1
|
Shen J, Lu L, Chen Z, Guo W, Wang S, Liu Z, Gong X, Qi Y, Jin R, Zhang C. Multi-omics analysis constructs a novel neuroendocrine prostate cancer classifier and classification system. Sci Rep 2025; 15:13901. [PMID: 40263498 PMCID: PMC12015331 DOI: 10.1038/s41598-025-96683-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/31/2025] [Indexed: 04/24/2025] Open
Abstract
Neuroendocrine prostate cancer (NEPC), a subtype of prostate cancer (PCa) with poor prognosis and high heterogeneity, currently lacks accurate markers. This study aims to identify a robust NEPC classifier and provide new perspectives for resolving intra- tumoral heterogeneity. Multi-omics analysis included 19 bulk transcriptomics, 14 single-cell transcriptomics, 1 spatial transcriptomics, 16 published NE signatures and 10 cellular experiments combined with multiple machine learning algorithms to construct a novel NEPC classifier and classification. A comprehensive single-cell atlas of prostate cancer was created from 70 samples, comprising 196,309 cells, among which 9% were identified as NE cells. Within this framework and in combination with bulk transcriptomics, a total of 100 high-quality NE-specific feature genes were identified and differentiated into NEPup sig and NEPdown sig. The random forest (RF) algorithm proved to be the most effective classifier for NEPC, leading to the establishment of the NEP100 model, which demonstrated robust validation across various datasets. In clinical settings, the use of the NEP100 model can greatly improve the diagnostic and prognostic prediction of NEPC. Hierarchical clustering based on NEP100 revealed four distinct NEPC subtypes, designated VR_O, Prol_N, Prol_P, and EMT_Y, each of which presented unique biological characteristics. This allows us to select different targeted therapeutic strategies for different subtypes of phenotypic pathways. Notably, NEP100 expression correlated positively with neuroendocrine differentiation and disease progression, while the VR-NE phenotype dominated by VR_O cells indicated a propensity for treatment resistance. Furthermore, AMIGO2, a component of the NEP100 signature, was associated with chemotherapy resistance and a poor prognosis, indicating that it is a pivotal target for future therapeutic strategies. This study used multi-omics analysis combined with machine learning to construct a novel NEPC classifier and classification system. NEP100 provides a clinically actionable framework for NEPC diagnosis and subtyping.
Collapse
Affiliation(s)
- Junxiao Shen
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Luyuan Lu
- Department of General Surgery, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Zujie Chen
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Wei Guo
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Shuwen Wang
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Ziqiao Liu
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xuke Gong
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yiming Qi
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Ruyi Jin
- Department of Dermatology, NHC Key Laboratory of Immunodermatology, The First Hospital of China Medical University, China Medical University, Shenyang, 110001, People's Republic of China
| | - Cheng Zhang
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| |
Collapse
|
2
|
Zhuang D, Kang J, Luo H, Tian Y, Liu X, Shao C. ARv7 promotes the escape of prostate cancer cells from androgen deprivation therapy-induced senescence by mediating the SKP2/p27 axis. BMC Biol 2025; 23:66. [PMID: 40022149 PMCID: PMC11871636 DOI: 10.1186/s12915-025-02172-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/13/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Androgen deprivation therapy (ADT) induces cellular senescence and tumor stasis, thus serving as the standard treatment for prostate cancer (PCa). However, continuous suppression of canonical androgen receptor signaling actually leads to the switch from androgen-responsive growth to androgen-independent growth, contributing to "escape" from this ADT-induced senescence (AIS) and, subsequently, the development of castration-resistant prostate cancer (CRPC). Unfortunately, the mechanism underlying this phenomenon remains elusive. RESULTS In this study, we demonstrated that androgen receptor splicing variant 7 (ARv7), a dominant factor mediating abnormal AR signaling and ADT resistance, is closely associated with outgrowth from AIS of PCa cells. Mechanistically, ARv7 binds to the promoter of SKP2, activating its transcription, and then promotes the proteasomal degradation of the cell cycle regulator p27 and G1/S transition. In addition, we applied bioinformatic and in vitro analyses to show that SKP2 expression level is dramatically inhibited upon ADT, but its reactivation is one key step during the establishment of CRPC. Finally, we also demonstrated that SKP2 inhibitor treatment can significantly inhibit the growth of androgen-independent cell lines and enhance the efficacy of ADT. CONCLUSIONS Our work reveals a novel role of ARv7 in regulating AIS and suggests that targeting the ARv7/SKP2/p27 axis could be a potential strategy to delay disease progression to the CRPC state during prolonged ADT.
Collapse
Affiliation(s)
- Dian Zhuang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Jinsong Kang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Haoge Luo
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Yu Tian
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Xiaoping Liu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Chen Shao
- Department of Biochemistry & Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.
| |
Collapse
|
3
|
Rahman M, Akter K, Ahmed KR, Fahim MMH, Aktary N, Park MN, Shin SW, Kim B. Synergistic Strategies for Castration-Resistant Prostate Cancer: Targeting AR-V7, Exploring Natural Compounds, and Optimizing FDA-Approved Therapies. Cancers (Basel) 2024; 16:2777. [PMID: 39199550 PMCID: PMC11352813 DOI: 10.3390/cancers16162777] [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: 07/01/2024] [Revised: 07/29/2024] [Accepted: 08/03/2024] [Indexed: 09/01/2024] Open
Abstract
Castration-resistant prostate cancer (CRPC) remains a significant therapeutic challenge due to its resistance to standard androgen deprivation therapy (ADT). The emergence of androgen receptor splice variant 7 (AR-V7) has been implicated in CRPC progression, contributing to treatment resistance. Current treatments, including first-generation chemotherapy, androgen receptor blockers, radiation therapy, immune therapy, and PARP inhibitors, often come with substantial side effects and limited efficacy. Natural compounds, particularly those derived from herbal medicine, have garnered increasing interest as adjunctive therapeutic agents against CRPC. This review explores the role of AR-V7 in CRPC and highlights the promising benefits of natural compounds as complementary treatments to conventional drugs in reducing CRPC and overcoming therapeutic resistance. We delve into the mechanisms of action underlying the anti-CRPC effects of natural compounds, showcasing their potential to enhance therapeutic outcomes while mitigating the side effects associated with conventional therapies. The exploration of natural compounds offers promising avenues for developing novel treatment strategies that enhance therapeutic outcomes and reduce the adverse effects of conventional CRPC therapies. These compounds provide a safer, more effective approach to managing CRPC, representing a significant advancement in improving patient care.
Collapse
Affiliation(s)
- Muntajin Rahman
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Khadija Akter
- Department of Plasma Bio Display, Kwangwoon University, Seoul 01897, Republic of Korea;
| | - Kazi Rejvee Ahmed
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Md. Maharub Hossain Fahim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Nahida Aktary
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Sang-Won Shin
- Department of Humanities & Social Medicine, School of Korean Medicine, Pusan National University, 49 Busandaehak-ro, Mulgeum-eup, Yangsan-si 50612, Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
- Department of Plasma Bio Display, Kwangwoon University, Seoul 01897, Republic of Korea;
| |
Collapse
|
4
|
Muralidhar A, Gamat-Huber M, Vakkalanka S, McNeel DG. Sequence of androgen receptor-targeted vaccination with androgen deprivation therapy affects anti-prostate tumor efficacy. J Immunother Cancer 2024; 12:e008848. [PMID: 38772685 PMCID: PMC11110578 DOI: 10.1136/jitc-2024-008848] [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] [Accepted: 05/07/2024] [Indexed: 05/23/2024] Open
Abstract
RATIONALE Androgen deprivation therapy (ADT) is the primary treatment for recurrent and metastatic prostate cancer. In addition to direct antitumor effects, ADT has immunomodulatory effects such as promoting T-cell infiltration and enhancing antigen processing/presentation. Previous studies in our laboratory have demonstrated that ADT also leads to increased expression of the androgen receptor (AR) and increased recognition of prostate tumor cells by AR-specific CD8+T cells. We have also demonstrated that ADT combined with a DNA vaccine encoding the AR significantly slowed tumor growth and improved the survival of prostate tumor-bearing mice. The current study aimed to investigate the impact of the timing and sequencing of ADT with vaccination on the tumor immune microenvironment in murine prostate cancer models to further increase the antitumor efficacy of vaccines. METHODS Male FVB mice implanted with Myc-CaP tumor cells, or male C57BL/6 mice implanted with TRAMP-C1 prostate tumor cells, were treated with a DNA vaccine encoding AR (pTVG-AR) and ADT. The sequence of administration was evaluated for its effect on tumor growth, and tumor-infiltrating immune populations were characterized. RESULTS Vaccination prior to ADT (pTVG-AR → ADT) significantly enhanced antitumor responses and survival. This was associated with increased tumor infiltration by CD4+ and CD8+ T cells, including AR-specific CD8+T cells. Depletion of CD8+T cells prior to ADT significantly worsened overall survival. Following ADT treatment, however, Gr1+ myeloid-derived suppressor cells (MDSCs) increased, and this was associated with fewer infiltrating T cells and reduced tumor growth. Inhibiting Gr1+MDSCs recruitment, either by using a CXCR2 antagonist or by cycling androgen deprivation with testosterone replacement, improved antitumor responses and overall survival. CONCLUSION Vaccination prior to ADT significantly improved antitumor responses, mediated in part by increased infiltration of CD8+T cells following ADT. Targeting MDSC recruitment following ADT further enhanced antitumor responses. These findings suggest logical directions for future clinical trials to improve the efficacy of prostate cancer vaccines.
Collapse
Affiliation(s)
- Anusha Muralidhar
- Cancer Biology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Melissa Gamat-Huber
- UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Sita Vakkalanka
- UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Douglas G McNeel
- Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
5
|
Han D, Labaf M, Zhao Y, Owiredu J, Zhang S, Patel K, Venkataramani K, Steinfeld JS, Han W, Li M, Liu M, Wang Z, Besschetnova A, Patalano S, Mulhearn MJ, Macoska JA, Yuan X, Balk SP, Nelson PS, Plymate SR, Gao S, Siegfried KR, Liu R, Stangis MM, Foxa G, Czernik PJ, Williams BO, Zarringhalam K, Li X, Cai C. Androgen receptor splice variants drive castration-resistant prostate cancer metastasis by activating distinct transcriptional programs. J Clin Invest 2024; 134:e168649. [PMID: 38687617 PMCID: PMC11142739 DOI: 10.1172/jci168649] [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: 01/06/2023] [Accepted: 04/12/2024] [Indexed: 05/02/2024] Open
Abstract
One critical mechanism through which prostate cancer (PCa) adapts to treatments targeting androgen receptor (AR) signaling is the emergence of ligand-binding domain-truncated and constitutively active AR splice variants, particularly AR-V7. While AR-V7 has been intensively studied, its ability to activate distinct biological functions compared with the full-length AR (AR-FL), and its role in regulating the metastatic progression of castration-resistant PCa (CRPC), remain unclear. Our study found that, under castrated conditions, AR-V7 strongly induced osteoblastic bone lesions, a response not observed with AR-FL overexpression. Through combined ChIP-seq, ATAC-seq, and RNA-seq analyses, we demonstrated that AR-V7 uniquely accesses the androgen-responsive elements in compact chromatin regions, activating a distinct transcription program. This program was highly enriched for genes involved in epithelial-mesenchymal transition and metastasis. Notably, we discovered that SOX9, a critical metastasis driver gene, was a direct target and downstream effector of AR-V7. Its protein expression was dramatically upregulated in AR-V7-induced bone lesions. Moreover, we found that Ser81 phosphorylation enhanced AR-V7's pro-metastasis function by selectively altering its specific transcription program. Blocking this phosphorylation with CDK9 inhibitors impaired the AR-V7-mediated metastasis program. Overall, our study has provided molecular insights into the role of AR splice variants in driving the metastatic progression of CRPC.
Collapse
Affiliation(s)
- Dong Han
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | - Maryam Labaf
- Center for Personalized Cancer Therapy
- Department of Mathematics, University of Massachusetts Boston, Boston, Massachusetts, USA
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Yawei Zhao
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Jude Owiredu
- Department of Cell & Developmental Biology, Weill Cornell Medical College, New York, New York, USA
| | - Songqi Zhang
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | - Krishna Patel
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | | | | | - Wanting Han
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Muqing Li
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | - Mingyu Liu
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | - Zifeng Wang
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | | | - Susan Patalano
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | | | - Jill A. Macoska
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | - Xin Yuan
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Steven P. Balk
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Peter S. Nelson
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Stephen R. Plymate
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Veterans Affairs Puget Sound Health Care System, Geriatric Research and Education Clinical Center (VAPSHCS-GRECC), Seattle, Washington, USA
| | - Shuai Gao
- Department of Cell Biology and Anatomy and
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York, USA
| | | | - Ruihua Liu
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Mary M. Stangis
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Gabrielle Foxa
- Department of Cell Biology, and Core Technologies and Services, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Piotr J. Czernik
- Department of Orthopaedic Surgery, MicroCT and Skeletal Research Core Facility, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Bart O. Williams
- Department of Cell Biology, and Core Technologies and Services, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Kourosh Zarringhalam
- Center for Personalized Cancer Therapy
- Department of Mathematics, University of Massachusetts Boston, Boston, Massachusetts, USA
| | - Xiaohong Li
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Changmeng Cai
- Center for Personalized Cancer Therapy
- Department of Biology, and
| |
Collapse
|
6
|
Yao Y, Chen X, Wang X, Li H, Zhu Y, Li X, Xiao Z, Zi T, Qin X, Zhao Y, Yang T, Wang L, Wu G, Fang X, Wu D. Glycolysis related lncRNA SNHG3 / miR-139-5p / PKM2 axis promotes castration-resistant prostate cancer (CRPC) development and enzalutamide resistance. Int J Biol Macromol 2024; 260:129635. [PMID: 38266860 DOI: 10.1016/j.ijbiomac.2024.129635] [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: 05/23/2023] [Revised: 12/11/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024]
Abstract
Although androgen deprivation therapy (ADT) by the anti-androgen drug enzalutamide (Enz) may improve the survival level of patients with castration-resistant prostate cancer (CRPC), most patients may eventually fail due to the acquired resistance. The reprogramming of glucose metabolism is one type of the paramount hallmarks of cancers. PKM2 (Pyruvate kinase isozyme typeM2) is a speed-limiting enzyme in the glycolytic mechanism, and has high expression in a variety of cancers. Emerging evidence has unveiled that microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) have impact on tumor development and therapeutic efficacy by regulating PKM2 expression. Herein, we found that lncRNA SNHG3, a highly expressed lncRNA in CRPC via bioinformatics analysis, promoted the invasive ability and the Enz resistance of the PCa cells. KEGG pathway enrichment analysis indicated that glucose metabolic process was tightly correlated with lncRNA SNHG3 level, suggesting lncRNA SNHG3 may affect glucose metabolism. Indeed, glucose uptake and lactate content determinations confirmed that lncRNA SNHG3 promoted the process of glycolysis. Mechanistic dissection demonstrated that lncRNA SNHG3 facilitated the advance of CRPC by adjusting the expression of PKM2. Further explorations unraveled the role of lncRNA SNHG3 as a 'sponge' of miR-139-5p and released its binding with PKM2 mRNA, leading to PKM2 up-regulation. Together, Our studies suggest that lncRNA SNHG3 / miR-139-5p / PKM2 pathway promotes the development of CRPC via regulating glycolysis process and provides valuable insight into a novel therapeutic approach for the disordered disease.
Collapse
Affiliation(s)
- Yicong Yao
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Xi Chen
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Xin'an Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Haopeng Li
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Yaru Zhu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Xilei Li
- School of Medicine, Tongji University, Shanghai 200092, China
| | - Zhihui Xiao
- School of Medicine, Tongji University, Shanghai 200092, China
| | - Tong Zi
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Xin Qin
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Yan Zhao
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Tao Yang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Licheng Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| | - Gang Wu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| | - Xia Fang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China.
| | - Denglong Wu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| |
Collapse
|
7
|
Jathal MK, Siddiqui S, Vasilatis DM, Durbin Johnson BP, Drake C, Mooso BA, D'Abronzo LS, Batra N, Mudryj M, Ghosh PM. Androgen receptor transcriptional activity is required for heregulin-1β-mediated nuclear localization of the HER3/ErbB3 receptor tyrosine kinase. J Biol Chem 2023; 299:104973. [PMID: 37380074 PMCID: PMC10407237 DOI: 10.1016/j.jbc.2023.104973] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 06/05/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023] Open
Abstract
Prostate cancer is initially regulated by the androgen receptor (AR), a ligand-activated, transcription factor, and is in a hormone-dependent state (hormone-sensitive prostate cancer (HSPC)), but eventually becomes androgen-refractory (castration-resistant prostate cancer (CRPC)) because of mechanisms that bypass the AR, including by activation of ErbB3, a member of the epidermal growth factor receptor family. ErbB3 is synthesized in the cytoplasm and transported to the plasma membrane for ligand binding and dimerization, where it regulates downstream signaling, but nuclear forms are reported. Here, we demonstrate in prostatectomy samples that ErbB3 nuclear localization is observed in malignant, but not benign prostate, and that cytoplasmic (but not nuclear) ErbB3 correlated positively with AR expression but negatively with AR transcriptional activity. In support of the latter, androgen depletion upregulated cytoplasmic, but not nuclear ErbB3, while in vivo studies showed that castration suppressed ErbB3 nuclear localization in HSPC, but not CRPC tumors. In vitro treatment with the ErbB3 ligand heregulin-1β (HRG) induced ErbB3 nuclear localization, which was androgen-regulated in HSPC but not in CRPC. In turn, HRG upregulated AR transcriptional activity in CRPC but not in HSPC cells. Positive correlation between ErbB3 and AR expression was demonstrated in AR-null PC-3 cells where stable transfection of AR restored HRG-induced ErbB3 nuclear transport, while AR knockdown in LNCaP reduced cytoplasmic ErbB3. Mutations of ErbB3's kinase domain did not affect its localization but was responsible for cell viability in CRPC cells. Taken together, we conclude that AR expression regulated ErbB3 expression, its transcriptional activity suppressed ErbB3 nuclear translocation, and HRG binding to ErbB3 promoted it.
Collapse
Affiliation(s)
- Maitreyee K Jathal
- Research Service, VA Northern California Health Care System, Mather, California, USA; Department of Medical Microbiology and Immunology, University of California Davis, Davis, California, USA
| | - Salma Siddiqui
- Research Service, VA Northern California Health Care System, Mather, California, USA
| | - Demitria M Vasilatis
- Research Service, VA Northern California Health Care System, Mather, California, USA; Department of Urologic Surgery, University of California Davis, Sacramento, California, USA
| | - Blythe P Durbin Johnson
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis, Davis, California, USA
| | - Christiana Drake
- Department of Statistics, University of California Davis, Davis, California, USA
| | - Benjamin A Mooso
- Research Service, VA Northern California Health Care System, Mather, California, USA
| | - Leandro S D'Abronzo
- Department of Urologic Surgery, University of California Davis, Sacramento, California, USA
| | - Neelu Batra
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, USA
| | - Maria Mudryj
- Research Service, VA Northern California Health Care System, Mather, California, USA; Department of Medical Microbiology and Immunology, University of California Davis, Davis, California, USA
| | - Paramita M Ghosh
- Research Service, VA Northern California Health Care System, Mather, California, USA; Department of Urologic Surgery, University of California Davis, Sacramento, California, USA; Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, USA.
| |
Collapse
|
8
|
Alebady ZAH, Azizyan M, Nakjang S, Lishman-Walker E, Al-Kharaif D, Walker S, Choo HX, Garnham R, Scott E, Johnson KL, Robson CN, Coffey K. CDC20 Is Regulated by the Histone Methyltransferase, KMT5A, in Castration-Resistant Prostate Cancer. Cancers (Basel) 2023; 15:3597. [PMID: 37509260 PMCID: PMC10377584 DOI: 10.3390/cancers15143597] [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: 05/26/2023] [Revised: 07/06/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
The methyltransferase KMT5A has been proposed as an oncogene in prostate cancer and therefore represents a putative therapeutic target. To confirm this hypothesis, we have performed a microarray study on a prostate cancer cell line model of androgen independence following KMT5A knockdown in the presence of the transcriptionally active androgen receptor (AR) to understand which genes and cellular processes are regulated by KMT5A in the presence of an active AR. We observed that 301 genes were down-regulated whilst 408 were up-regulated when KMT5A expression was reduced. KEGG pathway and gene ontology analysis revealed that apoptosis and DNA damage signalling were up-regulated in response to KMT5A knockdown whilst protein folding and RNA splicing were down-regulated. Under these conditions, the top non-AR regulated gene was found to be CDC20, a key regulator of the spindle assembly checkpoint with an oncogenic role in several cancer types. Further investigation revealed that KMT5A regulates CDC20 in a methyltransferase-dependent manner to modulate histone H4K20 methylation within its promoter region and indirectly via the p53 signalling pathway. A positive correlation between KMT5A and CDC20 expression was also observed in clinical prostate cancer samples, further supporting this association. Therefore, we conclude that KMT5A is a valid therapeutic target for the treatment of prostate cancer and CDC20 could potentially be utilised as a biomarker for effective therapeutic targeting.
Collapse
Affiliation(s)
- Zainab A H Alebady
- Biosciences Institute, Newcastle Cancer Centre, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Department of Laboratory and Clinical Science, College of Pharmacy, University of AL-Qadisiyah, Al-Diwaniya 58002, Iraq
| | - Mahsa Azizyan
- Biosciences Institute, Newcastle Cancer Centre, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Sirintra Nakjang
- Bioinformatics Support Unit, Newcastle University, Newcastle NE2 4HH, UK
| | - Emma Lishman-Walker
- Biosciences Institute, Newcastle Cancer Centre, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Dhuha Al-Kharaif
- Medical Laboratory Technology Department, College of Health Sciences, Public Authority of Applied Education and Training, Safat 13092, Kuwait
| | - Scott Walker
- School of Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Hui Xian Choo
- Biosciences Institute, Newcastle Cancer Centre, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Rebecca Garnham
- Biosciences Institute, Newcastle Cancer Centre, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Emma Scott
- Biosciences Institute, Newcastle Cancer Centre, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Katya L Johnson
- Biosciences Institute, Newcastle Cancer Centre, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Craig N Robson
- Translational and Clinical Research Institute, Newcastle Cancer Centre, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Kelly Coffey
- Biosciences Institute, Newcastle Cancer Centre, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
9
|
Raith F, O’Donovan DH, Lemos C, Politz O, Haendler B. Addressing the Reciprocal Crosstalk between the AR and the PI3K/AKT/mTOR Signaling Pathways for Prostate Cancer Treatment. Int J Mol Sci 2023; 24:ijms24032289. [PMID: 36768610 PMCID: PMC9917236 DOI: 10.3390/ijms24032289] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023] Open
Abstract
The reduction in androgen synthesis and the blockade of the androgen receptor (AR) function by chemical castration and AR signaling inhibitors represent the main treatment lines for the initial stages of prostate cancer. Unfortunately, resistance mechanisms ultimately develop due to alterations in the AR pathway, such as gene amplification or mutations, and also the emergence of alternative pathways that render the tumor less or, more rarely, completely independent of androgen activation. An essential oncogenic axis activated in prostate cancer is the phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway, as evidenced by the frequent alterations of the negative regulator phosphatase and tensin homolog (PTEN) and by the activating mutations in PI3K subunits. Additionally, crosstalk and reciprocal feedback loops between androgen signaling and the PI3K/AKT/mTOR signaling cascade that activate pro-survival signals and play an essential role in disease recurrence and progression have been evidenced. Inhibitors addressing different players of the PI3K/AKT/mTOR pathway have been evaluated in the clinic. Only a limited benefit has been reported in prostate cancer up to now due to the associated side effects, so novel combination approaches and biomarkers predictive of patient response are urgently needed. Here, we reviewed recent data on the crosstalk between AR signaling and the PI3K/AKT/mTOR pathway, the selective inhibitors identified, and the most advanced clinical studies, with a focus on combination treatments. A deeper understanding of the complex molecular mechanisms involved in disease progression and treatment resistance is essential to further guide therapeutic approaches with improved outcomes.
Collapse
Affiliation(s)
- Fabio Raith
- Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany
| | - Daniel H. O’Donovan
- Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany
| | - Clara Lemos
- Bayer Research and Innovation Center, Bayer US LLC, 238 Main Street, Cambridge, MA 02142, USA
| | - Oliver Politz
- Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany
| | - Bernard Haendler
- Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany
- Correspondence: ; Tel.: +49-30-2215-41198
| |
Collapse
|