1
|
Liang L, Shen Z, Zhang K, Zhang C, Dong L, Shi R, Hua L, Zhao R, Feng N. DLGAP5 upregulates E2F1 to promote prostate adenocarcinoma neuroendocrine differentiation. Life Sci 2025; 365:123442. [PMID: 39923838 DOI: 10.1016/j.lfs.2025.123442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/24/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025]
Abstract
AIMS DLGAP5 plays a significant role in promoting cancer progression across various cancers. However, the specific role of DLGAP5 in neuroendocrine differentiation (NED) of prostate cancer (PCa) remains elusive. Our objective is to explore the mechanism by which DLGAP5 mediates NED in PCa. MATERIALS AND METHODS Utilizing diverse public databases, we conducted bioinformatics analysis to examine DLGAP5 expression in PCa. We confirmed aberrant DLGAP5 expression in various PCa cell lines through Western blotting. Functional assays both in vivo and in vitro have validated the oncogenic role of DLGAP5 in PCa. Furthermore, we sought to identify downstream key genes to elucidate the mechanisms underlying DLGAP5-mediated NED in PCa. We also identified a small molecule drug, AAPK-25, which specifically targets DLGAP5. KEY FINDINGS DLGAP5 was highly expressed in NEPC. The suppression of AR signaling promoted DLGAP5 transcription. DLGAP5 endowed PCa cells with a robust ability to proliferate and migrate. E2F1 was an important downstream target of DLGAP5. DLGAP5 mediated the NED of PCa through E2F1. AAPK-25, as an inhibitor of DLGAP5, inhibited PRAD proliferation by repressing the DLGAP5/E2F1 axis both in vitro and in vivo. SIGNIFICANCE We identified the AR/DLGAP5/E2F1 signaling pathway as a pivotal mechanism that facilitates the transition of PCa towards a neuroendocrine phenotype. This pathway may represent a promising therapeutic target for NEPC patients.
Collapse
Affiliation(s)
- Linghui Liang
- Department of Urology, Affiliated Wuxi No.2 Hospital, Nanjing Medical University & Jiangnan University Medical Center, Wuxi, Jiangsu, China
| | - Zhiyi Shen
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kaiyu Zhang
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenglong Zhang
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lai Dong
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rongjie Shi
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lixin Hua
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ruizhe Zhao
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Ninghan Feng
- Department of Urology, Affiliated Wuxi No.2 Hospital, Nanjing Medical University & Jiangnan University Medical Center, Wuxi, Jiangsu, China.
| |
Collapse
|
2
|
Wei J, Wang J, Guan W, Li J, Pu T, Corey E, Lin TP, Gao AC, Wu BJ. PlexinD1 is a driver and a therapeutic target in advanced prostate cancer. EMBO Mol Med 2025; 17:336-364. [PMID: 39748059 PMCID: PMC11822115 DOI: 10.1038/s44321-024-00186-z] [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: 04/09/2024] [Revised: 12/06/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
Aggressive prostate cancer (PCa) variants associated with androgen receptor signaling inhibitor (ARSI) resistance and metastasis remain poorly understood. Here, we identify the axon guidance semaphorin receptor PlexinD1 as a crucial driver of cancer aggressiveness in metastatic castration-resistant prostate cancer (CRPC). High PlexinD1 expression in human PCa is correlated with adverse clinical outcomes. PlexinD1 critically maintains CRPC aggressive behaviors in vitro and in vivo, and confers stemness and cellular plasticity to promote multilineage differentiation including a neuroendocrine-like phenotype for ARSI resistance. Mechanistically, PlexinD1 is upregulated upon relief of AR-mediated transcriptional repression of PlexinD1 under ARSI treatment, and subsdquently transactivates ErbB3 and cMet via direct interaction, which triggers the ERK/AKT pathways to induce noncanonical Gli1-dictated Hedgehog signaling, facilitating the growth and plasticity of PCa cells. Blockade of PlexinD1 by the protein inhibitor D1SP restricted CRPC growth in multiple preclinical models. Collectively, these findings characterize PlexinD1's contribution to PCa progression and offer a potential PlexinD1-targeted therapy for advanced PCa.
Collapse
Affiliation(s)
- Jing Wei
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Jing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| | - Wen Guan
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Jingjing Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
- Engineering Research Center of Cell & Therapeutic Antibody, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Tianjie Pu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, 98195, USA
| | - Tzu-Ping Lin
- Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan, 11217, Republic of China
- Department of Urology, School of Medicine and Shu-Tien Urological Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, 11221, Republic of China
| | - Allen C Gao
- Department of Urologic Surgery, University of California, Davis, Sacramento, CA, 95817, USA
| | - Boyang Jason Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA.
| |
Collapse
|
3
|
Shang Z, Li X. Human cytomegalovirus: pathogenesis, prevention, and treatment. MOLECULAR BIOMEDICINE 2024; 5:61. [PMID: 39585514 PMCID: PMC11589059 DOI: 10.1186/s43556-024-00226-7] [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: 07/16/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024] Open
Abstract
Human cytomegalovirus (HCMV) infection remains a significant global health challenge, particularly for immunocompromised individuals and newborns. This comprehensive review synthesizes current knowledge on HCMV pathogenesis, prevention, and treatment strategies. We examine the molecular mechanisms of HCMV entry, focusing on the structure and function of key envelope glycoproteins (gB, gH/gL/gO, gH/gL/pUL128-131) and their interactions with cellular receptors such as PDGFRα, NRP2, and THBD. The review explores HCMV's sophisticated immune evasion strategies, including interference with pattern recognition receptor signaling, modulation of antigen presentation, and regulation of NK and T cell responses. We highlight recent advancements in developing neutralizing antibodies, various vaccine strategies (live-attenuated, subunit, vector-based, DNA, and mRNA), antiviral compounds (both virus-targeted and host-targeted), and emerging cellular therapies such as TCR-T cell approaches. By integrating insights from structural biology, immunology, and clinical research, we identify critical knowledge gaps and propose future research directions. This analysis aims to stimulate cross-disciplinary collaborations and accelerate the development of more effective prevention and treatment strategies for HCMV infections, addressing a significant unmet medical need.
Collapse
Affiliation(s)
- Zifang Shang
- Research Experiment Center, Meizhou Academy of Medical Sciences, Meizhou People's Hospital, Meizhou, 514031, Guangdong, China.
- Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, 514031, Guangdong, China.
| | - Xin Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101, China
| |
Collapse
|
4
|
Zhao F, Zhang T, Wei J, Chen L, Liu Z, Jin Y, Liu M, Zhou H, Hu Y, Sheng X. Integrated single-cell transcriptomic analyses identify a novel lineage plasticity-related cancer cell type involved in prostate cancer progression. EBioMedicine 2024; 109:105398. [PMID: 39418984 PMCID: PMC11530610 DOI: 10.1016/j.ebiom.2024.105398] [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: 04/24/2024] [Revised: 09/07/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Cancer cell plasticity is the ability of neoplastic cells to alter their identity and acquire new biological properties under microenvironmental pressures. In prostate cancer (PCa), lineage plasticity often results in therapy resistance and trans-differentiation to neuroendocrine (NE) lineage. However, identifying the cancer cells harboring lineage plasticity-related status remains challenging. METHODS Based on 13 multi-center human PCa bulk transcriptomic cohorts (samples = 3314) and 9 bulk transcriptomic datasets derived from PCa experimental models, we established an integrated lineage plasticity-related gene signature, termed LPSig. Leveraging this gene signature, AUCell enrichment analysis was applied to identify the cell population with high lineage plasticity from a comprehensive single-cell RNA-sequencing (scRNA-seq) meta-atlas assembled by us, which consisted of 10 public human PCa scRNA-seq datasets (samples = 93, cells = 222,529). Moreover, additional scRNA-seq dataset of human PCa, multiplex immunohistochemistry staining for human PCa tissues, in vitro and in vivo functional experiments, as well as qPCR and Western blot analyses were employed to validate our findings. FINDINGS We found that LPSig could finely capture the dynamics of tumor lineage plasticity throughout the progression of PCa, accurately estimating the status of lineage plasticity. Based on LPSig, we identified a previously undefined minority population of lineage plasticity-related PCa cells (LPCs) from the human PCa scRNA-seq meta-atlas assembled by this study. Furthermore, in-depth dissection revealed pivotal roles of LPCs in trans-differentiation, tumor recurrence, and poor patient survival during PCa progression. Furthermore, we identified HMMR as a representative cell surface marker for LPCs, which was validated using additional scRNA-seq datasets and multiplexed immunohistochemistry. Moreover, HMMR was transcriptionally inhibited by androgen receptor (AR), and was required for the aggressive adenocarcinoma features and NE phenotype. INTERPRETATION Our study uncovers a novel population of lineage plasticity-related cells with low AR activity, stemness-like traits, and elevated HMMR expression, that may facilitate poor prognosis in PCa. FUNDING This work was supported by National Key R&D Program of China (2022YFA0807000), National Natural Science Foundation of China (82160584), Advanced Prostate Cancer Diagnosis and Treatment Technology Innovation Team of Kunming Medical University (CXTD202216), and Reserve Talents of Young and Middle-aged Academic Leaders in Yunnan Province (202105AC160013).
Collapse
Affiliation(s)
- Faming Zhao
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA; School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tingting Zhang
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jinlan Wei
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Liang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yang Jin
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo 0316, Norway
| | - Mingsheng Liu
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing 655000, China
| | - Hongqing Zhou
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing 655000, China
| | - Yanxia Hu
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Xia Sheng
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
5
|
Niu X, Liu W, Zhang Y, Liu J, Zhang J, Li B, Qiu Y, Zhao P, Wang Z, Wang Z. Cancer plasticity in therapy resistance: Mechanisms and novel strategies. Drug Resist Updat 2024; 76:101114. [PMID: 38924995 DOI: 10.1016/j.drup.2024.101114] [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: 04/17/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
Therapy resistance poses a significant obstacle to effective cancer treatment. Recent insights into cell plasticity as a new paradigm for understanding resistance to treatment: as cancer progresses, cancer cells experience phenotypic and molecular alterations, corporately known as cell plasticity. These alterations are caused by microenvironment factors, stochastic genetic and epigenetic changes, and/or selective pressure engendered by treatment, resulting in tumor heterogeneity and therapy resistance. Increasing evidence suggests that cancer cells display remarkable intrinsic plasticity and reversibly adapt to dynamic microenvironment conditions. Dynamic interactions between cell states and with the surrounding microenvironment form a flexible tumor ecosystem, which is able to quickly adapt to external pressure, especially treatment. Here, this review delineates the formation of cancer cell plasticity (CCP) as well as its manipulation of cancer escape from treatment. Furthermore, the intrinsic and extrinsic mechanisms driving CCP that promote the development of therapy resistance is summarized. Novel treatment strategies, e.g., inhibiting or reversing CCP is also proposed. Moreover, the review discusses the multiple lines of ongoing clinical trials globally aimed at ameliorating therapy resistance. Such advances provide directions for the development of new treatment modalities and combination therapies against CCP in the context of therapy resistance.
Collapse
Affiliation(s)
- Xing Niu
- China Medical University, Shenyang, Liaoning 110122, China; Experimental Center of BIOQGene, YuanDong International Academy Of Life Sciences, 999077, Hong Kong, China
| | - Wenjing Liu
- Medical Oncology Department of Thoracic Cancer (2), Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Yinling Zhang
- Department of Oncology Radiotherapy 1, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong 266042, China
| | - Jing Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Jianjun Zhang
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Bo Li
- Department of Orthopedics, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
| | - Yue Qiu
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Peng Zhao
- Department of Medical Imaging, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Zhongmiao Wang
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| | - Zhe Wang
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| |
Collapse
|
6
|
Jamroze A, Liu X, Tang DG. Treatment-induced stemness and lineage plasticity in driving prostate cancer therapy resistance. CANCER HETEROGENEITY AND PLASTICITY 2024; 1:0005. [PMID: 39363904 PMCID: PMC11449474 DOI: 10.47248/chp2401010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
Most human cancers are heterogeneous consisting of cancer cells at different epigenetic and transcriptional states and with distinct phenotypes, functions, and drug sensitivities. This inherent cancer cell heterogeneity contributes to tumor resistance to clinical treatment, especially the molecularly targeted therapies such as tyrosine kinase inhibitors (TKIs) and androgen receptor signaling inhibitors (ARSIs). Therapeutic interventions, in turn, induce lineage plasticity (also called lineage infidelity) in cancer cells that also drives therapy resistance. In this Perspective, we focus our discussions on cancer cell lineage plasticity manifested as treatment-induced switching of epithelial cancer cells to basal/stem-like, mesenchymal, and neural lineages. We employ prostate cancer (PCa) as the prime example to highlight ARSI-induced lineage plasticity during and towards development of castration-resistant PCa (CRPC). We further discuss how the tumor microenvironment (TME) influences therapy-induced lineage plasticity. Finally, we offer an updated summary on the regulators and mechanisms driving cancer cell lineage infidelity, which should be therapeutically targeted to extend the therapeutic window and improve patients' survival.
Collapse
Affiliation(s)
- Anmbreen Jamroze
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Xiaozhuo Liu
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Dean G. Tang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Experimental Therapeutics (ET) Graduate Program, University at Buffalo & Roswell Park Comprehensive Cancer Center, NY 14263, USA
| |
Collapse
|
7
|
Chen B, Xu P, Yang JC, Nip C, Wang L, Shen Y, Ning S, Shang Y, Corey E, Gao AC, Gestwicki JE, Wei Q, Liu L, Liu C. Plexin D1 emerges as a novel target in the development of neural lineage plasticity in treatment-resistant prostate cancer. Oncogene 2024; 43:2325-2337. [PMID: 38877132 PMCID: PMC11286220 DOI: 10.1038/s41388-024-03081-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/16/2024]
Abstract
Treatment-induced neuroendocrine prostate cancer (t-NEPC) often arises from adenocarcinoma via lineage plasticity in response to androgen receptor signaling inhibitors, such as enzalutamide. However, the specific regulators and targets involved in the transition to NEPC are not well understood. Plexin D1 (PLXND1) is a cellular receptor of the semaphorin (SEMA) family that plays important roles in modulating the cytoskeleton and cell adhesion. Here, we found that PLXND1 was highly expressed and positively correlated with neuroendocrine markers in patients with NEPC. High PLXND1 expression was associated with poorer prognosis in prostate cancer patients. Additionally, PLXND1 was upregulated and negatively regulated by androgen receptor signaling in enzalutamide-resistant cells. Knockdown or knockout of PLXND1 inhibited neural lineage pathways, thereby suppressing NEPC cell proliferation, patient derived xenograft (PDX) tumor organoid viability, and xenograft tumor growth. Mechanistically, the heat shock protein 70 (HSP70) regulated PLXND1 protein stability through degradation, and inhibition of HSP70 decreased PLXND1 expression and NEPC organoid growth. In summary, our findings indicate that PLXND1 could serve as a promising therapeutic target and molecular marker for NEPC.
Collapse
Affiliation(s)
- Bo Chen
- Department of Urologic Surgery, University of California, Davis, CA, USA
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Pengfei Xu
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Joy C Yang
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Christopher Nip
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Leyi Wang
- Department of Urologic Surgery, University of California, Davis, CA, USA
- Graduate Group in Integrative Pathobiology, University of California, Davis, CA, USA
| | - Yuqiu Shen
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Shu Ning
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Yufeng Shang
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Eva Corey
- Department of Urology, University of Washington, Washington, WA, USA
| | - Allen C Gao
- Department of Urologic Surgery, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Qiang Wei
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Liangren Liu
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Chengfei Liu
- Department of Urologic Surgery, University of California, Davis, CA, USA.
- Graduate Group in Integrative Pathobiology, University of California, Davis, CA, USA.
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA.
| |
Collapse
|
8
|
Liu C, Chen B, Xu P, Yang J, Nip C, Wang L, Shen Y, Ning S, Shang Y, Corey E, Gao AC, Gestwicki J, Wei Q, Liu L. Plexin D1 emerges as a novel target in the development of neural lineage plasticity in treatment-resistant prostate cancer. RESEARCH SQUARE 2024:rs.3.rs-4095949. [PMID: 38585965 PMCID: PMC10996809 DOI: 10.21203/rs.3.rs-4095949/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Treatment-induced neuroendocrine prostate cancer (t-NEPC) often arises from adenocarcinoma via lineage plasticity in response to androgen receptor signaling inhibitors, such as enzalutamide. However, the specific regulators and targets involved in the transition to NEPC are not well understood. Plexin D1 (PLXND1) is a cellular receptor of the semaphorin (SEMA) family that plays important roles in modulating the cytoskeleton and cell adhesion. Here, we found that PLXND1 is highly expressed and positively correlated with neuroendocrine markers in patients with NEPC. High PLXND1 expression is associated with poorer prognosis in prostate cancer patients. Additionally, PLXND1 was upregulated and negatively regulated by androgen receptor signaling in enzalutamide-resistant cells. Knockdown or knockout of PLXND1 inhibit neural lineage pathways, suppressing NEPC cell proliferation, PDX tumor organoid viability, and xenograft tumor growth. Mechanistically, the chaperone protein HSP70 regulates PLXND1 protein stability through degradation, and inhibition of HSP70 decreases PLXND1 expression and NEPC organoid growth. In summary, our findings suggest that PLXND1 could be a new therapeutic target and molecular indicator for NEPC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Qiang Wei
- West China Hospital of Sichuan University
| | | |
Collapse
|
9
|
Nie J, Zhang P, Liang C, Yu Y, Wang X. ASCL1-mediated ferroptosis resistance enhances the progress of castration-resistant prostate cancer to neurosecretory prostate cancer. Free Radic Biol Med 2023; 205:318-331. [PMID: 37355053 DOI: 10.1016/j.freeradbiomed.2023.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 06/26/2023]
Abstract
Neuroendocrine prostate cancer (NEPC) is a multi-resistant variant of prostate cancer (PCa) that frequently emerges in castration-resistant prostate cancer (CRPC). NEPC is usually associated with tumor aggression, hormone therapy resistance, and poor clinical outcome. However, the mechanisms underlying the trans-differentiation from CRPC to NEPC have not been elucidated. Achaete-scute complex-like 1 (ASCL1) plays a role in neuronal commitment and differentiation and olfactory and autonomic neuron generation. This study revealed that ASCL1 was regulated by the SRY-box transcription factor 2 (SOX2) and highly expressed in NEPC cells, which was closely related to poor prognosis. Moreover, ASCL1 overexpression significantly enhanced CRPC progression to NEPC by resisting ferroptosis. Mechanically, ferroptosis resistance was mediated by CAMP-responsive element binding protein 1 (CREB1) phosphorylation, promoted by substantially upregulated ASCL1 in NEPC cells. In addition, upregulated SOX2 induced PCa cell differentiation into neuroendocrine tumors by mediating their lineage changes. In conclusion, inhibiting the ferroptosis resistance mediated by ASCL1 could provide a new NEPC therapeutic target and increase patient survival.
Collapse
Affiliation(s)
- Jiawei Nie
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Peng Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China.
| | - Chaoqi Liang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Ying Yu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China; Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China; Cancer Precision Diagnosis and Treatment and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China.
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China; Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China; Cancer Precision Diagnosis and Treatment and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China; Research Center of Wuhan for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, 430071, PR China.
| |
Collapse
|
10
|
Verlinden L, Doms S, Janssens I, Meyer MB, Pike JW, Carmeliet G, Verstuyf A. Neuropilin 2 in osteoblasts regulates trabecular bone mass in male mice. Front Endocrinol (Lausanne) 2023; 14:1223021. [PMID: 37600714 PMCID: PMC10436209 DOI: 10.3389/fendo.2023.1223021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/11/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Neuropilin 2 (NRP2) mediates the effects of class 3 semaphorins and vascular endothelial growth factor and is implicated in axonal guidance and angiogenesis. Moreover, NRP2 expression is suggested to be involved in the regulation of bone homeostasis. Indeed, osteoblasts and osteoclasts express NRP2 and male and female global Nrp2 knockout mice have a reduced bone mass accompanied by reduced osteoblast and increased osteoclast counts. Methods We first examined the in vitro effect of the calciotropic hormone 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] on Nrp2 transcription in osteoblasts. We next generated mice with a conditional deletion of Nrp2 in the osteoblast cell lineage under control of the paired related homeobox 1 promoter and mice with a conditional Nrp2 knockdown in osteoclasts under control of the Lysozyme promoter. Mice were examined under basal conditions or after treatment with either the bone anabolic vitamin D3 analog WY 1048 or with 1,25(OH)2D3. Results and discussion We show that Nrp2 expression is induced by 1,25(OH)2D3 in osteoblasts and is associated with enrichment of the vitamin D receptor in an intronic region of the Nrp2 gene. In male mice, conditional deletion of Nrp2 in osteoblast precursors and mature osteoblasts recapitulated the bone phenotype of global Nrp2 knockout mice, with a reduced cortical cross-sectional tissue area and lower trabecular bone content. However, female mice with reduced osteoblastic Nrp2 expression display a reduced cross-sectional tissue area but have a normal trabecular bone mass. Treatment with the vitamin D3 analog WY 1048 (0.4 μg/kg/d, 14 days, ip) resulted in a similar increase in bone mass in both genotypes and genders. Deleting Nrp2 from the osteoclast lineage did not result in a bone phenotype, even though in vitro osteoclastogenesis of hematopoietic cells derived from mutant mice was significantly increased. Moreover, treatment with a high dose of 1,25(OH)2D3 (0.5 μg/kg/d, 6 days, ip), to induce osteoclast-mediated bone resorption, resulted in a similar reduction in trabecular and cortical bone mass. In conclusion, osteoblastic Nrp2 expression is suggested to regulate bone homeostasis in a sex-specific manner.
Collapse
Affiliation(s)
- Lieve Verlinden
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Stefanie Doms
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Iris Janssens
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Mark B. Meyer
- Department of Nutritional Sciences, University of Wisconsin-Madision, Madison, WI, United States
| | - J. Wesley Pike
- Department of Biochemistry, University of Wisconsin-Madision, Madison, WI, United States
| | - Geert Carmeliet
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Annemieke Verstuyf
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| |
Collapse
|
11
|
Wang M, Wisniewski CA, Xiong C, Chhoy P, Goel HL, Kumar A, Zhu LJ, Li R, St Louis PA, Ferreira LM, Pakula H, Xu Z, Loda M, Jiang Z, Brehm MA, Mercurio AM. Therapeutic blocking of VEGF binding to neuropilin-2 diminishes PD-L1 expression to activate antitumor immunity in prostate cancer. Sci Transl Med 2023; 15:eade5855. [PMID: 37134151 DOI: 10.1126/scitranslmed.ade5855] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Prostate cancers are largely unresponsive to immune checkpoint inhibitors (ICIs), and there is strong evidence that programmed death-ligand 1 (PD-L1) expression itself must be inhibited to activate antitumor immunity. Here, we report that neuropilin-2 (NRP2), which functions as a vascular endothelial growth factor (VEGF) receptor on tumor cells, is an attractive target to activate antitumor immunity in prostate cancer because VEGF-NRP2 signaling sustains PD-L1 expression. NRP2 depletion increased T cell activation in vitro. In a syngeneic model of prostate cancer that is resistant to ICI, inhibition of the binding of VEGF to NRP2 using a mouse-specific anti-NRP2 monoclonal antibody (mAb) resulted in necrosis and tumor regression compared with both an anti-PD-L1 mAb and control immunoglobulin G. This therapy also decreased tumor PD-L1 expression and increased immune cell infiltration. We observed that the NRP2, VEGFA, and VEGFC genes are amplified in metastatic castration-resistant and neuroendocrine prostate cancer. We also found that individuals with NRP2High PD-L1High metastatic tumors had lower androgen receptor expression and higher neuroendocrine prostate cancer scores than other individuals with prostate cancer. In organoids derived from patients with neuroendocrine prostate cancer, therapeutic inhibition of VEGF binding to NRP2 using a high-affinity humanized mAb suitable for clinical use also diminished PD-L1 expression and caused a substantial increase in immune-mediated tumor cell killing, consistent with the animal studies. These findings provide justification for the initiation of clinical trials using this function-blocking NRP2 mAb in prostate cancer, especially for patients with aggressive disease.
Collapse
Affiliation(s)
- Mengdie Wang
- Departments of Molecular, Cell and Cancer Biology, University of Massachusetts Chan School of Medicine, Worcester, MA 01605, USA
| | - Christi A Wisniewski
- Departments of Molecular, Cell and Cancer Biology, University of Massachusetts Chan School of Medicine, Worcester, MA 01605, USA
| | - Choua Xiong
- Departments of Molecular, Cell and Cancer Biology, University of Massachusetts Chan School of Medicine, Worcester, MA 01605, USA
| | - Peter Chhoy
- Departments of Molecular, Cell and Cancer Biology, University of Massachusetts Chan School of Medicine, Worcester, MA 01605, USA
| | - Hira Lal Goel
- Departments of Molecular, Cell and Cancer Biology, University of Massachusetts Chan School of Medicine, Worcester, MA 01605, USA
| | - Ayush Kumar
- Departments of Molecular, Cell and Cancer Biology, University of Massachusetts Chan School of Medicine, Worcester, MA 01605, USA
| | - Lihua Julie Zhu
- Program in Molecular Medicine, University of Massachusetts Chan School of Medicine, Worcester, MA 01605, USA
| | - Rui Li
- Program in Molecular Medicine, University of Massachusetts Chan School of Medicine, Worcester, MA 01605, USA
| | - Pamela A St Louis
- Department of Neurology, University of Massachusetts Chan School of Medicine, Worcester, MA 01605, USA
| | - Lindsay M Ferreira
- Program in Molecular Medicine, University of Massachusetts Chan School of Medicine, Worcester, MA 01605, USA
| | - Hubert Pakula
- Department of Pathology, Cornell Weill School of Medicine, New York, NY 10065, USA
| | - Zhiwen Xu
- aTyr Pharma Inc., San Diego CA, 92121, USA
| | - Massimo Loda
- Department of Pathology, Cornell Weill School of Medicine, New York, NY 10065, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute (DFCI) and Harvard Medical School, Boston, MA 02215, USA
| | - Zhong Jiang
- Department of Pathology, University of Massachusetts Chan School of Medicine, Worcester, MA 01605, USA
| | - Michael A Brehm
- Program in Molecular Medicine, University of Massachusetts Chan School of Medicine, Worcester, MA 01605, USA
| | - Arthur M Mercurio
- Departments of Molecular, Cell and Cancer Biology, University of Massachusetts Chan School of Medicine, Worcester, MA 01605, USA
| |
Collapse
|
12
|
Verma P, Shukla N, Kumari S, Ansari M, Gautam NK, Patel GK. Cancer stem cell in prostate cancer progression, metastasis and therapy resistance. Biochim Biophys Acta Rev Cancer 2023; 1878:188887. [PMID: 36997008 DOI: 10.1016/j.bbcan.2023.188887] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/18/2023] [Accepted: 03/15/2023] [Indexed: 03/31/2023]
Abstract
Prostate cancer (PCa) is the most diagnosed malignancy in the men worldwide. Cancer stem cells (CSCs) are the sub-population of cells present in the tumor which possess unique properties of self-renewal and multilineage differentiation thus thought to be major cause of therapy resistance, disease relapse, and mortality in several malignancies including PCa. CSCs have also been shown positive for the common stem cells markers such as ALDH EZH2, OCT4, SOX2, c-MYC, Nanog etc. Therefore, isolation and characterization of CSCs specific markers which may discriminate CSCs and normal stem cells are critical to selectively eliminate CSCs. Rapid advances in the field offers a theoretical explanation for many of the enduring uncertainties encompassing the etiology and an optimism for the identification of new stem-cell targets, development of reliable and efficient therapies in the future. The emerging reports have also provided unprecedented insights into CSCs plasticity, quiescence, renewal, and therapeutic response. In this review, we discuss the identification of PCa stem cells, their unique properties, stemness-driving pathways, new diagnostics, and therapeutic interventions.
Collapse
|