1
|
Cicek C, Telkoparan-Akillilar P, Sertyel S, Bilgi C, Ozgun OD. Investigation of endoplasmic reticulum stress-regulated chaperones as biomarkers in idiopathic nonobstructive azoospermia. Cell Stress Chaperones 2024; 29:654-665. [PMID: 39237030 PMCID: PMC11424951 DOI: 10.1016/j.cstres.2024.08.004] [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: 08/11/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024] Open
Abstract
Azoospermia is a condition in which sperm cells are completely absent in a male's ejaculate. Typically, sperm production occurs in the testes and is regulated by a complex series of cellular and molecular interactions. Endoplasmic reticulum (ER) stress arises when there is a deviation from or damage to the normal functions of the ER within cells. In response to this stress, a cascade of response mechanisms is activated to regulate ER stress within cells. This study aims to investigate the role of ER stress-regulated chaperones as potential biomarkers in male infertility. ER stress associated with azoospermia can manifest in cells such as spermatogonia in the testes and can impact sperm production. As a result of ER stress, the expression and activity of a variety of proteins within cells can be altered. Among these proteins are chaperone proteins that regulate the ER stress response. The sample size was calculated to be a minimum of 36 patients in each group. In this preliminary study, we measured and compared serum levels of protein disulfide-isomerase A1, protein disulfide-isomerase A3 (PDIA3), mesencephalic astrocyte-derived neurotrophic factor (MANF), glucose regulatory protein 78 (GRP78), clusterin (CLU), calreticulin (CRT), and calnexin (CNX) between male subjects with idiopathic nonobstructive azoospermia and a control group of noninfertile males. Serum PDIA1 (P = 0.0004), MANF (P = 0.018), PDIA3 (P < 0.0001), GRP78 (P = 0.0027), and CRT (P = 0.0009) levels were higher in the infertile group compared to the control. In summary, this study presents novel findings in a cohort of male infertile patients, emphasizing the significance of incorporating diverse biomarkers. It underscores the promising role of ER stress-regulated proteins as potential serum indicators for male infertility. By elucidating the impact of ER stress on spermatogenic cells, the research illuminates the maintenance or disruption of cellular health. A deeper understanding of these results could open the door to novel treatment approaches for reproductive conditions, including azoospermia.
Collapse
Affiliation(s)
- Cigdem Cicek
- Department of Medical Biochemistry, Faculty of Medicine, Yuksek Ihtisas University, Ankara 06530, Turkey.
| | - Pelin Telkoparan-Akillilar
- Department of Medical Biology, Faculty of Medicine, Gazi University, Ankara 06500, Turkey; Department of Medical Biology, Faculty of Medicine, Yuksek Ihtisas University, Ankara 06530, Turkey
| | | | - Cumhur Bilgi
- Alife Hospital Biochemistry Laboratory, Ankara 06794, Turkey
| | | |
Collapse
|
2
|
Gross C, Guérin LP, Socol BG, Germain L, Guérin SL. The Ins and Outs of Clusterin: Its Role in Cancer, Eye Diseases and Wound Healing. Int J Mol Sci 2023; 24:13182. [PMID: 37685987 PMCID: PMC10488069 DOI: 10.3390/ijms241713182] [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: 06/30/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Clusterin (CLU) is a glycoprotein originally discovered in 1983 in ram testis fluid. Rapidly observed in other tissues, it was initially given various names based on its function in different tissues. In 1992, it was finally named CLU by consensus. Nearly omnipresent in human tissues, CLU is strongly expressed at fluid-tissue interfaces, including in the eye and in particular the cornea. Recent research has identified different forms of CLU, with the most prominent being a 75-80 kDa heterodimeric protein that is secreted. Another truncated version of CLU (55 kDa) is localized to the nucleus and exerts pro-apoptotic activities. CLU has been reported to be involved in various physiological processes such as sperm maturation, lipid transportation, complement inhibition and chaperone activity. CLU was also reported to exert important functions in tissue remodeling, cell-cell adhesion, cell-substratum interaction, cytoprotection, apoptotic cell death, cell proliferation and migration. Hence, this protein is sparking interest in tissue wound healing. Moreover, CLU gene expression is finely regulated by cytokines, growth factors and stress-inducing agents, leading to abnormally elevated levels of CLU in many states of cellular disturbance, including cancer and neurodegenerative conditions. In the eye, CLU expression has been reported as being severely increased in several pathologies, such as age-related macular degeneration and Fuch's corneal dystrophy, while it is depleted in others, such as pathologic keratinization. Nevertheless, the precise role of CLU in the development of ocular pathologies has yet to be deciphered. The question of whether CLU expression is influenced by these disorders or contributes to them remains open. In this article, we review the actual knowledge about CLU at both the protein and gene expression level in wound healing, and explore the possibility that CLU is a key factor in cancer and eye diseases. Understanding the expression and regulation of CLU could lead to the development of novel therapeutics for promoting wound healing.
Collapse
Affiliation(s)
- Christelle Gross
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec City, QC G1V 0A6, Canada; (C.G.); (B.G.S.); (L.G.)
- Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Québec City, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | | | - Bianca G. Socol
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec City, QC G1V 0A6, Canada; (C.G.); (B.G.S.); (L.G.)
| | - Lucie Germain
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec City, QC G1V 0A6, Canada; (C.G.); (B.G.S.); (L.G.)
- Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Québec City, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
- Département de Chirurgie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Sylvain L. Guérin
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec City, QC G1V 0A6, Canada; (C.G.); (B.G.S.); (L.G.)
- Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Québec City, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| |
Collapse
|
3
|
Huntoon K, Anderson SK, Ballman KV, Twohy E, Dooley K, Jiang W, An Y, Li J, von Roemeling C, Qie Y, Ross OA, Cerhan JH, Whitton AC, Greenspoon JN, Parney IF, Ashman JB, Bahary JP, Hadjipanayis C, Urbanic JJ, Farace E, Khuntia D, Laack NN, Brown PD, Roberge D, Kim BYS. Association of circulating markers with cognitive decline after radiation therapy for brain metastasis. Neuro Oncol 2023; 25:1123-1131. [PMID: 36472389 PMCID: PMC10237411 DOI: 10.1093/neuonc/noac262] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND A recent phase III trial (NCT01372774) comparing use of stereotactic radiosurgery [SRS] versus whole-brain radiation therapy [WBRT] after surgical resection of a single brain metastasis revealed that declines in cognitive function were more common with WBRT than with SRS. A secondary endpoint in that trial, and the primary objective in this secondary analysis, was to identify baseline biomarkers associated with cognitive impairment after either form of radiotherapy for brain metastasis. Here we report our findings on APOE genotype and serum levels of associated proteins and their association with radiation-induced neurocognitive decline. METHODS In this retrospective analysis of prospectively collected samples from a completed randomized clinical trial, patients provided blood samples every 3 months that were tested by genotyping and enzyme-linked immunosorbent assay, and results were analyzed in association with cognitive impairment. RESULTS The APOE genotype was not associated with neurocognitive impairment at 3 months. However, low serum levels of ApoJ, ApoE, or ApoA protein (all P < .01) and higher amyloid beta (Aβ 1-42) levels (P = .048) at baseline indicated a greater likelihood of neurocognitive decline at 3 months after SRS, whereas lower ApoJ levels were associated with decline after WBRT (P = .014). CONCLUSIONS Patients with these pretreatment serum markers should be counseled about radiation-related neurocognitive decline.
Collapse
Affiliation(s)
- Kristin Huntoon
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - S Keith Anderson
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Karla V Ballman
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biostatistics and Epidemiology, Weill Medical College of Cornell University, New York, New York, USA
| | - Erin Twohy
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Katharine Dooley
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas,USA
| | - Yi An
- Department of Therapeutic Radiology, Yale-New Haven Hospital, North Haven, Connecticut, USA
| | - Jing Li
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas,USA
| | | | - Yaqing Qie
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Jane H Cerhan
- Department of Psychiatry and Psychology Mayo Clinic, Rochester, Minnesota, USA
| | - Anthony C Whitton
- Department of Radiation Oncology, McMaster University, Hamilton, Ontario, Canada
| | - Jeffrey N Greenspoon
- Department of Radiation Oncology, McMaster University, Hamilton, Ontario, Canada
| | - Ian F Parney
- Department of Neurosurgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Jonathan B Ashman
- Department of Radiation Oncology, Mayo Clinic, Phoenix/Scottsdale, Arizona, USA
| | - Jean-Paul Bahary
- Department of Radiation Oncology, CHUM, Montreal, Quebec, Canada
| | | | - James J Urbanic
- Department of Radiation Oncology, University of California San Diego, Moores Cancer Center, La Jolla, California, USA
| | - Elana Farace
- Department of Public Health Sciences, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| | - Deepak Khuntia
- Department of Radiation Oncology, Precision Cancer Specialists and Varian Medical Systems, Palo Alto, California, USA
| | - Nadia N Laack
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Paul D Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - David Roberge
- Department of Radiation Oncology, CHUM, Montreal, Quebec, Canada
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
4
|
Das L, Shekhar S, Chandrani P, Varma AK. In silico structural analysis of secretory clusterin to assess pathogenicity of mutations identified in the evolutionarily conserved regions. J Biomol Struct Dyn 2023; 41:469-478. [PMID: 34821197 DOI: 10.1080/07391102.2021.2007791] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Clusterin (CLU) is a secreted glycoprotein, heterodimeric in nature, and is expressed in a wide variety of tissues and body fluids such as serum and plasma. CLU has also been known to be a promising biomarker for cell death, malignancy, cancer progression, and resistance development. However, the lack of a CLU crystal structure obstructs understanding the possible role of reported mutations on the structure, and the subsequent effects on downstream signaling pathways and cancer progression. Considering the importance of crystal structure, a model structure of the pre-secretory isoform of CLU was built to predict the effect of mutations at the molecular level. Ab initio model was built using RaptorX, and loop refinement and energy minimization were carried out with ModLoop, ModRefiner, and GalaxyWeb servers. The cancer associated mutational spectra of CLU was retrieved from the cBioPortal server and 117 unique missense mutations were identified. Evolutionarily conserved regions and pathogenicity of mutations identified in CLU were analyzed using ConSurf and Rhapsody, respectively. Furthermore, sequence and structure-based mutational analysis were carried out with iSTABLE, DynaMut and PremPS servers. Molecular dynamics simulations were carried out with GROMACS for 50 ns to determine the stability of the wild type and mutant protein structures. A dynamically stable model structure of pre-secretory CLU (psCLU) which has high concurrence with the sequence based secondary structure predictions has been explored. Changes in the intra-atomic interactions and folding pattern between wild type and mutant structures were observed. To our conclusion, eleven mutations with the highest structural and functional significance have been predicted to have pathogenic and deleterious effects.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Lipi Das
- Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, India.,Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - Shashank Shekhar
- Supercomputing Facility for Bioinformatics & Computational Biology, Indian Institute of Technology, New Delhi, India
| | - Pratik Chandrani
- Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, India.,Homi Bhabha National Institute, Training School Complex, Mumbai, India.,Medical Oncology Molecular Lab, Tata Memorial Hospital, Mumbai, India
| | - Ashok K Varma
- Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, India.,Homi Bhabha National Institute, Training School Complex, Mumbai, India
| |
Collapse
|
5
|
Zhang F, Biswas M, Massah S, Lee J, Lingadahalli S, Wong S, Wells C, Foo J, Khan N, Morin H, Saxena N, Kung SY, Sun B, Parra Nuñez A, Sanchez C, Chan N, Ung L, Altıntaş U, Bui J, Wang Y, Fazli L, Oo H, Rennie P, Lack N, Cherkasov A, Gleave M, Gsponer J, Lallous N. Dynamic phase separation of the androgen receptor and its coactivators key to regulate gene expression. Nucleic Acids Res 2022; 51:99-116. [PMID: 36535377 PMCID: PMC9841400 DOI: 10.1093/nar/gkac1158] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 10/27/2022] [Accepted: 11/21/2022] [Indexed: 12/24/2022] Open
Abstract
Numerous cancers, including prostate cancer (PCa), are addicted to transcription programs driven by specific genomic regions known as super-enhancers (SEs). The robust transcription of genes at such SEs is enabled by the formation of phase-separated condensates by transcription factors and coactivators with intrinsically disordered regions. The androgen receptor (AR), the main oncogenic driver in PCa, contains large disordered regions and is co-recruited with the transcriptional coactivator mediator complex subunit 1 (MED1) to SEs in androgen-dependent PCa cells, thereby promoting oncogenic transcriptional programs. In this work, we reveal that full-length AR forms foci with liquid-like properties in different PCa models. We demonstrate that foci formation correlates with AR transcriptional activity, as this activity can be modulated by changing cellular foci content chemically or by silencing MED1. AR ability to phase separate was also validated in vitro by using recombinant full-length AR protein. We also demonstrate that AR antagonists, which suppress transcriptional activity by targeting key regions for homotypic or heterotypic interactions of this receptor, hinder foci formation in PCa cells and phase separation in vitro. Our results suggest that enhanced compartmentalization of AR and coactivators may play an important role in the activation of oncogenic transcription programs in androgen-dependent PCa.
Collapse
Affiliation(s)
- Fan Zhang
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | | | | | - Joseph Lee
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Shreyas Lingadahalli
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Samantha Wong
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Christopher Wells
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Jane Foo
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Nabeel Khan
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Helene Morin
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Neetu Saxena
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Sonia H Y Kung
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Bei Sun
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Ana Karla Parra Nuñez
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Christophe Sanchez
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Novia Chan
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Lauren Ung
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Umut Berkay Altıntaş
- School of Medicine, Koç University, Rumelifeneri Yolu, Istanbul 34450, Turkey,Koç University Research Centre for Translational Medicine (KUTTAM), Koç University, Rumelifeneri Yolu, Istanbul 34450, Turkey
| | - Jennifer M Bui
- Michael Smith Laboratories, Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Htoo Zarni Oo
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Paul S Rennie
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Nathan A Lack
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada,School of Medicine, Koç University, Rumelifeneri Yolu, Istanbul 34450, Turkey,Koç University Research Centre for Translational Medicine (KUTTAM), Koç University, Rumelifeneri Yolu, Istanbul 34450, Turkey
| | - Artem Cherkasov
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Martin E Gleave
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6, Canada
| | - Jörg Gsponer
- Correspondence may also be addressed to Jörg Gsponer.
| | - Nada Lallous
- To whom correspondence should be addressed. Tel: +1 604 875 4111; Fax: +1 604 875 5654;
| |
Collapse
|
6
|
Shaashua L, Ben-Shmuel A, Pevsner-Fischer M, Friedman G, Levi-Galibov O, Nandakumar S, Barki D, Nevo R, Brown LE, Zhang W, Stein Y, Lior C, Kim HS, Bojmar L, Jarnagin WR, Lecomte N, Mayer S, Stok R, Bishara H, Hamodi R, Levy-Lahad E, Golan T, Porco JA, Iacobuzio-Donahue CA, Schultz N, Tuveson DA, Lyden D, Kelsen D, Scherz-Shouval R. BRCA mutational status shapes the stromal microenvironment of pancreatic cancer linking clusterin expression in cancer associated fibroblasts with HSF1 signaling. Nat Commun 2022; 13:6513. [PMID: 36316305 PMCID: PMC9622893 DOI: 10.1038/s41467-022-34081-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 10/13/2022] [Indexed: 11/12/2022] Open
Abstract
Tumors initiate by mutations in cancer cells, and progress through interactions of the cancer cells with non-malignant cells of the tumor microenvironment. Major players in the tumor microenvironment are cancer-associated fibroblasts (CAFs), which support tumor malignancy, and comprise up to 90% of the tumor mass in pancreatic cancer. CAFs are transcriptionally rewired by cancer cells. Whether this rewiring is differentially affected by different mutations in cancer cells is largely unknown. Here we address this question by dissecting the stromal landscape of BRCA-mutated and BRCA Wild-type pancreatic ductal adenocarcinoma. We comprehensively analyze pancreatic cancer samples from 42 patients, revealing different CAF subtype compositions in germline BRCA-mutated vs. BRCA Wild-type tumors. In particular, we detect an increase in a subset of immune-regulatory clusterin-positive CAFs in BRCA-mutated tumors. Using cancer organoids and mouse models we show that this process is mediated through activation of heat-shock factor 1, the transcriptional regulator of clusterin. Our findings unravel a dimension of stromal heterogeneity influenced by germline mutations in cancer cells, with direct implications for clinical research.
Collapse
Affiliation(s)
- Lee Shaashua
- grid.13992.300000 0004 0604 7563Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Aviad Ben-Shmuel
- grid.13992.300000 0004 0604 7563Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Meirav Pevsner-Fischer
- grid.13992.300000 0004 0604 7563Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Gil Friedman
- grid.13992.300000 0004 0604 7563Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Oshrat Levi-Galibov
- grid.13992.300000 0004 0604 7563Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Subhiksha Nandakumar
- grid.51462.340000 0001 2171 9952Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Debra Barki
- grid.13992.300000 0004 0604 7563Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Reinat Nevo
- grid.13992.300000 0004 0604 7563Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Lauren E. Brown
- grid.189504.10000 0004 1936 7558Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA USA
| | - Wenhan Zhang
- grid.189504.10000 0004 1936 7558Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA USA
| | - Yaniv Stein
- grid.13992.300000 0004 0604 7563Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Chen Lior
- grid.13992.300000 0004 0604 7563Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Han Sang Kim
- grid.5386.8000000041936877XChildren’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY USA ,grid.15444.300000 0004 0470 5454Yonsei Cancer Center, Division of Medical Oncology, Department of Internal Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Linda Bojmar
- grid.5386.8000000041936877XChildren’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY USA ,grid.5640.70000 0001 2162 9922Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - William R. Jarnagin
- grid.51462.340000 0001 2171 9952Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Nicolas Lecomte
- grid.51462.340000 0001 2171 9952David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Shimrit Mayer
- grid.13992.300000 0004 0604 7563Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Roni Stok
- grid.13992.300000 0004 0604 7563Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Hend Bishara
- grid.13992.300000 0004 0604 7563Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Rawand Hamodi
- grid.13992.300000 0004 0604 7563Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Ephrat Levy-Lahad
- grid.415593.f0000 0004 0470 7791The Fuld Family Medical Genetics Institute, Shaare Zedek Medical Center, Jerusalem, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Talia Golan
- grid.12136.370000 0004 1937 0546Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - John A. Porco
- grid.189504.10000 0004 1936 7558Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA USA
| | - Christine A. Iacobuzio-Donahue
- grid.51462.340000 0001 2171 9952David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Nikolaus Schultz
- grid.51462.340000 0001 2171 9952Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - David A. Tuveson
- grid.225279.90000 0004 0387 3667Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY USA
| | - David Lyden
- grid.5386.8000000041936877XChildren’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY USA
| | - David Kelsen
- grid.5386.8000000041936877XGastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY USA
| | - Ruth Scherz-Shouval
- grid.13992.300000 0004 0604 7563Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
7
|
Abstract
The brain, as one of the most lipid-rich organs, heavily relies on lipid transport and distribution to maintain homeostasis and neuronal function. Lipid transport mediated by lipoprotein particles, which are complex structures composed of apolipoproteins and lipids, has been thoroughly characterized in the periphery. Although lipoproteins in the central nervous system (CNS) were reported over half a century ago, the identification of APOE4 as the strongest genetic risk factor for Alzheimer's disease has accelerated investigation of the biology and pathobiology of lipoproteins in the CNS. This review provides an overview of the different components of lipoprotein particles, in particular apolipoproteins, and their involvements in both physiological functions and pathological mechanisms in the CNS.
Collapse
Affiliation(s)
| | - Yuka A Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA;
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA;
| |
Collapse
|
8
|
CircSTK40 contributes to recurrent implantation failure via modulating the HSP90/AKT/FOXO1 axis. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:208-221. [PMID: 34513305 PMCID: PMC8413673 DOI: 10.1016/j.omtn.2021.06.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 06/25/2021] [Indexed: 11/21/2022]
Abstract
Increasing evidence has revealed a close relationship between non-coding RNAs and recurrent implantation failure (RIF). However, the role of circular RNAs (circRNAs) in RIF pathogenesis remains largely unknown. Microarray analyses were used to identify the differentially expressed circRNA-circSTK40. Functional experiments, including decidualization induction and terminal deoxynucleotidyl transferase-mediated nick end labeling (TUNEL) assay, were performed to determine the effects of circSTK40 on human endometrial stromal cells (ESCs). The interactions between circSTK40 and proteins were investigated by RNA pull-down, RNA immunoprecipitation, and co-immunoprecipitation (coIP) assays. We observed that circSTK40 expression was upregulated in the RIF midluteal-phase endometrial samples. circSTK40 overexpression in ESCs inhibited the decidualization process but concurrently enhanced cell survival during stress. Mechanistically, circSTK40 directly bound to HSP90 and CLU, thus functioning as a scaffold to block their interactions and hinder the proteasomal degradation of HSP90. The resulting high levels of HSP90 led to the activation of the AKT pathway and downregulation of FOXO1 expression. Inhibitors of AKT (MK-2206) and HSP90 (17AAG) both abolished the effects of circSTK40 overexpression in ESCs and increased the decidualization levels in a dose-dependent manner. Our findings indicate a novel epigenetic mechanism for RIF pathogenesis involving circSTK40 activity and provide a foundation for targeted treatments in patients with low endometrial receptivity.
Collapse
|
9
|
Westaby D, Maza MDLDFDL, Paschalis A, Jimenez-Vacas JM, Welti J, de Bono J, Sharp A. A New Old Target: Androgen Receptor Signaling and Advanced Prostate Cancer. Annu Rev Pharmacol Toxicol 2021; 62:131-153. [PMID: 34449248 DOI: 10.1146/annurev-pharmtox-052220-015912] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Owing to the development of multiple novel therapies, there has been major progress in the treatment of advanced prostate cancer over the last two decades; however, the disease remains invariably fatal. Androgens and the androgen receptor (AR) play a critical role in prostate carcinogenesis, and targeting the AR signaling axis with abiraterone, enzalutamide, darolutamide, and apalutamide has improved outcomes for men with this lethal disease. Targeting the AR and elucidating mechanisms of resistance to these agents remains central to drug development efforts. This review provides an overview of the evolution and current approaches for targeting the AR in advanced prostate cancer. It describes the biology of AR signaling, explores AR-targeting resistance mechanisms, and discusses future perspectives and promising novel therapeutic strategies. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Daniel Westaby
- The Institute of Cancer Research, London SM2 5NG, United Kingdom; .,The Royal Marsden Hospital, London SM2 5PT, United Kingdom
| | | | - Alec Paschalis
- The Institute of Cancer Research, London SM2 5NG, United Kingdom; .,The Royal Marsden Hospital, London SM2 5PT, United Kingdom
| | | | - Jon Welti
- The Institute of Cancer Research, London SM2 5NG, United Kingdom;
| | - Johann de Bono
- The Institute of Cancer Research, London SM2 5NG, United Kingdom; .,The Royal Marsden Hospital, London SM2 5PT, United Kingdom
| | - Adam Sharp
- The Institute of Cancer Research, London SM2 5NG, United Kingdom; .,The Royal Marsden Hospital, London SM2 5PT, United Kingdom
| |
Collapse
|
10
|
Single-cell RNA sequencing reveals that targeting HSP90 suppresses PDAC progression by restraining mitochondrial bioenergetics. Oncogenesis 2021; 10:22. [PMID: 33658487 PMCID: PMC7930118 DOI: 10.1038/s41389-021-00311-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers, which lacks effective treatment strategies. There is an urgent need for the development of new strategies for PDAC therapy. The genetic and phenotypic heterogeneity of PDAC cancer cell populations poses further challenges in the clinical management of PDAC. In this study, we performed single-cell RNA sequencing to characterize PDAC tumors from KPC mice. Functional studies and clinical analysis showed that PDAC cluster 2 cells with highly Hsp90 expression is much more aggressive than the other clusters. Genetic and pharmacologic inhibition of Hsp90 impaired tumor cell growth both in vitro and in vivo. Further mechanistic study revealed that HSP90 inhibition disrupted the interaction between HSP90 and OPA1, leading to a reduction in mitochondrial cristae amount and mitochondrial energy production. Collectively, our study reveals that HSP90 might be a potential therapeutic target for PDAC.
Collapse
|
11
|
Praharaj PP, Patra S, Panigrahi DP, Patra SK, Bhutia SK. Clusterin as modulator of carcinogenesis: A potential avenue for targeted cancer therapy. Biochim Biophys Acta Rev Cancer 2020; 1875:188500. [PMID: 33385484 DOI: 10.1016/j.bbcan.2020.188500] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/14/2020] [Accepted: 12/24/2020] [Indexed: 12/30/2022]
Abstract
Clusterin (CLU) is an evolutionary conserved molecular chaperone present in different human tissues and fluids and established to be a significant cancer regulator. It controls several cancer-associated cellular events, including cancer cell proliferation, stemness, survival, metastasis, epithelial-mesenchymal transition, therapy resistance, and inhibition of programmed cell death to support cancer growth and recurrence. This multifunctional role of CLU makes it an ideal target for cancer control. More importantly, genetic and antisense-mediated (OGX-011) inhibition of CLU enhances the anticancer potential of different FDA-approved chemotherapeutic drugs at the clinical level, improving patient's survival. In this review, we have discussed the detailed mechanism of CLU-mediated modulation of different cancer-associated signaling pathways. We have also provided updated information on the current preclinical and clinical findings that drive trials in various cancer types for potential targeted cancer therapy.
Collapse
Affiliation(s)
- Prakash Priyadarshi Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Debasna Pritimanjari Panigrahi
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| |
Collapse
|
12
|
Yao M, Sai W, Zheng W, Wang L, Dong Z, Yao D. Secretory Clusterin as a Novel Molecular-targeted Therapy for Inhibiting Hepatocellular Carcinoma Growth. Curr Med Chem 2020; 27:3290-3301. [PMID: 31232234 DOI: 10.2174/0929867326666190624161158] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 03/19/2019] [Accepted: 05/28/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Although secretory clusterin (sCLU) plays a crucial role in Hepatocellular Carcinoma (HCC) cells proliferation, Multiple Drug Resistance (MDR), metastasis and so on, its targeted effects and exact mechanism are still unknown. This review summarizes some new progress in sCLU as a molecular-targeted therapy in the treatment of HCC. METHODS A systematic review of the published English-language literature about sCLU and HCC has been performed using the PubMed and bibliographic databases. Some valuable studies on sCLU in HCC progression were searched for relevant articles with the keywords: HCC, diagnosis, MDR, as molecular-targeted in treatment, and so on. RESULTS The incidence of the positive rate of sCLU was significantly higher in HCC tissues as compared to the surrounding tissues at mRNA or protein level, gradually increasing with tumor-nodemetastasis staging (P<0.05). Also, the abnormal level of sCLU was related to poor differentiation degree, and considered as a useful marker for HCC diagnosis or independent prognosis for patients. Hepatic sCLU could be silenced at mRNA level by specific sCLU-shRNA or by OGX-011 to inhibit cancer cell proliferation with an increase in apoptosis, cell cycle arrest, reversal MDR, alteration of cell migration or invasion behaviors, and a decrease in GSK-3β or AKT phosphorylation in vitro, as well as significant suppression of the xenograft growth by down-regulating β-catenin, p-GSK3β, and cyclinD1 expression in vivo. CONCLUSION Abnormal hepatic sCLU expression should not only be a new diagnostic biomarker but also a novel promising target for inhibiting HCC growth.
Collapse
Affiliation(s)
- Min Yao
- Medical School of Nantong University, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Wenli Sai
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Wenjie Zheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Li Wang
- Medical School of Nantong University, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Zhizhen Dong
- Department of Diagnostics, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Dengfu Yao
- Medical School of Nantong University, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| |
Collapse
|
13
|
Wei ZD, Sun YZ, Tu CX, Qi RQ, Huo W, Chen HD, Gao XH. DNAJA4 deficiency augments hyperthermia-induced Clusterin and ERK activation: two critical protective factors of human keratinocytes from hyperthermia-induced injury. J Eur Acad Dermatol Venereol 2020; 34:2308-2317. [PMID: 32277496 DOI: 10.1111/jdv.16432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/28/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Hyperthermia upregulates DNAJA4, a member of heat shock proteins (HSPs) 40 family, in human keratinocytes and HPV-infected tissue. DNAJA4 deficiency enhances growth arrest induced by hyperthermia. Clusterin (CLU) and phosphorylated ERK (p-ERK) play a role in regulating cell proliferation and apoptosis, under environmental stress. OBJECTIVES To examine the downstream molecules and signalling pathways of DNAJA4 and assess their roles in cell cycle and apoptosis of keratinocytes in response to hyperthermia. METHODS Wild-type and DNAJA4-knockout (KO) HaCaT cells were exposed to either 44 °C (hyperthermia) or 37 °C (control) for 30 min. The expression levels of CLU and p-ERK were determined by RT-PCR and Western blotting. RNAi and PD98059 were used to inhibit the expression of CLU and p-ERK, respectively. Cell viability, cell cycle and apoptosis were analysed by MTS assay and flow cytometry. Fresh biopsy samples of human normal foreskin or condyloma acuminatum (CA) were utilized to examine the expression of CLU and p-ERK after ex vivo culture at 44 °C. RESULTS The expression of CLU and p-ERK was significantly increased by hyperthermia treatment at 44 °C in HaCaT cells, foreskin and HPV-infected tissues. In HaCaT cells subjected to hyperthermia, DNAJA4 deficiency further augmented the expression of CLU and p-ERK. CLU deficiency enhanced the p-ERK expression. Hyperthermia-induced CLU and p-ERK exerted protective roles mainly through inhibiting apoptosis and maintaining cell cycle, respectively. CONCLUSIONS In keratinocytes, CLU and p-ERK are induced by hyperthermia, an effect which can be further enhanced by DNAJA4 deficiency. CLU deficiency also increases p-ERK expression. Both CLU and p-ERK are critical protective factors of human keratinocytes from hyperthermia-induced injury.
Collapse
Affiliation(s)
- Z-D Wei
- China Medical University, Shenyang, China.,Key Laboratory of Immunodermatology, Department of Dermatology, The First Hospital of China Medical University, Ministry of Health and Ministry of Education, China and National Engineering Research Center for Immunodermatoloigcal Theranostics, Shenyang, China.,Department of Dermatology, The 2nd Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Y-Z Sun
- Key Laboratory of Immunodermatology, Department of Dermatology, The First Hospital of China Medical University, Ministry of Health and Ministry of Education, China and National Engineering Research Center for Immunodermatoloigcal Theranostics, Shenyang, China
| | - C-X Tu
- Department of Dermatology, The 2nd Affiliated Hospital of Dalian Medical University, Dalian, China
| | - R-Q Qi
- Key Laboratory of Immunodermatology, Department of Dermatology, The First Hospital of China Medical University, Ministry of Health and Ministry of Education, China and National Engineering Research Center for Immunodermatoloigcal Theranostics, Shenyang, China
| | - W Huo
- Key Laboratory of Immunodermatology, Department of Dermatology, The First Hospital of China Medical University, Ministry of Health and Ministry of Education, China and National Engineering Research Center for Immunodermatoloigcal Theranostics, Shenyang, China
| | - H-D Chen
- Key Laboratory of Immunodermatology, Department of Dermatology, The First Hospital of China Medical University, Ministry of Health and Ministry of Education, China and National Engineering Research Center for Immunodermatoloigcal Theranostics, Shenyang, China
| | - X-H Gao
- China Medical University, Shenyang, China.,Key Laboratory of Immunodermatology, Department of Dermatology, The First Hospital of China Medical University, Ministry of Health and Ministry of Education, China and National Engineering Research Center for Immunodermatoloigcal Theranostics, Shenyang, China
| |
Collapse
|
14
|
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
|
15
|
Tian Y, Wang C, Chen S, Liu J, Fu Y, Luo Y. Extracellular Hsp90α and clusterin synergistically promote breast cancer epithelial-to-mesenchymal transition and metastasis via LRP1. J Cell Sci 2019; 132:jcs.228213. [PMID: 31273033 DOI: 10.1242/jcs.228213] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 06/28/2019] [Indexed: 12/18/2022] Open
Abstract
Extracellular heat shock protein 90 alpha (eHsp90α, also known as HSP90AA1) has been widely reported to promote tumor cell motility and tumor metastasis in various types of cancer. Several extracellular proteins and membrane receptors have been identified as interacting proteins of eHsp90α and mediate its pro-metastasis function. However, the regulatory mechanism of eHsp90α activity remains largely unknown. Here, we report that clusterin, a protein newly demonstrated to interact with eHsp90α, modulates eHsp90α signaling. We found that clusterin potentiated the effects of eHsp90α on activation of the AKT, ERK and NF-κB protein families, epithelial-to-mesenchymal transition (EMT) and migration in breast cancer cells. Furthermore, in vivo investigations demonstrated similar synergistic effects of eHsp90α and clusterin on tumor metastasis. Notably, the effects of eHsp90α and clusterin were mediated by low-density lipoprotein receptor-related protein 1 (LRP1). Proximity ligation assay and co-immunoprecipitation experiments demonstrated that clusterin participated in eHsp90α-LRP1 complex formation, which enhanced the binding affinity of eHsp90α to LRP1. Collectively, our data establish a role of clusterin as a newly discovered modulator of eHsp90α, and unravel detailed molecular mechanisms underlying the synergistic metastasis-promoting effects of clusterin and eHsp90α.
Collapse
Affiliation(s)
- Yang Tian
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China.,Beijing Key Laboratory for Protein Therapeutics, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China
| | - Chunying Wang
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China.,Beijing Key Laboratory for Protein Therapeutics, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China
| | - Shuohua Chen
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China.,Beijing Key Laboratory for Protein Therapeutics, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China
| | - Jie Liu
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China.,Beijing Key Laboratory for Protein Therapeutics, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China
| | - Yan Fu
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China.,Beijing Key Laboratory for Protein Therapeutics, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China
| | - Yongzhang Luo
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Beijing, Haidian district, 100084, China .,Beijing Key Laboratory for Protein Therapeutics, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, Haidian district, 100084, China
| |
Collapse
|
16
|
Foster EM, Dangla-Valls A, Lovestone S, Ribe EM, Buckley NJ. Clusterin in Alzheimer's Disease: Mechanisms, Genetics, and Lessons From Other Pathologies. Front Neurosci 2019; 13:164. [PMID: 30872998 PMCID: PMC6403191 DOI: 10.3389/fnins.2019.00164] [Citation(s) in RCA: 217] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/12/2019] [Indexed: 01/10/2023] Open
Abstract
Clusterin (CLU) or APOJ is a multifunctional glycoprotein that has been implicated in several physiological and pathological states, including Alzheimer's disease (AD). With a prominent extracellular chaperone function, additional roles have been discussed for clusterin, including lipid transport and immune modulation, and it is involved in pathways common to several diseases such as cell death and survival, oxidative stress, and proteotoxic stress. Although clusterin is normally a secreted protein, it has also been found intracellularly under certain stress conditions. Multiple hypotheses have been proposed regarding the origin of intracellular clusterin, including specific biogenic processes leading to alternative transcripts and protein isoforms, but these lines of research are incomplete and contradictory. Current consensus is that intracellular clusterin is most likely to have exited the secretory pathway at some point or to have re-entered the cell after secretion. Clusterin's relationship with amyloid beta (Aβ) has been of great interest to the AD field, including clusterin's apparent role in altering Aβ aggregation and/or clearance. Additionally, clusterin has been more recently identified as a mediator of Aβ toxicity, as evidenced by the neuroprotective effect of CLU knockdown and knockout in rodent and human iPSC-derived neurons. CLU is also the third most significant genetic risk factor for late onset AD and several variants have been identified in CLU. Although the exact contribution of these variants to altered AD risk is unclear, some have been linked to altered CLU expression at both mRNA and protein levels, altered cognitive and memory function, and altered brain structure. The apparent complexity of clusterin's biogenesis, the lack of clarity over the origin of the intracellular clusterin species, and the number of pathophysiological functions attributed to clusterin have all contributed to the challenge of understanding the role of clusterin in AD pathophysiology. Here, we highlight clusterin's relevance to AD by discussing the evidence linking clusterin to AD, as well as drawing parallels on how the role of clusterin in other diseases and pathways may help us understand its biological function(s) in association with AD.
Collapse
Affiliation(s)
| | | | | | | | - Noel J. Buckley
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Bouaouiche S, Magadoux L, Dondaine L, Reveneau S, Isambert N, Bettaieb A, Jeannin JF, Laurens V, Plenchette S. Glyceryl trinitrate‑induced cytotoxicity of docetaxel‑resistant prostatic cancer cells is associated with differential regulation of clusterin. Int J Oncol 2019; 54:1446-1456. [PMID: 30720069 DOI: 10.3892/ijo.2019.4708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/21/2018] [Indexed: 11/05/2022] Open
Abstract
Metastatic castration resistant prostate cancer (mCRPC) relapse due to acquired resistance to chemotherapy, such as docetaxel, remains a major threat to patient survival. Resistance of mCRPC to docetaxel can be associated with elevated levels of soluble clusterin (sCLU) and growth differentiation factor‑15 (GDF‑15). Any strategies aiming to modulate sCLU and/or GDF‑15 in docetaxel‑resistant prostate cancer cells present a therapeutic interest. The present study reports the cytotoxic effect of a nitric oxide donor, glyceryl trinitrate (GTN), on docetaxel‑resistant mCRPC human cell lines and demonstrates that GTN displays greater inhibition of cell viability toward docetaxel‑resistant mCRPC cells than on mCRPC cells. It is also demonstrated that GTN modulates the level of expression of clusterin (CLU) which is dependent of GDF‑15, two markers associated with docetaxel resistance in prostate cancer. The results indicate that GTN represses the level of expression of the cytoprotective isoform of CLU (sCLU) and can increase the level of expression of the cytotoxic isoform (nuclear CLU) in docetaxel resistant cells. Furthermore, it was observed that GTN differentially regulates the level of the precursor form of GDF‑15 between resistant and parental cells, and that recombinant GDF‑15 can modulate the expression of CLU isoforms and counteract GTN‑induced cytotoxicity in resistant cells. A link was established between GDF‑15 and the expression of CLU isoforms. The present study thus revealed GTN as a potential therapeutic strategy to overcome docetaxel‑resistant mCRPC.
Collapse
Affiliation(s)
- Sarra Bouaouiche
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Lea Magadoux
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Lucile Dondaine
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Sylvie Reveneau
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | | | - Ali Bettaieb
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Jean-François Jeannin
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Veronique Laurens
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| | - Stephanie Plenchette
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, F‑75000 Paris, France
| |
Collapse
|
18
|
Centenera MM, Selth LA, Ebrahimie E, Butler LM, Tilley WD. New Opportunities for Targeting the Androgen Receptor in Prostate Cancer. Cold Spring Harb Perspect Med 2018; 8:a030478. [PMID: 29530945 PMCID: PMC6280715 DOI: 10.1101/cshperspect.a030478] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Recent genomic analyses of metastatic prostate cancer have provided important insight into adaptive changes in androgen receptor (AR) signaling that underpin resistance to androgen deprivation therapies. Novel strategies are required to circumvent these AR-mediated resistance mechanisms and thereby improve prostate cancer survival. In this review, we present a summary of AR structure and function and discuss mechanisms of AR-mediated therapy resistance that represent important areas of focus for the development of new therapies.
Collapse
Affiliation(s)
- Margaret M Centenera
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide SA 5005, Australia
- South Australian Health and Medical Research Institute, Adelaide SA 5001, Australia
| | - Luke A Selth
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide SA 5005, Australia
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide SA 5005, Australia
| | - Esmaeil Ebrahimie
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide SA 5005, Australia
| | - Lisa M Butler
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide SA 5005, Australia
- South Australian Health and Medical Research Institute, Adelaide SA 5001, Australia
| | - Wayne D Tilley
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide SA 5005, Australia
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide SA 5005, Australia
| |
Collapse
|
19
|
Kim JH, Jung YJ, Choi B, Lee NL, Lee HJ, Kwak SY, Kwon Y, Na Y, Lee YS. Overcoming HSP27-mediated resistance by altered dimerization of HSP27 using small molecules. Oncotarget 2018; 7:53178-53190. [PMID: 27449291 PMCID: PMC5288177 DOI: 10.18632/oncotarget.10629] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 07/06/2016] [Indexed: 12/15/2022] Open
Abstract
Heat shock protein 27 (HSP27, HSPB1) is an anti-apoptotic protein characterized for its tumorigenic and metastatic properties, and now referenced as a major therapeutic target in many types of cancer. The biochemical properties of HSP27 rely on a structural oligomeric and dynamic organization that is important for its chaperone activity. Down-regulation by small interfering RNA or inhibition with a dominant-negative mutant efficiently counteracts the anti-apoptotic and protective properties of HSP27. However, unlike other HSPs such as HSP90 and HSP70, small molecule approaches for neutralization of HSP27 are not well established because of the absence of an ATP binding domain. Previously, we found that a small molecule, zerumbone (ZER), induced altered dimerization of HSP27 by cross linking the cysteine residues required to build a large oligomer, led to sensitization in combination with radiation. In this study, we identified another small molecule, a xanthone compound, more capable of altering dimeric HSP27 than ZER and yielding sensitization in human lung cancer cells when combined with HSP90 inhibitors or standard anticancer modalities such as irradiation and cytotoxic anticancer drugs. Therefore, altered dimerization of HSP27 represents a good strategy for anticancer therapy in HSP27-overexpressing cancer cells.
Collapse
Affiliation(s)
- Jee Hye Kim
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-720, Korea
| | - Ye Jin Jung
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-720, Korea
| | - Byeol Choi
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-720, Korea
| | - Na Lim Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-720, Korea
| | - Hae Jun Lee
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul, 139-706, Korea
| | - Soo Yeon Kwak
- College of Pharmacy, CHA University, Pocheon, 487-010, Korea
| | - Youngjoo Kwon
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-720, Korea
| | - Younghwa Na
- College of Pharmacy, CHA University, Pocheon, 487-010, Korea
| | - Yun-Sil Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-720, Korea
| |
Collapse
|
20
|
Vlachostergios PJ, Paddock M, Molina AM. Molecular Targeted Therapies of Prostate Cancer. MOLECULAR PATHOLOGY LIBRARY 2018. [DOI: 10.1007/978-3-319-64096-9_29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
21
|
Nishikawa M, Miyake H, Gleave M, Fujisawa M. Effect of Targeting Clusterin Using OGX-011 on Antitumor Activity of Temsirolimus in a Human Renal Cell Carcinoma Model. Target Oncol 2017; 12:69-79. [PMID: 27526062 DOI: 10.1007/s11523-016-0448-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND It has not been well documented that the modulation of stress response mediates the efficacy of the mammalian target of rapamycin (mTOR) inhibitor in renal cell carcinoma (RCC). OBJECTIVE The objective of this study was to investigate whether the activity of the mTOR inhibitor temsirolimus against RCC could be enhanced by OGX-011, an antisense oligodeoxynucleotide (ODN) targeting the stress-activated chaperone clusterin. METHODS We investigated the efficacy of combined treatment with temsirolimus plus OGX-011 in a human RCC Caki-1 model focusing on the effects on apoptotic and autophagic pathways. RESULTS Although clusterin expression was increased by temsirolims, additional treatment of Caki-1 with OGX-011 significantly inhibited clusterin upregulation (p < 0.05). Combined treatment of temsirolimus and OGX-011 synergistically enhanced the sensitivity of Caki-1 to temsirolimus (p < 0.01), reducing the IC50 by approximately 50 %. Apoptotic changes were marked in Caki-1 following combined treatment with a sublethal dose of temsirolimus and OGX-011, accompanying the significant downregulation of Mcl-1 (p < 0.05), but not with either agent alone. Furthermore, this combined treatment markedly blocked the temsirolimus-induced activation of autophagy in Caki-1 (p < 0.01). In-vivo systemic administration of temsirolimus plus OGX-011 significantly inhibited the growth of Caki-1 tumors compared with that of temsirolimus plus control ODN (p < 0.05). CONCLUSIONS Silencing of clusterin using OGX-011 resulted in the further enhancement of proapoptotic activity as well as the marked attenuation of the autophagic pathway induced by temsirolimus in a human RCC model. Thus, the combined use of OGX-011 could be a promising strategy through the enhanced cytotoxic activity of temsirolimus against RCC.
Collapse
Affiliation(s)
- Masatomo Nishikawa
- Division of Urology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Hideaki Miyake
- Department of Urology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-Ku, Hamamatsu, 431-3192, Japan.
| | - Martin Gleave
- Vancouver Prostate Centre and University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada
| | - Masato Fujisawa
- Division of Urology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| |
Collapse
|
22
|
Shiota M, Yokomizo A, Takeuchi A, Kashiwagi E, Dejima T, Inokuchi J, Tatsugami K, Uchiumi T, Eto M. Protein kinase C regulates Twist1 expression via NF-κB in prostate cancer. Endocr Relat Cancer 2017; 24:171-180. [PMID: 28223364 DOI: 10.1530/erc-16-0384] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 02/21/2017] [Indexed: 11/08/2022]
Abstract
The progression of prostate cancer to metastatic and castration-resistant disease represents a critical step. We previously showed that protein kinase C (PKC) activation followed by Twist1 and androgen receptor (AR) induction played a critical role in castration resistance, but the precise molecular mechanism remains unknown. This study aimed to elucidate the relevant molecular mechanism, focusing on NF-κB transcription factor. We examined the activity of NF-κB after PKC inhibition, and the expression of Twist1 and AR after inhibition of NF-κB in human prostate cancer cells. We also investigated the status of PKC/NF-κB after inhibition of AR signaling in cells resistant to hormonal therapy. As a result, inhibition of PKC signaling using knockdown and small-molecule inhibition of PKC suppressed RelA activity, while blocking NF-κB suppressed Twist1 and AR expression. Conversely, inhibition of AR signaling by androgen depletion and the novel antiandrogen enzalutamide induced PKC and RelA activation, resulting in Twist1/AR induction at the transcript level. Moreover, inhibition of NF-κB signaling prevented enzalutamide-induced Twist1 and AR induction. Finally, NF-κB was activated in both castration-resistant and enzalutamide-resistant cells. In conclusion, NF-κB signaling was responsible for Twist1 upregulation by PKC in response to AR inhibition, resulting in aberrant activation of AR. NF-κB signaling thus appears to play a critical role in promoting both castration resistance and enzalutamide resistance in PKC/Twist1 signaling in prostate cancer.
Collapse
Affiliation(s)
- Masaki Shiota
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akira Yokomizo
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ario Takeuchi
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Kashiwagi
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takashi Dejima
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Junichi Inokuchi
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsunori Tatsugami
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Uchiumi
- Department of Clinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masatoshi Eto
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
23
|
Woo JK, Jang JE, Kang JH, Seong JK, Yoon YS, Kim HC, Lee SJ, Oh SH. Lectin, Galactoside-Binding Soluble 3 Binding Protein Promotes 17-N-Allylamino-17-demethoxygeldanamycin Resistance through PI3K/Akt Pathway in Lung Cancer Cell Line. Mol Cancer Ther 2017; 16:1355-1365. [PMID: 28336809 DOI: 10.1158/1535-7163.mct-16-0574] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/21/2016] [Accepted: 02/23/2017] [Indexed: 11/16/2022]
Abstract
Heat shock protein 90 (HSP90) stabilizing oncoproteins has been an attractive target in cancer therapy. 17-N-Allylamino-17-demethoxygeldanamycin (17-AAG), an HSP90 inhibitor, was tested in phase II/III clinical trials, but due to lack of efficacy, clinical evaluation of 17-AAG has achieved limited success, which led to resistance to 17-AAG. However, the mechanism of 17-AAG resistance has not clearly been identified. Here, we identified LGALS3BP (Lectin, galactoside-binding soluble 3 binding protein), a secretory glycoprotein, as a 17-AAG resistance factor. In the clinical reports, it was suggested that LGALS3BP was associated with low survival rate, development of cancer progression, and enhancement of metastasis in human cancers. As we confirmed that the LGALS3BP level was increased in 17-AAG-resistant cells (H1299_17R) compared with that of the parental cell line (H1299_17P), knockdown of LGALS3BP expression increased sensitivity to 17-AAG in H1299_17R cells. Overexpression of LGALS3BP also augmented PI3K/Akt and ERK signaling pathways. Furthermore, we determined that the PI3K/Akt signaling pathway was involved in LGALS3BP-mediated 17-AAG resistance in vitro and in vivo, demonstrating that LGALS3BP mediates the resistance against 17-AAG through PI3K/Akt activation rather than ERK activation. These findings suggest that LGALS3BP would be a target to overcome resistance to 17-AAG in lung cancer. For example, the combination of 17-AAG and PI3K/Akt inhibitor would effectively suppress acquired resistance to 17-AAG. In conclusion, targeting of LGALS3BP-mediated-specific survival signaling pathway in resistant cells may provide a novel therapeutic model for the cancer therapy. Mol Cancer Ther; 16(7); 1355-65. ©2017 AACR.
Collapse
Affiliation(s)
- Jong Kyu Woo
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Jeong-Eun Jang
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Ju-Hee Kang
- Research Institute National Cancer Center, Goyang-si, Republic of Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yeo Sung Yoon
- Laboratory of Anatomy and Cell Biology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hyoung-Chin Kim
- Biomedical Mouse Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Republic of Korea
| | - Sang-Jin Lee
- Research Institute National Cancer Center, Goyang-si, Republic of Korea.
| | - Seung Hyun Oh
- College of Pharmacy, Gachon University, Incheon, Republic of Korea.
| |
Collapse
|
24
|
Dejima T, Imada K, Takeuchi A, Shiota M, Leong J, Tombe T, Tam K, Fazli L, Naito S, Gleave ME, Ong CJ. Suppression of LIM and SH3 Domain Protein 1 (LASP1) Negatively Regulated by Androgen Receptor Delays Castration Resistant Prostate Cancer Progression. Prostate 2017; 77:309-320. [PMID: 27775154 DOI: 10.1002/pros.23269] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/11/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND LIM and SH3 domain protein 1 (LASP1) has been implicated in several human malignancies and has been shown to predict PSA recurrence in prostate cancer. However, the anti-tumor effect of LASP1 knockdown and the association between LASP1 and the androgen receptor (AR) remains unclear. The aim of this study is to clarify the significance of LASP1 as a target for prostate cancer, and to test the effect of silencing LASP1 in vivo using antisense oligonucleotides (ASO). METHODS A tissue microarray (TMA) was performed to characterize the differences in LASP1 expression in prostate cancer treated after hormone deprivation therapy. Flow cytometry was used to analyze cell cycle. We designed LASP1 ASO for knockdown of LASP1 in vivo studies. RESULTS The expression of LASP1 in TMA was increased after androgen ablation and persisted in castration resistant prostate cancer (CRPC). Also in TMA, compared with LNCaP cell, LASP1 expression is elevated in CRPC cell lines (C4-2 and VehA cells). Interestingly, suppression of AR elevated LASP1 expression conversely, AR activation decreased LASP1 expression. Silencing of LASP1 reduced cell growth through G1 arrest which was accompanied by a decrease of cyclin D1. Forced overexpression of LASP1 promoted cell cycle and induced cell growth which was accompanied by an increase of cyclin D1. Systemic administration of LASP1 ASO with athymic mice significantly inhibited tumor growth in CRPC xenografts. CONCLUSIONS These results indicate that LASP1 is negatively regulated by AR at the transcriptional level and promotes tumor growth through induction of cell cycle, ultimately suggesting that LASP1 may be a potential target in prostate cancer treatment. Prostate 77:309-320, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Takashi Dejima
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenjiro Imada
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ario Takeuchi
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jeffrey Leong
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tabitha Tombe
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kevin Tam
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Seiji Naito
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Martin E Gleave
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher J Ong
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
25
|
Abstract
INTRODUCTION Clusterin (CLU) is a stress-activated, ATP-independent molecular chaperone, normally secreted from cells, that is up-regulated in Alzheimer disease and in many cancers. It plays important roles in protein homeostasis/proteostasis, inhibition of cell death pathways, and modulation of pro-survival signalling and transcriptional networks. Changes in the CLU gene locus are highly associated with Alzheimer disease, and many therapy-resistant cancers over-express CLU. The extensive post-translational processing and heterogeneous oligomerization of CLU have so far prevented any definitive structure determination. This in turn has meant that targeting CLU with small molecule inhibitors is challenging. Therefore, inhibiting CLU at the gene-expression level using siRNA or antisense is a valid approach to inhibit its function. Areas covered: This article reviews recent advances regarding the role of CLU in proteostasis, cellular trafficking, human diseases, and signalling pathways involved in oncogenesis. It addresses the rationale for CLU as a therapeutic target in cancer, and the current status of pre-clinical and clinical studies using CLU antisense inhibitor OGX011. Expert opinion: Discusses challenges facing the therapeutic targeting of CLU including rapid changes in the treatment landscape for prostate cancer with multiple new FDA approved drugs, selection of windows of intervention, and potential side effects when silencing CLU expression.
Collapse
Affiliation(s)
- Mark R Wilson
- a School of Biological Sciences , University of Wollongong , Wollongong , Australia
| | - Amina Zoubeidi
- b Department of Urologic Sciences, Vancouver Prostate Centre , University of British Columbia and Vancouver General Hospital , Vancouver , Canada
| |
Collapse
|
26
|
Armstrong HK, Koay YC, Irani S, Das R, Nassar ZD, Selth LA, Centenera MM, McAlpine SR, Butler LM. A Novel Class of Hsp90 C-Terminal Modulators Have Pre-Clinical Efficacy in Prostate Tumor Cells Without Induction of a Heat Shock Response. Prostate 2016; 76:1546-1559. [PMID: 27526951 DOI: 10.1002/pros.23239] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/15/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND While there is compelling rationale to use heat shock protein 90 (Hsp90) inhibitors for treatment of advanced prostate cancer, agents that target the N-terminal ATP-binding site of Hsp90 have shown little clinical benefit. These N-terminal binding agents induce a heat shock response that activates compensatory heat shock proteins, which is believed to contribute in part to the agents' lack of efficacy. Here, we describe the functional characterization of two novel agents, SM253 and SM258, that bind the N-middle linker region of Hsp90, resulting in reduced client protein activation and preventing C-terminal co-chaperones and client proteins from binding to Hsp90. METHODS Inhibition of Hsp90 activity in prostate cancer cells by SM253 and SM 258 was assessed by pull-down assays. Cell viability, proliferation and apoptosis were assayed in prostate cancer cell lines (LNCaP, 22Rv1, PC-3) cultured with N-terminal Hsp90 inhibitors (AUY922, 17-AAG), SM253 or SM258. Expression of HSR heat shock proteins, Hsp90 client proteins and co-chaperones was assessed by immunoblotting. Efficacy of the SM compounds was evaluated in human primary prostate tumors cultured ex vivo by immunohistochemical detection of Hsp70 and Ki67. RESULTS SM253 and SM258 exhibit antiproliferative and pro-apoptotic activity in multiple prostate cancer cell lines (LNCaP, 22Rv1, and PC-3) at low micromolar concentrations. Unlike the N-terminal inhibitors AUY922 and 17-AAG, these SM agents do not induce expression of Hsp27, Hsp40, or Hsp70, proteins that are characteristic of the heat shock response, in any of the prostate cell lines analyzed. Notably, SM258 significantly reduced proliferation within 2 days in human primary prostate tumors cultured ex vivo, without the significant induction of Hsp70 that was caused by AUY922 in the tissues. CONCLUSIONS Our findings provide the first evidence of efficacy of this class of C-terminal modulators of Hsp90 in human prostate tumors, and indicate that further evaluation of these promising new agents is warranted. Prostate 76:1546-1559, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Heather K Armstrong
- School of Medicine and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Yen Chin Koay
- School of Chemistry, University of New South Wales, Sydney, New South Wales, Australia
| | - Swati Irani
- School of Medicine and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Rajdeep Das
- School of Medicine and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia, Australia
- Dame Roma Mitchell Cancer Research Laboratories, School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Zeyad D Nassar
- School of Medicine and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Luke A Selth
- School of Medicine and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia, Australia
- Dame Roma Mitchell Cancer Research Laboratories, School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Margaret M Centenera
- School of Medicine and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Shelli R McAlpine
- School of Chemistry, University of New South Wales, Sydney, New South Wales, Australia.
| | - Lisa M Butler
- School of Medicine and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia, Australia.
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.
| |
Collapse
|
27
|
Hayashi T, Gust KM, Wyatt AW, Goriki A, Jäger W, Awrey S, Li N, Oo HZ, Altamirano-Dimas M, Buttyan R, Fazli L, Matsubara A, Black PC. Not all NOTCH Is Created Equal: The Oncogenic Role of NOTCH2 in Bladder Cancer and Its Implications for Targeted Therapy. Clin Cancer Res 2016; 22:2981-92. [PMID: 26769750 DOI: 10.1158/1078-0432.ccr-15-2360] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/31/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Recent molecular analyses of bladder cancer open the door to significant advances in targeted therapies. NOTCH has been identified as a tumor suppressor in bladder cancer, but prior reports have focused on NOTCH1 Here we hypothesized that NOTCH2 is an oncogene suitable for therapeutic targeting in bladder cancer. EXPERIMENTAL DESIGN We studied genomic aberrations of NOTCH, compared survival and tumor progression according to NOTCH2 expression levels, and studied NOTCH2 function in vitro and vivo RESULTS We report a high rate of NOTCH2 copy number gain in bladder cancer. High NOTCH2 expression was identified especially in the basal subtype and in mesenchymal tumors. NOTCH2 activation correlated with adverse disease parameters and worse prognosis by immunohistochemistry. Forced overexpression of the intracellular domain of NOTCH2 (N2ICD) induced cell growth and invasion by cell-cycle progression, maintenance of stemness and epithelial-to-mesenchymal transition (EMT). These effects were abrogated by silencing of CSL, indicating that the effects were mediated through the canonical NOTCH signaling pathway. In an orthotopic xenograft model, forced overexpression of N2ICD increased growth, invasion, and metastasis. To explore the potential for therapeutic targeting of NOTCH2, we first silenced the receptor with shRNA and subsequently treated with a specific inhibitory antibody. Both interventions decreased cell growth, invasion, and metastasis in vitro and in the orthotopic xenograft model. CONCLUSIONS We have demonstrated that NOTCH2 acts as an oncogene that promotes bladder cancer growth and metastasis through EMT, cell-cycle progression, and maintenance of stemness. Inhibition of NOTCH2 is a rational novel treatment strategy for invasive bladder cancer. Clin Cancer Res; 22(12); 2981-92. ©2016 AACR.
Collapse
Affiliation(s)
- Tetsutaro Hayashi
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada. Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada. Department of Urology, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Kilian M Gust
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada. Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Alexander W Wyatt
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada. Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Akihiro Goriki
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada. Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada. Department of Urology, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Wolfgang Jäger
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada. Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Shannon Awrey
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada. Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Na Li
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada. Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Htoo Zarni Oo
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada. Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Manuel Altamirano-Dimas
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada. Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ralph Buttyan
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada. Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ladan Fazli
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada. Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Akio Matsubara
- Department of Urology, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Peter C Black
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada. Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
28
|
Ory B, Baud'huin M, Verrecchia F, Royer BBL, Quillard T, Amiaud J, Battaglia S, Heymann D, Redini F, Lamoureux F. Blocking HSP90 Addiction Inhibits Tumor Cell Proliferation, Metastasis Development, and Synergistically Acts with Zoledronic Acid to Delay Osteosarcoma Progression. Clin Cancer Res 2015; 22:2520-33. [PMID: 26712686 DOI: 10.1158/1078-0432.ccr-15-1925] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 12/04/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Despite recent improvements in therapeutic management of osteosarcoma, ongoing challenges in improving the response to chemotherapy warrants the development of new strategies to improve overall patient survival. Among them, HSP90 is a molecular chaperone involved in the maturation and stability of various oncogenic proteins leading to tumor cells survival and disease progression. We assessed the antitumor properties of a synthetic HSP90 inhibitor, PF4942847, alone or in combination with zoledronic acid in osteosarcoma. EXPERIMENTAL DESIGN The effects of PF4942847 were evaluated on human osteosarcoma cells growth and apoptosis. Signaling pathways were analyzed by Western blotting. The consequence of HSP90 therapy combined or not with zoledronic acid was evaluated in mice bearing HOS-MNNG xenografts on tumor growth, associated bone lesions, and pulmonary metastasis. The effect of PF4942847 on osteoclastogenesis was assessed on human CD14(+) monocytes. RESULTS In osteosarcoma cell lines, PF4942847 inhibited cell growth in a dose-dependent manner (IC50 ±50 nmol/L) and induced apoptosis with an increase of sub-G1 fraction and cleaved PARP. These biologic events were accompanied by decreased expression of Akt, p-ERK, c-Met, and c-RAF1. When administered orally to mice bearing osteosarcoma tumors, PF4942847 significantly inhibited tumor growth by 80%, prolonged survival compared with controls, and inhibited pulmonary metastases by blocking c-Met, FAK, and MMP9 signaling. In contrast to 17-allylamino-17-demethoxygeldanamycin (17-AAG), PF4942847 did not induce osteoclast differentiation, and synergistically acted with zoledronic acid to delay osteosarcoma progression and prevent bone lesions. CONCLUSIONS All these data provide a strong rationale for clinical evaluation of PF4942847 alone or in combination with zoledronic acid in osteosarcoma. Clin Cancer Res; 22(10); 2520-33. ©2015 AACR.
Collapse
Affiliation(s)
- Benjamin Ory
- Université de Nantes, Nantes atlantique universités, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France. INSERM, UMR 957, Nantes, France. LUNAM Université, Nantes, France. Equipe labellisée LIGUE 2012, Nantes, France
| | - Marc Baud'huin
- Université de Nantes, Nantes atlantique universités, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France. INSERM, UMR 957, Nantes, France. LUNAM Université, Nantes, France. Equipe labellisée LIGUE 2012, Nantes, France. CHU de Nantes, Nantes, France
| | - Franck Verrecchia
- Université de Nantes, Nantes atlantique universités, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France. INSERM, UMR 957, Nantes, France. LUNAM Université, Nantes, France. Equipe labellisée LIGUE 2012, Nantes, France
| | - Bénédicte Brounais-Le Royer
- Université de Nantes, Nantes atlantique universités, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France. INSERM, UMR 957, Nantes, France. LUNAM Université, Nantes, France. Equipe labellisée LIGUE 2012, Nantes, France
| | - Thibaut Quillard
- Université de Nantes, Nantes atlantique universités, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France. INSERM, UMR 957, Nantes, France. LUNAM Université, Nantes, France. Equipe labellisée LIGUE 2012, Nantes, France
| | - Jérôme Amiaud
- Université de Nantes, Nantes atlantique universités, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France. INSERM, UMR 957, Nantes, France. LUNAM Université, Nantes, France. Equipe labellisée LIGUE 2012, Nantes, France
| | - Séverine Battaglia
- Université de Nantes, Nantes atlantique universités, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France. INSERM, UMR 957, Nantes, France. LUNAM Université, Nantes, France. Equipe labellisée LIGUE 2012, Nantes, France
| | - Dominique Heymann
- Université de Nantes, Nantes atlantique universités, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France. INSERM, UMR 957, Nantes, France. LUNAM Université, Nantes, France. Equipe labellisée LIGUE 2012, Nantes, France. CHU de Nantes, Nantes, France
| | - Francoise Redini
- Université de Nantes, Nantes atlantique universités, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France. INSERM, UMR 957, Nantes, France. LUNAM Université, Nantes, France. Equipe labellisée LIGUE 2012, Nantes, France
| | - Francois Lamoureux
- Université de Nantes, Nantes atlantique universités, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France. INSERM, UMR 957, Nantes, France. LUNAM Université, Nantes, France. Equipe labellisée LIGUE 2012, Nantes, France.
| |
Collapse
|
29
|
Shiota M, Itsumi M, Takeuchi A, Imada K, Yokomizo A, Kuruma H, Inokuchi J, Tatsugami K, Uchiumi T, Oda Y, Naito S. Crosstalk between epithelial-mesenchymal transition and castration resistance mediated by Twist1/AR signaling in prostate cancer. Endocr Relat Cancer 2015; 22:889-900. [PMID: 26311513 DOI: 10.1530/erc-15-0225] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 08/26/2015] [Indexed: 02/04/2023]
Abstract
Although invasive and metastatic progression via the epithelial-mesenchymal transition (EMT) and acquisition of resistance to castration are both critical steps in prostate cancer, the molecular mechanism of this interaction remains unclear. In this study, we aimed to elucidate the interaction of signaling between castration resistance and EMT, and to apply this information to the development of a novel therapeutic concept using transforming growth factor-β (TGF-β) inhibitor SB525334 combined with androgen-deprivation therapy against prostate cancer using an in vivo model. This study revealed that an EMT inducer (TGF-β) induced full-length androgen receptor (AR) and AR variant expression. In addition, a highly invasive clone showed augmented full-length AR and AR variant expression as well as acquisition of castration resistance. Conversely, full-length AR and AR as well as Twist1 and mesenchymal molecules variant expression were up-regulated in castration-resistant LNCaP xenograft. Finally, TGF-β inhibitor suppressed Twist1 and AR expression as well as prostate cancer growth combined with castration. Taken together, these results demonstrate that Twist1/AR signaling was augmented in castration resistant as well as mesenchymal-phenotype prostate cancer, indicating the molecular mechanism of mutual and functional crosstalk between EMT and castration resistance, which may play a crucial role in prostate carcinogenesis and progression.
Collapse
MESH Headings
- Adenocarcinoma/drug therapy
- Adenocarcinoma/genetics
- Adenocarcinoma/pathology
- Adenocarcinoma/physiopathology
- Adenocarcinoma/surgery
- Androgens
- Animals
- Cell Line, Tumor
- Cell Transformation, Neoplastic
- Combined Modality Therapy
- Epithelial-Mesenchymal Transition/physiology
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Imidazoles/pharmacology
- Imidazoles/therapeutic use
- Male
- Mice
- Mice, Nude
- Neoplasm Invasiveness
- Neoplasm Proteins/physiology
- Neoplasms, Hormone-Dependent/drug therapy
- Neoplasms, Hormone-Dependent/genetics
- Neoplasms, Hormone-Dependent/pathology
- Neoplasms, Hormone-Dependent/physiopathology
- Neoplasms, Hormone-Dependent/surgery
- Nuclear Proteins/physiology
- Orchiectomy
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms/physiopathology
- Prostatic Neoplasms/surgery
- Quinoxalines/pharmacology
- Quinoxalines/therapeutic use
- RNA Interference
- RNA, Small Interfering/genetics
- Random Allocation
- Receptors, Androgen/biosynthesis
- Receptors, Androgen/chemistry
- Receptors, Androgen/genetics
- Receptors, Androgen/physiology
- Signal Transduction
- Transforming Growth Factor beta/antagonists & inhibitors
- Twist-Related Protein 1/physiology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Masaki Shiota
- Departments of UrologyAnatomic PathologyGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, JapanDepartment of UrologySchool of Medicine, Jikei University, Tokyo 105-0003, JapanDepartment of Clinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Momoe Itsumi
- Departments of UrologyAnatomic PathologyGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, JapanDepartment of UrologySchool of Medicine, Jikei University, Tokyo 105-0003, JapanDepartment of Clinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ario Takeuchi
- Departments of UrologyAnatomic PathologyGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, JapanDepartment of UrologySchool of Medicine, Jikei University, Tokyo 105-0003, JapanDepartment of Clinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kenjiro Imada
- Departments of UrologyAnatomic PathologyGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, JapanDepartment of UrologySchool of Medicine, Jikei University, Tokyo 105-0003, JapanDepartment of Clinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan Departments of UrologyAnatomic PathologyGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, JapanDepartment of UrologySchool of Medicine, Jikei University, Tokyo 105-0003, JapanDepartment of Clinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Akira Yokomizo
- Departments of UrologyAnatomic PathologyGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, JapanDepartment of UrologySchool of Medicine, Jikei University, Tokyo 105-0003, JapanDepartment of Clinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hidetoshi Kuruma
- Departments of UrologyAnatomic PathologyGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, JapanDepartment of UrologySchool of Medicine, Jikei University, Tokyo 105-0003, JapanDepartment of Clinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Junichi Inokuchi
- Departments of UrologyAnatomic PathologyGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, JapanDepartment of UrologySchool of Medicine, Jikei University, Tokyo 105-0003, JapanDepartment of Clinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Katsunori Tatsugami
- Departments of UrologyAnatomic PathologyGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, JapanDepartment of UrologySchool of Medicine, Jikei University, Tokyo 105-0003, JapanDepartment of Clinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takeshi Uchiumi
- Departments of UrologyAnatomic PathologyGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, JapanDepartment of UrologySchool of Medicine, Jikei University, Tokyo 105-0003, JapanDepartment of Clinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoshinao Oda
- Departments of UrologyAnatomic PathologyGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, JapanDepartment of UrologySchool of Medicine, Jikei University, Tokyo 105-0003, JapanDepartment of Clinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Seiji Naito
- Departments of UrologyAnatomic PathologyGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, JapanDepartment of UrologySchool of Medicine, Jikei University, Tokyo 105-0003, JapanDepartment of Clinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
30
|
Wang C, Jin G, Jin H, Wang N, Luo Q, Zhang Y, Gao D, Jiang K, Gu D, Shen Q, Huo X, Hu F, Ge T, Zhao F, Chu W, Shu H, Yao M, Cong W, Qin W. Clusterin facilitates metastasis by EIF3I/Akt/MMP13 signaling in hepatocellular carcinoma. Oncotarget 2015; 6:2903-16. [PMID: 25609201 PMCID: PMC4413626 DOI: 10.18632/oncotarget.3093] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 12/25/2014] [Indexed: 12/12/2022] Open
Abstract
Clusterin (CLU) is a stress-induced chaperone that confers proliferative and survival advantages to cancer cells. However, effects and molecular mechanisms of CLU in hepatocellular carcinoma (HCC) metastasis are still unknown. In this study, HCC tissue array (n = 198) was utilized to investigate correlation between CLU expression and clinicopathological features. Overexpression of CLU in HCC tissues was correlated with shorter overall survival and higher tumor recurrence. In vitro and in vivo assays demonstrated that silencing CLU attenuated the invasion and metastasis of HCC cells, whereas ectopic overexpression of CLU resulted in the forced metastasis of HCC cells. We also revealed that CLU activated Akt signaling through complexing with eukaryotic translation initiation factor 3 subunit I (EIF3I), which in turn promoted matrix metalloproteinase 13 (MMP13) expression and HCC metastasis. Positive correlations between CLU and MMP13, p-Akt, or EIF3I were found in HCC tissues. We further observed that CLU knockdown using the CLU inhibitor OGX-011 significantly suppressed HCC metastasis in two metastatic models through inhibiting EIF3I/Akt/MMP13 signaling. These findings indicate that CLU is an independent predictive factor for prognosis of HCC and it facilitates metastasis through EIF3I/Akt/MMP13 signaling. CLU suppression using OGX-011 may represent a promising therapeutic option for suppressing HCC metastasis.
Collapse
Affiliation(s)
- Cun Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guangzhi Jin
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Haojie Jin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Luo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yurong Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongmei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Kai Jiang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Dishui Gu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiujing Shen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xisong Huo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangyuan Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianxiang Ge
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangyu Zhao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Chu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huiqun Shu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenming Cong
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Wenxin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
31
|
Wang C, Zhang Y, Guo K, Wang N, Jin H, Liu Y, Qin W. Heat shock proteins in hepatocellular carcinoma: Molecular mechanism and therapeutic potential. Int J Cancer 2015; 138:1824-34. [PMID: 26853533 DOI: 10.1002/ijc.29723] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 07/06/2015] [Accepted: 08/03/2015] [Indexed: 12/30/2022]
Abstract
Heat shock proteins (HSPs) are highly conserved proteins, which are expressed at low levels under normal conditions, but significantly induced in response to cellular stresses. As molecular chaperones, HSPs play crucial roles in protein homeostasis, apoptosis, invasion and cellular signaling transduction. The induction of HSPs is an important part of heat shock response, which could help cancer cells to adapt to stress conditions. Because of the constant stress condition in tumor microenvironment, HSPs overexpression is widely reported in many human cancers. In light of the significance of HSPs for cancer cells to survive and obtain invasive phenotype under stress condition, HSPs are often associated with poor prognosis and treatment resistance in many types of human cancers. It has been described that upregulation of HSPs may serve as diagnostic and prognostic markers in hepatocellular carcinoma (HCC). Targeting HSPs with specific inhibitor alone or in combination with chemotherapy regimens holds promise for the improvement of outcomes for HCC patients. In this review, we summarize the expression profiles, functions and molecular mechanisms of HSPs (HSP27, HSP70 and HSP90) as well as a HSP-like protein (clusterin) in HCC. In addition, we address progression and challenges in targeting these HSPs as novel therapeutic strategies in HCC.
Collapse
Affiliation(s)
- Cun Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yurong Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun Guo
- Liver Cancer Institute, Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai, China
| | - Ning Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haojie Jin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinkun Liu
- Liver Cancer Institute, Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai, China
- Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wenxin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
32
|
Lamoureux F, Baud'huin M, Ory B, Guiho R, Zoubeidi A, Gleave M, Heymann D, Rédini F. Clusterin inhibition using OGX-011 synergistically enhances zoledronic acid activity in osteosarcoma. Oncotarget 2015; 5:7805-19. [PMID: 25138053 PMCID: PMC4202162 DOI: 10.18632/oncotarget.2308] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose Despite recent improvements in therapeutic management of osteosarcoma, ongoing challenges in improving the response to chemotherapy warrants new strategies still needed to improve overall patient survival. Among new therapeutic approaches, zoledronic acid (ZOL) represents a promising adjuvant molecule to chemotherapy to limit the osteolytic component of bone tumors. However, ZOL triggers the elevation of heat shock proteins (Hsp), including Hsp27 and clusterin (CLU), which could enhance tumor cell survival and treatment resistance. We hypothesized that targeting CLU using siRNA or the antisense drug, OGX-011, will suppress treatment-induced CLU induction and enhance ZOL-induced cell death in osteosarcoma (OS) cells. Methods The combined effects of OGX-011 and ZOL were investigated in vitro on cell growth, viability, apoptosis and cell cycle repartition of ZOL-sensitive or -resistant human OS cell lines (SaOS2, U2OS, MG63 and MNNG/HOS). Results In OS cell lines, ZOL increased levels of HSPs, especially CLU, in a dose- and time-dependent manner by mechanism including increased HSF1 transcription activity. The OS resistant cells to ZOL exhibited higher CLU expression level than the sensitive cells. Moreover, CLU overexpression protects OS sensitive cells to ZOL-induced cell death by modulating the MDR1 and farnesyl diphosphate synthase expression. OGX-011 suppressed treatment-induced increases in CLU and synergistically enhanced the activity of ZOL on cell growth and apoptosis. These biologic events were accompanied by decreased expression of HSPs, MDR1 and HSF1 transcriptional activity. In vivo, OGX-011, administered 3 times a week (IP, 20mg/kg), potentiated the effect of ZOL (s.c; 50μg/kg), significantly inhibiting tumor growth by 50% and prolonging survival in MNNG/HOS xenograft model compared to ZOL alone. Conclusion These results indicate that ZOL-mediated induction of CLU can be attenuated by OGX-011, with synergistic effects on delaying progression of osteosarcoma.
Collapse
Affiliation(s)
- Francois Lamoureux
- Université de Nantes, Nantes atlantique universités, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes F-44035, France. INSERM, UMR 957, Nantes F-44035, France. LUNAM Université. Equipe labellisée LIGUE 2012, Nantes, cedex
| | - Marc Baud'huin
- Université de Nantes, Nantes atlantique universités, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes F-44035, France. INSERM, UMR 957, Nantes F-44035, France. LUNAM Université. CHU de Nantes, Nantes F-44035, France. Equipe labellisée LIGUE 2012, Nantes, cedex
| | - Benjamin Ory
- Université de Nantes, Nantes atlantique universités, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes F-44035, France. INSERM, UMR 957, Nantes F-44035, France. LUNAM Université. Equipe labellisée LIGUE 2012, Nantes, cedex
| | - Romain Guiho
- Université de Nantes, Nantes atlantique universités,Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes F-44035, France. INSERM, UMR 957, Nantes F-44035, France. LUNAM Université. Equipe labellisée LIGUE 2012, Nantes, cedex
| | - Amina Zoubeidi
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Martin Gleave
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Dominique Heymann
- Université de Nantes, Nantes atlantique universités, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes F-44035, France. INSERM, UMR 957, Nantes F-44035, France. LUNAM Université. CHU de Nantes, Nantes F-44035, France. Equipe labellisée LIGUE 2012, Nantes, cedex
| | - Françoise Rédini
- Université de Nantes, Nantes atlantique universités, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes F-44035, France. INSERM, UMR 957, Nantes F-44035, France. LUNAM Université. Equipe labellisée LIGUE 2012, Nantes, cedex
| |
Collapse
|
33
|
Wyatt AW, Gleave ME. Targeting the adaptive molecular landscape of castration-resistant prostate cancer. EMBO Mol Med 2015; 7:878-94. [PMID: 25896606 PMCID: PMC4520654 DOI: 10.15252/emmm.201303701] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 03/12/2015] [Accepted: 03/26/2015] [Indexed: 12/19/2022] Open
Abstract
Castration and androgen receptor (AR) pathway inhibitors induce profound and sustained responses in advanced prostate cancer. However, the inevitable recurrence is associated with reactivation of the AR and progression to a more aggressive phenotype termed castration-resistant prostate cancer (CRPC). AR reactivation can occur directly through genomic modification of the AR gene, or indirectly via co-factor and co-chaperone deregulation. This mechanistic heterogeneity is further complicated by the stress-driven induction of a myriad of overlapping cellular survival pathways. In this review, we describe the heterogeneous and evolvable molecular landscape of CRPC and explore recent successes and failures of therapeutic strategies designed to target AR reactivation and adaptive survival pathways. We also discuss exciting areas of burgeoning anti-tumour research, and their potential to improve the survival and management of patients with CRPC.
Collapse
Affiliation(s)
- Alexander W Wyatt
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Martin E Gleave
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
34
|
Yamamoto Y, Lin PJC, Beraldi E, Zhang F, Kawai Y, Leong J, Katsumi H, Fazli L, Fraser R, Cullis PR, Gleave M. siRNA Lipid Nanoparticle Potently Silences Clusterin and Delays Progression When Combined with Androgen Receptor Cotargeting in Enzalutamide-Resistant Prostate Cancer. Clin Cancer Res 2015; 21:4845-55. [PMID: 26106075 DOI: 10.1158/1078-0432.ccr-15-0866] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/15/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Lipid nanoparticle (LNP) formulations facilitate tumor uptake and intracellular processing through an enhanced permeation and retention effect (EPR), and currently multiple products are undergoing clinical evaluation. Clusterin (CLU) is a cytoprotective chaperone induced by androgen receptor (AR) pathway inhibition to facilitate adaptive survival pathway signaling and treatment resistance. In our study, we investigated the efficacy of siRNA tumor delivery using LNP systems in an enzalutamide-resistant (ENZ-R) castration-resistant prostate cancer (CRPC) model. EXPERIMENTAL DESIGN Gene silencing of a luciferase reporter gene in the PC-3M-luc stable cell line was first assessed in subcutaneous and metastatic PC-3 xenograft tumors. Upon validation, the effect of LNP siRNA targeting CLU in combination with AR antisense oligonucleotides (ASO) was assessed in ENZ-R CRPC LNCaP in vitro and in vivo models. RESULTS LNP LUC-siRNA silenced luciferase expression in PC-3M-luc subcutaneous xenograft and metastatic models. LNP CLU-siRNA potently suppressed CLU and AR ASO-induced CLU and AKT and ERK phosphorylation in ENZ-R LNCaP cells in vitro, more potently inhibiting ENZ-R cell growth rates and increased apoptosis when compared with AR-ASO monotherapy. In subcutaneous ENZ-R LNCaP xenografts, combinatory treatment of LNP CLU-siRNA plus AR-ASO significantly suppressed tumor growth and serum PSA levels compared with LNP LUC-siRNA (control) and AR-ASO. CONCLUSIONS LNP siRNA can silence target genes in vivo and enable inhibition of traditionally non-druggable genes like CLU and other promising cotargeting approaches in ENZ-R CRPC therapeutics.
Collapse
Affiliation(s)
- Yoshiaki Yamamoto
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada. Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Paulo J C Lin
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Eliana Beraldi
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fan Zhang
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yoshihisa Kawai
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada. Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Jeffrey Leong
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hidemasa Katsumi
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Ladan Fazli
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Robert Fraser
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Martin Gleave
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
35
|
Lelj-Garolla B, Kumano M, Beraldi E, Nappi L, Rocchi P, Ionescu DN, Fazli L, Zoubeidi A, Gleave ME. Hsp27 Inhibition with OGX-427 Sensitizes Non-Small Cell Lung Cancer Cells to Erlotinib and Chemotherapy. Mol Cancer Ther 2015; 14:1107-16. [PMID: 25740245 DOI: 10.1158/1535-7163.mct-14-0866] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 02/21/2015] [Indexed: 11/16/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the most frequent cause of death from cancer worldwide. Despite the availability of active chemotherapy regimens and EGFR tyrosine kinase inhibitors, all advanced patients develop recurrent disease after first-line therapy. Although Hsp27 is a stress-induced chaperone that promotes acquired resistance in several cancers, its relationship to treatment resistance in NSCLC has not been defined. Understanding adaptive responses of acquired resistance will help guide new strategies to control NSCLC. Hsp27 levels were evaluated in an HCC827 erlotinib-resistant-derived cell line (HCC-827Resistant), and sensitivity to erlotinib was examined in Hsp27-overexpressing A549 cells. The role of Hsp27 in both erlotinib and cytotoxic treatment resistance was evaluated in HCC-827 and A549 NSCLC cells using the Hsp27 antisense drug OGX-427. The effect of OGX-427 in combination with erlotinib was also assessed in mice bearing A549 xenografts. Hsp27 is induced by erlotinib and protects NSCLC cells from treatment-induced apoptosis, whereas OGX-427 sensitizes NSCLC cells to erlotinib. Interestingly, increased resistance to erlotinib was observed when Hsp27 was increased either in HCC827 erlotinib-resistant or overexpressing A549 cells. Combining OGX-427 with erlotinib significantly enhanced antitumor effects in vitro and delayed A549 xenograft growth in vivo. OGX-427 also significantly enhanced the activity of cytotoxic drugs used for NSCLC. These data indicate that treatment-induced Hsp27 contributes to the development of resistance, and provides preclinical proof-of-principle that inhibition of stress adaptive pathways mediated by Hsp27 enhances the activity of erlotinib and chemotherapeutics.
Collapse
Affiliation(s)
- Barbara Lelj-Garolla
- The Vancouver Prostate Centre and Department of Urological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Masafumi Kumano
- The Vancouver Prostate Centre and Department of Urological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Eliana Beraldi
- The Vancouver Prostate Centre and Department of Urological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lucia Nappi
- The Vancouver Prostate Centre and Department of Urological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Palma Rocchi
- Cancer Research Center of Marseille, INSERM, Institut Paoli-Calmettes, and Aix-Marseille Université, Marseille, France
| | - Diana N Ionescu
- Pathology Department, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Ladan Fazli
- The Vancouver Prostate Centre and Department of Urological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amina Zoubeidi
- The Vancouver Prostate Centre and Department of Urological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin E Gleave
- The Vancouver Prostate Centre and Department of Urological Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
36
|
Yamamoto Y, Loriot Y, Beraldi E, Zhang F, Wyatt AW, Al Nakouzi N, Mo F, Zhou T, Kim Y, Monia BP, MacLeod AR, Fazli L, Wang Y, Collins CC, Zoubeidi A, Gleave M. Generation 2.5 antisense oligonucleotides targeting the androgen receptor and its splice variants suppress enzalutamide-resistant prostate cancer cell growth. Clin Cancer Res 2015; 21:1675-87. [PMID: 25634993 DOI: 10.1158/1078-0432.ccr-14-1108] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 01/08/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Enzalutamide (ENZ) is a potent androgen receptor (AR) antagonist with activity in castration-resistant prostate cancer (CRPC); however, progression to ENZ-resistant (ENZ-R) CRPC frequently occurs with rising serum PSA levels, implicating AR full-length (ARFL) or variants (AR-Vs) in disease progression. EXPERIMENTAL DESIGN To define functional roles of ARFL and AR-Vs in ENZ-R CRPC, we designed 3 antisense oligonucleotides (ASO) targeting exon-1, intron-1, and exon-8 in AR pre-mRNA to knockdown ARFL alone or with AR-Vs, and examined their effects in three CRPC cell lines and patient-derived xenografts. RESULTS ENZ-R-LNCaP cells express high levels of both ARFL and AR-V7 compared with CRPC-LNCaP; in particular, ARFL levels were approximately 12-fold higher than AR-V7. Both ARFL and AR-V7 are highly expressed in the nuclear fractions of ENZ-R-LNCaP cells even in the absence of exogenous androgens. In ENZ-R-LNCaP cells, knockdown of ARFL alone, or ARFL plus AR-Vs, similarly induced apoptosis, suppressed cell growth and AR-regulated gene expression, and delayed tumor growth in vivo. In 22Rv1 cells that are inherently ENZ-resistant, knockdown of both ARFL and AR-Vs more potently suppressed cell growth, AR transcriptional activity, and AR-regulated gene expression than knockdown of ARFL alone. Exon-1 AR-ASO also inhibited tumor growth of LTL-313BR patient-derived CRPC xenografts. CONCLUSIONS These data identify the AR as an important driver of ENZ resistance, and while the contributions of ARFL and AR-Vs can vary across cell systems, ARFL is the key driver in the ENZ-R LNCaP model. AR targeting strategies against both ARFL and AR-Vs is a rational approach for AR-dependent CRPC.
Collapse
Affiliation(s)
- Yoshiaki Yamamoto
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada. Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Yohann Loriot
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Eliana Beraldi
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fan Zhang
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alexander W Wyatt
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nader Al Nakouzi
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fan Mo
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tianyuan Zhou
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., Carlsbad, California
| | - Youngsoo Kim
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., Carlsbad, California
| | - Brett P Monia
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., Carlsbad, California
| | - A Robert MacLeod
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., Carlsbad, California
| | - Ladan Fazli
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yuzhuo Wang
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Colin C Collins
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amina Zoubeidi
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin Gleave
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
37
|
Azad AA, Zoubeidi A, Gleave ME, Chi KN. Targeting heat shock proteins in metastatic castration-resistant prostate cancer. Nat Rev Urol 2014; 12:26-36. [DOI: 10.1038/nrurol.2014.320] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
38
|
Clusterin facilitates stress-induced lipidation of LC3 and autophagosome biogenesis to enhance cancer cell survival. Nat Commun 2014; 5:5775. [PMID: 25503391 PMCID: PMC4275590 DOI: 10.1038/ncomms6775] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 11/06/2014] [Indexed: 02/06/2023] Open
Abstract
We define stress-induced adaptive survival pathways linking autophagy with the molecular chaperone clusterin (CLU) that function to promote anticancer treatment resistance. During treatment stress, CLU co-localizes with LC3 via an LIR-binding sequence within autophagosome membranes, functioning to facilitate LC3–Atg3 heterocomplex stability and LC3 lipidation, and thereby enhance autophagosome biogenesis and autophagy activation. Stress-induced autophagy is attenuated with CLU silencing in CLU−/− mice and human prostate cancer cells. CLU-enhanced cell survival occurs via autophagy-dependent pathways, and is reduced following autophagy inhibition. Combining CLU inhibition with anticancer treatments attenuates autophagy activation, increases apoptosis and reduces prostate cancer growth. This study defines a novel adaptor protein function for CLU under stress conditions, and highlights how co-targeting CLU and autophagy can amplify proteotoxic stress to delay cancer progression. The induction of autophagy under stress conditions such as chemotherapy is a contributing factor towards resistance to anticancer therapy. Here, Zhang et al. identify the molecular chaperone clusterin as an adaptor that facilitates lipidation of LC3 and autophagosome biogenesis.
Collapse
|
39
|
Solárová Z, Mojžiš J, Solár P. Hsp90 inhibitor as a sensitizer of cancer cells to different therapies (review). Int J Oncol 2014; 46:907-26. [PMID: 25501619 DOI: 10.3892/ijo.2014.2791] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 10/22/2014] [Indexed: 11/06/2022] Open
Abstract
Hsp90 is a molecular chaperone that maintains the structural and functional integrity of various client proteins involved in signaling and many other functions of cancer cells. The natural inhibitors, ansamycins influence the Hsp90 chaperone function by preventing its binding to client proteins and resulting in their proteasomal degradation. N- and C-terminal inhibitors of Hsp90 and their analogues are widely tested as potential anticancer agents in vitro, in vivo as well as in clinical trials. It seems that Hsp90 competitive inhibitors target different tumor types at nanomolar concentrations and might have therapeutic benefit. On the contrary, some Hsp90 inhibitors increased toxicity and resistance of cancer cells induced by heat shock response, and through the interaction of survival signals, that occured as side effects of treatments, could be very effectively limited via combination of therapies. The aim of our review was to collect the data from experimental and clinical trials where Hsp90 inhibitor was combined with other therapies in order to prevent resistance as well as to potentiate the cytotoxic and/or antiproliferative effects.
Collapse
Affiliation(s)
- Zuzana Solárová
- Department of Pharmacology, Faculty of Medicine, P.J. Šafárik University, 040 01 Košice, Slovak Republic
| | - Ján Mojžiš
- Department of Pharmacology, Faculty of Medicine, P.J. Šafárik University, 040 01 Košice, Slovak Republic
| | - Peter Solár
- Laboratory of Cell Biology, Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University, 040 01 Košice, Slovak Republic
| |
Collapse
|
40
|
Mazaira GI, Lagadari M, Erlejman AG, Galigniana MD. The Emerging Role of TPR-Domain Immunophilins in the Mechanism of Action of Steroid Receptors. NUCLEAR RECEPTOR RESEARCH 2014. [DOI: 10.11131/2014/101094] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- G. I. Mazaira
- Departamento de Química Biológica-IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - M. Lagadari
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - A. G. Erlejman
- Departamento de Química Biológica-IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - M. D. Galigniana
- Departamento de Química Biológica-IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| |
Collapse
|
41
|
Zardan A, Nip KM, Thaper D, Toren P, Vahid S, Beraldi E, Fazli L, Lamoureux F, Gust KM, Cox ME, Bishop JL, Zoubeidi A. Lyn tyrosine kinase regulates androgen receptor expression and activity in castrate-resistant prostate cancer. Oncogenesis 2014; 3:e115. [PMID: 25133482 PMCID: PMC5189960 DOI: 10.1038/oncsis.2014.30] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 05/22/2014] [Accepted: 06/03/2014] [Indexed: 12/27/2022] Open
Abstract
Castrate-resistant prostate cancer (CRPC) progression is a complex process by which prostate cells acquire the ability to survive and proliferate in the absence or under very low levels of androgens. Most CRPC tumors continue to express the androgen receptor (AR) as well as androgen-responsive genes owing to reactivation of AR. Protein tyrosine kinases have been implicated in supporting AR activation under castrate conditions. Here we report that Lyn tyrosine kinase expression is upregulated in CRPC human specimens compared with hormone naive or normal tissue. Lyn overexpression enhanced AR transcriptional activity both in vitro and in vivo and accelerated CRPC. Reciprocally, specific targeting of Lyn resulted in a decrease of AR transcriptional activity in vitro and in vivo and prolonged time to castration. Mechanistically, we found that targeting Lyn kinase induces AR dissociation from the molecular chaperone Hsp90, leading to its ubiquitination and proteasomal degradation. This work indicates a novel mechanism of regulation of AR stability and transcriptional activity by Lyn and justifies further investigation of the Lyn tyrosine kinase as a therapeutic target for the treatment of CRPC.
Collapse
Affiliation(s)
- A Zardan
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - K M Nip
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - D Thaper
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - P Toren
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - S Vahid
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - E Beraldi
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - L Fazli
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - F Lamoureux
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - K M Gust
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - M E Cox
- 1] The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada [2] Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - J L Bishop
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - A Zoubeidi
- 1] The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada [2] Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
42
|
Downregulating sCLU enhances the sensitivity of hepatocellular carcinoma cells to gemcitabine by activating the intrinsic apoptosis pathway. Dig Dis Sci 2014; 59:1798-809. [PMID: 24671452 DOI: 10.1007/s10620-014-3111-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 03/10/2014] [Indexed: 12/12/2022]
Abstract
PURPOSE The purpose of this study was to investigate whether the therapeutic activity of gemcitabine (GCB) in hepatocellular carcinoma (HCC) could be increased by the down-regulation of secretory clusterin (sCLU), a glycoprotein that is considered to play a cytoprotective role in the resistance to chemotherapy. METHODS The expression of sCLU was detected in HCC tumor tissues and cell lines. A cell viability and apoptosis assay were performed in parental HCC cells or the same cells transfected with sCLU shRNA and treated with or without GCB. The potential downstream pathways were investigated using the Human Apoptosis RT(2) Profiler™ PCR Array. RESULTS The expression levels of sCLU in HCC tissues were significantly higher than in adjacent non-tumor liver tissues and were associated with the histological grade and transarterial chemoembolization. sCLU overexpression was also found in three HCC cell lines and hepatocytes. The depletion of sCLU synergistically increased GCB sensitivity in Bel7402 and SMMC7721 cells and induced cell apoptosis. Based on the PCR array analysis, sCLU depletion also resulted in the up-regulation of BNIP1, GADD45A, TNFRSF10A, and TRADD and down-regulation of AKT1 in Bel7402 and SMMC7721 cells compared with the parental controls. These results were further supported by a Western blot analysis, which showed increased GADD45a protein expression and the decreased expression of phosphorylated AKT. GADD45a overexpression also increased the sensitivity to GCB in the Bel7402 and SMMC7721 cells. CONCLUSION Targeting sCLU may be a useful method to enhance the cytotoxic effect of GCB in hepatocellular carcinoma.
Collapse
|
43
|
Ku S, Lasorsa E, Adelaiye R, Ramakrishnan S, Ellis L, Pili R. Inhibition of Hsp90 augments docetaxel therapy in castrate resistant prostate cancer. PLoS One 2014; 9:e103680. [PMID: 25072314 PMCID: PMC4114978 DOI: 10.1371/journal.pone.0103680] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 07/06/2014] [Indexed: 11/18/2022] Open
Abstract
First line treatment of patients with castrate resistant prostate cancer (CRPC) primarily involves administration of docetaxel chemotherapy. Unfortunately, resistance to docetaxel therapy is an ultimate occurrence. Alterations in androgen receptor (AR) expression and signaling are associated mechanisms underlying resistance to docetaxel treatment in CRPC. Heat shock protein 90 (Hsp90) is a molecular chaperone, which regulates the activation, maturation and stability of critical signaling proteins involved in prostate cancer, including the AR. This knowledge and recent advances in compound design and development have highlighted Hsp90 as an attractive therapeutic target for the treatment of CRPC. We recently reported the development of a MYC-CaP castrate resistant (MYC-CaP/CR) transplant tumor model, which expresses amplified wild type AR. Within, we report that a second generation Hsp90 inhibitor, NVP-AUY922, inhibits cell growth and significantly induces cell death in MYC-CaP/CR and Pten-CaP/cE2 cell lines. NVP-AUY922 induced proteasome degradation of AR, though interestingly does not require loss of AR protein to inhibit AR transcriptional activity. Further, NVP-AUY922 increased docetaxel toxicity in MYC-CaP/CR and Pten-CaP/cE2 cell lines in vitro. Finally, NVP-AUY922/docetaxel combination therapy in mice bearing MYC-CaP/CR tumors resulted in greater anti-tumor activity compared to single treatment. This study demonstrates that NVP-AUY922 elicits potent activity towards AR signaling and augments chemotherapy response in a mouse model of CRPC, providing rationale for the continued clinical development of Hsp90 inhibitors in clinical trials for treatment of CRPC patients.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Apoptosis/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Docetaxel
- Drug Resistance, Neoplasm
- Drug Therapy, Combination
- HSP90 Heat-Shock Proteins/antagonists & inhibitors
- HSP90 Heat-Shock Proteins/metabolism
- Isoxazoles/pharmacology
- Isoxazoles/therapeutic use
- Male
- Mice
- Mice, SCID
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/pathology
- Proteasome Endopeptidase Complex/metabolism
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Resorcinols/pharmacology
- Resorcinols/therapeutic use
- Taxoids/pharmacology
- Taxoids/therapeutic use
- Transcription, Genetic
- Transplantation, Heterologous
Collapse
Affiliation(s)
- ShengYu Ku
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York, United States of America
- Department of Cancer Prevention and Pathology, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Elena Lasorsa
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York, United States of America
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Remi Adelaiye
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York, United States of America
- Department of Cancer Prevention and Pathology, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Swathi Ramakrishnan
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York, United States of America
- Department of Cancer Prevention and Pathology, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Leigh Ellis
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York, United States of America
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York, United States of America
- * E-mail: (LE); (RP)
| | - Roberto Pili
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York, United States of America
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York, United States of America
- * E-mail: (LE); (RP)
| |
Collapse
|
44
|
Koay YC, McConnell JR, Wang Y, Kim SJ, Buckton LK, Mansour F, McAlpine SR. Chemically accessible hsp90 inhibitor that does not induce a heat shock response. ACS Med Chem Lett 2014; 5:771-6. [PMID: 25050163 DOI: 10.1021/ml500114p] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/13/2014] [Indexed: 02/01/2023] Open
Abstract
Recent cancer therapies have focused on targeting biology networks through a single regulatory protein. Heat shock protein 90 (hsp90) is an ideal oncogenic target as it regulates over 400 client proteins and cochaperones. However, clinical inhibitors of hsp90 have had limited success; the primary reason being that they induce a heat shock response. We describe the synthesis and biological evaluation of a new hsp90 inhibitor, SM253. The previous generation on which SM253 is based (SM145) has poor overall synthetic yields, low solubility, and micromolar cytotoxicity. By comparison SM253 has relatively high overall yields, good aqueous solubility, and is more cytotoxic than its parent compound. Verification that hsp90 is SM253's target was accomplished using pull-down and protein folding assays. SM253 is superior to both SM145 and the clinical candidate 17-AAG as it decreases proteins related to the heat shock response by 2-fold, versus a 2-4-fold increase observed when cells are treated with 17-AAG.
Collapse
Affiliation(s)
- Yen Chin Koay
- The University of New South Wales, Sydney, NSW 2052, Australia
| | | | - Yao Wang
- The University of New South Wales, Sydney, NSW 2052, Australia
| | - Seong Jong Kim
- The University of New South Wales, Sydney, NSW 2052, Australia
| | | | - Flora Mansour
- The University of New South Wales, Sydney, NSW 2052, Australia
| | | |
Collapse
|
45
|
Abstract
INTRODUCTION The androgen receptor (AR) is a ligand-activated transcription factor that is expressed in primary and metastatic prostate cancers. There are advances in endocrine therapy for prostate cancer that are based on improved understanding of AR function. AREAS COVERED PubMed has been used to include most important publications on targeting the AR in prostate cancer. AR expression may be downregulated by agents used for chemoprevention of prostate cancer or, in models of advanced prostate cancer, by antisense oligonucleotides. New drugs that inhibit the steroidogenic enzyme CYP17A1 (abiraterone acetate) or diminish nuclear translocation of the AR (enzalutamide) have been shown to improve patients' survival in prostate cancer. However, it is clear that there is a development of resistance to these novel therapies. They may include increased expression of truncated, constitutively active AR or activation of the signaling pathway of signal transducers and activators of transcription. EXPERT OPINION Although introduction of novel drugs have improved patients' survival, there is a need to investigate the mechanisms of resistance further. The role of truncated AR and compensatory activation of signaling pathways as well as the development of scientifically justified combination therapies seems to be issues of a high priority.
Collapse
Affiliation(s)
- Zoran Culig
- Innsbruck Medical University, Experimental Urology, Department of Urology , Anichstrasse 35, A-6020 Innsbruck , Austria +43 512 504 24717 ; +43 512 504 24817 ;
| |
Collapse
|
46
|
Androgen receptor (AR) inhibitor ErbB3-binding protein-1 (Ebp1) is not targeted by the newly identified AR controlling signaling axis heat-shock protein HSP27 and microRNA miR-1 in prostate cancer cells. World J Urol 2014; 33:323-7. [DOI: 10.1007/s00345-014-1307-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 04/22/2014] [Indexed: 10/25/2022] Open
|
47
|
Takeuchi A, Shiota M, Beraldi E, Thaper D, Takahara K, Ibuki N, Pollak M, Cox ME, Naito S, Gleave ME, Zoubeidi A. Insulin-like growth factor-I induces CLU expression through Twist1 to promote prostate cancer growth. Mol Cell Endocrinol 2014; 384:117-25. [PMID: 24491388 DOI: 10.1016/j.mce.2014.01.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 12/27/2013] [Accepted: 01/14/2014] [Indexed: 11/16/2022]
Abstract
Clusterin (CLU) is cytoprotective molecular chaperone that is highly expressed in castrate-resistant prostate cancer (CRPC). CRPC is also characterized by increased insulin-like growth factor (IGF)-I responsiveness which induces prostate cancer survival and CLU expression. However, how IGF-I induces CLU expression and whether CLU is required for IGF-mediated growth signaling remain unknown. Here we show that IGF-I induced CLU via STAT3-Twist1 signaling pathway. In response to IGF-I, STAT3 was phosphorylated, translocated to the nucleus and bound to the Twist1 promoter to activate Twist1 transcription. In turn, Twist1 bound to E-boxes on the CLU promoter and activated CLU transcription. Inversely, we demonstrated that knocking down Twist1 abrogated IGF-I induced CLU expression, indicating that Twist1 mediated IGF-I-induced CLU expression. When PTEN knockout mice were crossed with lit/lit mice, the resultant IGF-I deficiency suppressed Twist1 as well as CLU gene expression in mouse prostate glands. Moreover, both Twist1 and CLU knockdown suppressed prostate cancer growth accelerated by IGF-I, suggesting the relevance of this signaling not only in an in vitro, but also in an in vivo. Collectively, this study indicates that IGF-I induces CLU expression through sequential activation of STAT3 and Twist1, and suggests that this signaling cascade plays a critical role in prostate cancer pathogenesis.
Collapse
Affiliation(s)
- Ario Takeuchi
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Masaki Shiota
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Eliana Beraldi
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daksh Thaper
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kiyoshi Takahara
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Naokazu Ibuki
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael Pollak
- Department of Medicine and Oncology, McGill University, Montreal, Quebec, Canada
| | - Michael E Cox
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Seiji Naito
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Martin E Gleave
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amina Zoubeidi
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
48
|
Abstract
Clusterin is a heterodimeric disulfide-linked glycoprotein (449 amino acids) isolated in the rat prostate after castration. It is widely distributed in different tissues and highly conserved in species. There are two isoforms (1 and 2) with antagonistic actions regarding apoptosis. Clusterin is implicated in a number of biological processes, including lipid transport, membrane recycling, cell adhesion, programmed cell death, and complement cascade, representing a truly multifunctional protein. Isoform 2 is overexpressed under cellular stress conditions and protects cells from apoptosis by impeding Bax actions on the mitochondrial membrane and exerts other protumor activities, like phosphatidylinositol 3-kinase/protein kinase B pathway activation, modulation of extracellular signal-regulated kinase 1/2 signaling and matrix metallopeptidase-9 expression, increased angiogenesis, modulation of the nuclear factor kappa B pathway, among others. Its overexpression should be considered as a nonspecific cellular response to a wide variety of tissue insults like cytotoxic chemotherapy, radiation, excess of free oxygen radicals, androgen or estrogen deprivation, etc. A review of the recent literature strongly suggests potential roles for custirsen in particular, and proapoptosis treatments in general, as novel modalities in cancer management. Inhibition of clusterin is known to increase the cytotoxic effects of chemotherapeutic agents, and custirsen, a second-generation antisense oligonucleotide that blocks clusterin, is being tested in a Phase III clinical trial after successful results were achieved in Phase II studies. A major issue in cancer evolution that remains unanswered is whether clusterin represents a driving force of tumorigenesis or a late phenomenon after chemotherapy. This review presents preclinical data that encourages trials in various types of cancer other than advanced castration-resistance prostate cancer and discusses briefly the appropriate timing for clusterin inhibition in the clinical context.
Collapse
Affiliation(s)
- Tomas Koltai
- Gerencia de Efectores Sanitarios Propios, Instituto Nacional de la Seguridad Social para Jubilados y Pensionados, Buenos Aires, República Argentina
| |
Collapse
|
49
|
Suppression of heat shock protein 27 using OGX-427 induces endoplasmic reticulum stress and potentiates heat shock protein 90 inhibitors to delay castrate-resistant prostate cancer. Eur Urol 2013; 66:145-55. [PMID: 24411988 DOI: 10.1016/j.eururo.2013.12.019] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/13/2013] [Indexed: 01/03/2023]
Abstract
BACKGROUND Although prostate cancer responds initially to androgen ablation therapies, progression to castration-resistant prostate cancer (CRPC) frequently occurs. Heat shock protein (Hsp) 90 inhibition is a rational therapeutic strategy for CRPC that targets key proteins such as androgen receptor (AR) and protein kinase B (Akt); however, most Hsp90 inhibitors trigger elevation of stress proteins like Hsp27 that confer tumor cell survival and treatment resistance. OBJECTIVE We hypothesized that cotargeting the cytoprotective chaperone Hsp27 and Hsp90 would amplify endoplasmic reticulum (ER) stress and treatment-induced cell death in cancer. DESIGN, SETTING, AND PARTICIPANTS Inducible and constitutive Hsp27 and other HSPs were measured by real-time reverse transcription-polymerase chain reaction and immunoblot assays. The combinations of OGX-427 with Hsp90 inhibitors were evaluated in vitro for LNCaP cell growth and apoptosis and in vivo in CRPC LNCaP xenograft models. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Tumor volumes were compared using the Kruskal-Wallis test. Overall survival was analyzed using Kaplan-Meier curves, and statistical significance was assessed with the log-rank test. RESULTS AND LIMITATIONS Hsp90 inhibitors induced expression of HSPs in tumor cells and tissues in a dose- and time-dependent manner; in particular, Hsp27 mRNA and protein levels increased threefold. In vitro, OGX-427 synergistically enhanced Hsp90 inhibitor-induced suppression of cell growth and induced apoptosis by 60% as measured by increased sub-G1 fraction and poly(ADP-ribose) polymerase cleavage. These biologic events were accompanied by decreased expression of HSPs, Akt, AR, and prostate-specific antigen, and induction of ER stress markers (cleaved activating transcription factor 6, glucose-regulated protein 78, and DNA-damage-inducible transcript 3). In vivo, OGX-427 potentiated the anticancer effects of Hsp90 inhibitor PF-04929113 (orally, 25mg/kg) to inhibit tumor growth and prolong survival in CRPC LNCaP xenografts. CONCLUSIONS HSP90 inhibitor-mediated induction of Hsp27 expression can be attenuated by OGX-427, resulting in increased ER stress and apoptosis, and synergistic inhibition of CRPC tumor growth. PATIENT SUMMARY This study supports the development of targeted strategies using OGX-427 in combination with Hsp90 inhibitors to improve patient outcome in CRPC.
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Although the treatment of castration-resistant prostate cancer (CRPC) has benefited from the use of increasingly potent androgen synthesis inhibitors and androgen receptor (AR) antagonists, it is only marginally effective. There is therefore a critical need for a better understanding of the mechanisms underlying the CRPC development and more effective therapeutic approaches. Here, we focus on the advancements reported in the last 18 months, particularly with regard to the mechanisms of castration resistance and potential therapeutic targets emerging from the studies with in-vivo models. RECENT FINDINGS Recent findings indicate that AR-dependent mechanisms, for example, increased expression of CYP17A1 and AR splice variants, play important roles in in-vivo castration resistance to new antiandrogens and androgen synthesis inhibitors. Whereas current therapeutic approaches focus on AR-dependent CRPC, studies based on genetically engineered mouse models indicate that castration resistance can develop in the absence of robust AR signaling. Furthermore, increasing evidence suggests that cellular plasticity of prostate adenocarcinoma allows AR-independent CRPC development via various adaptive mechanisms. SUMMARY Significant progress has been made in the understanding of AR-dependent and AR-independent mechanisms involved in the development of CRPC. This may lead to identification of new therapeutic targets and improved therapy.
Collapse
|