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77205, USA
| | - Amy E. Whitener
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77205, USA
| | - Ping Yi
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77205, USA
| |
Collapse
|
5
|
Ren J, Yu P, Liu S, Li R, Niu X, Chen Y, Zhang Z, Zhou F, Zhang L. Deubiquitylating Enzymes in Cancer and Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303807. [PMID: 37888853 PMCID: PMC10754134 DOI: 10.1002/advs.202303807] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/30/2023] [Indexed: 10/28/2023]
Abstract
Deubiquitylating enzymes (DUBs) maintain relative homeostasis of the cellular ubiquitome by removing the post-translational modification ubiquitin moiety from substrates. Numerous DUBs have been demonstrated specificity for cleaving a certain type of ubiquitin linkage or positions within ubiquitin chains. Moreover, several DUBs perform functions through specific protein-protein interactions in a catalytically independent manner, which further expands the versatility and complexity of DUBs' functions. Dysregulation of DUBs disrupts the dynamic equilibrium of ubiquitome and causes various diseases, especially cancer and immune disorders. This review summarizes the Janus-faced roles of DUBs in cancer including proteasomal degradation, DNA repair, apoptosis, and tumor metastasis, as well as in immunity involving innate immune receptor signaling and inflammatory and autoimmune disorders. The prospects and challenges for the clinical development of DUB inhibitors are further discussed. The review provides a comprehensive understanding of the multi-faced roles of DUBs in cancer and immunity.
Collapse
Affiliation(s)
- Jiang Ren
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033P. R. China
| | - Peng Yu
- Zhongshan Institute for Drug DiscoveryShanghai Institute of Materia MedicaChinese Academy of SciencesZhongshanGuangdongP. R. China
| | - Sijia Liu
- International Biomed‐X Research CenterSecond Affiliated Hospital of Zhejiang University School of MedicineZhejiang UniversityHangzhouP. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhou310058China
| | - Ran Li
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033P. R. China
| | - Xin Niu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
| | - Yan Chen
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033P. R. China
| | - Zhenyu Zhang
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450003P. R. China
| | - Fangfang Zhou
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Long Zhang
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033P. R. China
- International Biomed‐X Research CenterSecond Affiliated Hospital of Zhejiang University School of MedicineZhejiang UniversityHangzhouP. R. China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058P. R. China
| |
Collapse
|
6
|
Saha G, Roy S, Basu M, Ghosh MK. USP7 - a crucial regulator of cancer hallmarks. Biochim Biophys Acta Rev Cancer 2023; 1878:188903. [PMID: 37127084 DOI: 10.1016/j.bbcan.2023.188903] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 04/28/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023]
Abstract
Over the course of three decades of study, the deubiquitinase Herpesvirus associated Ubiquitin-Specific Protease/Ubiquitin-Specific Protease 7 (HAUSP/USP7) has gradually come to be recognized as a crucially important molecule in cellular physiology. The fact that USP7 is overexpressed in a number of cancers, including breast, prostate, colorectal, and lung cancers, supports the idea that USP7 is also an important regulator of tumorigenesis. In this review, we discuss USP7's function in relation to the cancer hallmarks described by Hanahan and Weinberg. This post-translational modifier can support increased proliferation, block unfavorable growth signals, stop cell death, and support an unstable cellular genome by manipulating key players in the pertinent signalling circuit. It is interesting to note that USP7 also aids in the stabilization of molecules that support angiogenesis and metastasis. Targeting USP7 has now emerged as a crucial component of USP7 research because pharmacological inhibition of USP7 supports p53-mediated cell cycle arrest and apoptosis. Efficacious USP7 inhibition is currently being investigated in both synthetic and natural compounds, but issues with selectivity and a lack of co-crystal structure have hindered USP7 inhibition from being tested in clinical settings. Moreover, the development of new, more effective USP7 inhibitors and their encouraging implications by numerous groups give us a glimmer of hope for USP7-targeting medications as effective substitutes for hazardous cancer chemotherapeutics.
Collapse
Affiliation(s)
- Gouranga Saha
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata-700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, PIN - 700032, India
| | - Srija Roy
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata-700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, PIN - 700032, India
| | - Malini Basu
- Department of Microbiology, Dhruba Chand Halder College, University of Calcutta, Kolkata, PIN - 743372, India
| | - Mrinal K Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata-700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, PIN - 700032, India.
| |
Collapse
|
7
|
Guo Y, Cui S, Chen Y, Guo S, Chen D. Ubiquitin specific peptidases and prostate cancer. PeerJ 2023; 11:e14799. [PMID: 36811009 PMCID: PMC9939025 DOI: 10.7717/peerj.14799] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/04/2023] [Indexed: 02/18/2023] Open
Abstract
Protein ubiquitination is an important post-translational modification mechanism, which regulates protein stability and activity. The ubiquitination of proteins can be reversed by deubiquitinating enzymes (DUBs). Ubiquitin-specific proteases (USPs), the largest DUB subfamily, can regulate cellular functions by removing ubiquitin(s) from the target proteins. Prostate cancer (PCa) is the second leading type of cancer and the most common cause of cancer-related deaths in men worldwide. Numerous studies have demonstrated that the development of PCa is highly correlated with USPs. The expression of USPs is either high or low in PCa cells, thereby regulating the downstream signaling pathways and causing the development or suppression of PCa. This review summarized the functional roles of USPs in the development PCa and explored their potential applications as therapeutic targets for PCa.
Collapse
Affiliation(s)
- Yunfei Guo
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Shuaishuai Cui
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Yuanyuan Chen
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Song Guo
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Dahu Chen
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| |
Collapse
|
8
|
Sengupta M, Pluciennik A, Merry DE. The role of ubiquitination in spinal and bulbar muscular atrophy. Front Mol Neurosci 2022; 15:1020143. [PMID: 36277484 PMCID: PMC9583669 DOI: 10.3389/fnmol.2022.1020143] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA) is a neurodegenerative and neuromuscular genetic disease caused by the expansion of a polyglutamine-encoding CAG tract in the androgen receptor (AR) gene. The AR is an important transcriptional regulator of the nuclear hormone receptor superfamily; its levels are regulated in many ways including by ubiquitin-dependent degradation. Ubiquitination is a post-translational modification (PTM) which plays a key role in both AR transcriptional activity and its degradation. Moreover, the ubiquitin-proteasome system (UPS) is a fundamental component of cellular functioning and has been implicated in diseases of protein misfolding and aggregation, including polyglutamine (polyQ) repeat expansion diseases such as Huntington's disease and SBMA. In this review, we discuss the details of the UPS system, its functions and regulation, and the role of AR ubiquitination and UPS components in SBMA. We also discuss aspects of the UPS that may be manipulated for therapeutic effect in SBMA.
Collapse
Affiliation(s)
| | | | - Diane E. Merry
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
9
|
Valles G, Pozhidaeva A, Korzhnev DM, Bezsonova I. Backbone and ILV side-chain NMR resonance assignments of the catalytic domain of human deubiquitinating enzyme USP7. BIOMOLECULAR NMR ASSIGNMENTS 2022; 16:197-203. [PMID: 35536398 PMCID: PMC9529858 DOI: 10.1007/s12104-022-10079-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/16/2022] [Indexed: 06/14/2023]
Abstract
Ubiquitin specific protease 7 (USP7) is a deubiquitinating enzyme, which removes ubiquitin tag from numerous protein substrates involved in diverse cellular processes such as cell cycle regulation, apoptosis and DNA damage response. USP7 affects stability, interaction network and cellular localization of its cellular and viral substrates by controlling their ubiquitination status. The large 41 kDa catalytic domain of USP7 harbors the active site of the enzyme. Here we present a nearly complete (93%) NMR resonance assignment of isoleucine, leucine and valine (ILV) side-chains of the USP7 catalytic domain along with a refined nearly complete (93%) assignment of its backbone resonances. The reported ILV methyl group assignment will facilitate further NMR investigations of structure, interactions and conformational dynamics of the USP7 enzyme.
Collapse
Affiliation(s)
- Gabrielle Valles
- Department of Molecular Biology and Biophysics, UConn Health, 263 Farmington Avenue, Farmington, CT, 06032, USA
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
| | - Alexandra Pozhidaeva
- Department of Molecular Biology and Biophysics, UConn Health, 263 Farmington Avenue, Farmington, CT, 06032, USA
| | - Dmitry M Korzhnev
- Department of Molecular Biology and Biophysics, UConn Health, 263 Farmington Avenue, Farmington, CT, 06032, USA.
| | - Irina Bezsonova
- Department of Molecular Biology and Biophysics, UConn Health, 263 Farmington Avenue, Farmington, CT, 06032, USA.
| |
Collapse
|
10
|
LIU J, LEUNG CT, LIANG L, WANG Y, CHEN J, LAI KP, TSE WKF. Deubiquitinases in Cancers: Aspects of Proliferation, Metastasis, and Apoptosis. Cancers (Basel) 2022; 14:cancers14143547. [PMID: 35884607 PMCID: PMC9323628 DOI: 10.3390/cancers14143547] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 12/24/2022] Open
Abstract
Simple Summary This review summarizes the current DUBs findings that correlate with the most common cancers in the world (liver, breast, prostate, colorectal, pancreatic, and lung cancers). The DUBs were further classified by their biological functions in terms of proliferation, metastasis, and apoptosis. The work provides an updated of the current findings, and could be used as a quick guide for researchers to identify target DUBs in cancers. Abstract Deubiquitinases (DUBs) deconjugate ubiquitin (UBQ) from ubiquitylated substrates to regulate its activity and stability. They are involved in several cellular functions. In addition to the general biological regulation of normal cells, studies have demonstrated their critical roles in various cancers. In this review, we evaluated and grouped the biological roles of DUBs, including proliferation, metastasis, and apoptosis, in the most common cancers in the world (liver, breast, prostate, colorectal, pancreatic, and lung cancers). The current findings in these cancers are summarized, and the relevant mechanisms and relationship between DUBs and cancers are discussed. In addition to highlighting the importance of DUBs in cancer biology, this study also provides updated information on the roles of DUBs in different types of cancers.
Collapse
Affiliation(s)
- Jiaqi LIU
- Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, Guilin 541004, China; (J.L.); (L.L.); (Y.W.); (K.P.L.)
| | - Chi Tim LEUNG
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China;
| | - Luyun LIANG
- Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, Guilin 541004, China; (J.L.); (L.L.); (Y.W.); (K.P.L.)
| | - Yuqin WANG
- Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, Guilin 541004, China; (J.L.); (L.L.); (Y.W.); (K.P.L.)
| | - Jian CHEN
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541004, China
- Correspondence: (J.C.); (W.K.F.T.); Tel.: +86-773-5895860 (J.C.); +81-92-802-4767 (W.K.F.T.)
| | - Keng Po LAI
- Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, Guilin 541004, China; (J.L.); (L.L.); (Y.W.); (K.P.L.)
| | - William Ka Fai TSE
- Laboratory of Developmental Disorders and Toxicology, Center for Promotion of International Education and Research, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
- Correspondence: (J.C.); (W.K.F.T.); Tel.: +86-773-5895860 (J.C.); +81-92-802-4767 (W.K.F.T.)
| |
Collapse
|
11
|
Guo J, Zhao J, Sun L, Yang C. Role of ubiquitin specific proteases in the immune microenvironment of prostate cancer: A new direction. Front Oncol 2022; 12:955718. [PMID: 35924159 PMCID: PMC9339679 DOI: 10.3389/fonc.2022.955718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 06/28/2022] [Indexed: 11/23/2022] Open
Abstract
Regulation of ubiquitination is associated with multiple processes of tumorigenesis and development, including regulation of the tumor immune microenvironment. Deubiquitinating enzymes (DUBs) can remove ubiquitin chains from substrates, thereby stabilizing target proteins and altering and remodeling biological processes. During tumorigenesis, deubiquitination-altered biological processes are closely related to tumor metabolism, stemness, and the immune microenvironment. Recently, tumor microenvironment (TME) modulation strategies have attracted considerable attention in cancer immunotherapy. Targeting immunosuppressive mechanisms in the TME has revolutionized cancer therapy. Prostate cancer (PC) is one of the most common cancers and the second most common cause of cancer-related death in men worldwide. While immune checkpoint inhibition has produced meaningful therapeutic effects in many cancer types, clinical trials of anti-CTLA4 or anti-PD1 have not shown a clear advantage in PC patients. TME affects PC progression and also enables tumor cell immune evasion by activating the PD-1/PD-L1 axis. Over the past few decades, an increasing number of studies have demonstrated that deubiquitination in PC immune microenvironment may modulate the host immune system’s response to the tumor. As the largest and most diverse group of DUBs, ubiquitin-specific proteases (USPs) play an important role in regulating T cell development and function. According to current studies, USPs exhibit a high expression signature in PC and may promote tumorigenesis. Elevated expression of USPs often indicates poor tumor prognosis, suggesting that USPs are expected to develop as the markers of tumor prognosis and even potential drug targets for anti-tumor therapy. Herein, we first summarized recent advances of USPs in PC and focused on the relationship between USPs and immunity. Additionally, we clarified the resistance mechanisms of USPs to targeted drugs in PC. Finally, we reviewed the major achievement of targeting USPs in cancers.
Collapse
Affiliation(s)
- Jinhui Guo
- Cancer Center, Institute of clinical medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Jie Zhao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Litao Sun
- Cancer Center, Department of Ultrasound, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Litao Sun, ; Chen Yang,
| | - Chen Yang
- Cancer Center, Department of Ultrasound, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Litao Sun, ; Chen Yang,
| |
Collapse
|
12
|
Dahiya UR, Heemers HV. Analyzing the Androgen Receptor Interactome in Prostate Cancer: Implications for Therapeutic Intervention. Cells 2022; 11:936. [PMID: 35326387 PMCID: PMC8946651 DOI: 10.3390/cells11060936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 12/29/2022] Open
Abstract
The androgen receptor (AR) is a member of the ligand-activated nuclear receptor family of transcription factors. AR's transactivation activity is turned on by the binding of androgens, the male sex steroid hormones. AR is critical for the development and maintenance of the male phenotype but has been recognized to also play an important role in human diseases. Most notably, AR is a major driver of prostate cancer (CaP) progression, which remains the second leading cause of cancer deaths in American men. Androgen deprivation therapies (ADTs) that interfere with interactions between AR and its activating androgen ligands have been the mainstay for treatment of metastatic CaP. Although ADTs are effective and induce remissions, eventually they fail, while the growth of the majority of ADT-resistant CaPs remains under AR's control. Alternative approaches to inhibit AR activity and bypass resistance to ADT are being sought, such as preventing the interaction between AR and its cofactors and coregulators that is needed to execute AR-dependent transcription. For such strategies to be efficient, the 3D conformation of AR complexes needs to be well-understood and AR-regulator interaction sites resolved. Here, we review current insights into these 3D structures and the protein interaction sites in AR transcriptional complexes. We focus on methods and technological approaches used to identify AR interactors and discuss challenges and limitations that need to be overcome for efficient therapeutic AR complex disruption.
Collapse
Affiliation(s)
| | - Hannelore V. Heemers
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, NB-40, 9500 Euclid Avenue, Cleveland, OH 44195, USA;
| |
Collapse
|
13
|
Li J, Xiao X, Wang H, Wang W, Ou Y, Wang Z, Jiang H, Liu Y, Zhang Z, Yang S. CDK4/6-USP51 axis regulates lung adenocarcinoma metastasis through ZEB1. Cancer Gene Ther 2022; 29:1181-1192. [DOI: 10.1038/s41417-021-00420-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/19/2021] [Accepted: 12/15/2021] [Indexed: 12/23/2022]
|
14
|
Zhou L, Ouyang T, Li M, Hong T, Mhs A, Meng W, Zhang N. Ubiquitin-Specific Peptidase 7: A Novel Deubiquitinase That Regulates Protein Homeostasis and Cancers. Front Oncol 2021; 11:784672. [PMID: 34869041 PMCID: PMC8640129 DOI: 10.3389/fonc.2021.784672] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/26/2021] [Indexed: 12/23/2022] Open
Abstract
Ubiquitin-Specific Peptidase 7 (USP7), or herpes virus-associated protease (HAUSP), is the largest family of the deubiquitinating enzymes (DUBs). Recent studies have shown that USP7 plays a vital role in regulating various physiological and pathological processes. Dysregulation of these processes mediated by USP7 may contribute to many diseases, such as cancers. Moreover, USP7 with aberrant expression levels and abnormal activity are found in cancers. Therefore, given the association between USP7 and cancers, targeting USP7 could be considered as an attractive and potential therapeutic approach in cancer treatment. This review describes the functions of USP7 and the regulatory mechanisms of its expression and activity, aiming to emphasize the necessity of research on USP7, and provide a better understanding of USP7-related biological processes and cancer.
Collapse
Affiliation(s)
- Lin Zhou
- First Clinical Medical College, Nanchang University, Nanchang, China
| | - Taohui Ouyang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Meihua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tao Hong
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Alriashy Mhs
- Department of Neurosurgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Wei Meng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Na Zhang
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
15
|
Chen YX, Tan LM, Gong JP, Huang MS, Yin JY, Zhang W, Zhou HH, Liu ZQ. Response prediction biomarkers and drug combinations of PARP inhibitors in prostate cancer. Acta Pharmacol Sin 2021; 42:1970-1980. [PMID: 33589795 PMCID: PMC8632930 DOI: 10.1038/s41401-020-00604-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 12/20/2020] [Indexed: 01/31/2023]
Abstract
PARP inhibitors are a group of inhibitors targeting poly(ADP-ribose) polymerases (PARP1 or PARP2) involved in DNA repair and transcriptional regulation, which may induce synthetic lethality in BRCAness tumors. Systematic analyzes of genomic sequencing in prostate cancer show that ~10%-19% of patients with primary prostate cancer have inactivated DNA repair genes, with a notably higher proportion of 23%-27% in patients with metastatic castration-resistant prostate cancer (mCRPC). These characteristic genomic alterations confer possible vulnerability to PARP inhibitors in patients with mCRPC who benefit only modestly from other therapies. However, only a small proportion of patients with mCRPC shows sensitivity to PARP inhibitors, and these sensitive patients cannot be fully identified by existing response prediction biomarkers. In this review, we provide an overview of the potential response prediction biomarkers and synergistic combinations studied in the preclinical and clinical stages, which may expand the population of patients with prostate cancer who may benefit from PARP inhibitors.
Collapse
Affiliation(s)
- Yi-Xin Chen
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, China
| | - Li-Ming Tan
- Department of Pharmacy, The Second People's Hospital of Huaihua City, Huaihua, 418000, China
| | - Jian-Ping Gong
- Department of Pharmacy, The Second People's Hospital of Huaihua City, Huaihua, 418000, China
| | - Ma-Sha Huang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, China.
| |
Collapse
|
16
|
Vélot L, Lessard F, Bérubé-Simard FA, Tav C, Neveu B, Teyssier V, Boudaoud I, Dionne U, Lavoie N, Bilodeau S, Pouliot F, Bisson N. Proximity-dependent Mapping of the Androgen Receptor Identifies Kruppel-like Factor 4 as a Functional Partner. Mol Cell Proteomics 2021; 20:100064. [PMID: 33640491 PMCID: PMC8050775 DOI: 10.1016/j.mcpro.2021.100064] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer (PCa) is the most frequently diagnosed cancer in men and the third cause of cancer mortality. PCa initiation and growth are driven by the androgen receptor (AR). The AR is activated by androgens such as testosterone and controls prostatic cell proliferation and survival. Here, we report an AR signaling network generated using BioID proximity labeling proteomics in androgen-dependent LAPC4 cells. We identified 31 AR-associated proteins in nonstimulated cells. Strikingly, the AR signaling network increased to 182 and 200 proteins, upon 24 h or 72 h of androgenic stimulation, respectively, for a total of 267 nonredundant AR-associated candidates. Among the latter group, we identified 213 proteins that were not previously reported in databases. Many of these new AR-associated proteins are involved in DNA metabolism, RNA processing, and RNA polymerase II transcription. Moreover, we identified 44 transcription factors, including the Kru¨ppel-like factor 4 (KLF4), which were found interacting in androgen-stimulated cells. Interestingly, KLF4 repressed the well-characterized AR-dependent transcription of the KLK3 (PSA) gene; AR and KLF4 also colocalized genome-wide. Taken together, our data report an expanded high-confidence proximity network for AR, which will be instrumental to further dissect the molecular mechanisms underlying androgen signaling in PCa cells. BioID proteomics identifies 267 androgen receptor (AR)-associated candidates Krüppel-like factor 4 (KLF4) is a new AR interaction partner AR and KLF4 colocalize genome-wide on >4000 genes, including KLK3 (PSA) KLF4 acts as a repressor for the AR target gene KLK3 (PSA)
Collapse
Affiliation(s)
- Lauriane Vélot
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, Quebec, Canada; PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, Quebec, Canada
| | - Frédéric Lessard
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, Quebec, Canada; PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, Quebec, Canada
| | - Félix-Antoine Bérubé-Simard
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, Quebec, Canada
| | - Christophe Tav
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, Quebec, Canada; Centre de recherche en données massives de l'Université Laval, Québec, Québec, Canada
| | - Bertrand Neveu
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, Quebec, Canada
| | - Valentine Teyssier
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, Quebec, Canada; PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, Quebec, Canada
| | - Imène Boudaoud
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, Quebec, Canada
| | - Ugo Dionne
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, Quebec, Canada; PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, Quebec, Canada
| | - Noémie Lavoie
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, Quebec, Canada; PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, Quebec, Canada
| | - Steve Bilodeau
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, Quebec, Canada; Centre de recherche en données massives de l'Université Laval, Québec, Québec, Canada; Department of Molecular Biology, Medical Biochemistry and Pathology, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
| | - Frédéric Pouliot
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, Quebec, Canada; Department of Surgery, Faculté de Médecine, Université Laval, Québec, Quebec, Canada.
| | - Nicolas Bisson
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, Quebec, Canada; PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, Quebec, Canada; Department of Molecular Biology, Medical Biochemistry and Pathology, Faculté de Médecine, Université Laval, Québec, Quebec, Canada.
| |
Collapse
|
17
|
Liu Z, Liu C, Yan K, Liu J, Fang Z, Fan Y. Huaier Extract Inhibits Prostate Cancer Growth via Targeting AR/AR-V7 Pathway. Front Oncol 2021; 11:615568. [PMID: 33708629 PMCID: PMC7940541 DOI: 10.3389/fonc.2021.615568] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/07/2021] [Indexed: 01/05/2023] Open
Abstract
The androgen receptor (AR) plays a pivotal role in prostatic carcinogenesis, and it also affects the transition from hormone sensitive prostate cancer (HSPC) to castration-resistant prostate cancer (CRPC). Particularly, the persistent activation of the androgen receptor and the appearance of androgen receptor splicing variant 7 (AR-V7), could partly explain the failure of androgen deprivation therapy (ADT). In the present study, we reported that huaier extract, derived from officinal fungi, has potent antiproliferative effects in both HSPC and CRPC cells. Mechanistically, huaier extract downregulated both full length AR (AR-FL) and AR-V7 mRNA levels via targeting the SET and MYND domain-containing protein 3 (SMYD3) signaling pathway. Huaier extract also enhanced proteasome-mediated protein degradation of AR-FL and AR-V7 by downregulating proteasome-associated deubiquitinase ubiquitin-specific protease 14 (USP14). Furthermore, huaier extract inhibited AR-FL/AR-V7 transcriptional activity and their nuclear translocation. More importantly, our data demonstrated that huaier extract could re-sensitize enzalutamide-resistant prostate cancer cells to enzalutamide treatment in vitro and in vivo models. Our work revealed that huaier extract could be effective for treatment of prostate cancer either as monotherapy or in combination with enzalutamide.
Collapse
Affiliation(s)
- Zhengfang Liu
- Department of Urology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Cheng Liu
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Keqiang Yan
- Department of Urology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Jikai Liu
- Department of Urology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Zhiqing Fang
- Department of Urology, Qilu Hospital of Shandong University, Ji'nan, China.,Department of Medicine, Center for Molecular Medicine (CMM) and Bioclinicum, Karolinska Institute and Karolinska University Hospital Solna, Solna, Sweden
| | - Yidong Fan
- Department of Urology, Qilu Hospital of Shandong University, Ji'nan, China
| |
Collapse
|
18
|
Pandareesh MD, Kameshwar VH, Byrappa K. Prostate Carcinogenesis: Insights in Relation to Epigenetics and Inflammation. Endocr Metab Immune Disord Drug Targets 2021; 21:253-267. [PMID: 32682386 DOI: 10.2174/1871530320666200719020709] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/17/2020] [Accepted: 04/29/2020] [Indexed: 12/24/2022]
Abstract
Prostate cancer is a multifactorial disease that mainly occurs due to the accumulation of somatic, genetic, and epigenetic changes, resulting in the inactivation of tumor-suppressor genes and activation of oncogenes. Mutations in genes, specifically those that control cell growth and division or the repair of damaged DNA, make the cells grow and divide uncontrollably to form a tumor. The risk of developing prostate cancer depends upon the gene that has undergone the mutation. Identifying such genetic risk factors for prostate cancer poses a challenge for the researchers. Besides genetic mutations, many epigenetic alterations, including DNA methylation, histone modifications (methylation, acetylation, ubiquitylation, sumoylation, and phosphorylation) nucleosomal remodeling, and chromosomal looping, have significantly contributed to the onset of prostate cancer as well as the prognosis, diagnosis, and treatment of prostate cancer. Chronic inflammation also plays a major role in the onset and progression of human cancer, via modifications in the tumor microenvironment by initiating epithelialmesenchymal transition and remodeling the extracellular matrix. In this article, the authors present a brief history of the mechanisms and potential links between the genetic aberrations, epigenetic changes, inflammation, and inflammasomes that are known to contribute to the prognosis of prostate cancer. Furthermore, the authors examine and discuss the clinical potential of prostate carcinogenesis in relation to epigenetics and inflammation for its diagnosis and treatment..
Collapse
Affiliation(s)
- Mirazkar D Pandareesh
- Center for Research and Innovation, BGSIT Campus, Adichunchanagiri University, B.G. Nagara, Mandya District, Karnataka 571448, India
| | - Vivek H Kameshwar
- Center for Research and Innovation, BGSIT Campus, Adichunchanagiri University, B.G. Nagara, Mandya District, Karnataka 571448, India
| | - Kullaiah Byrappa
- Center for Research and Innovation, BGSIT Campus, Adichunchanagiri University, B.G. Nagara, Mandya District, Karnataka 571448, India
| |
Collapse
|
19
|
Ge J, Yu W, Li J, Ma H, Wang P, Zhou Y, Wang Y, Zhang J, Shi G. USP16 regulates castration-resistant prostate cancer cell proliferation by deubiquitinating and stablizing c-Myc. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:59. [PMID: 33546726 PMCID: PMC7866668 DOI: 10.1186/s13046-021-01843-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/13/2021] [Indexed: 12/14/2022]
Abstract
Background c-Myc, a well-established oncogene, plays an important role in the initiation and progression of various cancers, including prostate cancer. However, its mechanism in cancer cell remains largely unknown and whether there exist a deubiquitinase targeting c-Myc also remains elusive. Methods Bioinformatic analysis and shRNA screening methods were used to identify potential deubiquitinases that correlate with c-Myc gene signature. Cell proliferation and viability were measured by Cell-Counting-Kit 8 and colony formation assays. A mouse xenograft model of PC3 cells was established to confirm the function of USP16 in vivo. The interaction between USP16 and c-Myc protein was assessed by co-immunoprecipitation and protein co-localization assays. Immunohistochemistry staining was performed to detect the expression of USP16, Ki67, and c-Myc in xenograft tissues and clinical tumour tissues. Furthermore, the correlation between USP16 and c-Myc was confirmed by RNA sequencing. Results Functional analyses identified USP16, known as a deubiquitinase, was strongly correlated with the c-Myc gene signature. Depletion of USP16 was shown to significantly suppress the growth of PCa cells both in vitro and in vivo. Co-immunoprecipitation and ubiquitination assays confirmed that USP16 served as a novel deubiquitinase of c-Myc and overexpression of c-Myc significantly rescued the effects of USP16 disruption. Immunohistochemistry staining and RNA-seq tactics were further used to confirm the positive correlation between USP16 and c-Myc expression. Expression of USP16 in human PCa tissues was higher than that seen in normal prostate tissues and its high expression was found associated with poor prognosis. Conclusions USP16 serves as a novel deubiquitinase of c-Myc. Downregulation of USP16 markedly suppressed PCa cell growth both in vitro and in vivo. USP16 regulates PCa cell proliferation by deubiquitinating and stabilizing c-Myc, making it a potential therapeutic candidate for the treatment of PCa. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01843-8.
Collapse
Affiliation(s)
- Jianchao Ge
- Department of Urology, The Fifth People's Hospital of Shanghai, Fudan University, No. 801, Heqing Road,Minhang District, Shanghai, 200240, People's Republic of China
| | - Wandong Yu
- Department of Urology, The Fifth People's Hospital of Shanghai, Fudan University, No. 801, Heqing Road,Minhang District, Shanghai, 200240, People's Republic of China
| | - Junhong Li
- Department of Urology, The Fifth People's Hospital of Shanghai, Fudan University, No. 801, Heqing Road,Minhang District, Shanghai, 200240, People's Republic of China
| | - Hangbin Ma
- Department of Urology, The Fifth People's Hospital of Shanghai, Fudan University, No. 801, Heqing Road,Minhang District, Shanghai, 200240, People's Republic of China
| | - Pengyu Wang
- Department of Urology, The Fifth People's Hospital of Shanghai, Fudan University, No. 801, Heqing Road,Minhang District, Shanghai, 200240, People's Republic of China
| | - Yinghao Zhou
- Department of Urology, The Fifth People's Hospital of Shanghai, Fudan University, No. 801, Heqing Road,Minhang District, Shanghai, 200240, People's Republic of China
| | - Yang Wang
- Department of Urology, The Fifth People's Hospital of Shanghai, Fudan University, No. 801, Heqing Road,Minhang District, Shanghai, 200240, People's Republic of China
| | - Jun Zhang
- Department of Urology, The Fifth People's Hospital of Shanghai, Fudan University, No. 801, Heqing Road,Minhang District, Shanghai, 200240, People's Republic of China.
| | - Guowei Shi
- Department of Urology, The Fifth People's Hospital of Shanghai, Fudan University, No. 801, Heqing Road,Minhang District, Shanghai, 200240, People's Republic of China.
| |
Collapse
|
20
|
Pluciennik A, Liu Y, Molotsky E, Marsh GB, Ranxhi B, Arnold FJ, St.-Cyr S, Davidson B, Pourshafie N, Lieberman AP, Gu W, Todi SV, Merry DE. Deubiquitinase USP7 contributes to the pathogenicity of spinal and bulbar muscular atrophy. J Clin Invest 2021; 131:134565. [PMID: 33170804 PMCID: PMC7773404 DOI: 10.1172/jci134565] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 10/29/2020] [Indexed: 12/24/2022] Open
Abstract
Polyglutamine (polyQ) diseases are devastating, slowly progressing neurodegenerative conditions caused by expansion of polyQ-encoding CAG repeats within the coding regions of distinct, unrelated genes. In spinal and bulbar muscular atrophy (SBMA), polyQ expansion within the androgen receptor (AR) causes progressive neuromuscular toxicity, the molecular basis of which is unclear. Using quantitative proteomics, we identified changes in the AR interactome caused by polyQ expansion. We found that the deubiquitinase USP7 preferentially interacts with polyQ-expanded AR and that lowering USP7 levels reduced mutant AR aggregation and cytotoxicity in cell models of SBMA. Moreover, USP7 knockdown suppressed disease phenotypes in SBMA and spinocerebellar ataxia type 3 (SCA3) fly models, and monoallelic knockout of Usp7 ameliorated several motor deficiencies in transgenic SBMA mice. USP7 overexpression resulted in reduced AR ubiquitination, indicating the direct action of USP7 on AR. Using quantitative proteomics, we identified the ubiquitinated lysine residues on mutant AR that are regulated by USP7. Finally, we found that USP7 also differentially interacts with mutant Huntingtin (HTT) protein in striatum and frontal cortex of a knockin mouse model of Huntington's disease. Taken together, our findings reveal a critical role for USP7 in the pathophysiology of SBMA and suggest a similar role in SCA3 and Huntington's disease.
Collapse
Affiliation(s)
- Anna Pluciennik
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Yuhong Liu
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Elana Molotsky
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Gregory B. Marsh
- Department of Pharmacology and Neurology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Bedri Ranxhi
- Department of Pharmacology and Neurology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Frederick J. Arnold
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Sophie St.-Cyr
- Department of Pathology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Beverly Davidson
- Department of Pathology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Naemeh Pourshafie
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
- George Washington University, Institute of Biomedical Sciences, Washington, DC, USA
| | - Andrew P. Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Wei Gu
- Department of Pathology and Cell Biology and Institute for Cancer Genetics, Columbia University, New York, New York, USA
| | - Sokol V. Todi
- Department of Pharmacology and Neurology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Diane E. Merry
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
Dwane L, O'Connor AE, Das S, Moran B, Mulrane L, Pinto-Fernandez A, Ward E, Blümel AM, Cavanagh BL, Mooney B, Dirac AM, Jirström K, Kessler BM, Ní Chonghaile T, Bernards R, Gallagher WM, O'Connor DP. A Functional Genomic Screen Identifies the Deubiquitinase USP11 as a Novel Transcriptional Regulator of ERα in Breast Cancer. Cancer Res 2020; 80:5076-5088. [PMID: 33004351 DOI: 10.1158/0008-5472.can-20-0214] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 06/26/2020] [Accepted: 09/15/2020] [Indexed: 01/23/2023]
Abstract
Approximately 70% of breast cancers express estrogen receptor α (ERα) and depend on this key transcriptional regulator for proliferation and differentiation. While patients with this disease can be treated with targeted antiendocrine agents, drug resistance remains a significant issue, with almost half of patients ultimately relapsing. Elucidating the mechanisms that control ERα function may further our understanding of breast carcinogenesis and reveal new therapeutic opportunities. Here, we investigated the role of deubiquitinases (DUB) in regulating ERα in breast cancer. An RNAi loss-of-function screen in breast cancer cells targeting all DUBs identified USP11 as a regulator of ERα transcriptional activity, which was further validated by assessment of direct transcriptional targets of ERα. USP11 expression was induced by estradiol, an effect that was blocked by tamoxifen and not observed in ERα-negative cells. Mass spectrometry revealed a significant change to the proteome and ubiquitinome in USP11-knockdown (KD) cells in the presence of estradiol. RNA sequencing in LCC1 USP11-KD cells revealed significant suppression of cell-cycle-associated and ERα target genes, phenotypes that were not observed in LCC9 USP11-KD, antiendocrine-resistant cells. In a breast cancer patient cohort coupled with in silico analysis of publicly available cohorts, high expression of USP11 was significantly associated with poor survival in ERα-positive (ERα+) patients. Overall, this study highlights a novel role for USP11 in the regulation of ERα activity, where USP11 may represent a prognostic marker in ERα+ breast cancer. SIGNIFICANCE: A newly identified role for USP11 in ERα transcriptional activity represents a novel mechanism of ERα regulation and a pathway to be exploited for the management of ER-positive breast cancer.
Collapse
Affiliation(s)
- Lisa Dwane
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons Ireland, Dublin, Ireland
| | - Aisling E O'Connor
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Sudipto Das
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons Ireland, Dublin, Ireland
| | - Bruce Moran
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Laoighse Mulrane
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Adan Pinto-Fernandez
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Headington, Oxford, United Kingdom
| | - Elspeth Ward
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons Ireland, Dublin, Ireland
| | - Anna M Blümel
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons Ireland, Dublin, Ireland
| | - Brenton L Cavanagh
- Cellular and Molecular Imaging Core, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Brian Mooney
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons Ireland, Dublin, Ireland
| | - Annette M Dirac
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Karin Jirström
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Benedikt M Kessler
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Headington, Oxford, United Kingdom
| | - Tríona Ní Chonghaile
- Department of Physiology and Medical Physics, Royal College of Surgeons Ireland, Dublin, Ireland
| | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - William M Gallagher
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Darran P O'Connor
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons Ireland, Dublin, Ireland.
| |
Collapse
|
22
|
Lee JE, Park CM, Kim JH. USP7 deubiquitinates and stabilizes EZH2 in prostate cancer cells. Genet Mol Biol 2020; 43:e20190338. [PMID: 32453339 PMCID: PMC7252518 DOI: 10.1590/1678-4685-gmb-2019-0338] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/11/2020] [Indexed: 12/20/2022] Open
Abstract
Regulation of target proteins by the ubiquitin-proteasome system (UPS) is common in a wide range of cellular events, including transcriptional regulation, cell cycle progression, differentiation, and tumorigenesis. Ubiquitin-specific protease 7 (USP7) has been implicated in tumor development and metastasis in various malignancies through the regulation of target protein stability. In this study, we found that the enhancer of zeste homolog 2 (EZH2), which catalyzes the methylation at lysine 27 of histone H3, is a target of USP7 and is stabilized by USP7-mediated deubiquitination. In prostate cancer cells, the transcriptional repression function of EZH2 was inhibited by USP7-knockdown. Furthermore, ectopic introduction of EZH2 restored the cell migration, invasion, and sphere-forming potential of prostate cancer cells, which had been decreased by USP7-knockdown. Moreover, combined treatment with the USP7-specific inhibitor P5091 and EZH2 inhibitors, such as GSK126, EPZ6438, and DZNep, induced synergistic inhibitory effects on cell migration, invasion, and sphere-forming potential in prostate cancer cells. Collectively, our findings revealed that the promotion of the malignancy-associated characteristics of prostate cancer cells by USP7 was in part due to EZH2 stabilization. Thus, we suggest that simultaneous treatment with a USP7 inhibitor and an EZH2 inhibitor could be a rational strategy for treating EZH2-dependent cancers.
Collapse
Affiliation(s)
- Jae Eun Lee
- Inha University, Department of Biological Sciences, Incheon 22212,
South Korea
| | - Chan Mi Park
- Inha University, Department of Biological Sciences, Incheon 22212,
South Korea
| | - Jung Hwa Kim
- Inha University, Department of Biological Sciences, Incheon 22212,
South Korea
| |
Collapse
|
23
|
Preclinical Study Using ABT263 to Increase Enzalutamide Sensitivity to Suppress Prostate Cancer Progression Via Targeting BCL2/ROS/USP26 Axis Through Altering ARv7 Protein Degradation. Cancers (Basel) 2020; 12:cancers12040831. [PMID: 32235588 PMCID: PMC7226306 DOI: 10.3390/cancers12040831] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/15/2020] [Accepted: 03/23/2020] [Indexed: 12/22/2022] Open
Abstract
Background: The recently developed antiandrogen, Enzalutamide (Enz), has reformed the standard of care for castration resistant prostate cancer (CRPC) patients. However, Enz-resistance inevitably emerges despite success of Enz in prolonging CRPC patients’ survival. Here we found that Enz-resistant prostate cancer (PCa) cells had higher BCL2 expression. We aimed to test whether targeting BCL2 would influence Enz sensitivity of prostate cancer (PCa) and identify the potential mechanism. Methods: The study was designed to target Enz-induced BCL2 with inhibitor ABT263 and test Enz sensitivity in Enz-resistant PCa cells by MTT assay. Cellular reactive oxygen species (ROS) levels were detected with dihydroethidium staining, and in vitro deubiquitinating enzyme activity assay was used to evaluate ubiquitin specific protease 26 (USP26) activity. Results: ABT263 could increase Enz sensitivity in both Enz-sensitive and Enz-resistant PCa cells via inducing ROS generation. Elevated cellular ROS levels might then inhibit USP26 activity to increase the ubiquitination of androgen receptor (AR) and AR splice variant 7 (ARv7) and their ubiquitin/proteasome-dependent degradation, which contributed to the increase of Enz sensitivity. In vivo mouse model also demonstrates that ABT263 will suppress the PCa progression. Conclusion: This study demonstrated that targeting Enz-induced BCL2 with inhibitor ABT263 could increase Enz sensitivity in both Enz-sensitive and Enz-resistant PCa cells through induction of cellular ROS levels and suppression of USP26 activity with a consequent increase of ubiquitin/proteasome-dependent degradation of AR and ARv7 protein expression.
Collapse
|
24
|
Ballar Kirmizibayrak P, Erbaykent-Tepedelen B, Gozen O, Erzurumlu Y. Divergent Modulation of Proteostasis in Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1233:117-151. [PMID: 32274755 DOI: 10.1007/978-3-030-38266-7_5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Proteostasis regulates key cellular processes such as cell proliferation, differentiation, transcription, and apoptosis. The mechanisms by which proteostasis is regulated are crucial and the deterioration of cellular proteostasis has been significantly associated with tumorigenesis since it specifically targets key oncoproteins and tumor suppressors. Prostate cancer (PCa) is the second most common cause of cancer death in men worldwide. Androgens mediate one of the most central signaling pathways in all stages of PCa via the androgen receptor (AR). In addition to their regulation by hormones, PCa cells are also known to be highly secretory and are particularly prone to ER stress as proper ER function is essential. Alterations in various complex signaling pathways and cellular processes including cell cycle control, transcription, DNA repair, apoptosis, cell adhesion, epithelial-mesenchymal transition (EMT), and angiogenesis are critical factors influencing PCa development through key molecular changes mainly by posttranslational modifications in PCa-related proteins, including AR, NKX3.1, PTEN, p53, cyclin D1, and p27. Several ubiquitin ligases like MDM2, Siah2, RNF6, CHIP, and substrate-binding adaptor SPOP; deubiquitinases such as USP7, USP10, USP26, and USP12 are just some of the modifiers involved in the regulation of these key proteins via ubiquitin-proteasome system (UPS). Some ubiquitin-like modifiers, especially SUMOs, have been also closely associated with PCa. On the other hand, the proteotoxicity resulting from misfolded proteins and failure of ER adaptive capacity induce unfolded protein response (UPR) that is an indispensable signaling mechanism for PCa development. Lastly, ER-associated degradation (ERAD) also plays a crucial role in prostate tumorigenesis. In this section, the relationship between prostate cancer and proteostasis will be discussed in terms of UPS, UPR, SUMOylation, ERAD, and autophagy.
Collapse
Affiliation(s)
| | | | - Oguz Gozen
- Faculty of Medicine, Department of Physiology, Ege University, Izmir, Turkey
| | - Yalcin Erzurumlu
- Faculty of Pharmacy, Department of Biochemistry, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
25
|
Functional analysis of deubiquitylating enzymes in tumorigenesis and development. Biochim Biophys Acta Rev Cancer 2019; 1872:188312. [DOI: 10.1016/j.bbcan.2019.188312] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/16/2019] [Accepted: 08/16/2019] [Indexed: 02/06/2023]
|
26
|
Ducker C, Chow LKY, Saxton J, Handwerger J, McGregor A, Strahl T, Layfield R, Shaw PE. De-ubiquitination of ELK-1 by USP17 potentiates mitogenic gene expression and cell proliferation. Nucleic Acids Res 2019; 47:4495-4508. [PMID: 30854565 PMCID: PMC6511843 DOI: 10.1093/nar/gkz166] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 02/26/2019] [Accepted: 03/01/2019] [Indexed: 01/06/2023] Open
Abstract
ELK-1 is a transcription factor involved in ERK-induced cellular proliferation. Here, we show that its transcriptional activity is modulated by ubiquitination at lysine 35 (K35). The level of ubiquitinated ELK-1 rises in mitogen-deprived cells and falls upon mitogen stimulation or oncogene expression. Ectopic expression of USP17, a cell cycle-dependent deubiquitinase, decreases ELK-1 ubiquitination and up-regulates ELK-1 target-genes with a concomitant increase in cyclin D1 expression. In contrast, USP17 depletion attenuates ELK-1-dependent gene expression and slows cell proliferation. The reduced rate of proliferation upon USP17 depletion appears to be a direct effect of ELK-1 ubiquitination because it is rescued by an ELK-1(K35R) mutant refractory to ubiquitination. Overall, our results show that ubiquitination of ELK-1 at K35, and its reversal by USP17, are important mechanisms in the regulation of nuclear ERK signalling and cellular proliferation. Our findings will be relevant for tumours that exhibit elevated USP17 expression and suggest a new target for intervention.
Collapse
Affiliation(s)
- Charles Ducker
- Transcription and Molecular Signalling Laboratory, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Leo Kam Yuen Chow
- Transcription and Molecular Signalling Laboratory, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Janice Saxton
- Transcription and Molecular Signalling Laboratory, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Jürgen Handwerger
- Transcription and Molecular Signalling Laboratory, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Alexander McGregor
- Transcription and Molecular Signalling Laboratory, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Thomas Strahl
- Transcription and Molecular Signalling Laboratory, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Robert Layfield
- Transcription and Molecular Signalling Laboratory, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Peter E Shaw
- Transcription and Molecular Signalling Laboratory, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| |
Collapse
|
27
|
Deubiquitination and stabilization of estrogen receptor α by ubiquitin-specific protease 7 promotes breast tumorigenesis. Cancer Lett 2019; 465:118-128. [DOI: 10.1016/j.canlet.2019.09.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/03/2019] [Accepted: 09/06/2019] [Indexed: 01/19/2023]
|
28
|
Islam MT, Zhou X, Chen F, Khan MA, Fu J, Chen H. Targeting the signalling pathways regulated by deubiquitinases for prostate cancer therapeutics. Cell Biochem Funct 2019; 37:304-319. [PMID: 31062387 DOI: 10.1002/cbf.3401] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/06/2019] [Accepted: 03/14/2019] [Indexed: 12/22/2022]
Abstract
Prostate cancer (PCa) is the most common cancer diagnosed and the second most common cause of cancer-related death in men worldwide. The current androgen deprivation therapy for PCa cannot fully cure this disease. Moreover, androgen receptor gene amplification and mutation are associated with PCa to develop castration-resistant prostate cancer (CRPC). This review focuses on the deubiquitinases (DUBs) involved in PCa development and progression. For PCa development and progression, several cellular pathways are regulated by specific DUBs which are also highlighted in here. The ubiquitin-specific proteases (USPs), a family member of DUBs mostly involved in the regulation of cellular pathways for PCa development, and the ubiquitin C-terminal hydrolases (UCHs), another family member of DUBs, are responsible for PCa metastasis. Small molecular inhibitors against DUBs can inhibit or reduce the level of specific DUBs through the regulation of cellular pathway to treat this disease. Some small molecular inhibitors are already identified against some of the DUBs, but very few of them are clinically proved in PCa. So, to find out other DUBs involving in the regulation of PCa-related pathways and to develop more effective small molecule inhibitors with greater potency would be a great idea to target PCa cells for future therapeutics and drug development with or without the combination of other anticancer drugs. SIGNIFICANCE OF THE STUDY: This review is targeting DUB proteins which are responsible for PCa induction, proliferation, and metastasis by highlighting their signalling pathway so that the readers can get information about other mechanisms for PCa besides androgen receptor pathway and helps to find other oncogenic DUBs involving in these signalling pathways. This review also hopes to find other oncogenic DUBs involving in PCa-related signalling pathways or to find the DUBs that can regulate multiple oncogenic signalling pathways which might be a good target for PCa therapeutics. In addition, there are some small molecule inhibitors that can inhibit the oncogenic DUBs and thus able to control the oncogenic pathways which would be a novel strategy to treat CRPC by using DUB inhibitor combined with or without other anticancer drugs.
Collapse
Affiliation(s)
- Md Tariqul Islam
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, China
| | - Xi Zhou
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, China
| | - Fangzhi Chen
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Md Asaduzzaman Khan
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Hanchun Chen
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
29
|
Identification of Novel Biomarkers of Homologous Recombination Defect in DNA Repair to Predict Sensitivity of Prostate Cancer Cells to PARP-Inhibitors. Int J Mol Sci 2019; 20:ijms20123100. [PMID: 31242618 PMCID: PMC6627216 DOI: 10.3390/ijms20123100] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/07/2019] [Accepted: 06/20/2019] [Indexed: 12/19/2022] Open
Abstract
One of the most common malignancies in men is prostate cancer, for which androgen deprivation is the standard therapy. However, prostate cancer cells become insensitive to anti-androgen treatment and proceed to a castration-resistant state with limited therapeutic options. Therefore, besides the androgen deprivation approach, novel biomarkers are urgently required for specific targeting in this deadly disease. Recently, germline or somatic mutations in the homologous recombination (HR) DNA repair genes have been identified in at least 20–25% of metastatic castration-resistant prostate cancers (mCRPC). Defects in genes involved in HR DNA repair can sensitize cancer cells to poly(ADP-ribose) polymerase (PARP) inhibitors, a class of drugs already approved by the Food and Drug Administration (FDA) for breast and ovarian cancer carrying germline mutations in BRCA1/2 genes. For advanced prostate cancer carrying Breast cancer1/2 (BRCA1/2) or ataxia telengiectasia mutated (ATM) mutations, preclinical studies and clinical trials support the use of PARP-inhibitors, which received breakthrough therapy designation by the FDA. Based on these assumptions, several trials including DNA damage response and repair (DDR) targeting have been launched and are ongoing for prostate cancer. Here, we review the state-of-the-art potential biomarkers that could be predictive of cancer cell synthetic lethality with PARP inhibitors. The identification of key molecules that are affected in prostate cancer could be assayed in future clinical studies to better stratify prostate cancer patients who might benefit from target therapy.
Collapse
|
30
|
Xia X, Huang C, Liao Y, Liu Y, He J, Guo Z, Jiang L, Wang X, Liu J, Huang H. Inhibition of USP14 enhances the sensitivity of breast cancer to enzalutamide. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:220. [PMID: 31126320 PMCID: PMC6534920 DOI: 10.1186/s13046-019-1227-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/13/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Androgen receptor (AR) is expressed in approximately 70% of breast tumors. Recent studies increasingly support AR as a potential therapeutic target of AR-positive breast cancer. We have previously reported that deubiquitinase USP14 stabilizes AR proteins by deubiquitination and USP14 inhibition results in inhibition of cell growth and tumor progression in AR-positive prostate cancer and breast cancer. The current study aims to explore the anticancer effect of a treatment combining AR antagonist enzalutamide with USP14 inhibition on breast cancer cells. METHODS The combining effects of enzalutamide and USP14 inhibition on breast cancer cell proliferation and apoptosis and associated cell signaling were evaluated in vitro and in vivo. RESULTS USP14 inhibition via administration of IU1 or USP14-specific siRNA/shRNA enhanced cell growth inhibition and apoptosis induction by enzalutamide in breast cancer cell lines in vitro and in vivo. Additionally, the combination of enzalutamide with USP14 inhibition/knockdown induced significant downregulation of AR proteins and suppression of AR-related signaling pathways, including Wnt/β-catenin and PI3K/AKT pathways. Moreover, AKT inhibition via MK2206 increased the antiproliferative and proapoptotic effects of enzalutamide+IU1 combined treatment. CONCLUSION Collectively, our data suggest that USP14 inhibition in combination with enzalutamide represents a potentially new therapeutic strategy for breast cancer.
Collapse
Affiliation(s)
- Xiaohong Xia
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Chuyi Huang
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Yuning Liao
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Yuan Liu
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Jinchan He
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Zhiqiang Guo
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Lili Jiang
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Xuejun Wang
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD, 57069, USA
| | - Jinbao Liu
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China.
| | - Hongbiao Huang
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China.
| |
Collapse
|
31
|
Rawat R, Starczynowski DT, Ntziachristos P. Nuclear deubiquitination in the spotlight: the multifaceted nature of USP7 biology in disease. Curr Opin Cell Biol 2019; 58:85-94. [PMID: 30897496 DOI: 10.1016/j.ceb.2019.02.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 01/23/2019] [Accepted: 02/20/2019] [Indexed: 12/11/2022]
Abstract
Ubiquitination is a versatile and tightly regulated post-translational protein modification with many distinct outcomes affecting protein stability, localization, interactions, and activity. Ubiquitin chain linkages anchored on substrates can be further modified by additional post-translational modifications, including phosphorylation and SUMOylation. Deubiquitinases (DUBs) reverse these ubiquitin marks with matched levels of precision. Over hundred known DUBs regulate a wide variety of cellular events. In this review, we focus on ubiquitin-specific protease 7 (USP7, also known as herpesvirus-associated ubiquitin-specific protease, or HAUSP) as one of the best studied, disease-associated DUBs. By highlighting the functions of USP7, particularly in the nucleus, and the emergence of the newest generation of USP7 inhibitors, we illustrate the importance of individual DUBs in the nucleus, and the therapeutic prospects of DUB targeting in human disease.
Collapse
Affiliation(s)
- Radhika Rawat
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Chicago, IL 60611, USA
| | - Daniel T Starczynowski
- Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Cancer Biology, University of Cincinnati, Cincinnati, OH
| | - Panagiotis Ntziachristos
- Simpson Querrey Center for Epigenetics; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL.
| |
Collapse
|
32
|
Auranofin lethality to prostate cancer includes inhibition of proteasomal deubiquitinases and disrupted androgen receptor signaling. Eur J Pharmacol 2019; 846:1-11. [DOI: 10.1016/j.ejphar.2019.01.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 12/19/2018] [Accepted: 01/08/2019] [Indexed: 01/03/2023]
|
33
|
Jin Q, Martinez CA, Arcipowski KM, Zhu Y, Gutierrez-Diaz BT, Wang KK, Johnson MR, Volk AG, Wang F, Wu J, Grove C, Wang H, Sokirniy I, Thomas PM, Goo YA, Abshiru NA, Hijiya N, Peirs S, Vandamme N, Berx G, Goosens S, Marshall SA, Rendleman EJ, Takahashi YH, Wang L, Rawat R, Bartom ET, Collings CK, Van Vlierberghe P, Strikoudis A, Kelly S, Ueberheide B, Mantis C, Kandela I, Bourquin JP, Bornhauser B, Serafin V, Bresolin S, Paganin M, Accordi B, Basso G, Kelleher NL, Weinstock J, Kumar S, Crispino JD, Shilatifard A, Ntziachristos P. USP7 Cooperates with NOTCH1 to Drive the Oncogenic Transcriptional Program in T-Cell Leukemia. Clin Cancer Res 2019; 25:222-239. [PMID: 30224337 PMCID: PMC6320313 DOI: 10.1158/1078-0432.ccr-18-1740] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/18/2018] [Accepted: 09/11/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive disease, affecting children and adults. Chemotherapy treatments show high response rates but have debilitating effects and carry risk of relapse. Previous work implicated NOTCH1 and other oncogenes. However, direct inhibition of these pathways affects healthy tissues and cancer alike. Our goal in this work has been to identify enzymes active in T-ALL whose activity could be targeted for therapeutic purposes. EXPERIMENTAL DESIGN To identify and characterize new NOTCH1 druggable partners in T-ALL, we coupled studies of the NOTCH1 interactome to expression analysis and a series of functional analyses in cell lines, patient samples, and xenograft models. RESULTS We demonstrate that ubiquitin-specific protease 7 (USP7) interacts with NOTCH1 and controls leukemia growth by stabilizing the levels of NOTCH1 and JMJD3 histone demethylase. USP7 is highly expressed in T-ALL and is transcriptionally regulated by NOTCH1. In turn, USP7 controls NOTCH1 levels through deubiquitination. USP7 binds oncogenic targets and controls gene expression through stabilization of NOTCH1 and JMJD3 and ultimately H3K27me3 changes. We also show that USP7 and NOTCH1 bind T-ALL superenhancers, and inhibition of USP7 leads to a decrease of the transcriptional levels of NOTCH1 targets and significantly blocks T-ALL cell growth in vitro and in vivo. CONCLUSIONS These results provide a new model for USP7 deubiquitinase activity through recruitment to oncogenic chromatin loci and regulation of both oncogenic transcription factors and chromatin marks to promote leukemia. Our studies also show that targeting USP7 inhibition could be a therapeutic strategy in aggressive leukemia.
Collapse
Affiliation(s)
- Qi Jin
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Carlos A Martinez
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Kelly M Arcipowski
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Yixing Zhu
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Blanca T Gutierrez-Diaz
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Kenneth K Wang
- Master of Science in Biotechnology Graduate Program, Northwestern University, Evanston, Illinois
| | - Megan R Johnson
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Andrew G Volk
- Division of Hematology/Oncology, Department of Medicine, Northwestern University, Chicago, Illinois
| | - Feng Wang
- Progenra Inc., Malvern, Pennsylvania
| | - Jian Wu
- Progenra Inc., Malvern, Pennsylvania
| | | | - Hui Wang
- Progenra Inc., Malvern, Pennsylvania
| | | | - Paul M Thomas
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois
| | - Young Ah Goo
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois
| | - Nebiyu A Abshiru
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois
| | - Nobuko Hijiya
- Ann & Robert H. Lurie Children's Hospital, Northwestern University, Chicago, Illinois
| | - Sofie Peirs
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Niels Vandamme
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Molecular Cellular Oncology Lab, Department for Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Geert Berx
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Molecular Cellular Oncology Lab, Department for Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Steven Goosens
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Molecular Cellular Oncology Lab, Department for Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Stacy A Marshall
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Emily J Rendleman
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Yoh-Hei Takahashi
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Lu Wang
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Radhika Rawat
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Elizabeth T Bartom
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Clayton K Collings
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Pieter Van Vlierberghe
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | | | - Stephen Kelly
- Department of Pathology, New York University, New York, New York
| | - Beatrix Ueberheide
- Department of Biochemistry and Molecular Pharmacology, New York University, New York, New York
| | - Christine Mantis
- Center for Developmental Therapeutics, Northwestern University, Evanston, Illinois
| | - Irawati Kandela
- Center for Developmental Therapeutics, Northwestern University, Evanston, Illinois
| | - Jean-Pierre Bourquin
- University Children's Hospital, Division of Pediatric Oncology, University of Zurich, Switzerland
| | - Beat Bornhauser
- University Children's Hospital, Division of Pediatric Oncology, University of Zurich, Switzerland
| | - Valentina Serafin
- Oncohematology Laboratory, Department of Woman's and Child's Health, University of Padova, Padova, Italy
| | - Silvia Bresolin
- Oncohematology Laboratory, Department of Woman's and Child's Health, University of Padova, Padova, Italy
| | - Maddalena Paganin
- Oncohematology Laboratory, Department of Woman's and Child's Health, University of Padova, Padova, Italy
| | - Benedetta Accordi
- Oncohematology Laboratory, Department of Woman's and Child's Health, University of Padova, Padova, Italy
| | - Giuseppe Basso
- Oncohematology Laboratory, Department of Woman's and Child's Health, University of Padova, Padova, Italy
| | - Neil L Kelleher
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois
- Department of Chemistry, Northwestern University, Chicago, Illinois
| | | | | | - John D Crispino
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
- Division of Hematology/Oncology, Department of Medicine, Northwestern University, Chicago, Illinois
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| | - Ali Shilatifard
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| | - Panagiotis Ntziachristos
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois.
- Division of Hematology/Oncology, Department of Medicine, Northwestern University, Chicago, Illinois
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| |
Collapse
|
34
|
Wang S, Ekoue DN, Raj GV, Kittler R. Targeting the turnover of oncoproteins as a new avenue for therapeutics development in castration-resistant prostate cancer. Cancer Lett 2018; 438:86-96. [PMID: 30217566 PMCID: PMC6186492 DOI: 10.1016/j.canlet.2018.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/23/2018] [Accepted: 09/03/2018] [Indexed: 12/19/2022]
Abstract
The current therapeutic armamentarium for castration-resistant prostate cancer (CRPC) includes second-generation agents such as the Androgen Receptor (AR) inhibitor enzalutamide and the androgen synthesis inhibitor abiraterone acetate, immunotherapies like sipuleucel-T, chemotherapies including docetaxel and cabazitaxel and the radiopharmaceutical radium 223 dichloride. However, relapse of CRPC resistant to these therapeutic modalities occur rapidly. The mechanisms of resistance to these treatments are complex, including specific mutations or alternative splicing of oncogenic proteins. An alternative approach to treating CRPC may be to target the turnover of these molecular drivers of CRPC. In this review, the mechanisms by which protein stability of several oncoproteins such as AR, ERG, GR, CYP17A1 and MYC, will be discussed, as well as how these findings could be translated into novel therapeutic agents.
Collapse
Affiliation(s)
- Shan Wang
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Dede N Ekoue
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ganesh V Raj
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ralf Kittler
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA; Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
35
|
Inhibiting Multiple Deubiquitinases to Reduce Androgen Receptor Expression in Prostate Cancer Cells. Sci Rep 2018; 8:13146. [PMID: 30177856 PMCID: PMC6120934 DOI: 10.1038/s41598-018-31567-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 08/20/2018] [Indexed: 01/27/2023] Open
Abstract
Prostate cancer (PCa), a leading cause of cancer-related death in men, becomes resistant to androgen deprivation therapy by inducing androgen receptor (AR) activity, which is known as castration-resistant PCa (CRPC). Enzalutamide is an approved drug that inhibits AR activity and increases overall survival. However, resistance to enzalutamide develops rapidly often by increasing AR activity, suggesting that new therapies are required for CRPC. We investigated whether betulinic acid (BA), a small molecule from plants that inhibits multiple deubiquitinases (DUBs), reduces AR, and selectively kills PCa cells, can provide an adjuvant strategy for CRPC. Our data indicated that BA reduced AR protein stability and mRNA expression, making it an attractive agent for CRPC. BA decreased AR mRNA possibly by inhibiting a histone 2A DUB thereby increasing ubiquitinated histone 2A, a transcriptional repressor. We identified multiple and specific DUBs inhibited by BA either in PCa cells or using recombinant DUBs. Similar results were obtained using another multi-DUB inhibitor WP1130, suggesting that these DUB inhibitors can decrease AR expression and increase PCa-specific death. Our results also suggest that combining multi-DUB inhibitors BA or WP1130 with enzalutamide may provide a novel strategy for CRPC by further decreasing AR expression and increasing apoptotic cell death.
Collapse
|
36
|
Relevance of poly (ADP-ribose) polymerase inhibitors in prostate cancer. Curr Opin Support Palliat Care 2018; 12:339-343. [DOI: 10.1097/spc.0000000000000358] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
37
|
Emerging insights into HAUSP (USP7) in physiology, cancer and other diseases. Signal Transduct Target Ther 2018; 3:17. [PMID: 29967688 PMCID: PMC6023882 DOI: 10.1038/s41392-018-0012-y] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/13/2018] [Accepted: 02/23/2018] [Indexed: 12/13/2022] Open
Abstract
Herpesvirus-associated ubiquitin-specific protease (HAUSP) is a USP family deubiquitinase. HAUSP is a protein of immense biological importance as it is involved in several cellular processes, including host-virus interactions, oncogenesis and tumor suppression, DNA damage and repair processes, DNA dynamics and epigenetic modulations, regulation of gene expression and protein function, spatio-temporal distribution, and immune functions. Since its discovery in the late 1990s as a protein interacting with a herpes virus regulatory protein, extensive studies have assessed its complex roles in p53-MDM2-related networks, identified numerous additional interacting partners, and elucidated the different roles of HAUSP in the context of cancer, development, and metabolic and neurological pathologies. Recent analyses have provided new insights into its biochemical and functional dynamics. In this review, we provide a comprehensive account of our current knowledge about emerging insights into HAUSP in physiology and diseases, which shed light on fundamental biological questions and promise to provide a potential target for therapeutic intervention. Improved understandings of a molecular-tag-removing enzyme could lead to the development of therapies for many diseases. Dr. Mrinal K Ghosh of the Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB) and colleagues reviewed 20 years of research on herpesvirus-associated ubiquitin-specific protease (HAUSP), involved in a wide range of cellular processes through its role in removing the ubiquitin from molecules, thus signaling their fate. It was first discovered in/as a herpes virus infected cells, ultimately enhancing infection. It was later found to have a wide range of functions depending on the molecules it interacts with under normal physiological and disease conditions. Targeting HAUSP with drugs shows promise for suppressing prostate, lung, colon, breast, blood, and other cancers. It could also impact treatment of neurological conditions such as Huntington’s disease, and metabolic disorders, such as diabetes.
Collapse
|
38
|
Yeasmin Khusbu F, Chen FZ, Chen HC. Targeting ubiquitin specific protease 7 in cancer: A deubiquitinase with great prospects. Cell Biochem Funct 2018; 36:244-254. [PMID: 29781103 DOI: 10.1002/cbf.3336] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/21/2018] [Accepted: 04/23/2018] [Indexed: 12/20/2022]
Abstract
Deubiquitinase (DUB)-mediated cleavage of ubiquitin chain balances ubiquitination and deubiquitination for determining protein fate. USP7 is one of the best characterized DUBs and functionally important. Numerous proteins have been identified as potential substrates and binding partners of USP7; those play crucial roles in diverse array of cellular and biological processes including tumour suppression, cell cycle, DNA repair, chromatin remodelling, and epigenetic regulation. This review aims at summarizing the current knowledge of this wide association of USP7 with many cellular processes that enlightens the possibility of abnormal USP7 activity in promoting oncogenesis and the importance of identification of specific inhibitors.
Collapse
Affiliation(s)
- Farjana Yeasmin Khusbu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Fang-Zhi Chen
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Han-Chun Chen
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| |
Collapse
|
39
|
Liao Y, Xia X, Liu N, Cai J, Guo Z, Li Y, Jiang L, Dou QP, Tang D, Huang H, Liu J. Growth arrest and apoptosis induction in androgen receptor-positive human breast cancer cells by inhibition of USP14-mediated androgen receptor deubiquitination. Oncogene 2018; 37:1896-1910. [PMID: 29353883 PMCID: PMC5886989 DOI: 10.1038/s41388-017-0069-z] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/23/2017] [Accepted: 09/19/2017] [Indexed: 11/10/2022]
Abstract
It has been well known that androgen receptor (AR) is critical to prostate cancer development and progression. It has also been documented that AR is expressed in more than 60% of breast tumors, which promotes the growth of estrogen receptor-negative (ER-)/AR-positive (AR+) breast cancer cells. Thus, AR might be a potential therapeutic target for AR-positive/ER-negative breast cancer patients. Previously we reported that in prostate cancer cells proteasome-associated deubiquitinase ubiquitin-specific protease 14 (USP14) stabilized AR protein level by removing its ubiquitin chain. In the current study, we studied the USP14-AR protein interaction and cell proliferation status after USP14 reduction or inhibition in breast cancer cells, and our results support the conclusion that targeting USP14 is a novel strategy for treating AR-responsive breast cancer. We found that inhibition of USP14 accelerated the K48-ubiquitination and proteasome-mediated degradation of AR protein. Additionally, both genetic and pharmacological inhibition of USP14 significantly suppressed cell proliferation in AR-responsive breast cancer cells by blocking G0/G1 to S phase transition and inducing apoptosis. Moreover, AR overexpression inhibited USP14 inhibition-induced events, suggesting that AR deubiquitination by USP14 is critical for breast cancer growth and USP14 inhibition is a possible strategy to treat AR-positive breast cancer.
Collapse
Affiliation(s)
- Yuning Liao
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaohong Xia
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ningning Liu
- Guangzhou Institute of Cardiovascular Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jianyu Cai
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhiqiang Guo
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yanling Li
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lili Jiang
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Q Ping Dou
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- The Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
- Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Daolin Tang
- The Third Affiliated Hospital, Center for DAMP Biology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Protein Modification and Degradation Laboratory, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hongbiao Huang
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Jinbao Liu
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
40
|
Jing B, Liu M, Yang L, Cai HY, Chen JB, Li ZX, Kou X, Wu YZ, Qin DJ, Zhou L, Jin J, Lei H, Xu HZ, Wang WW, Wu YL. Characterization of naturally occurring pentacyclic triterpenes as novel inhibitors of deubiquitinating protease USP7 with anticancer activity in vitro. Acta Pharmacol Sin 2018; 39:492-498. [PMID: 29168472 PMCID: PMC5843828 DOI: 10.1038/aps.2017.119] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/21/2017] [Indexed: 12/23/2022]
Abstract
Deubiquitinating protease USP7 is a promising therapeutic target for cancer treatment, and interest in developing USP7 inhibitors has greatly increased. In the present study, we reported a series of natural pentacyclic triterpenes with USP7 inhibitory activity in vitro. Among them, both the ursane triterpenes and oleanane triterpenes were more active than the lupine triterpenes, whereas ursolic acid was the most potent with IC50 of 7.0±1.5 μmol/L. Molecular docking studies showed that ursolic acid might occupy the ubiquitin binding pocket of USP7, with the 17-carboxyl group and 3-hydroxyl group playing a vital role in the USP7-ursolic acid interaction. Using the cellular thermal shift assay, we demonstrated that ursolic acid interacted with USP7 in RPMI8226 human myeloma cells. Ursolic acid dose-dependently inhibited the proliferation of the myeloma cells with IC50 of 6.56 μmol/L, accompanied by reductions in USP7 substrates such as MDM2, UHRF1 and DNMT1. Overexpression of USP7 partially, but significantly attenuated ursolic acid-induced cell death as well as downregulation of MDM2, UHRF1 and DNMT1. In conclusion, we demonstrate for the first time that pentacyclic triterpenes represent a novel scaffold for developing USP7 inhibitors and that USP7 inhibition contributes to the anti-cancer effect of ursolic acid.
Collapse
Affiliation(s)
- Bo Jing
- Hongqiao International Institute of
Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology
Division of Shanghai Universities E-Institutes, Key Laboratory of Cell
Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao
Tong University School of Medicine, Shanghai
200025, China
| | - Meng Liu
- Hongqiao International Institute of
Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology
Division of Shanghai Universities E-Institutes, Key Laboratory of Cell
Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao
Tong University School of Medicine, Shanghai
200025, China
| | - Li Yang
- Hongqiao International Institute of
Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology
Division of Shanghai Universities E-Institutes, Key Laboratory of Cell
Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao
Tong University School of Medicine, Shanghai
200025, China
| | - Hai-yan Cai
- Hongqiao International Institute of
Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology
Division of Shanghai Universities E-Institutes, Key Laboratory of Cell
Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao
Tong University School of Medicine, Shanghai
200025, China
| | - Jie-bo Chen
- Hongqiao International Institute of
Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology
Division of Shanghai Universities E-Institutes, Key Laboratory of Cell
Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao
Tong University School of Medicine, Shanghai
200025, China
| | - Ze-xi Li
- Hongqiao International Institute of
Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology
Division of Shanghai Universities E-Institutes, Key Laboratory of Cell
Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao
Tong University School of Medicine, Shanghai
200025, China
| | - Xi Kou
- Hongqiao International Institute of
Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology
Division of Shanghai Universities E-Institutes, Key Laboratory of Cell
Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao
Tong University School of Medicine, Shanghai
200025, China
| | - Yun-zhao Wu
- Hongqiao International Institute of
Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology
Division of Shanghai Universities E-Institutes, Key Laboratory of Cell
Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao
Tong University School of Medicine, Shanghai
200025, China
| | - Dong-jun Qin
- Hongqiao International Institute of
Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology
Division of Shanghai Universities E-Institutes, Key Laboratory of Cell
Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao
Tong University School of Medicine, Shanghai
200025, China
| | - Li Zhou
- Department of Hematology, Rui-Jin
Hospital, Shanghai Jiao-Tong University School of Medicine,
Shanghai
200025, China
| | - Jin Jin
- Hongqiao International Institute of
Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology
Division of Shanghai Universities E-Institutes, Key Laboratory of Cell
Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao
Tong University School of Medicine, Shanghai
200025, China
| | - Hu Lei
- Hongqiao International Institute of
Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology
Division of Shanghai Universities E-Institutes, Key Laboratory of Cell
Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao
Tong University School of Medicine, Shanghai
200025, China
| | - Han-zhang Xu
- Hongqiao International Institute of
Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology
Division of Shanghai Universities E-Institutes, Key Laboratory of Cell
Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao
Tong University School of Medicine, Shanghai
200025, China
| | - Wei-wei Wang
- Hongqiao International Institute of
Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology
Division of Shanghai Universities E-Institutes, Key Laboratory of Cell
Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao
Tong University School of Medicine, Shanghai
200025, China
| | - Ying-li Wu
- Hongqiao International Institute of
Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology
Division of Shanghai Universities E-Institutes, Key Laboratory of Cell
Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao
Tong University School of Medicine, Shanghai
200025, China
| |
Collapse
|
41
|
CDDO-Me reveals USP7 as a novel target in ovarian cancer cells. Oncotarget 2018; 7:77096-77109. [PMID: 27780924 PMCID: PMC5363571 DOI: 10.18632/oncotarget.12801] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 10/14/2016] [Indexed: 12/19/2022] Open
Abstract
Deubiquitinating enzyme USP7 has been involved in the pathogenesis and progression of several cancers. Targeting USP7 is becoming an attractive strategy for cancer therapy. In this study, we identified synthetic triterpenoid C-28 methyl ester of 2-cyano-3, 12-dioxoolen-1, 9-dien-28-oic acid (CDDO-Me) as a novel inhibitor of USP7 but not of other cysteine proteases such as cathepsin B and cathepsin D. CDDO-Me inhibits USP7 activity via a mechanism that is independent of the presence of α, β-unsaturated ketones. Molecular docking studies showed that CDDO-Me fits well in the ubiquitin carboxyl terminus-binding pocket on USP7. Given that CDDO-Me is known to be effective against ovarian cancer cells, we speculated that CDDO-Me may target USP7 in ovarian cancer cells. We demonstrated that ovarian cancer cells have higher USP7 expression than their normal counterparts. Knockdown of USP7 inhibits the proliferation of ovarian cancer cells both in vitro and in vivo. Using the cellular thermal shift assay and the drug affinity responsive target stability assay, we further demonstrated that CDDO-Me directly binds to USP7 in cells, which leads to the decrease of its substrates such as MDM2, MDMX and UHRF1. CDDO-Me suppresses ovarian cancer tumor growth in an xenograft model. In conclusion, we demonstrate that USP7 is a novel target of ovarian cancer cells; targeting USP7 may contribute to the anti-cancer effect of CDDO-Me. The development of novel USP7 selective compounds based on the CDDO-Me-scaffold warrants further investigation.
Collapse
|
42
|
Cerrato A, Merolla F, Morra F, Celetti A. CCDC6: the identity of a protein known to be partner in fusion. Int J Cancer 2017; 142:1300-1308. [PMID: 29044514 DOI: 10.1002/ijc.31106] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/07/2017] [Accepted: 10/05/2017] [Indexed: 12/19/2022]
Abstract
Coiled Coil Domain Containing 6 gene, CCDC6, was initially isolated as part of a tumorigenic DNA originated by the fusion of CCDC6 with the tyrosine kinase of RET receptor, following a paracentric inversion of chromosome 10. For a long time, CCDC6 has been considered as an accidental partner of the RET protooncogene, providing the promoter and the first 101 aa necessary for the constitutive activation of the oncogenic Tyrosine Kinase (TK) RET in thyroid cells. With the advent of more refined diagnostic tools and bioinformatic algorithms, an exponential growth in fusion genes discoveries has allowed the identification of CCDC6 as partner of genes other than RET in different tumor types. CCDC6 gene product has a proper role in sustaining the DNA damage checkpoints in response to DNA damage. The inactivation of CCDC6 secondary to chromosomal rearrangements or gene mutations could enhance tumor progression by impairing the apoptotic response upon the DNA damage exposure, contributing to the generation of radio- and chemoresistance. Preclinical studies indicate that the attenuation of CCDC6 in cancer, while conferring a resistance to cisplatinum, sensitizes the cancer cells to the small molecule inhibitors of Poly (ADP-ribose) polymerase (PARP1/2) with a synthetic lethal effect. Several CCDC6 mutations and gene rearrangements have been described so far in different types of cancer and CCDC6 may represent a possible predictive biomarker of tumor resistance to the conventional anticancer treatments. Nevertheless, the detection of a CCDC6 impairment in cancer patients may help to select, in future clinical trials, those patients who could benefit of PARP-inhibitors treatment alone or in combination with other treatments.
Collapse
Affiliation(s)
- Aniello Cerrato
- Institute for Experimental Endocrinology and Oncology, Research National Council, Naples, Italy
| | - Francesco Merolla
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Francesco Morra
- Institute for Experimental Endocrinology and Oncology, Research National Council, Naples, Italy
| | - Angela Celetti
- Institute for Experimental Endocrinology and Oncology, Research National Council, Naples, Italy
| |
Collapse
|
43
|
Baumgart SJ, Haendler B. Exploiting Epigenetic Alterations in Prostate Cancer. Int J Mol Sci 2017; 18:ijms18051017. [PMID: 28486411 PMCID: PMC5454930 DOI: 10.3390/ijms18051017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/04/2017] [Accepted: 05/04/2017] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer affects an increasing number of men worldwide and is a leading cause of cancer-associated deaths. Beside genetic mutations, many epigenetic alterations including DNA and histone modifications have been identified in clinical prostate tumor samples. They have been linked to aberrant activity of enzymes and reader proteins involved in these epigenetic processes, leading to the search for dedicated inhibitory compounds. In the wake of encouraging anti-tumor efficacy results in preclinical models, epigenetic modulators addressing different targets are now being tested in prostate cancer patients. In addition, the assessment of microRNAs as stratification biomarkers, and early clinical trials evaluating suppressor microRNAs as potential prostate cancer treatment are being discussed.
Collapse
Affiliation(s)
- Simon J Baumgart
- Drug Discovery, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany.
| | - Bernard Haendler
- Drug Discovery, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany.
| |
Collapse
|
44
|
Morra F, Merolla F, Napolitano V, Ilardi G, Miro C, Paladino S, Staibano S, Cerrato A, Celetti A. The combined effect of USP7 inhibitors and PARP inhibitors in hormone-sensitive and castration-resistant prostate cancer cells. Oncotarget 2017; 8:31815-31829. [PMID: 28415632 PMCID: PMC5458250 DOI: 10.18632/oncotarget.16463] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/15/2017] [Indexed: 11/27/2022] Open
Abstract
PURPOSE OF THE STUDY Reduced levels of the tumor suppressor protein CCDC6 sensitize cancer cells to the treatment with PARP-inhibitors. The turnover of CCDC6 protein is regulated by the de-ubiquitinase USP7, which also controls the androgen receptor (AR) stability. Here, we correlated the expression levels of CCDC6 and USP7 proteins in primary prostate cancers (PC). Moreover, we tested the efficacy of the USP7 inhibitors, in combination with PARP-inhibitors as a novel therapeutic option in advanced prostate cancer.Experimental techniques: PC cells were exposed to USP7 inhibitor, P5091, together with cycloheximide, to investigate the turnover of the USP7 substrates, AR and CCDC6. As outcome of the AR downregulation, transcription targets of AR and its variant V7 were examined by qPCR. As a result of CCDC6 degradation, the induction of PARP inhibitors sensitivity was evaluated by analyzing PC cells viability and foci formation. We scored and correlated CCDC6 and USP7 expression levels in a prostate cancer tissue microarray (TMA). RESULTS P5091 accelerated the degradation of AR and V7 isoform affecting PSA, UBE2C, CDC20 transcription and PC cells proliferation. Moreover, P5091 accelerated the degradation of CCDC6 sensitizing the cells to PARP-inhibitors, that acted sinergistically with genotoxic agents. The immunohistochemical analysis of both CCDC6 and USP7 proteins exhibited significant correlation for the intensity of staining (p ≤ 0.05).Data interpretation: Thus, CCDC6 and USP7 represent predictive markers for the combined treatment of the USP7-inhibitors and PARP-inhibitors in advanced prostate cancer.
Collapse
Affiliation(s)
- Francesco Morra
- Institute for Experimental Endocrinology and Oncology, Research National Council, Naples, Italy
| | - Francesco Merolla
- Department of Advanced Biomedical Sciences, University “Federico II”, Naples, Italy
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Virginia Napolitano
- Institute for Experimental Endocrinology and Oncology, Research National Council, Naples, Italy
- Department of Advanced Biomedical Sciences, University “Federico II”, Naples, Italy
| | - Gennaro Ilardi
- Department of Advanced Biomedical Sciences, University “Federico II”, Naples, Italy
| | - Caterina Miro
- Institute for Experimental Endocrinology and Oncology, Research National Council, Naples, Italy
| | - Simona Paladino
- Department of Molecular Medicine and Medical Biotechnology, University “Federico II”, Naples, Italy
| | - Stefania Staibano
- Department of Advanced Biomedical Sciences, University “Federico II”, Naples, Italy
| | - Aniello Cerrato
- Institute for Experimental Endocrinology and Oncology, Research National Council, Naples, Italy
| | - Angela Celetti
- Institute for Experimental Endocrinology and Oncology, Research National Council, Naples, Italy
| |
Collapse
|
45
|
Qiu GZ, Sun W, Jin MZ, Lin J, Lu PG, Jin WL. The bad seed gardener: Deubiquitinases in the cancer stem-cell signaling network and therapeutic resistance. Pharmacol Ther 2017; 172:127-138. [DOI: 10.1016/j.pharmthera.2016.12.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
46
|
Zhang L, Wang H, Tian L, Li H. Expression of USP7 and MARCH7 Is Correlated with Poor Prognosis in Epithelial Ovarian Cancer. TOHOKU J EXP MED 2017; 239:165-75. [PMID: 27302477 DOI: 10.1620/tjem.239.165] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Epithelial ovarian cancer (EOC) is one of the worst malignancies in females with poor overall survival due to the rapid metastasis and the absence of ideal biomarkers. Ubiquitin-specific protease 7 (USP7), an important deubiquitinating enzyme, was reported to be upregulated in several cancers, including liver, prostate and colon cancers. Membrane associated RING-CH protein 7 (MARCH7) belongs to the member of the E3 ubiquitin ligases. In addition, MARCH7 regulates T cell proliferation and the neuronal development and participates in the membrane trafficking and protein degradation. Importantly, MARCH7 itself is ubiquitinated and acts as a potential substrate of USP7. However, the roles of USP7 and MARCH7 in EOC remain to be investigated. We collected 121 EOC patients and analyzed the expression levels of USP7 and MARCH7 in tumor tissues with immunohistochemical staining. We found that the high expression of the two proteins was correlated with lymph node metastasis in EOC patients. Univariate and multivariate analyses revealed that the patients with high expression of the two proteins showed poorer prognosis compared with other patients. Subsequently, using SKOV3 human ovarian adenocarcinoma cells, we showed that either USP7 or MARCH7 enhanced the proliferation and invasion abilities. Moreover, USP7 could regulate the expression levels of E-cadherin and β-catenin through the MARCH7 signaling pathway. Our findings indicate that USP7 and MARCH7 are involved in the progression of EOC. In conclusion, analyzing the expression of USP7 and MARCH7 has high prognostic value in predicting EOC prognosis.
Collapse
Affiliation(s)
- Li Zhang
- Yidu Central Hospital of Weifang
| | | | | | | |
Collapse
|
47
|
Nath SR, Lieberman AP. The Ubiquitination, Disaggregation and Proteasomal Degradation Machineries in Polyglutamine Disease. Front Mol Neurosci 2017; 10:78. [PMID: 28381987 PMCID: PMC5360718 DOI: 10.3389/fnmol.2017.00078] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 03/06/2017] [Indexed: 12/14/2022] Open
Abstract
Polyglutamine disorders are chronic, progressive neurodegenerative diseases caused by expansion of a glutamine tract in widely expressed genes. Despite excellent models of disease, a well-documented clinical history and progression, and established genetic causes, there are no FDA approved, disease modifying treatments for these disorders. Downstream of the mutant protein, several divergent pathways of toxicity have been identified over the last several decades, supporting the idea that targeting only one of these pathways of toxicity is unlikely to robustly alleviate disease progression. As a result, a vast body of research has focused on eliminating the mutant protein to broadly prevent downstream toxicity, either by silencing mutant protein expression or leveraging the endogenous protein quality control machinery. In the latter approach, a focus has been placed on four critical components of mutant protein degradation that are active in the nucleus, a key site of toxicity: disaggregation, ubiquitination, deubiquitination, and proteasomal activity. These machineries have unique functional components, but work together as a cellular defense system that can be successfully leveraged to alleviate disease phenotypes in several models of polyglutamine toxicity. This review will highlight recent advances in understanding both the potential and role of these components of the protein quality control machinery in polyglutamine disease pathophysiology.
Collapse
Affiliation(s)
- Samir R Nath
- Medical Scientist Training Program, University of Michigan Medical SchoolAnn Arbor, MI, USA; Cellular and Molecular Biology Graduate Program, University of Michigan Medical SchoolAnn Arbor, MI, USA; Department of Pathology, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School Ann Arbor, MI, USA
| |
Collapse
|
48
|
Liao Y, Liu N, Hua X, Cai J, Xia X, Wang X, Huang H, Liu J. Proteasome-associated deubiquitinase ubiquitin-specific protease 14 regulates prostate cancer proliferation by deubiquitinating and stabilizing androgen receptor. Cell Death Dis 2017; 8:e2585. [PMID: 28151478 PMCID: PMC5386460 DOI: 10.1038/cddis.2016.477] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 11/07/2016] [Accepted: 11/09/2016] [Indexed: 12/14/2022]
Abstract
Androgen receptor (AR) is frequently over-expressed and plays a critical role in the growth and progression of human prostate cancer. The therapy attempting to target AR signalling was established in decades ago but the treatment of prostate cancer is far from being satisfactory. The assignable cause is that our understanding of the mechanism of AR regulation and re-activation remains incomplete. Increasing evidence suggests that deubiquitinases are involved in the regulation of cancer development and progression but the specific underlying mechanism often is not elucidated. In the current study, we have identified ubiquitin-specific protease 14 (USP14) as a novel regulator of AR, inhibiting the degradation of AR via deubiquitinating this oncoprotein in the androgen-responsive prostate cancer cells. We found that (i) USP14 could bind to AR, and additionally, both genetic and pharmacological inhibition of USP14 accelerated the ubiquitination and degradation of AR; (ii) downregulation or inhibition of USP14 suppressed cell proliferation and colony formation of LNcap cells and, conversely, overexpression of USP14 promoted the proliferation; and (iii) reduction or inhibition of USP14 induced G0/G1 phase arrest in LNcap prostate cancer cells. Hence, we conclude that USP14 promotes prostate cancer progression likely through stabilization of AR, suggesting that USP14 could be a promising therapeutic target for prostate cancer.
Collapse
Affiliation(s)
- Yuning Liao
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Ningning Liu
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China.,Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xianliang Hua
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Jianyu Cai
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Xiaohong Xia
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Xuejun Wang
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China.,Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, USA
| | - Hongbiao Huang
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Jinbao Liu
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| |
Collapse
|
49
|
Ma M, Yu N. Ubiquitin-specific protease 7 expression is a prognostic factor in epithelial ovarian cancer and correlates with lymph node metastasis. Onco Targets Ther 2016; 9:1559-69. [PMID: 27051296 PMCID: PMC4803273 DOI: 10.2147/ott.s100050] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Objective Ubiquitin-specific protease 7 (USP7) is a common target of herpesviruses and is important in the DNA damage response, which is also upregulated in several cancers, including prostate, colon, liver, and lung cancers. However, less is known about its expression in ovarian cancer tissues. The role of USP7 in epithelial ovarian cancer (EOC) has not yet been investigated. Materials and methods We recruited 141 patients from Linyi People’s Hospital between June 1999 and June 2013, all pathologically diagnosed with primary EOC. Their clinical data were collected, and the expression of USP7 in the tumor tissues was determined using immunohistochemistry. The correlations between USP7 expression and the clinicopathological variables of patients with EOC were assessed using Spearman’s rank correlation test. Kaplan–Meier analysis and Cox regression analysis were used to identify the prognosis value of USP7. The function of USP7 in the EOC cells was also detected in vitro. Results Among the 141 cases, USP7 expression was high in 59 EOC samples (41.8%), and was significantly correlated with lymphatic invasion; USP7 can act as independent prognostic indicator for the overall survival (OS) of EOC, and its high expression was associated with poor OS rate. The RNA inteference and overexpression assays indicated that USP7 can positively regulate the ovarian cell vitality and invasion process. Conclusion Patients with EOC expressing high level of USP7 have worse OS compared with those with low USP7 expression. USP7 may be involved in the proliferation and invasion of EOC cells, and USP7 expression can serve as an independent predictor of EOC.
Collapse
Affiliation(s)
- Ming Ma
- Department of Oncology, Linyi People's Hospital, Linyi, People's Republic of China
| | - Nina Yu
- Department of Gynecology and Obstetrics, Linyi People's Hospital, Linyi, People's Republic of China
| |
Collapse
|