1
|
Liyanage C, Fernando A, Chamberlain A, Moradi A, Batra J. RNA m6a Methylation Regulator Expression in Castration-Resistant Prostate Cancer Progression and Its Genetic Associations. Cancers (Basel) 2024; 16:1303. [PMID: 38610981 PMCID: PMC11011207 DOI: 10.3390/cancers16071303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
N6-methyladenosine (m6A) methylation, a prevalent epitranscriptomic modification, plays a crucial role in regulating mRNA expression, stability, and translation in mammals. M6A regulators have gained attention for their potential implications in tumorigenesis and clinical applications, such as cancer diagnosis and therapeutics. The existing literature predominantly addresses m6A regulators in the context of primary prostate cancer (PCa). However, a notable gap in the knowledge emerges regarding the dynamic expression patterns of these regulators as PCa progresses towards the castration-resistant stage (CRPC). Employing sequential window acquisition of all theoretical mass spectra (SWATH-MS) and RNAseq analysis, we comprehensively profiled the expression of 27 m6A regulators in hormone/androgen-dependent and -independent PCa cell lines, revealing distinct clustering between tumor and adjacent normal prostate tissues. High-grade PCa tumors demonstrated the upregulation of METTL3, RBM15B, and HNRNAPA2B1 and the downregulation of ZC3H13, NUDT21, and FTO. Notably, we identified six m6A regulators associated with PCa survival. Additionally, association analysis of the PCa-associated risk loci in the cancer genome atlas program (TCGA) data unveiled genetic variations near the WTAP, HNRNPA2B1, and FTO genes as significant expression quantitative trait loci. In summary, our study unraveled abnormalities in m6A regulator expression in PCa progression, elucidating their association with PCa risk loci. Considering the heterogeneity within the PCa phenotypes and treatment responses, our findings suggest that prognostic stratification based on m6A regulator expression could enhance PCa diagnosis and prognosis.
Collapse
Affiliation(s)
- Chamikara Liyanage
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Center for Genomics and Personalized Health, Translational Research Institute, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Achala Fernando
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Center for Genomics and Personalized Health, Translational Research Institute, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Audrey Chamberlain
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Center for Genomics and Personalized Health, Translational Research Institute, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Afshin Moradi
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Center for Genomics and Personalized Health, Translational Research Institute, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Center for Genomics and Personalized Health, Translational Research Institute, Queensland University of Technology, Brisbane, QLD 4059, Australia
| |
Collapse
|
2
|
Granata I, Barboro P. Identification of Molecular Markers Associated with Prostate Cancer Subtypes: An Integrative Bioinformatics Approach. Biomolecules 2024; 14:87. [PMID: 38254687 PMCID: PMC10813078 DOI: 10.3390/biom14010087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Prostate cancer (PCa) is characterised by androgen dependency. Unfortunately, under anti-androgen treatment pressure, castration-resistant prostate cancer (CRPC) emerges, characterised by heterogeneous cell populations that, over time, lead to the development of different androgen-dependent or -independent phenotypes. Despite important advances in therapeutic strategies, CRPC remains incurable. Context-specific essential genes represent valuable candidates for targeted anti-cancer therapies. Through the investigation of gene and protein annotations and the integration of published transcriptomic data, we identified two consensus lists to stratify PCa patients' risk and discriminate CRPC phenotypes based on androgen receptor activity. ROC and Kaplan-Meier survival analyses were used for gene set validation in independent datasets. We further evaluated these genes for their association with cancer dependency. The deregulated expression of the PCa-related genes was associated with overall and disease-specific survival, metastasis and/or high recurrence risk, while the CRPC-related genes clearly discriminated between adeno and neuroendocrine phenotypes. Some of the genes showed context-specific essentiality. We further identified candidate drugs through a computational repositioning approach for targeting these genes and treating lethal variants of PCa. This work provides a proof-of-concept for the use of an integrative approach to identify candidate biomarkers involved in PCa progression and CRPC pathogenesis within the goal of precision medicine.
Collapse
Affiliation(s)
- Ilaria Granata
- High Performance Computing and Networking Institute (ICAR), National Council of Research (CNR), Via Pietro Castellino 111, 80131 Naples, Italy
| | - Paola Barboro
- Proteomic and Mass Spectrometry Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genoa, Italy;
| |
Collapse
|
3
|
Ben Toumia I, Bachetti T, Chekir-Ghedira L, Profumo A, Ponassi M, Di Domizio A, Izzotti A, Sciacca S, Puglisi C, Forte S, Giuffrida R, Colarossi C, Milardi D, Grasso G, Lanza V, Fiordoro S, Drago G, Tkachenko K, Cardinali B, Romano P, Iervasi E, Vargas GC, Barboro P, Kohnke FH, Rosano C. Fraisinib: a calixpyrrole derivative reducing A549 cell-derived NSCLC tumor in vivo acts as a ligand of the glycine-tRNA synthase, a new molecular target in oncology. Front Pharmacol 2024; 14:1258108. [PMID: 38235113 PMCID: PMC10791888 DOI: 10.3389/fphar.2023.1258108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/17/2023] [Indexed: 01/19/2024] Open
Abstract
Background and purpose: Lung cancer is the leading cause of death in both men and women, constituting a major public health problem worldwide. Non-small-cell lung cancer accounts for 85%-90% of all lung cancers. We propose a compound that successfully fights tumor growth in vivo by targeting the enzyme GARS1. Experimental approach: We present an in-depth investigation of the mechanism through which Fraisinib [meso-(p-acetamidophenyl)-calix(4)pyrrole] affects the human lung adenocarcinoma A549 cell line. In a xenografted model of non-small-cell lung cancer, Fraisinib was found to reduce tumor mass volume without affecting the vital parameters or body weight of mice. Through a computational approach, we uncovered that glycyl-tRNA synthetase is its molecular target. Differential proteomics analysis further confirmed that pathways regulated by Fraisinib are consistent with glycyl-tRNA synthetase inhibition. Key results: Fraisinib displays a strong anti-tumoral potential coupled with limited toxicity in mice. Glycyl-tRNA synthetase has been identified and validated as a protein target of this compound. By inhibiting GARS1, Fraisinib modulates different key biological processes involved in tumoral growth, aggressiveness, and invasiveness. Conclusion and implications: The overall results indicate that Fraisinib is a powerful inhibitor of non-small-cell lung cancer growth by exerting its action on the enzyme GARS1 while displaying marginal toxicity in animal models. Together with the proven ability of this compound to cross the blood-brain barrier, we can assess that Fraisinib can kill two birds with one stone: targeting the primary tumor and its metastases "in one shot." Taken together, we suggest that inhibiting GARS1 expression and/or GARS1 enzymatic activity may be innovative molecular targets for cancer treatment.
Collapse
Affiliation(s)
| | | | - Leila Chekir-Ghedira
- Unit of Bioactive Natural Substances and Biotechnology, Faculty of Dental Medicine of Monastir, University of Monastir, Monastir, Tunisia
| | - Aldo Profumo
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Marco Ponassi
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Alberto Izzotti
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | | | | | - Stefano Forte
- Istituto Oncologico del Mediterraneo, Viagrande, Italy
| | | | | | - Danilo Milardi
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Catania, Italy
| | - Giuseppe Grasso
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - Valeria Lanza
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Catania, Italy
| | - Stefano Fiordoro
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Giacomo Drago
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | | | | | - Paolo Romano
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Erika Iervasi
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Paola Barboro
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Franz Heinrich Kohnke
- Dipartimento di Scienze Chimiche, Farmaceutiche ed Ambientali (CHIBIOFARAM), University of Messina, Messina, Italy
| | | |
Collapse
|
4
|
Yu Y, Papukashvili D, Ren R, Rcheulishvili N, Feng S, Bai W, Zhang H, Xi Y, Lu X, Xing N. siRNA-based approaches for castration-resistant prostate cancer therapy targeting the androgen receptor signaling pathway. Future Oncol 2023; 19:2055-2073. [PMID: 37823367 DOI: 10.2217/fon-2023-0227] [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] [Indexed: 10/13/2023] Open
Abstract
Androgen deprivation therapy is a common treatment method for metastatic prostate cancer through lowering androgen levels; however, this therapy frequently leads to the development of castration-resistant prostate cancer (CRPC). This is attributed to the activation of the androgen receptor (AR) signaling pathway. Current treatments targeting AR are often ineffective mostly due to AR gene overexpression and mutations, as well as the presence of splice variants that accelerate CRPC progression. Thus there is a critical need for more specific medication to treat CRPC. Small interfering RNAs have shown great potential as a targeted therapy. This review discusses prostate cancer progression and the role of AR signaling in CRPC, and proposes siRNA-based targeted therapy as a promising strategy for CRPC.
Collapse
Affiliation(s)
- Yanling Yu
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | | | - Ruimin Ren
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Urology, Taiyuan, 030032, China
| | | | - Shunping Feng
- Southern University of Science & Technology, Shenzhen, 518000, China
| | - Wenqi Bai
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Huanhu Zhang
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Yanfeng Xi
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Xiaoqing Lu
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Nianzeng Xing
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
5
|
Coluccia M. Cyclooxygenase and Cancer: Fundamental Molecular Investigations. Int J Mol Sci 2023; 24:12342. [PMID: 37569718 PMCID: PMC10418830 DOI: 10.3390/ijms241512342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
The involvement of prostaglandins in cancer was first observed in human esophageal carcinoma cells, whose invasive and metastatic potential in nude mice was found to be related to PGE2 and PGF2a production [...].
Collapse
Affiliation(s)
- Mauro Coluccia
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125 Bari, Italy
| |
Collapse
|
6
|
Xia J, Li S, Liu S, Zhang L. Aldehyde dehydrogenase in solid tumors and other diseases: Potential biomarkers and therapeutic targets. MedComm (Beijing) 2023; 4:e195. [PMID: 36694633 PMCID: PMC9842923 DOI: 10.1002/mco2.195] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 01/18/2023] Open
Abstract
The family of aldehyde dehydrogenases (ALDHs) contains 19 isozymes and is involved in the oxidation of endogenous and exogenous aldehydes to carboxylic acids, which contributes to cellular and tissue homeostasis. ALDHs play essential parts in detoxification, biosynthesis, and antioxidants, which are of important value for cell proliferation, differentiation, and survival in normal body tissues. However, ALDHs are frequently dysregulated and associated with various diseases like Alzheimer's disease, Parkinson's disease, and especially solid tumors. Notably, the involvement of the ALDHs in tumor progression is responsible for the maintenance of the stem-cell-like phenotype, triggering rapid and aggressive clinical progressions. ALDHs have captured increasing attention as biomarkers for disease diagnosis and prognosis. Nevertheless, these require further longitudinal clinical studies in large populations for broad application. This review summarizes our current knowledge regarding ALDHs as potential biomarkers in tumors and several non-tumor diseases, as well as recent advances in our understanding of the functions and underlying molecular mechanisms of ALDHs in disease development. Finally, we discuss the therapeutic potential of ALDHs in diseases, especially in tumor therapy with an emphasis on their clinical implications.
Collapse
Affiliation(s)
- Jie Xia
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Siqin Li
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer MedicineNanjing Medical UniversityNanjingChina
| | - Lixing Zhang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
7
|
Granata I, Manipur I, Giordano M, Maddalena L, Guarracino MR. TumorMet: A repository of tumor metabolic networks derived from context-specific Genome-Scale Metabolic Models. Sci Data 2022; 9:607. [PMID: 36207341 PMCID: PMC9547001 DOI: 10.1038/s41597-022-01702-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 09/15/2022] [Indexed: 11/25/2022] Open
Abstract
Studies about the metabolic alterations during tumorigenesis have increased our knowledge of the underlying mechanisms and consequences, which are important for diagnostic and therapeutic investigations. In this scenario and in the era of systems biology, metabolic networks have become a powerful tool to unravel the complexity of the cancer metabolic machinery and the heterogeneity of this disease. Here, we present TumorMet, a repository of tumor metabolic networks extracted from context-specific Genome-Scale Metabolic Models, as a benchmark for graph machine learning algorithms and network analyses. This repository has an extended scope for use in graph classification, clustering, community detection, and graph embedding studies. Along with the data, we developed and provided Met2Graph, an R package for creating three different types of metabolic graphs, depending on the desired nodes and edges: Metabolites-, Enzymes-, and Reactions-based graphs. This package allows the easy generation of datasets for downstream analysis. Measurement(s) | gene expression, metabolic relationships | Technology Type(s) | Genome Scale Metabolic Models; Computational network biology | Sample Characteristic - Organism | Homo sapiens |
Collapse
|
8
|
Toumia IB, Ponassi M, Barboro P, Iervasi E, Vargas GC, Banelli B, Fiordoro S, Ghedira LC, Kohnke FH, Izzotti A, Rosano C. Two calix[4]pyrroles as potential therapeutics for castration-resistant prostate cancer. Invest New Drugs 2022; 40:1185-1193. [PMID: 35976541 DOI: 10.1007/s10637-022-01294-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/10/2022] [Indexed: 12/24/2022]
Abstract
Macrocyclic compounds meso-(p-acetamidophenyl)-calix[4]pyrrole and meso-(m-acetamidophenyl)-calix[4]pyrrole have previously been reported to exhibit cytotoxic properties towards lung cancer cells. Here, we report pre-clinical in vitro and in vivo studies showing that these calixpyrrole derivatives can inhibit cell growth in both PC3 and DU145 prostatic cancer cell lines. We explored the impact of these compounds on programmed cell death, as well as their ability to inhibit cellular invasion. In this study we have demonstrated the safety of these macrocyclic compounds by cytotoxicity tests on ex-vivo human peripheral blood mononuclear cells (PBMCs), and by in vivo subcutaneous administration. Preliminary in vivo tests demonstrated no hepato-, no nephro- and no genotoxicity in Balb/c mice compared to controls treated with cisplatin. These findings suggest these calixpyrroles might be novel therapeutic tools for the treatment of prostate cancer and of particular interest for the treatment of androgen-independent castration-resistant prostate cancer.
Collapse
Affiliation(s)
| | - Marco Ponassi
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Paola Barboro
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Erika Iervasi
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | | | | | - Leila Chekir Ghedira
- Unit of Bioactive Natural Substances and Biotechnology UR17ES47, Faculty of Dental Medicine of Monastir, University of Monastir, Monastir, Tunisia
| | | | - Alberto Izzotti
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Experimental Medicine, University of Genoa, 16132, Genoa, Italy
| | | |
Collapse
|
9
|
Zhou H, He Q, Li C, Alsharafi BLM, Deng L, Long Z, Gan Y. Focus on the tumor microenvironment: A seedbed for neuroendocrine prostate cancer. Front Cell Dev Biol 2022; 10:955669. [PMID: 35938167 PMCID: PMC9355504 DOI: 10.3389/fcell.2022.955669] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
The tumor microenvironment (TME) is a microecology consisting of tumor and mesenchymal cells and extracellular matrices. The TME plays important regulatory roles in tumor proliferation, invasion, metastasis, and differentiation. Neuroendocrine differentiation (NED) is a mechanism by which castration resistance develops in advanced prostate cancer (PCa). NED is induced after androgen deprivation therapy and neuroendocrine prostate cancer (NEPC) is established finally. NEPC has poor prognosis and short overall survival and is a major cause of death in patients with PCa. Both the cellular and non-cellular components of the TME regulate and induce NEPC formation through various pathways. Insights into the roles of the TME in NEPC evolution, growth, and progression have increased over the past few years. These novel insights will help refine the NEPC formation model and lay the foundation for the discovery of new NEPC therapies targeting the TME.
Collapse
Affiliation(s)
- Hengfeng Zhou
- Andrology Center, Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Qiangrong He
- Andrology Center, Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Chao Li
- Andrology Center, Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
| | | | - Liang Deng
- Andrology Center, Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Long
- Andrology Center, Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhi Long, ; Yu Gan,
| | - Yu Gan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhi Long, ; Yu Gan,
| |
Collapse
|
10
|
Kong P, Zhang L, Zhang Z, Feng K, Sang Y, Duan X, Liu C, Sun T, Tao Z, Liu W. Emerging Proteins in CRPC: Functional Roles and Clinical Implications. Front Oncol 2022; 12:873876. [PMID: 35756667 PMCID: PMC9226405 DOI: 10.3389/fonc.2022.873876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Prostate cancer (PCa) is the most common cancer in men in the western world, but the lack of specific and sensitive markers often leads to overtreatment of prostate cancer which eventually develops into castration-resistant prostate cancer (CRPC). Novel protein markers for diagnosis and management of CRPC will be promising. In this review, we systematically summarize and discuss the expression pattern of emerging proteins in tissue, cell lines, and serum when castration-sensitive prostate cancer (CSPC) progresses to CRPC; focus on the proteins involved in CRPC growth, invasion, metastasis, metabolism, and immune microenvironment; summarize the current understanding of the regulatory mechanisms of emerging proteins in CSPC progressed to CRPC at the molecular level; and finally summarize the clinical applications of emerging proteins as diagnostic marker, prognostic marker, predictive marker, and therapeutic marker.
Collapse
Affiliation(s)
- Piaoping Kong
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lingyu Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengliang Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Kangle Feng
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yiwen Sang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiuzhi Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Chunhua Liu
- Department of Blood Transfusion, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Sun
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Weiwei Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
11
|
Pre-Exposure to Stress-Inducing Agents Increase the Anticancer Efficacy of Focused Ultrasound against Aggressive Prostate Cancer Cells. Antioxidants (Basel) 2022; 11:antiox11020341. [PMID: 35204223 PMCID: PMC8868501 DOI: 10.3390/antiox11020341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 02/01/2023] Open
Abstract
Despite the initial success in treatment of localized prostate cancer (PCa) using surgery, radiation or hormonal therapy, recurrence of aggressive tumors dictates morbidity and mortality. Focused ultrasound (FUS) is being tested as a targeted, noninvasive approach to eliminate the localized PCa foci, and strategies to enhance the anticancer potential of FUS have a high translational value. Since aggressive cancer cells utilize oxidative stress (Ox-stress) and endoplasmic reticulum stress (ER-stress) pathways for their survival and recurrence, we hypothesized that pre-treatment with drugs that disrupt stress-signaling pathways in tumor cells may increase FUS efficacy. Using four different PCa cell lines, i.e., LNCaP, C4-2B, 22Rv1 and DU145, we tested the in vitro effects of FUS, alone and in combination with two clinically tested drugs that increase Ox-stress (i.e., CDDO-me) or ER-stress (i.e., nelfinavir). As compared to standalone FUS, significant (p < 0.05) suppressions in both survival and recurrence of PCa cells were observed following pre-sensitization with low-dose CDDO-me (100 nM) and/or nelfinavir (2 µM). In drug pre-sensitized cells, significant anticancer effects were evident at a FUS intensity of as low as 0.7 kW/cm2. This combined mechanochemical disruption (MCD) approach decreased cell proliferation, migration and clonogenic ability and increased apoptosis/necrosis and reactive oxygen species (ROS) production. Furthermore, although activated in cells that survived standalone FUS, pre-sensitization with CDDO-me and/or nelfinavir suppressed both total and activated (phosphorylated) NF-κB and Akt protein levels. Thus, a combined MCD therapy may be a safe and effective approach towards the targeted elimination of aggressive PCa cells.
Collapse
|
12
|
Pechlivanis M, Campbell BK, Hovens CM, Corcoran NM. Biomarkers of Response to Neoadjuvant Androgen Deprivation in Localised Prostate Cancer. Cancers (Basel) 2021; 14:cancers14010166. [PMID: 35008330 PMCID: PMC8750084 DOI: 10.3390/cancers14010166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/15/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Prostate cancer is the second leading cause of cancer deaths in men. Attempts to improve patient outcomes include trials of neoadjuvant androgen deprivation therapy for patients with high-risk disease. Neoadjuvant treatment refers to androgen deprivation therapy that is administered prior to surgery (or radiation therapy). Patients typically respond well to this treatment regimen, showing a decrease in tumour size, but a significant proportion of patients eventually relapse and progress to metastatic disease. The mechanisms driving this resistance to neoadjuvant treatment are currently unknown. This review explores theories of resistance broadly, and their possible applications in the prostate cancer setting. Additionally, this review draws comparisons between breakthrough resistance and neoadjuvant resistance, and lastly investigates the current biomarkers for treatment sensitivity. Abstract Prostate cancer (PCa) is a hormone driven cancer, characterised by defects in androgen receptor signalling which drive the disease process. As such, androgen targeted therapies have been the mainstay for PCa treatment for over 70 years. High-risk PCa presents unique therapeutic challenges, namely in minimising the primary tumour, and eliminating any undetected micro metastases. Trials of neoadjuvant androgen deprivation therapy aim to address these challenges. Patients typically respond well to neoadjuvant treatment, showing regression of the primary tumour and negative surgical margins at the time of resection, however the majority of patients relapse and progress to metastatic disease. The mechanisms affording this resistance are largely unknown. This commentary attempts to explore theories of resistance more broadly, namely, clonal evolution, cancer stem cells, cell persistence, and drug tolerance. Moreover, it aims to explore the application of these theories in the PCa setting. This commentary also highlights the distinction between castration resistant PCa, and neoadjuvant resistant disease, and identifies the markers and characteristics of neoadjuvant resistant disease presented by current literature.
Collapse
Affiliation(s)
- Maree Pechlivanis
- Department of Surgery, University of Melbourne, Parkville, VIC 3050, Australia; (B.K.C.); (C.M.H.); (N.M.C.)
- Correspondence: ; Tel.: +61-3-9342-7294; Fax: +61-3-9342-8928
| | - Bethany K. Campbell
- Department of Surgery, University of Melbourne, Parkville, VIC 3050, Australia; (B.K.C.); (C.M.H.); (N.M.C.)
| | - Christopher M. Hovens
- Department of Surgery, University of Melbourne, Parkville, VIC 3050, Australia; (B.K.C.); (C.M.H.); (N.M.C.)
| | - Niall M. Corcoran
- Department of Surgery, University of Melbourne, Parkville, VIC 3050, Australia; (B.K.C.); (C.M.H.); (N.M.C.)
- Department of Urology, Royal Melbourne Hospital, Parkville, VIC 3050, Australia
- Department of Urology, Western Health, Footscray, VIC 3011, Australia
- Victorian Comprehensive Cancer Centre, Melbourne, VIC 3000, Australia
| |
Collapse
|
13
|
Fu X, Zhang Z, Liu M, Li J, A J, Fu L, Huang C, Dong JT. AR imposes different effects on ZFHX3 transcription depending on androgen status in prostate cancer cells. J Cell Mol Med 2021; 26:800-812. [PMID: 34953044 PMCID: PMC8817138 DOI: 10.1111/jcmm.17125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/11/2021] [Accepted: 11/29/2021] [Indexed: 12/24/2022] Open
Abstract
Both androgen receptor (AR) and the ZFHX3 transcription factor modulate prostate development. While AR drives prostatic carcinogenesis, ZFHX3 is a tumour suppressor whose loss activates the PI3K/AKT signalling in advanced prostate cancer (PCa). However, it is unknown whether ZFHX3 and AR are functionally related in PCa cells and, if so, how. Here, we report that in AR-positive LNCaP and C4-2B PCa cells, androgen upregulates ZFHX3 transcription via androgen-induced AR binding to the androgen-responsive elements (AREs) of the ZFHX3 promoter. Androgen also upregulated ZFHX3 transcription in vivo, as castration dramatically reduced Zfhx3 mRNA and protein levels in mouse prostates, and ZFHX3 mRNA levels correlated with AR activities in human PCa. Interestingly, the binding of AR to one ARE occurred in the absence of androgen, and the binding repressed ZFHX3 transcription as this repressive binding was interrupted by androgen treatment. The enzalutamide antiandrogen prevented androgen from inducing ZFHX3 transcription and caused excess ZFHX3 protein degradation. In human PCa, ZFHX3 was downregulated and the downregulation correlated with worse patient survival. These findings establish a regulatory relationship between AR and ZFHX3, suggest a role of ZFHX3 in AR function and implicate ZFHX3 loss in the antiandrogen therapies of PCa.
Collapse
Affiliation(s)
- Xing Fu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China.,Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Zhiqian Zhang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Mingcheng Liu
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Juan Li
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jun A
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Liya Fu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Chenyang Huang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jin-Tang Dong
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
14
|
Püschel J, Dubrovska A, Gorodetska I. The Multifaceted Role of Aldehyde Dehydrogenases in Prostate Cancer Stem Cells. Cancers (Basel) 2021; 13:4703. [PMID: 34572930 PMCID: PMC8472046 DOI: 10.3390/cancers13184703] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/27/2021] [Accepted: 09/13/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) are the only tumor cells possessing self-renewal and differentiation properties, making them an engine of tumor progression and a source of tumor regrowth after treatment. Conventional therapies eliminate most non-CSCs, while CSCs often remain radiation and drug resistant, leading to tumor relapse and metastases. Thus, targeting CSCs might be a powerful tool to overcome tumor resistance and increase the efficiency of current cancer treatment strategies. The identification and isolation of the CSC population based on its high aldehyde dehydrogenase activity (ALDH) is widely accepted for prostate cancer (PCa) and many other solid tumors. In PCa, several ALDH genes contribute to the ALDH activity, which can be measured in the enzymatic assay by converting 4, 4-difluoro-4-bora-3a, 4a-diaza-s-indacene (BODIPY) aminoacetaldehyde (BAAA) into the fluorescent product BODIPY-aminoacetate (BAA). Although each ALDH isoform plays an individual role in PCa biology, their mutual functional interplay also contributes to PCa progression. Thus, ALDH proteins are markers and functional regulators of CSC properties, representing an attractive target for cancer treatment. In this review, we discuss the current state of research regarding the role of individual ALDH isoforms in PCa development and progression, their possible therapeutic targeting, and provide an outlook for the future advances in this field.
Collapse
Affiliation(s)
- Jakob Püschel
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany;
| | - Anna Dubrovska
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany;
- National Center for Tumor Diseases (NCT), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Ielizaveta Gorodetska
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany;
| |
Collapse
|
15
|
Photodynamic therapy of prostate cancer using porphyrinic formulations. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 223:112301. [PMID: 34492530 DOI: 10.1016/j.jphotobiol.2021.112301] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/01/2021] [Accepted: 08/24/2021] [Indexed: 01/21/2023]
Abstract
Prostate cancer (PCa) is the second most frequent cancer diagnosed in men worldwide. Among the common treatment options, photodynamic therapy (PDT) is being considered a promising local therapy to treat this cancer. Although PDT is an established treatment modality approved for several types of cancer, the low solubility, the reduced tumor selectivity, the absorption in the therapeutic window and the poor clearance from the body of the currently approved photosensitizers (PS) hampers its wide clinical application. In this regard, herein we synthesized three fluorinated porphyrinoid derivatives and entrapped them into polyvinylpyrrolidone (PVP) to prevent their aggregation and preserve their desirable photophysical properties under the physiological environment. In vitro studies revealed the negligible dark cytotoxicity of all PVP formulations (PS1@PVP, PS2@PVP and PS3@PVP) at the tested concentrations (5.0 to 20 μM), but also confirmed the significant photodynamic effect of PS2@PVP and PS3@PVP towards the PCa cell line PC-3, upon red light irradiation at an irradiance of 17.6 mW.cm-2. To provide insight into the underlying mechanisms of cell death under PDT treatment induced by PS2@PVP and PS3@PVP, their intracellular localization in PC-3 cells was firstly investigated by confocal microscopy. Since both PS2@PVP and PS3@PVP nanoparticles were mainly localized in mitochondria, the involvement of this organelle in PDT-induced apoptosis mediated by both formulations was further explored. Western blot analysis revealed that PDT treatment of PC-3 cells with either PS2@PVP or PS3@PVP resulted in the reduction of the expression level of the anti-apoptotic protein Bcl-2. As the photodamage to Bcl-2 after PDT with PS2@PVP and PS3@PVP was accompanied by the further activation of pro-caspase-3, we assumed that upon irradiation the photogenerated reactive oxygen species (ROS) were able to activate a caspase-dependent apoptotic response as a consequence of a post-mitochondrial event. Taken together, these findings demonstrate that among the tested fluorinated porphyrinoids, PS2@PVP and, particularly, PS3@PVP, are significantly more effective in overall PC-3 cell killing than PS1@PVP, thus highlighting their great potential as therapeutic agents for PCa.
Collapse
|
16
|
Barboro P, Benelli R, Tosetti F, Costa D, Capaia M, Astigiano S, Venè R, Poggi A, Ferrari N. Aspartate β-hydroxylase targeting in castration-resistant prostate cancer modulates the NOTCH/HIF1α/GSK3β crosstalk. Carcinogenesis 2021; 41:1246-1252. [PMID: 32525968 DOI: 10.1093/carcin/bgaa053] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 05/17/2020] [Accepted: 06/06/2020] [Indexed: 12/14/2022] Open
Abstract
Castration-resistant prostate cancer (CRPC) is an incurable stage of the disease. A multivariate principal component analysis on CRPC in vitro models identified aspartyl (asparaginyl) β hydrolase (ASPH) as the most relevant molecule associated with the CRPC phenotype. ASPH is overexpressed in various malignant neoplasms and catalyzes the hydroxylation of aspartyl and asparaginyl residues in the epidermal growth factor (EGF)-like domains of proteins like NOTCH receptors and ligands, enhancing cell motility, invasion and metastatic spread. Bioinformatics analyses of ASPH in prostate cancer (PCa) and CRPC datasets indicate that ASPH gene alterations have prognostic value both in PCa and CRPC patients. In CRPC cells, inhibition of ASPH expression obtained through specific small interfering RNA or culturing cells in hypoxic conditions, reduced cell proliferation, invasion and cyclin D1 expression through modulation of the NOTCH signaling. ASPH and HIF1α crosstalk, within a hydroxylation-regulated signaling pathway, might be transiently driven by the oxidative stress evidenced inside CRPC cells. In addition, increased phosphorylation of GSK3β by ASPH silencing demonstrates that ASPH regulates GSK3β activity inhibiting its interactions with upstream kinases. These findings demonstrate the critical involvement of ASPH in CRPC development and may represent an attractive molecular target for therapy.
Collapse
Affiliation(s)
- Paola Barboro
- Department of Oncology and Hematology, Academic Unit of Medical Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Roberto Benelli
- Department of Scientific Direction, Molecular Oncology & Angiogenesis, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesca Tosetti
- Department of Scientific Direction, Molecular Oncology & Angiogenesis, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Delfina Costa
- Department of Scientific Direction, Molecular Oncology & Angiogenesis, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Matteo Capaia
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, Genova, Italy
| | - Simonetta Astigiano
- Department of Scientific Direction, Immunology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Roberta Venè
- Department of Scientific Direction, Molecular Oncology & Angiogenesis, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Alessandro Poggi
- Department of Scientific Direction, Molecular Oncology & Angiogenesis, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Nicoletta Ferrari
- Department of Scientific Direction, Molecular Oncology & Angiogenesis, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
17
|
Identification and Characterization of Alternatively Spliced Transcript Isoforms of IRX4 in Prostate Cancer. Genes (Basel) 2021; 12:genes12050615. [PMID: 33919200 PMCID: PMC8143155 DOI: 10.3390/genes12050615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/06/2021] [Accepted: 04/13/2021] [Indexed: 01/19/2023] Open
Abstract
Alternative splicing (AS) is tightly regulated to maintain genomic stability in humans. However, tumor growth, metastasis and therapy resistance benefit from aberrant RNA splicing. Iroquois-class homeodomain protein 4 (IRX4) is a TALE homeobox transcription factor which has been implicated in prostate cancer (PCa) as a tumor suppressor through genome-wide association studies (GWAS) and functional follow-up studies. In the current study, we characterized 12 IRX4 transcripts in PCa cell lines, including seven novel transcripts by RT-PCR and sequencing. They demonstrate unique expression profiles between androgen-responsive and nonresponsive cell lines. These transcripts were significantly overexpressed in PCa cell lines and the cancer genome atlas program (TCGA) PCa clinical specimens, suggesting their probable involvement in PCa progression. Moreover, a PCa risk-associated SNP rs12653946 genotype GG was corelated with lower IRX4 transcript levels. Using mass spectrometry analysis, we identified two IRX4 protein isoforms (54.4 kDa, 57 kDa) comprising all the functional domains and two novel isoforms (40 kDa, 8.7 kDa) lacking functional domains. These IRX4 isoforms might induce distinct functional programming that could contribute to PCa hallmarks, thus providing novel insights into diagnostic, prognostic and therapeutic significance in PCa management.
Collapse
|
18
|
Liyanage C, Malik A, Abeysinghe P, Clements J, Batra J. SWATH-MS Based Proteomic Profiling of Prostate Cancer Cells Reveals Adaptive Molecular Mechanisms in Response to Anti-Androgen Therapy. Cancers (Basel) 2021; 13:715. [PMID: 33572476 PMCID: PMC7916382 DOI: 10.3390/cancers13040715] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/15/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer (PCa) is the second most common cancer affecting men worldwide. PCa shows a broad-spectrum heterogeneity in its biological and clinical behavior. Although androgen targeted therapy (ATT) has been the mainstay therapy for advanced PCa, it inevitably leads to treatment resistance and progression to castration resistant PCa (CRPC). Thus, greater understanding of the molecular basis of treatment resistance and CRPC progression is needed to improve treatments for this lethal phenotype. The current study interrogated both proteomics and transcriptomic alterations stimulated in AR antagonist/anti-androgen (Bicalutamide and Enzalutamide) treated androgen-dependent cell model (LNCaP) in comparison with androgen-independent/castration-resistant cell model (C4-2B). The analysis highlighted the activation of MYC and PSF/SFPQ oncogenic upstream regulators in response to the anti-androgen treatment. Moreover, the study revealed anti-androgen induced genes/proteins related to transcription/translation regulation, energy metabolism, cell communication and signaling cascades promoting tumor growth and proliferation. In addition, these molecules were found dysregulated in PCa clinical proteomic and transcriptomic datasets, suggesting their potential involvement in PCa progression. In conclusion, our study provides key molecular signatures and associated pathways that might contribute to CRPC progression despite treatment with anti-androgens. Such molecular signatures could be potential therapeutic targets to improve the efficacy of existing therapies and/or predictive/prognostic value in CRPC for treatment response.
Collapse
Affiliation(s)
- Chamikara Liyanage
- Faculty of Health, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4059, Australia; (C.L.); (A.M.); (P.A.); (J.C.)
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Queensland University of Technology, Brisbane, QLD 4012, Australia
| | - Adil Malik
- Faculty of Health, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4059, Australia; (C.L.); (A.M.); (P.A.); (J.C.)
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Queensland University of Technology, Brisbane, QLD 4012, Australia
| | - Pevindu Abeysinghe
- Faculty of Health, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4059, Australia; (C.L.); (A.M.); (P.A.); (J.C.)
| | - Judith Clements
- Faculty of Health, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4059, Australia; (C.L.); (A.M.); (P.A.); (J.C.)
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Queensland University of Technology, Brisbane, QLD 4012, Australia
| | - Jyotsna Batra
- Faculty of Health, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4059, Australia; (C.L.); (A.M.); (P.A.); (J.C.)
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Queensland University of Technology, Brisbane, QLD 4012, Australia
| |
Collapse
|
19
|
Etoposide and topoisomerase II inhibition for aggressive prostate cancer: Data from a translational study. Cancer Treat Res Commun 2020; 25:100221. [PMID: 33091733 DOI: 10.1016/j.ctarc.2020.100221] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/05/2020] [Accepted: 10/04/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Etoposide phosphate (VP-16) is a topoisomerase 2 (TOP2) inhibitor that demonstrated activity in patients with metastatic castration-resistant prostate cancer (mCRPC). We investigated the sensitivity of prostate cancer (PCa) cells (LNCaP, 22Rv1, PC3, DU145, PDB and MDB) to VP-16 and the possible relationship between VP-16 activity and TOP2 expression. The activity of VP-16 was compared with that of docetaxel, enzalutamide and olaparib. The prevalence and clinical significance of TOP2 genetic and transcriptomic alterations was also explored in mCRPC. METHODS Cell cultures and crystal violet cell proliferation assays were performed. Specific antibodies were used in western blots analyses of cell protein extracts. Datasets were analyzed in cBioportal. RESULTS VP-16 was active in all PCa cell lines analyzed and demonstrated increased activity in PC3 and DU145 cells. VP-16 was more cytotoxic compared to the other treatments, except for LNCaP and 22Rv1, which were more sensitive to docetaxel. Maintenance of antiandrogen treatment in MDB and PDB increased sensitivity to VP-16, docetaxel and enzalutamide. TOP2A was found overexpressed in 22Rv1, DU145 and PC3, whereas TOP2B was overexpressed in 22Rv1 and PDB. In the mCRPC datasets analysis, TOP2A mRNA overexpression was associated with worse patients' prognosis, with the molecular features of neuroendocrine prostate cancer (NEPC) and with lower androgen receptor (AR) score. Patients overexpressing TOP2A mRNA were more likely to harbor RB1 loss. CONCLUSIONS Specific subpopulations of patients with aggressive variant prostate cancer (AVPC) could benefit from VP-16 treatment. TOP2A overexpression, rather than TOP2B, might be a good biomarker to predict response to VP-16.
Collapse
|
20
|
Pérez-Velázquez J, Rejniak KA. Drug-Induced Resistance in Micrometastases: Analysis of Spatio-Temporal Cell Lineages. Front Physiol 2020; 11:319. [PMID: 32362836 PMCID: PMC7180185 DOI: 10.3389/fphys.2020.00319] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/20/2020] [Indexed: 12/16/2022] Open
Abstract
Resistance to anti-cancer drugs is a major cause of treatment failure. While several intracellular mechanisms of resistance have been postulated, the role of extrinsic factors in the development of resistance in individual tumor cells is still not fully understood. Here we used a hybrid agent-based model to investigate how sensitive tumor cells develop drug resistance in the heterogeneous tumor microenvironment. We characterized the spatio-temporal evolution of lineages of the resistant cells and examined how resistance at the single-cell level contributes to the overall tumor resistance. We also developed new methods to track tumor cell adaptation, to trace cell viability trajectories and to examine the three-dimensional spatio-temporal lineage trees. Our findings indicate that drug-induced resistance can result from cells adaptation to the changes in drug distribution. Two modes of cell adaptation were identified that coincide with microenvironmental niches—areas sheltered by cell micro-communities (protectorates) or regions with limited drug penetration (refuga or sanctuaries). We also recognized that certain cells gave rise to lineages of resistant cells (precursors of resistance) and pinpointed three temporal periods and spatial locations at which such cells emerged. This supports the hypothesis that tumor micrometastases do not need to harbor cell populations with pre-existing resistance, but that individual tumor cells can adapt and develop resistance induced by the drug during the treatment.
Collapse
Affiliation(s)
- Judith Pérez-Velázquez
- Mathematical Modeling of Biological Systems, Centre for Mathematical Science, Technical University of Munich, Garching, Germany
| | - Katarzyna A Rejniak
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States.,Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, Tampa, FL, United States
| |
Collapse
|
21
|
Qiao H, Tan X, Lv DJ, Xing RW, Shu FP, Zhong CF, Li C, Zou YG, Mao XM. Phosphoribosyl pyrophosphate synthetases 2 knockdown inhibits prostate cancer progression by suppressing cell cycle and inducing cell apoptosis. J Cancer 2020; 11:1027-1037. [PMID: 31956349 PMCID: PMC6959080 DOI: 10.7150/jca.37401] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 10/11/2019] [Indexed: 12/24/2022] Open
Abstract
Phosphoribosyl pyrophosphate synthetases 2 (PRPS2) protein function as nucleotide synthesis enzyme that plays vital roles in cancer biology. However, the expression profile and function of PRPS2 in prostate cancer (PCa) remain to be identified. Here we investigated the expression of PRPS2 protein in human PCa and paired normal tissues by immunohistochemistry, meanwhile the regulatory effects on cell proliferation, apoptosis and growth of xenograft tumors in nude mice were evaluated in PCa cells with PRPS2 depletion. Moreover, the signaling pathways were also explored by western blot analysis and quantitative polymerase chain reaction assays. We found that PRPS2 was dramatically upregulated in prostate adenocarcinoma tissues in comparison with normal tissues, and that increased PRPS2 was linked intimately to advanced clinical stage and pT status. Functional experiments showed that knockdown of PRPS2 significantly suppressed cell growth both in vitro and in vivo. In addition, depletion of PRPS2 induced G1 phase cell cycle arrest and elevated cell apoptosis. Silencing of PRPS2 resulted in the decreased expression of Bcl‑2 and cyclinD1 and increased levels of Bax, cleavage of caspases‑3, caspases‑9 and PARP. Furthermore, we also detected PRPS2 expression was significantly induced after DHT treatment, which implied the important role of PRPS2 in oncogenesis of PCa. Taken together, our findings elucidated that PRPS2 may be a potential novel candidate for PCa therapy.
Collapse
Affiliation(s)
- Hui Qiao
- Nursing Department, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong Province, China
| | - Xiao Tan
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong Province China
| | - Dao-Jun Lv
- Department of Urology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong Province, China.,Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong Province China
| | - Rong-Wei Xing
- Department of Urology, the Affiliated Weihai Second Municipal Hospital of Qingdao University, 264200, Weihai, Shandong Province, China
| | - Fang-Peng Shu
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong Province China
| | - Chuan-Fan Zhong
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong Province China
| | - Chun Li
- Nursing Department, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong Province, China
| | - Ya-Guang Zou
- Department of Stomatology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong Province, China
| | - Xiang-Ming Mao
- Department of Urology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong Province, China.,Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong Province China
| |
Collapse
|
22
|
Multifocal Signal Modulation Therapy by Celecoxib: A Strategy for Managing Castration-Resistant Prostate Cancer. Int J Mol Sci 2019; 20:ijms20236091. [PMID: 31816863 PMCID: PMC6929142 DOI: 10.3390/ijms20236091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/28/2019] [Accepted: 11/29/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a significant health concern throughout the world. Standard therapy for advanced disease consists of anti-androgens, however, almost all prostate tumors become castration resistant (CRPC). Progression from androgen-sensitive PCa to CRPC is promoted by inflammatory signaling through cyclooxygenase-2 (COX-2) expression and ErbB family receptors/AKT activation, compensating androgen receptor inactivity. METHODS Making use of CRPC cell lines, we investigated the effects of the anti-inflammatory drug celecoxib. Biochemical data obtained using immunoblotting, enzyme-linked immunosorbent assay (ELISA), invasion, and xenografts were further integrated by bioinformatic analyses. RESULTS Celecoxib reduced cell growth and induced apoptosis through AKT blockade, cleavage of poly (ADP-ribose) polymerase-1 (PARP-1), and proteasomal degradation of the anti-apoptotic protein Mcl-1. Epidermal growth factor receptor (EGFR), ErbB2, and ErbB3 degradation, and heterogeneous nuclear ribonucleoprotein K (hnRNP K) downregulation, further amplified the inhibition of androgen signaling. Celecoxib reduced the invasive phenotype of CRPC cells by modulating NF-κB activity and reduced tumor growth in mice xenografts when administered in association with the anti-EGFR receptor antibody cetuximab. Bioinformatic analyses on human prostate cancer datasets support the relevance of these pathways in PCa progression. CONCLUSIONS Signaling nodes at the intersection of pathways implicated in PCa progression are simultaneously modulated by celecoxib treatment. In combination therapies with cetuximab, celecoxib could represent a novel therapeutic strategy to curb signal transduction during CRPC progression.
Collapse
|
23
|
Granata I, Troiano E, Sangiovanni M, Guarracino MR. Integration of transcriptomic data in a genome-scale metabolic model to investigate the link between obesity and breast cancer. BMC Bioinformatics 2019; 20:162. [PMID: 30999849 PMCID: PMC6471692 DOI: 10.1186/s12859-019-2685-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Obesity is a complex disorder associated with an increased risk of developing several comorbid chronic diseases, including postmenopausal breast cancer. Although many studies have investigated this issue, the link between body weight and either risk or poor outcome of breast cancer is still to characterize. Systems biology approaches, based on the integration of multiscale models and data from a wide variety of sources, are particularly suitable for investigating the underlying molecular mechanisms of complex diseases. In this scenario, GEnome-scale metabolic Models (GEMs) are a valuable tool, since they represent the metabolic structure of cells and provide a functional scaffold for simulating and quantifying metabolic fluxes in living organisms through constraint-based mathematical methods. The integration of omics data into the structural information described by GEMs allows to build more accurate descriptions of metabolic states. RESULTS In this work, we exploited gene expression data of postmenopausal breast cancer obese and lean patients to simulate a curated GEM of the human adipocyte, available in the Human Metabolic Atlas database. To this aim, we used a published algorithm which exploits a data-driven approach to overcome the limitation of defining a single objective function to simulate the model. The flux solutions were used to build condition-specific graphs to visualise and investigate the reaction networks and their properties. In particular, we performed a network topology differential analysis to search for pattern differences and identify the principal reactions associated with significant changes across the two conditions under study. CONCLUSIONS Metabolic network models represent an important source to study the metabolic phenotype of an organism in different conditions. Here we demonstrate the importance of exploiting Next Generation Sequencing data to perform condition-specific GEM analyses. In particular, we show that the qualitative and quantitative assessment of metabolic fluxes modulated by gene expression data provides a valuable method for investigating the mechanisms associated with the phenotype under study, and can foster our interpretation of biological phenomena.
Collapse
Affiliation(s)
- Ilaria Granata
- High Performance Computing and Networking Institute, National Research Council of Italy, Via P. Castellino, 111, Napoli, 80131 Italy
| | - Enrico Troiano
- High Performance Computing and Networking Institute, National Research Council of Italy, Via P. Castellino, 111, Napoli, 80131 Italy
| | - Mara Sangiovanni
- Stazione Zoologica Anton Dohrn, Villa Comunale, Napoli, 80121 Italy
| | - Mario Rosario Guarracino
- High Performance Computing and Networking Institute, National Research Council of Italy, Via P. Castellino, 111, Napoli, 80131 Italy
| |
Collapse
|
24
|
Manipur I, Granata I, Guarracino MR. Exploiting single-cell RNA sequencing data to link alternative splicing and cancer heterogeneity: A computational approach. Int J Biochem Cell Biol 2019; 108:51-60. [PMID: 30633986 DOI: 10.1016/j.biocel.2018.12.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/21/2018] [Accepted: 12/24/2018] [Indexed: 02/09/2023]
Abstract
Cell heterogeneity studies using single-cell sequencing are gaining great significance in the era of personalized medicine. In particular, characterization of tumor heterogeneity is an emergent issue to improve clinical oncology, since both inter- and intra-tumor level heterogeneity influence the utility and application of molecular classifications through specific biomarkers. Majority of studies have exploited gene expression to discriminate cell types. However, to provide a more nuanced view of the underlying differences, isoform expression and alternative splicing events have to be analyzed in detail. In this study, we utilize publicly available single cell and bulk RNA sequencing datasets of breast cancer cells from primary tumors and immortalized cell lines. Breast cancer is very heterogeneous with well defined molecular subtypes and was therefore chosen for this study. RNA-seq data were explored in terms of genes, isoforms abundance and splicing events. The study was conducted from an average based approach (gene level expression) to detailed and deeper ones (isoforms abundance/splicing events) to perform a comparative analysis, and, thus, highlight the importance of the splicing machinery in defining the tumor heterogeneity. Moreover, here we demonstrate how the investigation of gene isoforms expression can help to identify the appropriate in vitro models. We furthermore extracted marker isoforms, and alternatively spliced genes between and within the different single cell populations to improve the classification of the breast cancer subtypes.
Collapse
Affiliation(s)
- Ichcha Manipur
- High Performance Computing and Networking Institute, National Research Council, Italy
| | - Ilaria Granata
- High Performance Computing and Networking Institute, National Research Council, Italy.
| | | |
Collapse
|
25
|
Li S, Li R, Ma Y, Zhang C, Huang T, Zhu S. Transcriptome analysis of differentially expressed genes and pathways associated with mitoxantrone treatment prostate cancer. J Cell Mol Med 2018; 23:1987-2000. [PMID: 30592148 PMCID: PMC6378179 DOI: 10.1111/jcmm.14100] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 12/12/2022] Open
Abstract
The global physiological function of specifically expressed genes of mitoxantrone (MTX)‐resistant prostate cancer (PCa) is unclear. In this study, gene expression pattern from microarray data was investigated for identifying differentially expressed genes (DEGs) in MTX‐resistant PCa xenografts. Human PCa cell lines DU145 and PC3 were cultured in vitro and xenografted into severe combined immunodeficiency (SCID) mice, treated with MTX intragastrically, three times a week until all mice relapsed. Gene expression profiles of the xenografts from castrated mice were performed with Affymetrix human whole genomic oligonucleotide microarray. The Cytoscape software was used to investigate the relationship between proteins and the signalling transduction network. A total of 355 overlapping genes were differentially expressed in MTX‐resistant DU145R and PC3R xenografts. Of these, 16 genes were selected to be validated by quantitative real‐time PCR (qRT‐PCR) in these xenografts, and further tested in a set of formalin‐fixed, paraffin‐embedded and optimal cutting temperature (OCT) clinical tumour samples. Functional and pathway enrichment analyses revealed that these DEGs were closely related to cellular activity, androgen synthesis, DNA damage and repair, also involved in the ERK/MAPK, PI3K/serine‐threonine protein kinase, also known as protein kinase B, PKB (AKT) and apoptosis signalling pathways. This exploratory analysis provides information about potential candidate genes and may bring new insights into the molecular cascade involvement in MTX‐resistant PCa.
Collapse
Affiliation(s)
- Sanqiang Li
- Key laboratory of Infection and Immunization, Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.,Medical College, Henan University of Science and Technology, Luoyang, Henan, China
| | - Ruifang Li
- College of Biological Engineering, Henan University of Technology, Zhengzhou, Henan, China
| | - Yu Ma
- Key laboratory of Infection and Immunization, Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Cong Zhang
- Key laboratory of Infection and Immunization, Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Tao Huang
- Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Sha Zhu
- Key laboratory of Infection and Immunization, Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.,Collaborative Innovation Center of Cancer Chemoprevention, Zhengzhou, Henan, China
| |
Collapse
|
26
|
Zhang L, Li X, Chao Y, He R, Liu J, Yuan Y, Zhao W, Han C, Song X. KLF4, a miR-32-5p targeted gene, promotes cisplatin-induced apoptosis by upregulating BIK expression in prostate cancer. Cell Commun Signal 2018; 16:53. [PMID: 30176890 PMCID: PMC6122640 DOI: 10.1186/s12964-018-0270-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/30/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Chemotherapeutic insensitivity remains a big challenge in prostate cancer treatment. Recently, increasing evidence has indicated that KLF4 plays a key role in prostate cancer. However, the potential biological role of KLF4 in Chemotherapeutic insensitivity of prostate cancer is still unknown. METHODS The role of KLF4 in cisplatin-induced apoptosis was detected by western blotting and a cell counting kit (CCK8). The potential molecular mechanism of KLF4 in regulating prostate cancer chemosensitivity was investigated by RNA sequencing analysis, q-RT-PCR, western blotting and chromatin immunoprecipitation (ChIP). The expression level of KLF4 mediated by miR-32-5p was confirmed by bioinformatic analysis and luciferase assays. RESULTS Here, we found that KLF4 was induced by cisplatin in prostate cancer cells and that the increase in KLF4 promoted cell apoptosis. Further mechanistic studies revealed that KLF4 directly bound to the promoter of BIK, facilitating its transcription. Additionally, we also found that the gene encoding KLF4 was a direct target of miR-32-5p. The downregulation of miR-32-5p in response to cisplatin treatment promoted KLF4 expression, which resulted in a increase in the chemosensitivity of prostate cancer. CONCLUSION Thus, our data revealed that KLF4 is an essential regulator in cisplatin-induced apoptosis, and the miR-32-5p-KLF4-BIK signalling axis plays an important role in prostate cancer chemosensitivity.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Urology of the First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044 People’s Republic of China
- Department of Orthopedics, Second Affiliated Hospital, Dalian Medical University, Dalian, 116044 China
| | - Xiaojie Li
- Department of Urology of the First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044 People’s Republic of China
- College of Stomatology, Dalian Medical University, Dalian, 116044 China
| | - Yulin Chao
- Department of Urology of the First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044 People’s Republic of China
| | - Ruiping He
- Department of Urology of the First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044 People’s Republic of China
| | - Junqiang Liu
- Department of Urology of the First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044 People’s Republic of China
| | - Yi Yuan
- Department of Urology of the First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044 People’s Republic of China
| | - Wenzhi Zhao
- Department of Orthopedics, Second Affiliated Hospital, Dalian Medical University, Dalian, 116044 China
| | - Chuanchun Han
- Department of Urology of the First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044 People’s Republic of China
| | - Xishuang Song
- Department of Urology of the First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044 People’s Republic of China
| |
Collapse
|
27
|
Capaia M, Granata I, Guarracino M, Petretto A, Inglese E, Cattrini C, Ferrari N, Boccardo F, Barboro P. A hnRNP K⁻AR-Related Signature Reflects Progression toward Castration-Resistant Prostate Cancer. Int J Mol Sci 2018; 19:ijms19071920. [PMID: 29966326 PMCID: PMC6073607 DOI: 10.3390/ijms19071920] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 06/27/2018] [Accepted: 06/29/2018] [Indexed: 12/21/2022] Open
Abstract
The major challenge in castration-resistant prostate cancer (CRPC) remains the ability to predict the clinical responses to improve patient selection for appropriate treatments. The finding that androgen deprivation therapy (ADT) induces alterations in the androgen receptor (AR) transcriptional program by AR coregulators activity in a context-dependent manner, offers the opportunity for identifying signatures discriminating different clinical states of prostate cancer (PCa) progression. Gel electrophoretic analyses combined with western blot showed that, in androgen-dependent PCa and CRPC in vitro models, the subcellular distribution of spliced and serine-phosphorylated heterogeneous nuclear ribonucleoprotein K (hnRNP K) isoforms can be associated with different AR activities. Using mass spectrometry and bioinformatic analyses, we showed that the protein sets of androgen-dependent (LNCaP) and ADT-resistant cell lines (PDB and MDB) co-immunoprecipitated with hnRNP K varied depending on the cell type, unravelling a dynamic relationship between hnRNP K and AR during PCa progression to CRPC. By comparing the interactome of LNCaP, PDB, and MDB cell lines, we identified 51 proteins differentially interacting with hnRNP K, among which KLK3, SORD, SPON2, IMPDH2, ACTN4, ATP1B1, HSPB1, and KHDRBS1 were associated with AR and differentially expressed in normal and tumor human prostate tissues. This hnRNP K–AR-related signature, associated with androgen sensitivity and PCa progression, may help clinicians to better manage patients with CRPC.
Collapse
Affiliation(s)
- Matteo Capaia
- Academic Unit of Medical Oncology, Ospedale Policlinico San Martino-IRCCS, L.go R. Benzi 10, 16132 Genova, Italy.
| | - Ilaria Granata
- Institute for High Performance Computing and Networking (ICAR), National Research Council (CNR), Via Pietro Castellino 111, 80131 Napoli, Italy.
| | - Mario Guarracino
- Institute for High Performance Computing and Networking (ICAR), National Research Council (CNR), Via Pietro Castellino 111, 80131 Napoli, Italy.
| | - Andrea Petretto
- Core Facilities-Proteomics Laboratory, Giannina Gaslini Institute, L.go G. Gaslini 5, 16147 Genova, Italy.
| | - Elvira Inglese
- Core Facilities-Proteomics Laboratory, Giannina Gaslini Institute, L.go G. Gaslini 5, 16147 Genova, Italy.
| | - Carlo Cattrini
- Academic Unit of Medical Oncology, Ospedale Policlinico San Martino-IRCCS, L.go R. Benzi 10, 16132 Genova, Italy.
- Department of Internal Medicine and Medical Specialties, School of Medicine, University of Genova, L.go R. Benzi 10, 16132 Genova, Italy.
| | - Nicoletta Ferrari
- Molecular Oncology and Angiogenesis, Ospedale Policlinico San Martino-IRCCS, L.go R. Benzi 10, 16132 Genova, Italy.
| | - Francesco Boccardo
- Academic Unit of Medical Oncology, Ospedale Policlinico San Martino-IRCCS, L.go R. Benzi 10, 16132 Genova, Italy.
- Department of Internal Medicine and Medical Specialties, School of Medicine, University of Genova, L.go R. Benzi 10, 16132 Genova, Italy.
| | - Paola Barboro
- Academic Unit of Medical Oncology, Ospedale Policlinico San Martino-IRCCS, L.go R. Benzi 10, 16132 Genova, Italy.
| |
Collapse
|