1
|
Miao G, Yang Y, Yang X, Chen D, Liu L, Lei X. The multifaceted potential of TPT1 as biomarker and therapeutic target. Heliyon 2024; 10:e38819. [PMID: 39397949 PMCID: PMC11471257 DOI: 10.1016/j.heliyon.2024.e38819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/17/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024] Open
Abstract
Tumor Protein Translationally-Controlled 1 (TPT1) is a highly conserved gene found across eukaryotic species. The protein encoded by TPT1 is ubiquitously expressed both intracellularly and extracellularly across various tissues, and its levels are influenced by various external factors. TPT1 interacts with several key proteins, including p53, MCL1, and immunoglobulins, highlighting its crucial role in cellular processes. The dysregulation of TPT1 expression has been documented in a wide range of diseases, indicating its potential as a valuable biomarker. Additionally, targeting TPT1 presents a promising approach for treating and preventing various conditions. This review will assess the potential of TPT1 as a biomarker and evaluate the effectiveness of current strategies designed to inhibit TPT1 in disease contexts.
Collapse
Affiliation(s)
- Gelan Miao
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Yulian Yang
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Xuelian Yang
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Dexiu Chen
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Li Liu
- Department of Anesthesiology, The First Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Xianying Lei
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| |
Collapse
|
2
|
Chinnapaka S, Bakthavachalam V, Dasari S, Kannan J, Sapkota S, Kumar R, Munirathinam G. Vitamin K3 derivative inhibits androgen receptor signaling in targeting aggressive prostate cancer cells. Biofactors 2024. [PMID: 39225404 DOI: 10.1002/biof.2117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024]
Abstract
Prostate cancer (PCa) is the second critical cause of cancer-related deaths, with African Americans dying at higher rates in the U.S. The main reasons for the higher mortality rate are ethnic differences and lack of understanding of prostate cancer biology and affordable treatments, as well as the financial burden of African American men to obtain the most effective and safe treatments. The effect of micronutrients, including Vitamin K, on various cancer cell lines has been widely studied, but the potential anticancer effect of VK3-OCH3, an analog of vitamin K3 (Menadione), on African American prostate cancer has not been evaluated. In this study, we compared the anticancer effect of VK3-OCH3 on targeting African American derived PCa cell lines namely RC77-T and MDA-PCa-2b. Our results show that VK3-OCH3 significantly inhibits the proliferation of both RC77-T and MDA-PCa-2b African American PCa cells and promotes apoptosis, and the underlying mechanism of cell death appears to be similar in both the cell lines. Notably, VK3-OCH3 inhibits colony-forming ability and induces apoptosis by blocking the cell cycle at G0 in African American PCa cells. VK3-OCH3 also acts as an anti-metastatic agent by inhibiting the migration ability of the metastatic properties of African American PCa cells. The cell death of African American PCa cells mediated by VK3-OCH3 is associated with the production of free radicals, such as intracellular and mitochondrial reactive oxygen species (ROS). Interestingly, antioxidants such as N-Acetylcysteine (NAC) and Glutathione (GSH) effectively negated the oxidative stress induced by VK3-OCH3 on PCa cell lines derived from African American patients. Of note, VK3-OCH3 reduces androgen receptor and prostate-specific antigen expression in these PCa cells. Furthermore, molecular dynamic studies reiterated that VK3-OCH3 strongly binds to the androgen receptor, suggesting that the androgen receptor is the potential molecular target of VK3-OCH3. In addition, Western blot analysis showed that VK3-OCH3 reduces the expression of androgen receptor, TRX2, and anti-apoptotic signaling molecules such as Bcl-2 and TCTP in the MDA-PCa-2b metastatic PCa cellular model. In conclusion, our results suggested that VK3-OCH3 is a promising anticancer agent that could potentially reduce the mortality rates of African American PCa patients, warranting further preclinical and translational studies.
Collapse
Affiliation(s)
- Somaiah Chinnapaka
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
| | - Velavan Bakthavachalam
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
| | | | - Jhishnuraj Kannan
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
| | - Sworaj Sapkota
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
| | - Raj Kumar
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Solan, Himachal Pradesh, India
| | - Gnanasekar Munirathinam
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
| |
Collapse
|
3
|
Liu AB, Liu J, Wang S, Ma L, Zhang JF. Biological role and expression of translationally controlled tumor protein (TCTP) in tumorigenesis and development and its potential for targeted tumor therapy. Cancer Cell Int 2024; 24:198. [PMID: 38835077 DOI: 10.1186/s12935-024-03355-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
Translationally controlled tumor protein (TCTP), also known as histamine-releasing factor (HRF) or fortilin, is a highly conserved protein found in various species. To date, multiple studies have demonstrated the crucial role of TCTP in a wide range of cellular pathophysiological processes, including cell proliferation and survival, cell cycle regulation, cell death, as well as cell migration and movement, all of which are major pathogenic mechanisms of tumorigenesis and development. This review aims to provide an in-depth analysis of the functional role of TCTP in tumor initiation and progression, with a particular focus on cell proliferation, cell death, and cell migration. It will highlight the expression and pathological implications of TCTP in various tumor types, summarizing the current prevailing therapeutic strategies that target TCTP.
Collapse
Affiliation(s)
- An-Bu Liu
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Jia Liu
- Medical Experimental Center, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Sheng Wang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Lei Ma
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China.
| | - Jun-Fei Zhang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China.
| |
Collapse
|
4
|
Van Lent J, Prior R, Pérez Siles G, Cutrupi AN, Kennerson ML, Vangansewinkel T, Wolfs E, Mukherjee-Clavin B, Nevin Z, Judge L, Conklin B, Tyynismaa H, Clark AJ, Bennett DL, Van Den Bosch L, Saporta M, Timmerman V. Advances and challenges in modeling inherited peripheral neuropathies using iPSCs. Exp Mol Med 2024; 56:1348-1364. [PMID: 38825644 PMCID: PMC11263568 DOI: 10.1038/s12276-024-01250-x] [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: 11/10/2023] [Revised: 02/21/2024] [Accepted: 03/18/2024] [Indexed: 06/04/2024] Open
Abstract
Inherited peripheral neuropathies (IPNs) are a group of diseases associated with mutations in various genes with fundamental roles in the development and function of peripheral nerves. Over the past 10 years, significant advances in identifying molecular disease mechanisms underlying axonal and myelin degeneration, acquired from cellular biology studies and transgenic fly and rodent models, have facilitated the development of promising treatment strategies. However, no clinical treatment has emerged to date. This lack of treatment highlights the urgent need for more biologically and clinically relevant models recapitulating IPNs. For both neurodevelopmental and neurodegenerative diseases, patient-specific induced pluripotent stem cells (iPSCs) are a particularly powerful platform for disease modeling and preclinical studies. In this review, we provide an update on different in vitro human cellular IPN models, including traditional two-dimensional monoculture iPSC derivatives, and recent advances in more complex human iPSC-based systems using microfluidic chips, organoids, and assembloids.
Collapse
Grants
- Wellcome Trust
- R01 NS119678 NINDS NIH HHS
- U01 ES032673 NIEHS NIH HHS
- DOC-PRO4 Universiteit Antwerpen (University of Antwerp)
- This work was supported in part by the University of Antwerp (DOC-PRO4 PhD fellowship to J.V.L. and TOP-BOF research grant no. 38694 to V.T.), the Association Française contre les Myopathies (AFM research grant no. 24063 to V.T.), Association Belge contre les Maladies Neuromusculaires (ABMM research grant no. 1 to J.V.L and V.T), the interuniversity research fund (iBOF project to. L.V.D.B, E.W. and V.T.). V.T. is part of the μNEURO Research Centre of Excellence of the University of Antwerp and is an active member of the European Network for Stem Cell Core Facilities (CorEUStem, COST Action CA20140). Work in the M.L.K group was supported by the NHMRC Ideas Grant (APP1186867), CMT Australia Grant awarded to M.L.K and G.P.-S and the Australian Medical Research Future Fund (MRFF) Genomics Health Futures Mission Grant 2007681. B.M.C. is supported by the American Academy of Neurology and the Passano Foundation. L.M.J. and B.R.C. are supported by the Charcot-Marie-Tooth Association, NINDS R01 NS119678, NIEHS U01 ES032673. H.T. is supported by Academy of Finland Centre of Excellence in Stem Cell Metabolism and Sigrid Juselius Foundation. Work in the D.L.B. group is supported by a Wellcome Investigator Grant (223149/Z/21/Z), the MRC (MR/T020113/1), and with funding from the MRC and Versus Arthritis to the PAINSTORM consortium as part of the Advanced Pain Discovery Platform (MR/W002388/1).
- Australian Medical Association (Australian Medical Association Limited)
- Universiteit Hasselt (UHasselt)
- American Academy of Neurology (AAN)
- Gladstone Institutes (J. David Gladstone Institutes)
- Academy of Finland (Suomen Akatemia)
- Academy of Medical Royal Colleges (AoMRC)
- Wellcome Trust (Wellcome)
- Oxford University Hospitals NHS Trust (Oxford University Hospitals National Health Service Trust)
- KU Leuven (Katholieke Universiteit Leuven)
- Vlaams Instituut voor Biotechnologie (Flanders Institute for Biotechnology)
- Miami University | Leonard M. Miller School of Medicine (Miller School of Medicine)
Collapse
Affiliation(s)
- Jonas Van Lent
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, 2610, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, 2610, Antwerp, Belgium
- Institute of Oncology Research (IOR), BIOS+, 6500, Bellinzona, Switzerland
- Università della Svizzera Italiana, 6900, Lugano, Switzerland
| | - Robert Prior
- Universitätsklinikum Bonn (UKB), University of Bonn, Bonn, Germany
| | - Gonzalo Pérez Siles
- Northcott Neuroscience Laboratory, ANZAC Research Institute Sydney Local Health District and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Anthony N Cutrupi
- Northcott Neuroscience Laboratory, ANZAC Research Institute Sydney Local Health District and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Marina L Kennerson
- Northcott Neuroscience Laboratory, ANZAC Research Institute Sydney Local Health District and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Molecular Medicine Laboratory, Concord Hospital, Sydney, NSW, Australia
| | - Tim Vangansewinkel
- UHasselt - Hasselt University, BIOMED, Laboratory for Functional Imaging and Research on Stem Cells (FIERCE Lab), Agoralaan, 3590, Diepenbeek, Belgium
- VIB-Center for Brain and Disease Research, Laboratory of Neurobiology, 3000, Leuven, Belgium
| | - Esther Wolfs
- UHasselt - Hasselt University, BIOMED, Laboratory for Functional Imaging and Research on Stem Cells (FIERCE Lab), Agoralaan, 3590, Diepenbeek, Belgium
| | | | | | - Luke Judge
- Gladstone Institutes, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce Conklin
- Gladstone Institutes, San Francisco, CA, USA
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Henna Tyynismaa
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00290, Helsinki, Finland
| | - Alex J Clark
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - David L Bennett
- Nuffield Department of Clinical Neuroscience, Oxford University, Oxford, UK
| | - Ludo Van Den Bosch
- VIB-Center for Brain and Disease Research, Laboratory of Neurobiology, 3000, Leuven, Belgium
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute, KU Leuven-University of Leuven, 3000, Leuven, Belgium
| | - Mario Saporta
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Vincent Timmerman
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, 2610, Antwerp, Belgium.
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, 2610, Antwerp, Belgium.
| |
Collapse
|
5
|
Xia D, Xu GP, Zhang YT, Yan WW, Pan XR, Tong JH. Targeting inhibition of TCTP could inhibit proliferation and induce apoptosis in AML cells. Cell Signal 2024; 117:111074. [PMID: 38309549 DOI: 10.1016/j.cellsig.2024.111074] [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: 11/26/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/05/2024]
Abstract
Translationally controlled tumor protein (TCTP) is a highly conserved multifunctional protein, which participates in many important physiological processes. Recently, the roles of TCTP in cell proliferation and apoptosis, especially its close relationship with various tumors, have attracted widespread attention. In this study, we found that the protein level of TCTP was significantly reduced in acute promyelocytic leukemia cell line NB4 transfected with retinoic acid-induced gene G (RIG-G). The RIG-G was found in our previous work as a key mediator of anti-proliferative activity in retinoid/interferon-related pathways. Here, we tried to further explore the function of TCTP in the development of acute myeloid leukemia (AML) from different levels. Our results showed that inhibiting TCTP expression could attenuate AML cells proliferation and induce apoptosis both in AML cell lines and in xenograft of NOD-SCID mice. In addition, either compared with patients in complete remission or non-leukemia patients, we detected that the expression of TCTP was generally high in the fresh bone marrow of AML patients, suggesting that there was a certain correlation between TCTP and AML disease progression. Taken together, our study revealed the role of TCTP in AML development, and provided a potential target for AML treatment.
Collapse
Affiliation(s)
- Di Xia
- Central Laboratory, Rui-jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui-jin Er Road, Shanghai 200025, China
| | - Gui-Ping Xu
- Transfusion Department, The Second Affiliated Hospital of Chongqing Medical University, No.74 Linjiang Road, Yuzhong District, Chongqing 400010, China
| | - Ying-Ting Zhang
- Central Laboratory, Rui-jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui-jin Er Road, Shanghai 200025, China
| | - Wei-Wei Yan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xiao-Rong Pan
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui-jin Er Road, Shanghai 200025, China.
| | - Jian-Hua Tong
- Central Laboratory, Rui-jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui-jin Er Road, Shanghai 200025, China.
| |
Collapse
|
6
|
Paul R, Shreya S, Pandey S, Shriya S, Abou Hammoud A, Grosset CF, Prakash Jain B. Functions and Therapeutic Use of Heat Shock Proteins in Hepatocellular Carcinoma. LIVERS 2024; 4:142-163. [DOI: 10.3390/livers4010011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/10/2024] Open
Abstract
Heat shock proteins are intracellular proteins expressed in prokaryotes and eukaryotes that help protect the cell from stress. They play an important role in regulating cell cycle and cell death, work as molecular chaperons during the folding of newly synthesized proteins, and also in the degradation of misfolded proteins. They are not only produced under stress conditions like acidosis, energy depletion, and oxidative stress but are also continuously synthesized as a result of their housekeeping functions. There are different heat shock protein families based on their molecular weight, like HSP70, HSP90, HSP60, HSP27, HSP40, etc. Heat shock proteins are involved in many cancers, particularly hepatocellular carcinoma, the main primary tumor of the liver in adults. Their deregulations in hepatocellular carcinoma are associated with metastasis, angiogenesis, cell invasion, and cell proliferation and upregulated heat shock proteins can be used as either diagnostic or prognostic markers. Targeting heat shock proteins is a relevant strategy for the treatment of patients with liver cancer. In this review, we provide insights into heat shock proteins and heat shock protein-like proteins (clusterin) in the progression of hepatocellular carcinoma and their use as therapeutic targets.
Collapse
Affiliation(s)
- Ramakrushna Paul
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Smriti Shreya
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, India
| | | | - Srishti Shriya
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Aya Abou Hammoud
- MIRCADE Team, U1312, Bordeaux Institute of Oncology, BRIC, INSERM, University of Bordeaux, 33000 Bordeaux, France
| | - Christophe F. Grosset
- MIRCADE Team, U1312, Bordeaux Institute of Oncology, BRIC, INSERM, University of Bordeaux, 33000 Bordeaux, France
| | - Buddhi Prakash Jain
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, India
| |
Collapse
|
7
|
Le TK, Duong QH, Baylot V, Fargette C, Baboudjian M, Colleaux L, Taïeb D, Rocchi P. Castration-Resistant Prostate Cancer: From Uncovered Resistance Mechanisms to Current Treatments. Cancers (Basel) 2023; 15:5047. [PMID: 37894414 PMCID: PMC10605314 DOI: 10.3390/cancers15205047] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/26/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Prostate cancer (PC) is the second most common cancer in men worldwide. Despite recent advances in diagnosis and treatment, castration-resistant prostate cancer (CRPC) remains a significant medical challenge. Prostate cancer cells can develop mechanisms to resist androgen deprivation therapy, such as AR overexpression, AR mutations, alterations in AR coregulators, increased steroidogenic signaling pathways, outlaw pathways, and bypass pathways. Various treatment options for CRPC exist, including androgen deprivation therapy, chemotherapy, immunotherapy, localized or systemic therapeutic radiation, and PARP inhibitors. However, more research is needed to combat CRPC effectively. Further investigation into the underlying mechanisms of the disease and the development of new therapeutic strategies will be crucial in improving patient outcomes. The present work summarizes the current knowledge regarding the underlying mechanisms that promote CRPC, including both AR-dependent and independent pathways. Additionally, we provide an overview of the currently approved therapeutic options for CRPC, with special emphasis on chemotherapy, radiation therapy, immunotherapy, PARP inhibitors, and potential combination strategies.
Collapse
Affiliation(s)
- Thi Khanh Le
- Centre de Recherche en Cancérologie de Marseille—CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, 13009 Marseille, France; (T.K.L.); (Q.H.D.); (V.B.); (M.B.); (D.T.)
- European Center for Research in Medical Imaging (CERIMED), Aix-Marseille University, 13005 Marseille, France;
| | - Quang Hieu Duong
- Centre de Recherche en Cancérologie de Marseille—CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, 13009 Marseille, France; (T.K.L.); (Q.H.D.); (V.B.); (M.B.); (D.T.)
- European Center for Research in Medical Imaging (CERIMED), Aix-Marseille University, 13005 Marseille, France;
- Vietnam Academy of Science and Technology (VAST), University of Science and Technology of Hanoi (USTH), Hanoi 10000, Vietnam
| | - Virginie Baylot
- Centre de Recherche en Cancérologie de Marseille—CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, 13009 Marseille, France; (T.K.L.); (Q.H.D.); (V.B.); (M.B.); (D.T.)
- European Center for Research in Medical Imaging (CERIMED), Aix-Marseille University, 13005 Marseille, France;
| | - Christelle Fargette
- European Center for Research in Medical Imaging (CERIMED), Aix-Marseille University, 13005 Marseille, France;
- Department of Nuclear Medicine, La Timone University Hospital, Aix-Marseille University, 13005 Marseille, France
| | - Michael Baboudjian
- Centre de Recherche en Cancérologie de Marseille—CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, 13009 Marseille, France; (T.K.L.); (Q.H.D.); (V.B.); (M.B.); (D.T.)
- European Center for Research in Medical Imaging (CERIMED), Aix-Marseille University, 13005 Marseille, France;
- Department of Urology AP-HM, Aix-Marseille University, 13005 Marseille, France
| | - Laurence Colleaux
- Faculté de Médecine Timone, INSERM, MMG, U1251, Aix-Marseille University, 13385 Marseille, France;
| | - David Taïeb
- Centre de Recherche en Cancérologie de Marseille—CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, 13009 Marseille, France; (T.K.L.); (Q.H.D.); (V.B.); (M.B.); (D.T.)
- European Center for Research in Medical Imaging (CERIMED), Aix-Marseille University, 13005 Marseille, France;
- Department of Nuclear Medicine, La Timone University Hospital, Aix-Marseille University, 13005 Marseille, France
| | - Palma Rocchi
- Centre de Recherche en Cancérologie de Marseille—CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, 13009 Marseille, France; (T.K.L.); (Q.H.D.); (V.B.); (M.B.); (D.T.)
- European Center for Research in Medical Imaging (CERIMED), Aix-Marseille University, 13005 Marseille, France;
| |
Collapse
|
8
|
Baldissera AB, Boia-Ferreira M, Basílio ABC, Resende JSDS, Castro MAA, Chaim OM, Gremski LH, Veiga SS, Senff-Ribeiro A. Sertraline as a potential cancer therapeutic approach: Biological relevance of TCTP in breast cancer cell lines and tumors. Adv Med Sci 2023; 68:227-237. [PMID: 37379765 DOI: 10.1016/j.advms.2023.06.001] [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: 11/11/2022] [Revised: 02/27/2023] [Accepted: 06/01/2023] [Indexed: 06/30/2023]
Abstract
PURPOSE This study aimed to evaluate the role of Translationally Controlled Tumor Protein (TCTP) in breast cancer (BC) and investigate the effects of sertraline, a serotonin selective reuptake inhibitor (SSRI), on BC cells. The objective was to assess the potential of sertraline as a therapeutic agent in BC treatment by examining its ability to inhibit TCTP expression and exert antitumor effects. MATERIAL AND METHODS We utilized five different BC cell lines representing the molecular heterogeneity and distinct subtypes of BC, including luminal, normal-like, HER2-positive, and triple-negative BC. These subtypes play a crucial role in determining clinical treatment strategies and prognosis. RESULTS The highest levels of TCTP were observed in triple-negative BC cell lines, known for their aggressive behavior. Sertraline treatment reduced TCTP expression in BC cell lines, significantly impacting cell viability, clonogenicity, and migration. Additionally, sertraline sensitized triple-negative BC cell lines to cytotoxic chemotherapeutic drugs (doxorubicin and cisplatin) suggesting its potential as an adjunctive therapy to enhance the chemotherapeutic response. Bioinformatic analysis of TCTP mRNA levels in TCGA BC data revealed a negative correlation between TCTP levels and patient survival, as well as between TCTP/tpt1 and Ki67. These findings contradict our data and previous studies indicating a correlation between TCTP protein levels and aggressiveness and poor prognosis in BC. CONCLUSIONS Sertraline shows a promise as a potential therapeutic option for BC, particularly in triple-negative BC. Its ability to inhibit TCTP expression, enhance chemotherapeutic response, highlights its potential clinical utility in BC treatment, specifically in triple-negative BC subtype.
Collapse
Affiliation(s)
| | | | - Alana B C Basílio
- Department of Cell Biology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Jean Silva de Souza Resende
- Bioinformatics and Systems Biology Laboratory, Federal University of Paraná, Curitiba, PR, Brazil; Pelé Pequeno Príncipe Research Institute, Oncology Division, Curitiba, PR, Brazil
| | | | - Olga M Chaim
- Department of Cell Biology, Federal University of Paraná, Curitiba, PR, Brazil
| | | | - Silvio S Veiga
- Department of Cell Biology, Federal University of Paraná, Curitiba, PR, Brazil
| | | |
Collapse
|
9
|
Santamaria G, Cioce M, Rizzuto A, Fazio VM, Viglietto G, Lucibello M. Harnessing the value of TCTP in breast cancer treatment resistance: an opportunity for personalized therapy. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:447-467. [PMID: 37842235 PMCID: PMC10571059 DOI: 10.20517/cdr.2023.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/25/2023] [Accepted: 06/15/2023] [Indexed: 10/17/2023]
Abstract
Early identification of breast cancer (BC) patients at a high risk of progression may aid in therapeutic and prognostic aims. This is especially true for metastatic disease, which is responsible for most cancer-related deaths. Growing evidence indicates that the translationally controlled tumor protein (TCTP) may be a clinically relevant marker for identifying poorly differentiated aggressive BC tumors. TCTP is an intriguing protein with pleiotropic functions, which is involved in multiple signaling pathways. TCTP may also be involved in stress response, cell growth and proliferation-related processes, underlying its potential role in the initiation of metastatic growth. Thus, TCTP marks specific cancer cell sub-populations with pronounced stress adaptation, stem-like and immune-evasive properties. Therefore, we have shown that in vivo phospho-TCTP levels correlate with the response of BC cells to anti-HER2 agents. In this review, we discuss the clinical relevance of TCTP for personalized therapy, specific TCTP-targeting strategies, and currently available therapeutic agents. We propose TCTP as an actionable clinically relevant target that could potentially improve patient outcomes.
Collapse
Affiliation(s)
- Gianluca Santamaria
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, Catanzaro 88100, Italy
- These authors contributed equally
| | - Mario Cioce
- Department of Medicine, Laboratory of Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, Rome 00128, Italy
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome 00133, Italy
- These authors contributed equally
| | - Antonia Rizzuto
- Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, Catanzaro 88100, Italy
| | - Vito Michele Fazio
- Department of Medicine, Laboratory of Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, Rome 00128, Italy
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome 00133, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, Catanzaro 88100, Italy
| | - Maria Lucibello
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, Catanzaro 88100, Italy
- Department of Biomedical Sciences, Institute for Biomedical Research and Innovation, National Research Council of Italy (CNR), Catanzaro 88100, Italy
| |
Collapse
|
10
|
Udu-Ituma S, Adélaïde J, Le TK, Omabe K, Finetti P, Paris C, Guille A, Bertucci F, Birnbaum D, Rocchi P, Chaffanet M. ZNF703 mRNA-Targeting Antisense Oligonucleotide Blocks Cell Proliferation and Induces Apoptosis in Breast Cancer Cell Lines. Pharmaceutics 2023; 15:1930. [PMID: 37514116 PMCID: PMC10384502 DOI: 10.3390/pharmaceutics15071930] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
The luminal B molecular subtype of breast cancers (BC) accounts for more than a third of BCs and is associated with aggressive clinical behavior and poor prognosis. The use of endocrine therapy in BC treatment has significantly contributed to the decrease in the number of deaths in recent years. However, most BC patients with prolonged exposure to estrogen receptor (ER) selective modulators such as tamoxifen develop resistance and become non-responsive over time. Recent studies have implicated overexpression of the ZNF703 gene in BC resistance to endocrine drugs, thereby highlighting ZNF703 inhibition as an attractive modality in BC treatment, especially luminal B BCs. However, there is no known inhibitor of ZNF703 due to its nuclear association and non-enzymatic activity. Here, we have developed an antisense oligonucleotide (ASO) against ZNF703 mRNA and shown that it downregulates ZNF703 protein expression. ZNF703 inhibition decreased cell proliferation and induced apoptosis. Combined with cisplatin, the anti-cancer effects of ZNF703-ASO9 were improved. Moreover, our work shows that ASO technology may be used to increase the number of targetable cancer genes.
Collapse
Affiliation(s)
- Sandra Udu-Ituma
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
- Department of Biology, Alex Ekwueme Federal University Ndufu-Alike Ikwo, Abakaliki P.M.B. 1010, Ebonyi State, Nigeria
- European Center for Research in Medical Imaging, Aix-Marseille University, 13005 Marseille, France
| | - José Adélaïde
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| | - Thi Khanh Le
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
- European Center for Research in Medical Imaging, Aix-Marseille University, 13005 Marseille, France
| | - Kenneth Omabe
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| | - Pascal Finetti
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| | - Clément Paris
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| | - Arnaud Guille
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| | - François Bertucci
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| | - Daniel Birnbaum
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| | - Palma Rocchi
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
- European Center for Research in Medical Imaging, Aix-Marseille University, 13005 Marseille, France
| | - Max Chaffanet
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| |
Collapse
|
11
|
Malard F, Sizun C, Thureau A, Carlier L, Lescop E. Structural transitions in TCTP tumor protein upon binding to the anti-apoptotic protein family member Mcl-1. J Biol Chem 2023:104830. [PMID: 37201583 PMCID: PMC10333598 DOI: 10.1016/j.jbc.2023.104830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/20/2023] Open
Abstract
Translationally Controlled Tumor Protein (TCTP) serves as a pro-survival factor in tumor cells, inhibiting the mitochondrial apoptosis pathway by enhancing the function of anti-apoptotic Bcl-2 family members Mcl-1 and Bcl-xL. TCTP specifically binds to Bcl-xL, preventing Bax-dependent Bcl-xL-induced cytochrome c release, and it reduces Mcl-1 turnover by inhibiting its ubiquitination, thereby decreasing Mcl-1-mediated apoptosis. TCTP harbors a BH3-like motif that forms a β-strand buried in the globular domain of the protein. In contrast, the crystal structure of the TCTP BH3-like peptide in complex with the Bcl-2 family member Bcl-xL reveals an α-helical conformation for the BH3-like motif, suggesting significant structural changes upon complex formation. Employing biochemical and biophysical methods, including limited proteolysis, circular dichroism NMR, and SAXS, we describe the TCTP complex with the Bcl-2 homolog Mcl-1. Our findings demonstrate that full-length TCTP binds to the BH3 binding groove of Mcl-1 via its BH3-like motif, experiencing conformational exchange at the interface on a micro- to milli-second timescale. Concurrently, the TCTP globular domain becomes destabilized, transitioning into a molten-globule state. Furthermore, we establish that the non-canonical residue D16 within TCTP BH3-like motif reduces stability while enhancing the dynamics of the intermolecular interface. In conclusion, we detail the structural plasticity of TCTP and discuss its implications for partner interactions and future anticancer drug design strategies aimed at targeting TCTP complexes.
Collapse
Affiliation(s)
- Florian Malard
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Université Paris-Saclay, LabEx LERMIT, 1 avenue de la Terrasse, 91190 Gif-sur-Yvette, France
| | - Christina Sizun
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Université Paris-Saclay, LabEx LERMIT, 1 avenue de la Terrasse, 91190 Gif-sur-Yvette, France
| | | | - Ludovic Carlier
- Laboratoire Des Biomolécules, LBM, Sorbonne Université, Ecole Normale Supérieure, PSL University, CNRS, Paris, France
| | - Ewen Lescop
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Université Paris-Saclay, LabEx LERMIT, 1 avenue de la Terrasse, 91190 Gif-sur-Yvette, France.
| |
Collapse
|
12
|
Le TK, Cherif C, Omabe K, Paris C, Lannes F, Audebert S, Baudelet E, Hamimed M, Barbolosi D, Finetti P, Bastide C, Fazli L, Gleave M, Bertucci F, Taïeb D, Rocchi P. DDX5 mRNA-targeting antisense oligonucleotide as a new promising therapeutic in combating castration-resistant prostate cancer. Mol Ther 2023; 31:471-486. [PMID: 35965411 PMCID: PMC9931527 DOI: 10.1016/j.ymthe.2022.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 06/26/2022] [Accepted: 08/09/2022] [Indexed: 02/07/2023] Open
Abstract
The heat shock protein 27 (Hsp27) has emerged as a principal factor of the castration-resistant prostate cancer (CRPC) progression. Also, an antisense oligonucleotide (ASO) against Hsp27 (OGX-427 or apatorsen) has been assessed in different clinical trials. Here, we illustrate that Hsp27 highly regulates the expression of the human DEAD-box protein 5 (DDX5), and we define DDX5 as a novel therapeutic target for CRPC treatment. DDX5 overexpression is strongly correlated with aggressive tumor features, notably with CRPC. DDX5 downregulation using a specific ASO-based inhibitor that acts on DDX5 mRNAs inhibits cell proliferation in preclinical models, and it particularly restores the treatment sensitivity of CRPC. Interestingly, through the identification and analysis of DDX5 protein interaction networks, we have identified some specific functions of DDX5 in CRPC that could contribute actively to tumor progression and therapeutic resistance. We first present the interactions of DDX5 and the Ku70/80 heterodimer and the transcription factor IIH, thereby uncovering DDX5 roles in different DNA repair pathways. Collectively, our study highlights critical functions of DDX5 contributing to CRPC progression and provides preclinical proof of concept that a combination of ASO-directed DDX5 inhibition with a DNA damage-inducing therapy can serve as a highly potential novel strategy to treat CRPC.
Collapse
Affiliation(s)
- Thi Khanh Le
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, 13273 Marseille, France; Department of Life Science, University of Science and Technology of Hanoi, Hanoi 000084, Vietnam
| | - Chaïma Cherif
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, 13273 Marseille, France
| | - Kenneth Omabe
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, 13273 Marseille, France
| | - Clément Paris
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, 13273 Marseille, France
| | - François Lannes
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, 13273 Marseille, France; Urology Deparment, AP-HM Hospital Nord, Aix-Marseille University, 13915 Marseille Cedex 20, France
| | - Stéphane Audebert
- Marseille Protéomique, Centre de Recherche en Cancérologie de Marseille, INSERM, CNRS, Institut Paoli-Calmettes, Aix-Marseille University, 13009 Marseille, France
| | - Emilie Baudelet
- Marseille Protéomique, Centre de Recherche en Cancérologie de Marseille, INSERM, CNRS, Institut Paoli-Calmettes, Aix-Marseille University, 13009 Marseille, France
| | - Mourad Hamimed
- Inria - Inserm team COMPO, COMPutational pharmacology and clinical Oncology, Centre Inria Sophia Antipolis - Méditerranée, Centre de Recherches en Cancérologie de Marseille, Inserm U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Boulevard Jean Moulin, 13005 Marseille, France
| | - Dominique Barbolosi
- Inria - Inserm team COMPO, COMPutational pharmacology and clinical Oncology, Centre Inria Sophia Antipolis - Méditerranée, Centre de Recherches en Cancérologie de Marseille, Inserm U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Boulevard Jean Moulin, 13005 Marseille, France
| | - Pascal Finetti
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, 13273 Marseille, France
| | - Cyrille Bastide
- Urology Deparment, AP-HM Hospital Nord, Aix-Marseille University, 13915 Marseille Cedex 20, France
| | - Ladan Fazli
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Martin Gleave
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - François Bertucci
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, 13273 Marseille, France
| | - David Taïeb
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, 13273 Marseille, France; La Timone University Hospital, Aix-Marseille University, 13005 Marseille, France; European Center for Research in Medical Imaging, Aix-Marseille University, 13005 Marseille, France
| | - Palma Rocchi
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, 13273 Marseille, France; European Center for Research in Medical Imaging, Aix-Marseille University, 13005 Marseille, France.
| |
Collapse
|
13
|
Gao J, Ma Y, Yang G, Li G. Translationally controlled tumor protein: the mediator promoting cancer invasion and migration and its potential clinical prospects. J Zhejiang Univ Sci B 2022; 23:642-654. [PMID: 35953758 DOI: 10.1631/jzus.b2100910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Translationally controlled tumor protein (TCTP) is a highly conserved multifunctional protein localized in the cytoplasm and nucleus of eukaryotic cells. It is secreted through exosomes and its degradation is associated with the ubiquitin-proteasome system (UPS), heat shock protein 27 (Hsp27), and chaperone-mediated autophagy (CMA). Its structure contains three α-helices and eleven β-strands, and features a helical hairpin as its hallmark. TCTP shows a remarkable similarity to the methionine-R-sulfoxide reductase B (MsrB) and mammalian suppressor of Sec4 (Mss4/Dss4) protein families, which exerts guanine nucleotide exchange factor (GEF) activity on small guanosine triphosphatase (GTPase) proteins, suggesting that some functions of TCTP may at least depend on its GEF action. Indeed, TCTP exerts GEF activity on Ras homolog enriched in brain (Rheb) to boost the growth and proliferation of Drosophila cells. TCTP also enhances the expression of cell division control protein 42 homolog (Cdc42) to promote cancer cell invasion and migration. Moreover, TCTP regulates cytoskeleton organization by interacting with actin microfilament (MF) and microtubule (MT) proteins and inducing the epithelial-mesenchymal transition (EMT) process. In essence, TCTP promotes cancer cell movement. It is usually highly expressed in cancerous tissues and thus reduces patient survival; meanwhile, drugs can target TCTP to reduce this effect. In this review, we summarize the mechanisms of TCTP in promoting cancer invasion and migration, and describe the current inhibitory strategy to target TCTP in cancerous diseases.
Collapse
Affiliation(s)
- Junying Gao
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Yan Ma
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Guiwen Yang
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life Sciences, Shandong Normal University, Jinan 250014, China.
| | - Guorong Li
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life Sciences, Shandong Normal University, Jinan 250014, China. ,
| |
Collapse
|
14
|
Bonhoure A, Henry L, Bich C, Blanc L, Bergeret B, Bousquet M, Coux O, Stoebner P, Vidal M. Extracellular
20S
proteasome secreted via microvesicles can degrade poorly folded proteins and inhibit Galectin‐3 agglutination activity. Traffic 2022; 23:287-304. [DOI: 10.1111/tra.12840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Anne Bonhoure
- Laboratory of Pathogen Host Interactions Université Montpellier, CNRS Montpellier France
| | - Laurent Henry
- Institut des Biomolécules Max Mousseron Université Montpellier, CNRS Montpellier France
| | - Claudia Bich
- Institut des Biomolécules Max Mousseron Université Montpellier, CNRS Montpellier France
| | - Lionel Blanc
- The Feinstein Institutes for Medical Research Manhasset New York USA
| | - Blanche Bergeret
- Institut des Biomolécules Max Mousseron Université Montpellier, CNRS Montpellier France
| | - Marie‐Pierre Bousquet
- Institut de Pharmacologie et de Biologie Structurale Université Toulouse, CNRS, UPS Toulouse France
| | - Olivier Coux
- Centre de Recherche en Biologie cellulaire de Montpellier Univ. Montpellier, CNRS Montpellier France
| | - Pierre‐Emmanuel Stoebner
- Service de Dermatologie, CHU Nîmes Nîmes France
- Institut de Recherche en Cancérologie de Montpellier (IRCM) Université Montpellier Montpellier France
| | - Michel Vidal
- Laboratory of Pathogen Host Interactions Université Montpellier, CNRS Montpellier France
| |
Collapse
|
15
|
Lu C, Li Z, Zhang W, Guo H, Lan W, Shen G, Xia Q, Zhao P. SUMOylation of Translationally Regulated Tumor Protein Modulates Its Immune Function. Front Immunol 2022; 13:807097. [PMID: 35197979 PMCID: PMC8858932 DOI: 10.3389/fimmu.2022.807097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/13/2022] [Indexed: 11/13/2022] Open
Abstract
Translationally controlled tumor protein (TCTP) is a highly conserved protein possessing numerous biological functions and molecular interactions, ranging from cell growth to immune responses. However, the molecular mechanism by which TCTP regulates immune function is largely unknown. Here, we found that knockdown of Bombyx mori translationally controlled tumor protein (BmTCTP) led to the increased susceptibility of silkworm cells to virus infection, whereas overexpression of BmTCTP significantly decreased the virus replication. We further demonstrated that BmTCTP could be modified by SUMOylation molecular BmSMT3 at the lysine 164 via the conjugating enzyme BmUBC9, and the stable SUMOylation of BmTCTP by expressing BmTCTP-BmSMT3 fusion protein exhibited strong antiviral activity, which confirmed that the SUMOylation of BmTCTP would contribute to its immune responses. Further work indicated that BmTCTP is able to physically interact with interleukin enhancer binding factor (ILF), one immune molecular, involved in antivirus, and also induce the expression of BmILF in response to virus infection, which in turn enhanced antiviral activity of BmTCTP. Altogether, our present study has provided a novel insight into defending against virus via BmTCTP SUMOylation signaling pathway and interacting with key immune molecular in silkworm.
Collapse
Affiliation(s)
- Chenchen Lu
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, China
| | - Zhiqing Li
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, China
- *Correspondence: Zhiqing Li,
| | - Wenchang Zhang
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, China
| | - Hao Guo
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, China
| | - Weiqun Lan
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, China
| | - Guanwang Shen
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, China
| | - Qingyou Xia
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, China
| | - Ping Zhao
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, China
| |
Collapse
|
16
|
Cherif C, Nguyen DT, Paris C, Le TK, Sefiane T, Carbuccia N, Finetti P, Chaffanet M, Kaoutari AE, Vernerey J, Fazli L, Gleave M, Manai M, Barthélémy P, Birnbaum D, Bertucci F, Taïeb D, Rocchi P. Menin inhibition suppresses castration-resistant prostate cancer and enhances chemosensitivity. Oncogene 2021; 41:125-137. [PMID: 34711954 PMCID: PMC8724010 DOI: 10.1038/s41388-021-02039-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 09/09/2021] [Accepted: 09/27/2021] [Indexed: 11/09/2022]
Abstract
Disease progression and therapeutic resistance of prostate cancer (PC) are linked to multiple molecular events that promote survival and plasticity. We previously showed that heat shock protein 27 (HSP27) acted as a driver of castration-resistant phenotype (CRPC) and developed an oligonucleotides antisense (ASO) against HSP27 with evidence of anti-cancer activity in men with CRPC. Here, we show that the tumor suppressor Menin (MEN1) is highly regulated by HSP27. Menin is overexpressed in high-grade PC and CRPC. High MEN1 mRNA expression is associated with decreased biochemical relapse-free and overall survival. Silencing Menin with ASO technology inhibits CRPC cell proliferation, tumor growth, and restores chemotherapeutic sensitivity. ChIP-seq analysis revealed differential DNA binding sites of Menin in various prostatic cells, suggesting a switch from tumor suppressor to oncogenic functions in CRPC. These data support the evaluation of ASO against Menin for CRPC. ![]()
Collapse
Affiliation(s)
- Chaïma Cherif
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France.,Laboratory of Biochemistry and Molecular Biology, Science University of Tunis, 2092, El Manar, Tunis, Tunisia
| | - Dang Tan Nguyen
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Clément Paris
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Thi Khanh Le
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Thibaud Sefiane
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Nadine Carbuccia
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Pascal Finetti
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Max Chaffanet
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Abdessamad El Kaoutari
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Julien Vernerey
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Ladan Fazli
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Martin Gleave
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Mohamed Manai
- Laboratory of Biochemistry and Molecular Biology, Science University of Tunis, 2092, El Manar, Tunis, Tunisia
| | - Philippe Barthélémy
- ARNA Laboratory, INSERM U1212, CNRS UMR 5320, University of Bordeaux, F-33076 Bordeaux, France
| | - Daniel Birnbaum
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - François Bertucci
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - David Taïeb
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France.,Biophysics and Nuclear Medicine Department, La Timone University Hospital, European Center for Research in Medical Imaging, Aix-Marseille University, F-13005 Marseille, France
| | - Palma Rocchi
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France.
| |
Collapse
|
17
|
Jeong M, Jeong MH, Kim JE, Cho S, Lee KJ, Park S, Sohn J, Park YG. TCTP protein degradation by targeting mTORC1 and signaling through S6K, Akt, and Plk1 sensitizes lung cancer cells to DNA-damaging drugs. Sci Rep 2021; 11:20812. [PMID: 34675258 PMCID: PMC8531033 DOI: 10.1038/s41598-021-00247-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 10/08/2021] [Indexed: 11/14/2022] Open
Abstract
Translationally controlled tumor protein (TCTP) is expressed in many tissues, particularly in human tumors. It plays a role in malignant transformation, apoptosis prevention, and DNA damage repair. The signaling mechanisms underlying TCTP regulation in cancer are only partially understood. Here, we investigated the role of mTORC1 in regulating TCTP protein levels, thereby modulating chemosensitivity, in human lung cancer cells and an A549 lung cancer xenograft model. The inhibition of mTORC1, but not mTORC2, induced ubiquitin/proteasome-dependent TCTP degradation without a decrease in the mRNA level. PLK1 activity was required for TCTP ubiquitination and degradation and for its phosphorylation at Ser46 upon mTORC1 inhibition. Akt phosphorylation and activation was indispensable for rapamycin-induced TCTP degradation and PLK1 activation, and depended on S6K inhibition, but not mTORC2 activation. Furthermore, the minimal dose of rapamycin required to induce TCTP proteolysis enhanced the efficacy of DNA-damaging drugs, such as cisplatin and doxorubicin, through the induction of apoptotic cell death in vitro and in vivo. This synergistic cytotoxicity of these drugs was induced irrespective of the functional status of p53. These results demonstrate a new mechanism of TCTP regulation in which the mTORC1/S6K pathway inhibits a novel Akt/PLK1 signaling axis and thereby induces TCTP protein stabilization and confers resistance to DNA-damaging agents. The results of this study suggest a new therapeutic strategy for enhancing chemosensitivity in lung cancers regardless of the functional status of p53.
Collapse
Affiliation(s)
- Mini Jeong
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mi Hyeon Jeong
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jung Eun Kim
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Serin Cho
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyoung Jin Lee
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Serkin Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jeongwon Sohn
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yun Gyu Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea.
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
18
|
Malard F, Jacquet E, Nhiri N, Sizun C, Chabrier A, Messaoudi S, Dejeu J, Betzi S, Zhang X, Thureau A, Lescop E. Revisiting the Molecular Interactions between the Tumor Protein TCTP and the Drugs Sertraline/Thioridazine. ChemMedChem 2021; 17:e202100528. [PMID: 34472703 DOI: 10.1002/cmdc.202100528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Indexed: 11/07/2022]
Abstract
TCTP protein is a pharmacological target in cancer and TCTP inhibitors such as sertraline have been evaluated in clinical trials. The direct interaction of TCTP with the drugs sertraline and thioridazine has been reported in vitro by SPR experiments to be in the ∼30-50 μM Kd range (Amson et al. Nature Med 2012), supporting a TCTP-dependent mode of action of the drugs on tumor cells. However, the molecular details of the interaction remain elusive although they are crucial to improve the efforts of on-going medicinal chemistry. In addition, TCTP can be phosphorylated by the Plk-1 kinase, which is indicative of poor prognosis in several cancers. The impact of phosphorylation on TCTP structure/dynamics and binding with therapeutical ligands remains unexplored. Here, we combined NMR, TSA, SPR, BLI and ITC techniques to probe the molecular interactions between TCTP with the drugs sertraline and thioridazine. We reveal that drug binding is much weaker than reported with an apparent ∼mM Kd and leads to protein destabilization that obscured the analysis of the published SPR data. We further demonstrate by NMR and SAXS that TCTP S46 phosphorylation does not promote tighter interaction between TCTP and sertraline. Accordingly, we question the supported model in which sertraline and thioridazine directly interact with isolated TCTP in tumor cells and discuss alternative modes of action for the drugs in light of current literature.
Collapse
Affiliation(s)
- Florian Malard
- Institut de Chimie des Substances Naturelles, CNRS, Université Paris-Saclay, 1 av. de la terrasse, 91198, Gif-sur-Yvette, France
| | - Eric Jacquet
- Institut de Chimie des Substances Naturelles, CNRS, Université Paris-Saclay, 1 av. de la terrasse, 91198, Gif-sur-Yvette, France
| | - Naima Nhiri
- Institut de Chimie des Substances Naturelles, CNRS, Université Paris-Saclay, 1 av. de la terrasse, 91198, Gif-sur-Yvette, France
| | - Christina Sizun
- Institut de Chimie des Substances Naturelles, CNRS, Université Paris-Saclay, 1 av. de la terrasse, 91198, Gif-sur-Yvette, France
| | - Amélie Chabrier
- Université Paris-Saclay, BioCIS, Faculté de Pharmacie, CNRS, 92290, Châtenay-Malabry, France
| | - Samir Messaoudi
- Université Paris-Saclay, BioCIS, Faculté de Pharmacie, CNRS, 92290, Châtenay-Malabry, France
| | - Jérôme Dejeu
- Univ. Grenoble Alpes, CNRS, DCM, 38000, Grenoble, France
| | - Stéphane Betzi
- Centre de Recherche en Cancérologie de Marseille (CRCM), CNRS, Aix-Marseille Université, Inserm, Institut Paoli-Calmettes, 27 bd Lei Roure, 13273, Marseille CEDEX 9, France
| | - Xu Zhang
- Centre de Recherche en Cancérologie de Marseille (CRCM), CNRS, Aix-Marseille Université, Inserm, Institut Paoli-Calmettes, 27 bd Lei Roure, 13273, Marseille CEDEX 9, France
| | | | - Ewen Lescop
- Institut de Chimie des Substances Naturelles, CNRS, Université Paris-Saclay, 1 av. de la terrasse, 91198, Gif-sur-Yvette, France
| |
Collapse
|
19
|
Topoisomerase II is regulated by translationally controlled tumor protein for cell survival during organ growth in Drosophila. Cell Death Dis 2021; 12:811. [PMID: 34453033 PMCID: PMC8397738 DOI: 10.1038/s41419-021-04091-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 07/28/2021] [Accepted: 08/12/2021] [Indexed: 11/08/2022]
Abstract
Regulation of cell survival is critical for organ development. Translationally controlled tumor protein (TCTP) is a conserved protein family implicated in the control of cell survival during normal development and tumorigenesis. Previously, we have identified a human Topoisomerase II (TOP2) as a TCTP partner, but its role in vivo has been unknown. To determine the significance of this interaction, we examined their roles in developing Drosophila organs. Top2 RNAi in the wing disc leads to tissue reduction and caspase activation, indicating the essential role of Top2 for cell survival. Top2 RNAi in the eye disc also causes loss of eye and head tissues. Tctp RNAi enhances the phenotypes of Top2 RNAi. The depletion of Tctp reduces Top2 levels in the wing disc and vice versa. Wing size is reduced by Top2 overexpression, implying that proper regulation of Top2 level is important for normal organ development. The wing phenotype of Tctp RNAi is partially suppressed by Top2 overexpression. This study suggests that mutual regulation of Tctp and Top2 protein levels is critical for cell survival during organ development.
Collapse
|
20
|
Nucleoside-Lipid-Based Nanoparticles for Phenazine Delivery: A New Therapeutic Strategy to Disrupt Hsp27-eIF4E Interaction in Castration Resistant Prostate Cancer. Pharmaceutics 2021; 13:pharmaceutics13050623. [PMID: 33925528 PMCID: PMC8146835 DOI: 10.3390/pharmaceutics13050623] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 11/25/2022] Open
Abstract
Heat shock protein 27 (Hsp27) has an established role in tumor progression and chemo-resistance of castration-resistant prostate cancer (CRPC). Hsp27 protects eukaryotic translation initiation factor 4E (eIF4E) from degradation, thereby maintaining survival during treatment. Phenazine derivative compound #14 was demonstrated to specifically disrupt Hsp27/eIF4E interaction and significantly delay castration-resistant tumor progression in prostate cancer xenografts. In the present work, various strategies of encapsulation of phenazine #14 with either DOTAU (N-[5′-(2′,3′-dioleoyl)uridine]-N′,N′,N′-trimethylammonium tosylate) and DOU-PEG2000 (5′-PEG2000-2′,3′-dioleoyluridine) nucleolipids (NLs) were developed in order to improve its solubilization, biological activity, and bioavailability. We observed that NLs-encapsulated phenazine #14-driven Hsp27-eIF4E interaction disruption increased cytotoxic effects on castration-resistant prostate cancer cell line and inhibited tumor growth in castration-resistant prostate cancer cell xenografted mice compared to phenazine #14 and NLs alone. Phenazine #14 NL encapsulation might represent an interesting nanostrategy for CRPC therapy.
Collapse
|
21
|
Omabe K, Paris C, Lannes F, Taïeb D, Rocchi P. Nanovectorization of Prostate Cancer Treatment Strategies: A New Approach to Improved Outcomes. Pharmaceutics 2021; 13:591. [PMID: 33919150 PMCID: PMC8143094 DOI: 10.3390/pharmaceutics13050591] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer (PC) is the most frequent male cancer in the Western world. Progression to Castration Resistant Prostate Cancer (CRPC) is a known consequence of androgen withdrawal therapy, making CRPC an end-stage disease. Combination of cytotoxic drugs and hormonal therapy/or genotherapy is a recognized modality for the treatment of advanced PC. However, this strategy is limited by poor bio-accessibility of the chemotherapy to tumor sites, resulting in an increased rate of collateral toxicity and incidence of multidrug resistance (MDR). Nanovectorization of these strategies has evolved to an effective approach to efficacious therapeutic outcomes. It offers the possibility to consolidate their antitumor activity through enhanced specific and less toxic active or passive targeting mechanisms, as well as enabling diagnostic imaging through theranostics. While studies on nanomedicine are common in other cancer types, only a few have focused on prostate cancer. This review provides an in-depth knowledge of the principles of nanotherapeutics and nanotheranostics, and how the application of this rapidly evolving technology can clinically impact CRPC treatment. With particular reference to respective nanovectors, we draw clinical and preclinical evidence, demonstrating the potentials and prospects of homing nanovectorization into CRPC treatment strategies.
Collapse
Affiliation(s)
- Kenneth Omabe
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
- Department of Biochemistry & Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike Ikwo, PMB 1010, Abakaliki 84001, Nigeria
| | - Clément Paris
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
| | - François Lannes
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
| | - David Taïeb
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
- Biophysics and Nuclear Medicine, La Timone University Hospital, European Center for Research in Medical Imaging, Aix-Marseille University, 13005 Marseille, France
| | - Palma Rocchi
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
| |
Collapse
|
22
|
A J, Zhang B, Zhang Z, Hu H, Dong JT. Novel Gene Signatures Predictive of Patient Recurrence-Free Survival and Castration Resistance in Prostate Cancer. Cancers (Basel) 2021; 13:cancers13040917. [PMID: 33671634 PMCID: PMC7927111 DOI: 10.3390/cancers13040917] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/10/2021] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Molecular signatures predictive of recurrence-free survival (RFS) and castration resistance are critical for treatment decision-making in prostate cancer (PCa), but the robustness of current signatures is limited. This study aims to identify castration-resistant PCa (CRPC)-associated genes and develop robust RFS and CRPC signatures. Among 287 genes differentially expressed between localized CRPC and hormone-sensitive PCa (HSPC) samples, 6 genes constituted a signature (CRPC-derived prognosis signature, CRPCPS) that predicted RFS. Moreover, a 3-gene panel derived from the 6 CRPCPS genes was capable of distinguishing CRPC from HSPC. The CRPCPS predicted RFS in 5/9 cohorts in the multivariate analysis and maintained prognostic in patients stratified by tumor stage, Gleason score, and lymph node metastasis status. It also predicted overall survival and metastasis-free survival. Notably, the signature was validated in another six independent cohorts. These findings suggest that these two signatures could be robust tools for predicting RFS and CRPC in clinical practice. Abstract Molecular signatures predictive of recurrence-free survival (RFS) and castration resistance are critical for treatment decision-making in prostate cancer (PCa), but the robustness of current signatures is limited. Here, we applied the Robust Rank Aggregation (RRA) method to PCa transcriptome profiles and identified 287 genes differentially expressed between localized castration-resistant PCa (CRPC) and hormone-sensitive PCa (HSPC). Least absolute shrinkage and selection operator (LASSO) and stepwise Cox regression analyses of the 287 genes developed a 6-gene signature predictive of RFS in PCa. This signature included NPEPL1, VWF, LMO7, ALDH2, NUAK1, and TPT1, and was named CRPC-derived prognosis signature (CRPCPS). Interestingly, three of these 6 genes constituted another signature capable of distinguishing CRPC from HSPC. The CRPCPS predicted RFS in 5/9 cohorts in the multivariate analysis and remained valid in patients stratified by tumor stage, Gleason score, and lymph node status. The signature also predicted overall survival and metastasis-free survival. The signature’s robustness was demonstrated by the C-index (0.55–0.74) and the calibration plot in all nine cohorts and the 3-, 5-, and 8-year area under the receiver operating characteristic curve (0.67–0.77) in three cohorts. The nomogram analyses demonstrated CRPCPS’ clinical applicability. The CRPCPS thus appears useful for RFS prediction in PCa.
Collapse
Affiliation(s)
- Jun A
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China;
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China;
| | - Baotong Zhang
- Emory Winship Cancer Institute, Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365-C Clifton Road, Atlanta, GA 30322, USA;
| | - Zhiqian Zhang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China;
| | - Hailiang Hu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China;
| | - Jin-Tang Dong
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China;
- Correspondence:
| |
Collapse
|
23
|
Kumar R, Saran S. Comparative modelling unravels the structural features of eukaryotic TCTP implicated in its multifunctional properties: an in silico approach. J Mol Model 2021; 27:20. [PMID: 33410974 DOI: 10.1007/s00894-020-04630-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 12/01/2020] [Indexed: 10/22/2022]
Abstract
Comparative modelling helps compare the structure and functions of a given protein, to track the path of its origin and evolution and also guide in structure-based drug discovery. Presently, this has been applied for modelling the tertiary structure of highly conserved eukaryotic TCTP (translationally controlled tumour protein) which is involved in a plethora of functions during growth and development and also acts as a biomarker for many cancers like lung, breast, and prostate cancer. The modelled TCTP structures of different organisms belonging to the eukaryotic group showed similar spatial arrangement of structural units except loops and similar patterns of root mean square deviation (RMSD), root mean square fluctuation, and radius of gyration (Rg) inspected through molecular dynamics simulations. Essential dynamics (ED) analyses revealed different domains that exhibited different motions for the assistance in its multifunctional properties. Construction of a free-energy landscape (FEL) based on Rg versus RMSD was employed to characterize the folding behaviours of structures and observe that all proteins had nearly similar conformation and topologies, indicating common thermodynamic/kinetic pathways. A physico-chemical interaction study demonstrated the helices and sheets were well stabilized with ample amounts of bonding compared to turns or loops and charged residues were more accessible to solvent molecules. Hence, the current study reveals the important structural features of TCTP that aid in diverse functions in a wide range of organisms, thus extending our knowledge of TCTP and also providing a venue for designing the potent inhibitors against it.
Collapse
Affiliation(s)
- Rakesh Kumar
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Shweta Saran
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
24
|
Hawner M, Ducho C. Cellular Targeting of Oligonucleotides by Conjugation with Small Molecules. Molecules 2020; 25:E5963. [PMID: 33339365 PMCID: PMC7766908 DOI: 10.3390/molecules25245963] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 12/20/2022] Open
Abstract
Drug candidates derived from oligonucleotides (ON) are receiving increased attention that is supported by the clinical approval of several ON drugs. Such therapeutic ON are designed to alter the expression levels of specific disease-related proteins, e.g., by displaying antigene, antisense, and RNA interference mechanisms. However, the high polarity of the polyanionic ON and their relatively rapid nuclease-mediated cleavage represent two major pharmacokinetic hurdles for their application in vivo. This has led to a range of non-natural modifications of ON structures that are routinely applied in the design of therapeutic ON. The polyanionic architecture of ON often hampers their penetration of target cells or tissues, and ON usually show no inherent specificity for certain cell types. These limitations can be overcome by conjugation of ON with molecular entities mediating cellular 'targeting', i.e., enhanced accumulation at and/or penetration of a specific cell type. In this context, the use of small molecules as targeting units appears particularly attractive and promising. This review provides an overview of advances in the emerging field of cellular targeting of ON via their conjugation with small-molecule targeting structures.
Collapse
Affiliation(s)
| | - Christian Ducho
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2 3, 66 123 Saarbrücken, Germany;
| |
Collapse
|
25
|
Enhanced Antisense Oligonucleotide Delivery Using Cationic Liposomes Grafted with Trastuzumab: A Proof-of-Concept Study in Prostate Cancer. Pharmaceutics 2020; 12:pharmaceutics12121166. [PMID: 33260460 PMCID: PMC7761013 DOI: 10.3390/pharmaceutics12121166] [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: 11/13/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 12/31/2022] Open
Abstract
Prostate cancer (PCa) is the second most common cancer in men worldwide and the fifth leading cause of death by cancer. The overexpression of TCTP protein plays an important role in castration resistance. Over the last decade, antisense technology has emerged as a rising strategy in oncology. Using antisense oligonucleotide (ASO) to silence TCTP protein is a promising therapeutic option—however, the pharmacokinetics of ASO does not always meet the requirements of proper delivery to the tumor site. In this context, developing drug delivery systems is an attractive strategy for improving the efficacy of ASO directed against TCTP. The liposome should protect and deliver ASO at the intracellular level in order to be effective. In addition, because prostate cancer cells express Her2, using an anti-Her2 targeting antibody will increase the affinity of the liposome for the cell and optimize the intratumoral penetration of the ASO, thus improving efficacy. Here, we have designed and developed pegylated liposomes and Her2-targeting immunoliposomes. Mean diameter was below 200 nm, thus ensuring proper enhanced permeation and retention (EPR) effect. Encapsulation rate for ASO was about 40%. Using human PC-3 prostate cancer cells as a canonical model, free ASO and ASO encapsulated into either liposomes or anti-Her2 immunoliposomes were tested for efficacy in vitro using 2D and 3D spheroid models. While the encapsulated forms of ASO were always more effective than free ASO, we observed differences in efficacy of encapsulated ASO. For short exposure times (i.e., 4 h) ASO liposomes (ASO-Li) were more effective than ASO-immunoliposomes (ASO-iLi). Conversely, for longer exposure times, ASO-iLi performed better than ASO-Li. This pilot study demonstrates that it is possible to encapsulate ASO into liposomes and to yield antiproliferative efficacy against PCa. Importantly, despite mild Her2 expression in this PC-3 model, using a surface mAb as targeting agent provides further efficacy, especially when exposure is longer. Overall, the development of third-generation ASO-iLi should help to take advantage of the expression of Her2 by prostate cancer cells in order to allow greater specificity of action in vivo and thus a gain in efficacy.
Collapse
|
26
|
Samy ALPA, Bakthavachalam V, Vudutha M, Vinjamuri S, Chinnapaka S, Munirathinam G. Eprinomectin, a novel semi-synthetic macrocylic lactone is cytotoxic to PC3 metastatic prostate cancer cells via inducing apoptosis. Toxicol Appl Pharmacol 2020; 401:115071. [PMID: 32454055 PMCID: PMC7716802 DOI: 10.1016/j.taap.2020.115071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/16/2020] [Accepted: 05/21/2020] [Indexed: 12/25/2022]
Abstract
Prostate Cancer (PCa) is the second most common cancer among men in United States after skin cancer. Conventional chemotherapeutic drugs available for PCa treatment are limited due to toxicity and resistance issues. Therefore, there is an urgent need to develop more effective treatment for advanced PCa. In this current study, we focused on evaluating the anti-cancer efficacy of Eprinomectin (EP), a novel avermectin analog against PC3 metastatic PCa cells. EP displayed robust inhibition of cell viability of PC3 cells in addition to suppressing the colony formation and wound healing capabilities. Our study showed that EP targets PC3 cells via inducing ROS and apoptosis activation. EP treatment enforces cell cycle arrest at G0/G1 phase via targeting cyclin-dependent kinase 4 (CDK4) and subsequent induction of apoptosis in PC3 cells. At the molecular level, EP effectively inhibited the expression of various cancer stem cell markers such as ALDH1, Sox-2, Nanog, Oct3/4 and CD44. Interestingly, EP also inhibited the activity of alkaline phosphatase, a maker of pluripotent stem cells. Of note, EP treatment resulted in the translocation of β-catenin from the nucleus to the cytoplasm indicating that EP antagonizes Wnt/β-catenin signaling pathway. Western blotting analysis revealed that EP downregulated the expression of key cell cycle markers such as cyclin D1, cyclin D3, CDK4, and c-Myc. In addition, EP inhibited the anti-apoptotic markers such as Mcl-1, XIAP, c-IAP1 and survivin in PC3 cells. On the other hand, EP treatment resulted in the activation of pH2A.X, Bad, caspase-9, caspase-3 and cleavage of PARP1. Taken together, our data suggests that EP is a potential agent to treat advanced PCa cells via modulating apoptosis signaling.
Collapse
Affiliation(s)
| | - Velavan Bakthavachalam
- Department of Biomedical Sciences, University of Illinois, Rockford, IL, United States of America
| | - Mona Vudutha
- Department of Biomedical Sciences, University of Illinois, Rockford, IL, United States of America
| | - Smita Vinjamuri
- Department of Biomedical Sciences, University of Illinois, Rockford, IL, United States of America
| | - Somaiah Chinnapaka
- Department of Biomedical Sciences, University of Illinois, Rockford, IL, United States of America
| | - Gnanasekar Munirathinam
- Department of Biomedical Sciences, University of Illinois, Rockford, IL, United States of America.
| |
Collapse
|
27
|
Dysregulation of TCTP in Biological Processes and Diseases. Cells 2020; 9:cells9071632. [PMID: 32645936 PMCID: PMC7407922 DOI: 10.3390/cells9071632] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022] Open
Abstract
Translationally controlled tumor protein (TCTP), also called histamine releasing factor (HRF) or fortilin, is a multifunctional protein present in almost all eukaryotic organisms. TCTP is involved in a range of basic cell biological processes, such as promotion of growth and development, or cellular defense in response to biological stresses. Cellular TCTP levels are highly regulated in response to a variety of physiological signals, and regulatory mechanism at various levels have been elucidated. Given the importance of TCTP in maintaining cellular homeostasis, it is not surprising that dysregulation of this protein is associated with a range of disease processes. Here, we review recent progress that has been made in the characterisation of the basic biological functions of TCTP, in the description of mechanisms involved in regulating its cellular levels and in the understanding of dysregulation of TCTP, as it occurs in disease processes such as cancer.
Collapse
|
28
|
Chinnapaka S, Bakthavachalam V, Munirathinam G. Repurposing antidepressant sertraline as a pharmacological drug to target prostate cancer stem cells: dual activation of apoptosis and autophagy signaling by deregulating redox balance. Am J Cancer Res 2020; 10:2043-2065. [PMID: 32775000 PMCID: PMC7407340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 05/14/2020] [Indexed: 06/11/2023] Open
Abstract
Cancer stem cells play a major role in tumor initiation, progression, and tumor relapse of prostate cancer (PCa). Recent studies suggest that Translationally Controlled Tumor Protein (TCTP) is a critical survival factor of stem cells including cancer stem cells. Here, we aimed to determine whether the TCTP inhibitor sertraline (STL) could target prostate cancer stem cells (PCSC). In colony formation, spheroidogenesis, angiogenesis, and wound healing assays STL showed a robust inhibition of tumorigenic (colony growth), angiogenic (endothelial tube formation) and metastatic (wound healing and migration) potential of PCSC. Interestingly, antioxidants such as N-acetyl cysteine (NAC), Glutathione (GSH) and catalase effectively blocked the cytotoxicity effect of STL on PCSC implicating oxidative stress as the underlying anti-PCSC targeting mechanism. Cell cycle analysis showed a robust G0 arrest in PCSC exposed to STL. Notably, STL induced both apoptosis and autophagy by activating free radical generation, hydrogen peroxide formation (H2O2), lipid peroxidation (LPO) and depleted the levels of glutathione (GSH). Moreover, surface marker expression analysis using confocal revealed that STL significantly down regulates the expression levels of aldehyde dehydrogenase 1 (ALDH1) and cluster of differentiation 44 (CD44) stem cell markers. Furthermore, in western blot analysis, STL treatment applied in a dose-dependent manner, caused a marked decrease in TCTP, phospho TCTP, anti-apoptotic markers survivin and cellular inhibitor of apoptosis protein 1 (cIAP1) expression as well as a significant increase in cleaved caspase3 and cleaved Poly [ADP-ribose] polymerase 1 (PARP-1) expression. Of note, STL also significantly down regulated the stem cell markers (ALDH1 and CD44) and epithelial to mesenchymal transition (EMT) markers such as transcription factor 8 (TCF8) and lymphoid enhancer-binding factor-1 (LEF1) expression levels. Concurrently, STL increased the levels of autophagy markers such as light chain (LC3), Beclin1 and autophagy-related gene (ATG5). Taken together, our study suggests that STL could be an effective therapeutic agent in eliminating prostate cancer stem cells.
Collapse
Affiliation(s)
- Somaiah Chinnapaka
- Department of Biomedical Sciences, College of Medicine, University of Illinois Rockford, IL, USA
| | - Velavan Bakthavachalam
- Department of Biomedical Sciences, College of Medicine, University of Illinois Rockford, IL, USA
| | - Gnanasekar Munirathinam
- Department of Biomedical Sciences, College of Medicine, University of Illinois Rockford, IL, USA
| |
Collapse
|
29
|
Lee JS, Jang EH, Woo HA, Lee K. Regulation of Autophagy Is a Novel Tumorigenesis-Related Activity of Multifunctional Translationally Controlled Tumor Protein. Cells 2020; 9:cells9010257. [PMID: 31968668 PMCID: PMC7017196 DOI: 10.3390/cells9010257] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 12/13/2022] Open
Abstract
Translationally controlled tumor protein (TCTP) is highly conserved in eukaryotic organisms and plays multiple roles regulating cellular growth and homeostasis. Because of its anti-apoptotic activity and its role in the regulation of cancer metastasis, TCTP has become a promising target for cancer therapy. Moreover, growing evidence points to its clinical role in cancer prognosis. How TCTP regulates cellular growth in cancer has been widely studied, but how it regulates cellular homeostasis has received relatively little attention. This review discusses how TCTP is related to cancer and its potential as a target in cancer therapeutics, including its novel role in the regulation of autophagy. Regulation of autophagy is essential for cell recycling and scavenging cellular materials to sustain cell survival under the metabolic stress that cancer cells undergo during their aggressive proliferation.
Collapse
|
30
|
Hoter A, Rizk S, Naim HY. The Multiple Roles and Therapeutic Potential of Molecular Chaperones in Prostate Cancer. Cancers (Basel) 2019; 11:cancers11081194. [PMID: 31426412 PMCID: PMC6721600 DOI: 10.3390/cancers11081194] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer (PCa) is one of the most common cancer types in men worldwide. Heat shock proteins (HSPs) are molecular chaperones that are widely implicated in the pathogenesis, diagnosis, prognosis, and treatment of many cancers. The role of HSPs in PCa is complex and their expression has been linked to the progression and aggressiveness of the tumor. Prominent chaperones, including HSP90 and HSP70, are involved in the folding and trafficking of critical cancer-related proteins. Other members of HSPs, including HSP27 and HSP60, have been considered as promising biomarkers, similar to prostate-specific membrane antigen (PSMA), for PCa screening in order to evaluate and monitor the progression or recurrence of the disease. Moreover, expression level of chaperones like clusterin has been shown to correlate directly with the prostate tumor grade. Hence, targeting HSPs in PCa has been suggested as a promising strategy for cancer therapy. In the current review, we discuss the functions as well as the role of HSPs in PCa progression and further evaluate the approach of inhibiting HSPs as a cancer treatment strategy.
Collapse
Affiliation(s)
- Abdullah Hoter
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Sandra Rizk
- School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Hassan Y Naim
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany.
| |
Collapse
|
31
|
Abstract
This chapter focuses on published studies specifically concerning TCTP and its involvement in degradation or stabilization of various proteins, and also in its own degradation in different ways. The first part relates to the inhibition of proteasomal degradation of proteins. This can be achieved by masking ubiquitination sites of specific partners, by favoring ubiquitin E3 ligase degradation, or by regulating proteasome activity. The second part addresses the ability of TCTP to favor degradation of specific proteins through proteasome or macroautophagic pathways. The third part discusses about the different ways by which TCTP has been shown to be degraded.
Collapse
|
32
|
Sun R, Lu X, Gong L, Jin F. TCTP promotes epithelial-mesenchymal transition in lung adenocarcinoma. Onco Targets Ther 2019; 12:1641-1653. [PMID: 30881019 PMCID: PMC6398409 DOI: 10.2147/ott.s184555] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Lung cancer is the most common and lethal malignancy worldwide. TCTP is highly expressed in various cancers including lung cancer. Epithelial–mesenchymal transition (EMT) could increase cancer cell invasion. Whether TCTP’s expression is associated with EMT in lung adenocarcinoma is largely unknown. Methods Several Gene Expression Omnibus datasets were used to analyze the correlation between TCTP expression and overall survival of lung adenocarcinoma patients by Kaplan–Meier survival analysis. Then, 24 surgically removed fresh lung adenocarcinoma tissue samples and paired paracancer tissue samples were used to analyze the correlation between TCTP expression and tumor stage by immunohistochemical analysis. Furthermore, stable cell lines were generated using lentiviral transduction systems to knock down or overexpress TCTP in A549 cells. Cell migration and invasion were measured by scratch and transwell assays, and EMT marker proteins such as α-SMA, ZEB1, and E-cadherin were quantitated by Western blot. The expression levels of miR-200a, miR-141, and miR-429 were determined by real-time quantitative PCR, and their target genes were predicted by an online database miRTarBase. The interaction between TCTP and these genes was analyzed by String database and visualized by Cytoscape. Results TCTP was highly expressed in tumor tissues compared to paracancer tissues. The expression of TCTP was associated with shorter overall survival. TCTP knockdown experiment in A549 cells suggested that TCTP knockdown could decrease the migration and invasion of lung cancer cells, and the expression level of ZEB1 and α-SMA, but increase the expression of E-cadherin and p53. Vice versa, overexpression of TCTP could increase the migration and invasion of cancer cells, and the expression level of ZEB1 and α-SMA, but decrease the expression of E-cadherin and p53. Furthermore, we found the expression of miR-200a, miR-141, and miR-429 was associated with TCTP expression. Conclusion TCTP promotes EMT in lung adenocarcinoma, and this effect may be associated with miR-200 family members like miR-200a, miR-141, and miR-429.
Collapse
Affiliation(s)
- Ruilin Sun
- Department of Respiratory Medicine, The Second Affiliated Hospital of Air Force Medical University, Xi'an, People's Republic of China,
| | - Xi Lu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Air Force Medical University, Xi'an, People's Republic of China,
| | - Li Gong
- Department of Pathology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, People's Republic of China
| | - Faguang Jin
- Department of Respiratory Medicine, The Second Affiliated Hospital of Air Force Medical University, Xi'an, People's Republic of China,
| |
Collapse
|
33
|
Benizri S, Gissot A, Martin A, Vialet B, Grinstaff MW, Barthélémy P. Bioconjugated Oligonucleotides: Recent Developments and Therapeutic Applications. Bioconjug Chem 2019; 30:366-383. [PMID: 30608140 PMCID: PMC6766081 DOI: 10.1021/acs.bioconjchem.8b00761] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Oligonucleotide-based agents have the potential to treat or cure almost any disease, and are one of the key therapeutic drug classes of the future. Bioconjugated oligonucleotides, a subset of this class, are emerging from basic research and being successfully translated to the clinic. In this Review, we first briefly describe two approaches for inhibiting specific genes using oligonucleotides-antisense DNA (ASO) and RNA interference (RNAi)-followed by a discussion on delivery to cells. We then summarize and analyze recent developments in bioconjugated oligonucleotides including those possessing GalNAc, cell penetrating peptides, α-tocopherol, aptamers, antibodies, cholesterol, squalene, fatty acids, or nucleolipids. These novel conjugates provide a means to enhance tissue targeting, cell internalization, endosomal escape, target binding specificity, resistance to nucleases, and more. We next describe those bioconjugated oligonucleotides approved for patient use or in clinical trials. Finally, we summarize the state of the field, describe current limitations, and discuss future prospects. Bioconjugation chemistry is at the centerpiece of this therapeutic oligonucleotide revolution, and significant opportunities exist for development of new modification chemistries, for mechanistic studies at the chemical-biology interface, and for translating such agents to the clinic.
Collapse
Affiliation(s)
- Sebastien Benizri
- Inserm U1212, F-33076 Bordeaux, France
- CNRS 5320, F-33076 Bordeaux, France
- Universitéde Bordeaux, 146 rue Léo Saignat, F-33076 Bordeaux Cedex, France
| | - Arnaud Gissot
- Inserm U1212, F-33076 Bordeaux, France
- CNRS 5320, F-33076 Bordeaux, France
- Universitéde Bordeaux, 146 rue Léo Saignat, F-33076 Bordeaux Cedex, France
| | - Andrew Martin
- Departments of Biomedical Engineering, Chemistry, and Medicine, Boston University, Boston, Massachusetts 02215, United States
| | - Brune Vialet
- Inserm U1212, F-33076 Bordeaux, France
- CNRS 5320, F-33076 Bordeaux, France
- Universitéde Bordeaux, 146 rue Léo Saignat, F-33076 Bordeaux Cedex, France
| | - Mark W. Grinstaff
- Departments of Biomedical Engineering, Chemistry, and Medicine, Boston University, Boston, Massachusetts 02215, United States
| | - Philippe Barthélémy
- Inserm U1212, F-33076 Bordeaux, France
- CNRS 5320, F-33076 Bordeaux, France
- Universitéde Bordeaux, 146 rue Léo Saignat, F-33076 Bordeaux Cedex, France
| |
Collapse
|
34
|
Neuhäuser K, Küper L, Christiansen H, Bogdanova N. Assessment of the role of translationally controlled tumor protein 1 (TPT1/TCTP) in breast cancer susceptibility and ATM signaling. Clin Transl Radiat Oncol 2019; 15:99-107. [PMID: 30815593 PMCID: PMC6378894 DOI: 10.1016/j.ctro.2019.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/19/2019] [Accepted: 01/23/2019] [Indexed: 01/08/2023] Open
Abstract
TPT1 sequencing identified one novel, potentially damaging mutation in 200 breast cancer patients. TPT1 is not required for the recognition of radiation-induced DNA damage. Phosphorylation of KAP1 and CHEK2 by ATM is not affected by silencing of TPT1. Nuclear localization and foci formation of TPT1 potentially depends on cell type. TPT1 knockdown might exert a marginally significant effect on residual γH2A.X foci.
Background and purpose The translationally controlled tumor protein 1 (TPT1/TCTP) has been implicated in the intracellular DNA damage response. We tested the role of TPT1 in breast cancer (BC) predisposition and re-evaluated its function in Ataxia-Telangiectasia mutated (ATM)-mediated damage recognition and DNA repair. Material and methods The TPT1 coding sequence was scanned for mutations in genomic DNA from 200 breast cancer patients. TPT1 was down-regulated through siRNA in breast epithelial and fibroblast cell cultures. ATM activation after irradiation (IR) was analyzed by western blotting, and γH2A.X foci were monitored by immunocytochemistry. Results The sequencing study identified a novel, potentially damaging missense mutation in a single patient. Silencing of TPT1 did not significantly affect ATM kinase activity and did not impair the initial formation of γH2A.X foci, while we observed a marginally significant effect on residual γH2A.X foci at 6–48 h after IR. Conclusions TPT1 does not harbor common mutations as BC susceptibility gene. Consistently, TPT1 protein is not required for the recognition of radiation-induced DNA damage via the ATM-dependent pathway and has only slight impact on timely repair. These results may be important when considering TPT1 as a DNA damage marker.
Collapse
Affiliation(s)
- Katharina Neuhäuser
- Radiation Oncology Research Unit, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Leonie Küper
- Radiation Oncology Research Unit, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany.,Gynaecology Research Unit, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Hans Christiansen
- Radiation Oncology Research Unit, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Natalia Bogdanova
- Radiation Oncology Research Unit, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| |
Collapse
|
35
|
Gouveia Roque C, Holt CE. Growth Cone Tctp Is Dynamically Regulated by Guidance Cues. Front Mol Neurosci 2018; 11:399. [PMID: 30459552 PMCID: PMC6232380 DOI: 10.3389/fnmol.2018.00399] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 10/12/2018] [Indexed: 12/19/2022] Open
Abstract
Translationally controlled tumor protein (Tctp) contributes to retinal circuitry formation by promoting axon growth and guidance, but it remains unknown to what extent axonal Tctp specifically influences axon development programs. Various genome-wide profiling studies have ranked tctp transcripts among the most enriched in the axonal compartment of distinct neuronal populations, including embryonic retinal ganglion cells (RGCs), suggesting its expression can be regulated locally and that this may be important during development. Here, we report that growth cone Tctp levels change rapidly in response to Netrin-1 and Ephrin-A1, two guidance cues encountered by navigating RGC growth cones. This regulation is opposite in effect, as we observed protein synthesis- and mTORC1-dependent increases in growth cone Tctp levels after acute treatment with Netrin-1, but a decline upon exposure to Ephrin-A1, an inhibitor of mTORC1. Live imaging with translation reporters further showed that Netrin-1-induced synthesis of Tctp in growth cones is driven by a short 3'untranslated region (3'UTR) tctp mRNA isoform. However, acute inhibition of de novo Tctp synthesis in axons did not perturb the advance of retinal projections through the optic tract in vivo, indicating that locally produced Tctp is not necessary for normal axon growth and guidance.
Collapse
Affiliation(s)
- Cláudio Gouveia Roque
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom.,Doctoral Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Christine E Holt
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
36
|
Some Biological Consequences of the Inhibition of Na,K-ATPase by Translationally Controlled Tumor Protein (TCTP). Int J Mol Sci 2018; 19:ijms19061657. [PMID: 29867020 PMCID: PMC6032315 DOI: 10.3390/ijms19061657] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 12/17/2022] Open
Abstract
Na,K-ATPase is an ionic pump that regulates the osmotic equilibrium and membrane potential of cells and also functions as a signal transducer. The interaction of Na,K-ATPase with translationally controlled tumor protein (TCTP) results, among others, in the inhibition of the former's pump activity and in the initiation of manifold biological and pathological phenomena. These phenomena include hypertension and cataract development in TCTP-overexpressing transgenic mice, as well as the induction of tumorigenesis signaling pathways and the activation of Src that ultimately leads to cell proliferation and migration. This review attempts to collate the biological effects of Na,K-ATPase and TCTP interaction and suggests that this interaction has the potential to serve as a possible therapeutic target for selected diseases.
Collapse
|
37
|
Wang K, Ruan H, Xu T, Liu L, Liu D, Yang H, Zhang X, Chen K. Recent advances on the progressive mechanism and therapy in castration-resistant prostate cancer. Onco Targets Ther 2018; 11:3167-3178. [PMID: 29881290 PMCID: PMC5983013 DOI: 10.2147/ott.s159777] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Background Although there have been great advances in mechanisms and therapeutic methods of prostate cancer, the mortality rate of prostate cancer remains high. The castration-resistant prostate cancer (CRPC), which develops from hormone-sensitive prostate cancer, foreshadows a more dismal outcome. Concomitant with the researches in the mechanism of CRPC and therapy for CRPC, more and more landmark progress has been made in recent years. Methods A number of clinical and experimental studies were reviewed to indicate the novel advancement in the progressive mechanism and therapy of CRPC. Results The androgen receptor (AR) is still a vital driver in the progression of CRPC, while other multiple mechanisms also contribute to this progression, such as tumor immunity, cancer stem cells, epithelial–mesenchymal transition and DNA repair disorder. In terms of the therapeutic methods of CRPC, chemotherapy with drugs, such as docetaxel, has been the first-line therapy for CRPC for many years. Besides, newer agents, which target some of the above mechanisms, show additional overall survival benefits for CRPC patients. These therapies include drugs targeting the androgen axis pathway (androgen synthesis, androgen receptor splice variants, coactivators of AR and so on), PI3K-AKT pathway, WNT pathway, DNA repair, rearrangement of ETS gene, novel chemotherapy and immunotherapy, bone metastasis therapy and so on. Understanding these novel findings on the mechanisms of CRPC and the latest potential CRPC therapies will direct us for further exploration of CRPC. Conclusion Through comprehensive consideration, the predominant mechanism of CRPC might be the AR signal axis concomitant with tumor microenvironment, stress, immunity, tumor microenvironment and so on. For CRPC therapy, targeting the AR axis pathway and chemotherapy are the first-line treatments at present. However, with the advancements in CRPC therapy made by the researchers, other novel potential methods will occupy more and more important position in the treatment of CRPC, especially the therapies targeting the tumor microenviroment, tumor immunity and DNA repair and so on.
Collapse
Affiliation(s)
- Keshan Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hailong Ruan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tianbo Xu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Di Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongmei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ke Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
38
|
Liang C, Xu Y, Ge H, Li G, Wu J. The clinicopathological and prognostic value of HSP27 in hepatocellular carcinoma: a systematic review and meta-analysis. Onco Targets Ther 2018; 11:1293-1303. [PMID: 29563808 PMCID: PMC5846765 DOI: 10.2147/ott.s154227] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background In the recent past, there is increasing evidence demonstrating that HSP27 plays a key role in tumor progression. However, the relationship between HSP27 expression and the clinicopathological features of hepatocellular carcinoma (HCC), as well as its prognostic value in HCC patients remain controversial. Accordingly, we conducted a meta-analysis to assess the correlation between HSP27 expression and HCC, and determine the prognostic value of HSP27 in HCC. Methods The data included clinicopathological features and survival information extracted from the published literature in the databases PubMed, EMBASE, Cochrane Library, Web of Science, CNKI, and Wan Fang. The pooled odds ratios and hazard ratios with 95% CIs were calculated using Forest plot analysis. Results The meta-analysis results indicated that the positive HSP27 expression was significantly correlated with HCC incidence, tumor differentiation, and α-fetoprotein level in patients with HCC. However, the expression of HSP27 was not associated with metastasis, hepatitis B virus surface antigen, gender, tumor size, TNM stage, and vascular invasion. Additionally, HSP27 expression indicated a poor overall survival rate, but it was not related to disease-free survival rate. Conclusion This meta-analysis revealed that HSP27 may play a key role in the development of HCC and could be a reliable biomarker for the prognosis of patients with HCC. However, additional high-quality research is needed to support the results.
Collapse
Affiliation(s)
- Chaojie Liang
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yingchen Xu
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hua Ge
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Guangming Li
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jixiang Wu
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
39
|
Goodman CA, Coenen AM, Frey JW, You JS, Barker RG, Frankish BP, Murphy RM, Hornberger TA. Insights into the role and regulation of TCTP in skeletal muscle. Oncotarget 2017; 8:18754-18772. [PMID: 27813490 PMCID: PMC5386645 DOI: 10.18632/oncotarget.13009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/28/2016] [Indexed: 01/07/2023] Open
Abstract
The translationally controlled tumor protein (TCTP) is upregulated in a range of cancer cell types, in part, by the activation of the mechanistic target of rapamycin (mTOR). Recently, TCTP has also been proposed to act as an indirect activator of mTOR. While it is known that mTOR plays a major role in the regulation of skeletal muscle mass, very little is known about the role and regulation of TCTP in this post-mitotic tissue. This study shows that muscle TCTP and mTOR signaling are upregulated in a range of mouse models (mdx mouse, mechanical load-induced hypertrophy, and denervation- and immobilization-induced atrophy). Furthermore, the increase in TCTP observed in the hypertrophic and atrophic conditions occurred, in part, via a rapamycin-sensitive mTOR-dependent mechanism. However, the overexpression of TCTP was not sufficient to activate mTOR signaling (or increase protein synthesis) and is thus unlikely to take part in a recently proposed positive feedback loop with mTOR. Nonetheless, TCTP overexpression was sufficient to induce muscle fiber hypertrophy. Finally, TCTP overexpression inhibited the promoter activity of the muscle-specific ubiquitin proteasome E3-ligase, MuRF1, suggesting that TCTP may play a role in inhibiting protein degradation. These findings provide novel data on the role and regulation of TCTP in skeletal muscle in vivo.
Collapse
Affiliation(s)
- Craig A Goodman
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA.,Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, Melbourne, Victoria, 8001, Australia.,Institute for Sport, Exercise and Active Living (ISEAL), Victoria University, Melbourne, Victoria, 8001, Australia
| | - Allison M Coenen
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
| | - John W Frey
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
| | - Jae-Sung You
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
| | - Robert G Barker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Barnaby P Frankish
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Robyn M Murphy
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Troy A Hornberger
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
| |
Collapse
|
40
|
Li Y, Sun H, Zhang C, Liu J, Zhang H, Fan F, Everley RA, Ning X, Sun Y, Hu J, Liu J, Zhang J, Ye W, Qiu X, Dai S, Liu B, Xu H, Fu S, Gygi SP, Zhou C. Identification of translationally controlled tumor protein in promotion of DNA homologous recombination repair in cancer cells by affinity proteomics. Oncogene 2017; 36:6839-6849. [PMID: 28846114 PMCID: PMC5735297 DOI: 10.1038/onc.2017.289] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 06/09/2017] [Accepted: 07/13/2017] [Indexed: 01/21/2023]
Abstract
Translationally controlled tumor protein(TCTP) has been implicated in the regulation of apoptosis, DNA repair and drug resistance. However, the underlying molecular mechanisms are poorly defined. To better understand the molecular mechanisms underlying TCTP involved in cellular processes, we performed an affinity purification-based proteomic profiling to identify proteins interacting with TCTP in human cervical cancer HeLa cells. We found that a group of proteins involved in DNA repair are enriched in the potential TCTP interactome. Silencing TCTP by short hairpin RNA in breast carcinoma MCF-7 cells leads to the declined repair efficiency for DNA double-strand breaks on the GFP-Pem1 reporter gene by homologous recombination, the persistent activation and the prolonged retention of γH2AX and Rad51 foci following ionizing radiation. Reciprocal immunoprecipitations indicated that TCTP forms complexes with Rad51 in vivo, and the stability maintenance of Rad51 requires TCTP in MCF-7 cells under normal cell culture conditions. Moreover, inactivation of TCTP by sertraline treatment enhances UVC irradiation-induced apoptosis in MCF-7 cells, and causes sensitization to DNA-damaging drug etoposide and DNA repair inhibitor olaparib. Thus, we have identified an important role of TCTP in promoting DNA double-stand break repair via facilitating DNA homologous recombination processes and highlighted the great potential of TCTP as a drug target to enhance conventional chemotherapy for cancer patients with high levels of TCTP expression.
Collapse
Affiliation(s)
- Y Li
- The Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - H Sun
- The Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - C Zhang
- The 2nd Affiliated Hospital, Harbin Medical University, Harbin, China
| | - J Liu
- The Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - H Zhang
- The Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - F Fan
- The Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - R A Everley
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - X Ning
- The Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Y Sun
- The Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - J Hu
- The Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - J Liu
- The 2nd Affiliated Hospital, Harbin Medical University, Harbin, China
| | - J Zhang
- The 2nd Affiliated Hospital, Harbin Medical University, Harbin, China
| | - W Ye
- The 2nd Affiliated Hospital, Harbin Medical University, Harbin, China
| | - X Qiu
- The 2nd Affiliated Hospital, Harbin Medical University, Harbin, China
| | - S Dai
- The Tumor Hospital, Harbin Medical University, Harbin, China
| | - B Liu
- The Tumor Hospital, Harbin Medical University, Harbin, China
| | - H Xu
- Department of Clinical Laboratory, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - S Fu
- The Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - S P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - C Zhou
- The Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| |
Collapse
|
41
|
Targeting Hsp27/eIF4E interaction with phenazine compound: a promising alternative for castration-resistant prostate cancer treatment. Oncotarget 2017; 8:77317-77329. [PMID: 29100389 PMCID: PMC5652782 DOI: 10.18632/oncotarget.20469] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/25/2017] [Indexed: 01/01/2023] Open
Abstract
The actual strategy to improve current therapies in advanced prostate cancer involves targeting genes activated by androgen withdrawal, either to delay or prevent the emergence of the castration-refractory phenotype. However, these genes are often implicated in several physiological processes, and long-term inhibition of survival proteins might be accompanied with cytotoxic effects. To avoid this problem, an alternative therapeutic strategy relies on the identification and use of compounds that disrupt specific protein-protein interactions involved in androgen withdrawal. Specifically, the interaction of the chaperone protein Hsp27 with the initiation factor eIF4E leads to the protection of protein synthesis initiation process and enhances cell survival during cell stress induced by castration or chemotherapy. Thus, in this work we aimed at i) identifying the interaction site of the Hsp27/eIF4E complex and ii) interfere with the relevant protein/protein association mechanism involved in castration-resistant progression of prostate cancer. By a combination of experimental and modeling techniques, we proved that eIF4E interacts with the C-terminal part of Hsp27, preferentially when Hsp27 is phosphorylated. We also observed that the loss of this interaction increased cell chemo-and hormone-sensitivity. In order to find a potential inhibitor of Hsp27/eIF4E interaction, BRET assays in combination with molecular simulations identified the phenazine derivative 14 as the compound able to efficiently interfere with this protein/protein interaction, thereby inhibiting cell viability and increasing cell death in chemo- and castration-resistant prostate cancer models in vitro and in vivo.
Collapse
|
42
|
Pinkaew D, Fujise K. Fortilin: A Potential Target for the Prevention and Treatment of Human Diseases. Adv Clin Chem 2017; 82:265-300. [PMID: 28939212 DOI: 10.1016/bs.acc.2017.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Fortilin is a highly conserved 172-amino-acid polypeptide found in the cytosol, nucleus, mitochondria, extracellular space, and circulating blood. It is a multifunctional protein that protects cells against apoptosis, promotes cell growth and cell cycle progression, binds calcium (Ca2+) and has antipathogen activities. Its role in the pathogenesis of human and animal diseases is also diverse. Fortilin facilitates the development of atherosclerosis, contributes to both systemic and pulmonary arterial hypertension, participates in the development of cancers, and worsens diabetic nephropathy. It is important for the adaptive expansion of pancreatic β-cells in response to obesity and increased insulin requirement, for the regeneration of liver after hepatectomy, and for protection of the liver against alcohol- and ER stress-induced injury. Fortilin is a viable surrogate marker for in vivo apoptosis, and it plays a key role in embryo and organ development in vertebrates. In fish and shrimp, fortilin participates in host defense against bacterial and viral pathogens. Further translational research could prove fortilin to be a viable molecular target for treatment of various human diseases including and not limited to atherosclerosis, hypertension, certain tumors, diabetes mellitus, diabetic nephropathy, hepatic injury, and aberrant immunity and host defense.
Collapse
Affiliation(s)
- Decha Pinkaew
- University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ken Fujise
- University of Texas Medical Branch at Galveston, Galveston, TX, United States; The Institute of Translational Sciences, University of Texas Medical Branch at Galveston, Galveston, TX, United States.
| |
Collapse
|
43
|
Boia-Ferreira M, Basílio AB, Hamasaki AE, Matsubara FH, Appel MH, Da Costa CRV, Amson R, Telerman A, Chaim OM, Veiga SS, Senff-Ribeiro A. TCTP as a therapeutic target in melanoma treatment. Br J Cancer 2017; 117:656-665. [PMID: 28751755 PMCID: PMC5572181 DOI: 10.1038/bjc.2017.230] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/27/2017] [Accepted: 06/23/2017] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Translationally controlled tumour protein (TCTP) is an antiapoptotic protein highly conserved through phylogeny. Translationally controlled tumour protein overexpression was detected in several tumour types. Silencing TCTP was shown to induce tumour reversion. There is a reciprocal repression between TCTP and P53. Sertraline interacts with TCTP and decreases its cellular levels. METHODS We evaluate the role of TCTP in melanoma using sertraline and siRNA. Cell viability, migration, and clonogenicity were assessed in human and murine melanoma cells in vitro. Sertraline was evaluated in a murine melanoma model and was compared with dacarbazine, a major chemotherapeutic agent used in melanoma treatment. RESULTS Inhibition of TCTP levels decreases melanoma cell viability, migration, clonogenicity, and in vivo tumour growth. Human melanoma cells treated with sertraline show diminished migration properties and capacity to form colonies. Sertraline was effective in inhibiting tumour growth in a murine melanoma model; its effect was stronger when compared with dacarbazine. CONCLUSIONS Altogether, these results indicate that sertraline could be effective against melanoma and TCTP can be a target for melanoma therapy.
Collapse
Affiliation(s)
- M Boia-Ferreira
- Department of Cell Biology, Centro Politécnico, Federal University of Paraná, UFPR, Jardim das Américas, CEP 81531-990, Curitiba, Paraná, Brazil
| | - A B Basílio
- Department of Cell Biology, Centro Politécnico, Federal University of Paraná, UFPR, Jardim das Américas, CEP 81531-990, Curitiba, Paraná, Brazil
| | - A E Hamasaki
- Department of Cell Biology, Centro Politécnico, Federal University of Paraná, UFPR, Jardim das Américas, CEP 81531-990, Curitiba, Paraná, Brazil
| | - F H Matsubara
- Department of Cell Biology, Centro Politécnico, Federal University of Paraná, UFPR, Jardim das Américas, CEP 81531-990, Curitiba, Paraná, Brazil
| | - M H Appel
- Department of Structural, Molecular Biology and Genetics, State University of Ponta Grossa, Ponta Grossa, Paraná, Brazil
| | - C R V Da Costa
- Department of Cell Biology, Centro Politécnico, Federal University of Paraná, UFPR, Jardim das Américas, CEP 81531-990, Curitiba, Paraná, Brazil
| | - R Amson
- Institute Gustave Roussy, Unité Inserm U981, Bâtiment B2M, Villejuif, France
| | - A Telerman
- Institute Gustave Roussy, Unité Inserm U981, Bâtiment B2M, Villejuif, France
| | - O M Chaim
- Department of Cell Biology, Centro Politécnico, Federal University of Paraná, UFPR, Jardim das Américas, CEP 81531-990, Curitiba, Paraná, Brazil
| | - S S Veiga
- Department of Cell Biology, Centro Politécnico, Federal University of Paraná, UFPR, Jardim das Américas, CEP 81531-990, Curitiba, Paraná, Brazil
| | - A Senff-Ribeiro
- Department of Cell Biology, Centro Politécnico, Federal University of Paraná, UFPR, Jardim das Américas, CEP 81531-990, Curitiba, Paraná, Brazil
| |
Collapse
|
44
|
Karaki S, Benizri S, Mejías R, Baylot V, Branger N, Nguyen T, Vialet B, Oumzil K, Barthélémy P, Rocchi P. Lipid-oligonucleotide conjugates improve cellular uptake and efficiency of TCTP-antisense in castration-resistant prostate cancer. J Control Release 2017; 258:1-9. [PMID: 28472637 DOI: 10.1016/j.jconrel.2017.04.042] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/25/2017] [Accepted: 04/29/2017] [Indexed: 01/08/2023]
Abstract
Translationally controlled tumor protein (TCTP) has been implicated in a plethora of important cellular processes related to cell growth, cell cycle progression, malignant transformation and inhibition of apoptosis. Therefore, TCTP is now recognized as a potential therapeutic target in several cancers including prostate, breast and lung cancers. We previously showed that TCTP is overexpressed in castration-resistant prostate cancer (CRPC), and it has been implicated resistance to treatment. Recently, we developed TCTP antisense oligonucleotides (ASOs) to inhibit TCTP expression. However, the intracellular delivery and silencing activity of these oligonucleotides remains a challenge, and depend on the use of transfection agents and delivery systems. Here we show that lipid-modified ASO (LASOs) has improved penetration and efficiency in inhibiting TCTP expression in the absence of additional transfection agents, both in vitro and in vivo. Transfection with TCTP-LASO led to rapid and prolonged internalization via macropinocytosis, TCTP downregulation and significant decreased cell viability. We also show that lipid-modification led to delayed tumor progression in CRPC xenografts models, with no significant toxic effects observed.
Collapse
Affiliation(s)
- Sara Karaki
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, 27 Bd. Lei Roure BP30059, 13273 Marseille, France; Institut Paoli-Calmettes, 13273 Marseille, France; Aix-Marseille Université, 13284 Marseille, France; CNRS UMR7258, 13009 Marseille, France
| | - Sebastien Benizri
- ARNA Laboratory, University of Bordeaux, F-33076 Bordeaux, France; INSERM U1212, F-33076, Bordeaux, France; UMR CNRS 5320, F-33076, Bordeaux, France
| | - Raquel Mejías
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, 27 Bd. Lei Roure BP30059, 13273 Marseille, France; Institut Paoli-Calmettes, 13273 Marseille, France; Aix-Marseille Université, 13284 Marseille, France; CNRS UMR7258, 13009 Marseille, France
| | - Virginie Baylot
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, 27 Bd. Lei Roure BP30059, 13273 Marseille, France; Institut Paoli-Calmettes, 13273 Marseille, France; Aix-Marseille Université, 13284 Marseille, France; CNRS UMR7258, 13009 Marseille, France
| | - Nicolas Branger
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, 27 Bd. Lei Roure BP30059, 13273 Marseille, France; Institut Paoli-Calmettes, 13273 Marseille, France; Aix-Marseille Université, 13284 Marseille, France; CNRS UMR7258, 13009 Marseille, France
| | - Tan Nguyen
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, 27 Bd. Lei Roure BP30059, 13273 Marseille, France; Institut Paoli-Calmettes, 13273 Marseille, France; Aix-Marseille Université, 13284 Marseille, France; CNRS UMR7258, 13009 Marseille, France
| | - Brune Vialet
- ARNA Laboratory, University of Bordeaux, F-33076 Bordeaux, France; UMR CNRS 5320, F-33076, Bordeaux, France; UMR CNRS 5320, F-33076, Bordeaux, France
| | - Khalid Oumzil
- ARNA Laboratory, University of Bordeaux, F-33076 Bordeaux, France; INSERM U1212, F-33076, Bordeaux, France
| | - Philippe Barthélémy
- ARNA Laboratory, University of Bordeaux, F-33076 Bordeaux, France; INSERM U1212, F-33076, Bordeaux, France; UMR CNRS 5320, F-33076, Bordeaux, France
| | - Palma Rocchi
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, 27 Bd. Lei Roure BP30059, 13273 Marseille, France; Institut Paoli-Calmettes, 13273 Marseille, France; Aix-Marseille Université, 13284 Marseille, France; CNRS UMR7258, 13009 Marseille, France.
| |
Collapse
|
45
|
Bommer UA, Vine KL, Puri P, Engel M, Belfiore L, Fildes K, Batterham M, Lochhead A, Aghmesheh M. Translationally controlled tumour protein TCTP is induced early in human colorectal tumours and contributes to the resistance of HCT116 colon cancer cells to 5-FU and oxaliplatin. Cell Commun Signal 2017; 15:9. [PMID: 28143584 PMCID: PMC5286767 DOI: 10.1186/s12964-017-0164-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 01/23/2017] [Indexed: 12/22/2022] Open
Abstract
Background Translationally controlled tumour protein TCTP is an anti-apoptotic protein frequently overexpressed in cancers, where high levels are often associated with poor patient outcome. TCTP may be involved in protecting cancer cells against the cytotoxic action of anti-cancer drugs. Here we study the early increase of TCTP levels in human colorectal cancer (CRC) and the regulation of TCTP expression in HCT116 colon cancer cells, in response to treatment with the anti-cancer drugs 5-FU and oxaliplatin. Methods Using immunohistochemistry, we assessed TCTP levels in surgical samples from adenomas and adenocarcinomas of the colon, compared to normal colon tissue. We also studied the regulation of TCTP in HCT116 colon cancer cells in response to 5-FU and oxaliplatin by western blotting. TCTP mRNA levels were assessed by RT-qPCR. We used mTOR kinase inhibitors to demonstrate mTOR-dependent translational regulation of TCTP under these conditions. Employing the Real-Time Cell Analysis (RTCA) System and the MTS assay, we investigated the effect of TCTP-knockdown on the sensitivity of HCT116 cells to the anti-cancer drugs 5-FU and oxaliplatin. Results 1. TCTP levels are significantly increased in colon adenomas and adenocarcinomas, compared to normal colon tissue. 2. TCTP protein levels are about 4-fold upregulated in HCT116 colon cancer cells, in response to 5-FU and oxaliplatin treatment, whereas TCTP mRNA levels are down regulated. 3. mTOR kinase inhibitors prevented the up-regulation of TCTP protein, indicating that TCTP is translationally regulated through the mTOR complex 1 signalling pathway under these conditions. 4. Using two cellular assay systems, we demonstrated that TCTP-knockdown sensitises HCT116 cells to the cytotoxicity caused by 5-FU and oxaliplatin. Conclusions Our results demonstrate that TCTP levels increase significantly in the early stages of CRC development. In colon cancer cells, expression of this protein is largely upregulated during treatment with the DNA-damaging anti-cancer drugs 5-FU and oxaliplatin, as part of the cellular stress response. TCTP may thus contribute to the development of anti-cancer drug resistance. These findings indicate that TCTP might be suitable as a biomarker and that combinatorial treatment using 5-FU/oxaliplatin, together with mTOR kinase inhibitors, could be a route to preventing the development of resistance to these drugs. Electronic supplementary material The online version of this article (doi:10.1186/s12964-017-0164-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ulrich-Axel Bommer
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia. .,Graduate School of Medicine, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia.
| | - Kara L Vine
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia.,School of Biological Sciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, 2522, NSW, Australia
| | - Prianka Puri
- Graduate School of Medicine, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia.,Present address: Southeast Sydney Illawarra Area Health Services, Sydney, NSW, Australia
| | - Martin Engel
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia.,School of Biological Sciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, 2522, NSW, Australia
| | - Lisa Belfiore
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia.,School of Biological Sciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, 2522, NSW, Australia
| | - Karen Fildes
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia.,Graduate School of Medicine, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia
| | - Marijka Batterham
- School of Mathematics and Applied Statistics, Faculty of Engineering and Information Sciences University of Wollongong, Wollongong, 2522, NSW, Australia
| | - Alistair Lochhead
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia.,Southern IML Pathology Wollongong, 2500, Wollongong, NSW, Australia.,Present address: Syd-Path, St. Vincent's Hospital Darlinghurst, Sydney, 2010, NSW, Australia
| | - Morteza Aghmesheh
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia.,Illawarra Cancer Care Centre, The Wollongong Hospital, Wollongong, 2500, NSW, Australia
| |
Collapse
|
46
|
Bonhoure A, Vallentin A, Martin M, Senff-Ribeiro A, Amson R, Telerman A, Vidal M. Acetylation of translationally controlled tumor protein promotes its degradation through chaperone-mediated autophagy. Eur J Cell Biol 2017; 96:83-98. [PMID: 28110910 DOI: 10.1016/j.ejcb.2016.12.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 12/13/2016] [Accepted: 12/13/2016] [Indexed: 12/22/2022] Open
Abstract
Translationally controlled tumor protein (Tpt1/TCTP) is a multi-functional cytosolic protein whose cellular levels are finely tuned. TCTP regulates protein behavior by favoring stabilization of protein partners or on the contrary by promoting degradation of others. TCTP has been shown to be transcriptionally and translationally regulated, but much less is known about its degradation process. In this study, we present evidence that chaperone-mediated autophagy (CMA) contributes to TCTP regulation. CMA allows lysosomal degradation of specific cytosolic proteins on a molecule-by-molecule basis. It contributes to cellular homeostasis especially by acting as a quality control for cytosolic proteins in response to stress and as a way of regulating the level of specific proteins. Using a variety of approaches, we show that CMA degradation of TCTP is Hsc70 and LAMP-2A dependent. Our data indicate that (i) TCTP directly interacts with Hsc70; (ii) silencing LAMP-2A in MEFs using siRNA leads to inhibition of TCTP downregulation; (iii) TCTP is relocalized from a diffuse cytosolic pattern to a punctate lysosomal pattern when CMA is upregulated; (iv) TCTP is degraded in vitro by purified lysosomes. Importantly, using lysine-mutated forms of TCTP, we show that acetylation of Lysine 19 generates a KFERQ-like motif and promotes binding to Hsc70, lysosome targeting and TCTP degradation by CMA. Altogether these results indicate that TCTP is degraded by chaperone-mediated autophagy in an acetylation dependent manner.
Collapse
Affiliation(s)
- Anne Bonhoure
- UMR 5235, CNRS, Université Montpellier, 34095 Montpellier, France
| | - Alice Vallentin
- UMR 5235, CNRS, Université Montpellier, 34095 Montpellier, France
| | - Marianne Martin
- UMR 5235, CNRS, Université Montpellier, 34095 Montpellier, France
| | - Andrea Senff-Ribeiro
- UMR 8113, École Normale Supérieure, 94235 Cachan, France; UMR 981, Institut Gustave Roussy, 94800 Villejuif, France
| | - Robert Amson
- UMR 8113, École Normale Supérieure, 94235 Cachan, France; UMR 981, Institut Gustave Roussy, 94800 Villejuif, France
| | - Adam Telerman
- UMR 8113, École Normale Supérieure, 94235 Cachan, France; UMR 981, Institut Gustave Roussy, 94800 Villejuif, France
| | - Michel Vidal
- UMR 5235, CNRS, Université Montpellier, 34095 Montpellier, France.
| |
Collapse
|
47
|
Baylot V, Karaki S, Rocchi P. TCTP Has a Crucial Role in the Different Stages of Prostate Cancer Malignant Progression. Results Probl Cell Differ 2017; 64:255-261. [PMID: 29149413 DOI: 10.1007/978-3-319-67591-6_13] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Prostate cancer (PC) is the second most common cause of cancer-related mortality in men in the western world after lung cancer. Many patients are not candidates for resection given the advanced stage of their cancer. The primary treatment for advanced PC is the castration therapy which supresses the production of androgens, hormone that promotes PC growth. Despite the efficiency of the castration therapy, most patients develop castration resistant disease which remains uncurable. Clearly, novel approaches are required to effectively treat castration resistant PC (CRPC). New strategies that identify the molecular mechanisms by which PC becomes resistant to conventional therapies may enable the identification of novel therapeutic targets that could improve clinical outcome. Recent studies have demonstrated the implication of TCTP's over-expression in PC and CRPC, and its role in resistance to treatment. TCTP's interaction with p53 and their negative feedback loop regulation have also been described to be causal for PC progression and invasion. A novel nanotherapy that inhibits TCTP has been developed as a new therapeutical strategy in CRPC. This chapter will highlight the role of TCTP as new therapeutic target in PC, in particular, therapy-resistant advanced PC and report the development of novel nanotherapy against TCTP that restore treatment-sensitivity in CRPC that deserve to be tested in clinical trial.
Collapse
Affiliation(s)
- Virginie Baylot
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Sara Karaki
- UMR 1068, Inserm, CRCM, BP30059, Cedex9, 27 Boulevard Leï Roure, Marseille, 13273, France
- Institut Paoli-Calmettes, Marseille, 13009, France
- Aix-Marseille Université, 13284, Marseille, France
- CNRS, UMR7258, CRCM, Marseille, 13009, France
| | - Palma Rocchi
- UMR 1068, Inserm, CRCM, BP30059, Cedex9, 27 Boulevard Leï Roure, Marseille, 13273, France.
- Institut Paoli-Calmettes, Marseille, 13009, France.
- Aix-Marseille Université, 13284, Marseille, France.
- CNRS, UMR7258, CRCM, Marseille, 13009, France.
| |
Collapse
|
48
|
Assrir N, Malard F, Lescop E. Structural Insights into TCTP and Its Interactions with Ligands and Proteins. Results Probl Cell Differ 2017; 64:9-46. [PMID: 29149402 DOI: 10.1007/978-3-319-67591-6_2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The 19-24 kDa Translationally Controlled Tumor Protein (TCTP) is involved in a wide range of molecular interactions with biological and nonbiological partners of various chemical compositions such as proteins, peptides, nucleic acids, carbohydrates, or small molecules. TCTP is therefore an important and versatile binding platform. Many of these protein-protein interactions have been validated, albeit only few received an in-depth structural characterization. In this chapter, we will focus on the structural analysis of TCTP and we will review the available literature regarding its interaction network from a structural perspective.
Collapse
Affiliation(s)
- Nadine Assrir
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Sud, Université Paris-Saclay, 1 avenue de la Terrasse, 91190, Gif-sur-Yvette, France
| | - Florian Malard
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Sud, Université Paris-Saclay, 1 avenue de la Terrasse, 91190, Gif-sur-Yvette, France
| | - Ewen Lescop
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Sud, Université Paris-Saclay, 1 avenue de la Terrasse, 91190, Gif-sur-Yvette, France.
| |
Collapse
|
49
|
Bommer UA. The Translational Controlled Tumour Protein TCTP: Biological Functions and Regulation. Results Probl Cell Differ 2017; 64:69-126. [PMID: 29149404 DOI: 10.1007/978-3-319-67591-6_4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The Translational Controlled Tumour Protein TCTP (gene symbol TPT1, also called P21, P23, Q23, fortilin or histamine-releasing factor, HRF) is a highly conserved protein present in essentially all eukaryotic organisms and involved in many fundamental cell biological and disease processes. It was first discovered about 35 years ago, and it took an extended period of time for its multiple functions to be revealed, and even today we do not yet fully understand all the details. Having witnessed most of this history, in this chapter, I give a brief overview and review the current knowledge on the structure, biological functions, disease involvements and cellular regulation of this protein.TCTP is able to interact with a large number of other proteins and is therefore involved in many core cell biological processes, predominantly in the response to cellular stresses, such as oxidative stress, heat shock, genotoxic stress, imbalance of ion metabolism as well as other conditions. Mechanistically, TCTP acts as an anti-apoptotic protein, and it is involved in DNA-damage repair and in cellular autophagy. Thus, broadly speaking, TCTP can be considered a cytoprotective protein. In addition, TCTP facilitates cell division through stabilising the mitotic spindle and cell growth through modulating growth signalling pathways and through its interaction with the proteosynthetic machinery of the cell. Due to its activities, both as an anti-apoptotic protein and in promoting cell growth and division, TCTP is also essential in the early development of both animals and plants.Apart from its involvement in various biological processes at the cellular level, TCTP can also act as an extracellular protein and as such has been involved in modulating whole-body defence processes, namely in the mammalian immune system. Extracellular TCTP, typically in its dimerised form, is able to induce the release of cytokines and other signalling molecules from various types of immune cells. There are also several examples, where TCTP was shown to be involved in antiviral/antibacterial defence in lower animals. In plants, the protein appears to have a protective effect against phytotoxic stresses, such as flooding, draught, too high or low temperature, salt stress or exposure to heavy metals. The finding for the latter stress condition is corroborated by earlier reports that TCTP levels are considerably up-regulated upon exposure of earthworms to high levels of heavy metals.Given the involvement of TCTP in many biological processes aimed at maintaining cellular or whole-body homeostasis, it is not surprising that dysregulation of TCTP levels may promote a range of disease processes, foremost cancer. Indeed a large body of evidence now supports a role of TCTP in at least the most predominant types of human cancers. Typically, this can be ascribed to both the anti-apoptotic activity of the protein and to its function in promoting cell growth and division. However, TCTP also appears to be involved in the later stages of cancer progression, such as invasion and metastasis. Hence, high TCTP levels in tumour tissues are often associated with a poor patient outcome. Due to its multiple roles in cancer progression, TCTP has been proposed as a potential target for the development of new anti-cancer strategies in recent pilot studies. Apart from its role in cancer, TCTP dysregulation has been reported to contribute to certain processes in the development of diabetes, as well as in diseases associated with the cardiovascular system.Since cellular TCTP levels are highly regulated, e.g. in response to cell stress or to growth signalling, and because deregulation of this protein contributes to many disease processes, a detailed understanding of regulatory processes that impinge on TCTP levels is required. The last section of this chapter summarises our current knowledge on the mechanisms that may be involved in the regulation of TCTP levels. Essentially, expression of the TPT1 gene is regulated at both the transcriptional and the translational level, the latter being particularly advantageous when a rapid adjustment of cellular TCTP levels is required, for example in cell stress responses. Other regulatory mechanisms, such as protein stability regulation, may also contribute to the regulation of overall TCTP levels.
Collapse
Affiliation(s)
- Ulrich-Axel Bommer
- School of Medicine, Graduate Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
50
|
Amson R, Auclair C, André F, Karp J, Telerman A. Targeting TCTP with Sertraline and Thioridazine in Cancer Treatment. Results Probl Cell Differ 2017; 64:283-290. [PMID: 29149415 DOI: 10.1007/978-3-319-67591-6_15] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We have initially demonstrated in knocking down experiments that decreasing TCTP in cancer cells leads in some tissues to cell death while in others to a complete reorganization of the tumor into architectural structures reminiscent of normal ones. Based on these experiments and a series of other findings confirming the key role of TCTP in cancer, it became important to find pharmacological compounds to inhibit its function, and this became for us a priority. In the present text, we explain in detail the experiments that were performed and the perspectives of sertraline in cancer treatment, as this became today a reality with a clinical study that started in collaboration with Columbia University and Johns Hopkins University.
Collapse
Affiliation(s)
- Robert Amson
- Bâtiment B2M, Institut Gustave Roussy, Unité Inserm U981, 114 rue Édouard-Vaillant, 94805, Villejuif, France.
| | - Christian Auclair
- CNRS-UMR 8113, LBPA, École Normale Supérieure, 61 avenue du Président Wilson, 94235, Cachan, France
| | - Fabrice André
- Institut Gustave Roussy, Unité Inserm U981, Bâtiment B2M, 114 rue Édouard-Vaillant, 94805, Villejuif, France
| | - Judith Karp
- Division of Hematologic Malignancies, The Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21231-1000, USA
| | - Adam Telerman
- Bâtiment B2M, Institut Gustave Roussy, Unité Inserm U981, 114 rue Édouard-Vaillant, 94805, Villejuif, France
| |
Collapse
|