1
|
Escudé Martinez de Castilla P, Estapé Senti M, Erkens S, van Weerden WM, Kooijmans SAA, Fens MH, Vader P, Schiffelers RM. Reticuloendothelial system blockade does not enhance siRNA-LNP circulation or tumor accumulation in mice. Int J Pharm X 2025; 9:100324. [PMID: 40115963 PMCID: PMC11925117 DOI: 10.1016/j.ijpx.2025.100324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/23/2025] Open
Abstract
One of the biggest challenges for siRNA-based therapeutics is intracellular delivery into the target cell, which can be facilitated by encapsulating siRNA in lipid nanoparticles (LNPs). In this study, we formulated D-Lin-MC3-DMA-LNPs encapsulating siRNA against the androgen receptor (AR), a key driver in prostate cancer. We effectively knocked down AR expression at both the mRNA as well as protein levels in vitro in AR-expressing prostate cancer cell lines. However, when moving to in vivo studies, siRNA-LNP efficacy is hindered by rapid clearance by the reticuloendothelial system (RES) in the liver and spleen. We evaluated whether transient RES blockade through systemic pre-administration of dextran sulfate or liposomes could extend the circulation time and enhance tumor accumulation of siRNA-LNPs in tumor-bearing mice. In two different mouse prostate cancer (PCa) xenograft models, we observed that, upon systemic administration, LNPs still predominantly accumulated in the liver and spleen, with only limited tumor uptake. Our findings demonstrate that pre-treatment with dextran sulfate or liposomes did not enhance siRNA-LNP blood circulation time or tumor accumulation in vivo, indicating the need for alternative strategies to enhance siRNA-LNP delivery to tumors.
Collapse
Affiliation(s)
| | | | - Sigrun Erkens
- Department of Urology, Erasmus MC, Rotterdam, the Netherlands
| | | | | | - Marcel H Fens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, the Netherlands
| | - Pieter Vader
- CDL Research, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | |
Collapse
|
2
|
Li GX, Ma B, Zhang S, Liu R, Siddiqi IN, Sali A, El-Khoueiry A, Gross M, Salhia B, Sadeghi S, Gill PS. EphB4-ephrin-B2 are targets in castration resistant prostate cancer. Br J Cancer 2025; 132:679-689. [PMID: 40044981 PMCID: PMC11997055 DOI: 10.1038/s41416-025-02942-5] [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: 06/01/2024] [Revised: 10/31/2024] [Accepted: 01/14/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND PI3K pathway activation is a common and early event in prostate cancer, from loss of function mutations in PTEN, or activating mutations in PIK3Ca or AKT leading to constitutive activation, induction of growth factor-receptors kinase EphB4 and its ligand ephrin-B2. We hypothesized that induction of EphB4 is an early event required for tumor initiation. Secondly, we hypothesized that EphB4 remains relevant when prostate cancer becomes androgen independent. METHODS Genetic mouse model of conditional PTEN deletion in prostate epithelium induces tumor in all mice. We tested this model against EPHB4 wild type and deleted in prostate epithelium. This allowed us to test its role in tumor initiation. We also tested an orthogonal approach by using decoy soluble EphB4 to block bidirectional signaling resulting from EphB4-ephrin-B2 interaction. Role of EphB4-ephrin-B2 in androgen deprived mice was tested for role in refractory cancer model. RESULTS PTEN deletion induces EphB4 and ephrin-B2 in prostate cancer which was substantially reduced when EPHB4 is deleted in the same prostate epithelial cells. sEphB4-alb fusion protein with improved pharmacokinetics similarly inhibited tumor formation, thus establishing the role in tumor initiation. sEphB4-alb retained the efficacy in castration resistant androgen independent prostate cancer. We have thus observed that induction of EphB4 is required for the initiation of prostate cancer in PTEN null mouse and that signaling downstream from EphB4 is required in androgen deprivation and thus castration resistant prostate cancer. Pharmacological inhibition of EphB4 pathway reproduced the results. Targeting EphB4 should be tested in prostate cancer especially those resistant to androgen deprivation therapy. CONCLUSIONS EphB4 and ephrin-B2 receptor ligand pair is induced in PTEN null prostate cancer, which significantly contributes to the tumor initiation. Secondly, EphB4-ephrin-B2 pathway continue to promote tumor progression even in androgen deprivation and thus hormone refractory tumor. EphB4-ephrin-B2 may be candidates for precision medicine with biomarker-based patient selection with and without concurrent standard of care.
Collapse
Affiliation(s)
- Grace Xiuqing Li
- Department of Medicine, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Binyun Ma
- Department of Medicine, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shaobing Zhang
- Department of Medicine, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ren Liu
- Merck Pharmaceutical Inc. Previous: Department of Medicine, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Imran N Siddiqi
- Department of Pathology, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Akash Sali
- Catherine & Joseph Aresty Department of Urology, USC Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anthony El-Khoueiry
- Department of Medicine, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mitchell Gross
- Department of Medicine, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Bodour Salhia
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sarmad Sadeghi
- Department of Medicine, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Parkash S Gill
- Department of Medicine, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Karatzas P, Brotzakis ZF, Sarimveis H. Small Molecules Targeting the Structural Dynamics of AR-V7 Partially Disordered Proteins Using Deep Ensemble Docking. J Chem Theory Comput 2025. [PMID: 40231860 DOI: 10.1021/acs.jctc.5c00171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
The extensive conformational dynamics of partially disordered proteins hinders the efficiency of traditional in-silico structure-based drug discovery approaches due to the challenge of screening large chemical spaces of compounds, albeit with an excessive number of transient binding sites, quickly making this problem intractable. In this study, using the monomer of the AR-V7 transcription factor splicing variant related to prostate cancer as a test case, we present a deep ensemble docking pipeline that accelerates the screening of small molecule binders targeting partially disordered proteins at functional regions. By swiftly identifying the conformational ensemble of AR-V7 and reducing the dimension of binding sites by a factor of 90, we identify functionally relevant binding sites along the AR-V7 structural ensemble at phase separation-prone regions that have been experimentally shown to contribute to enhanced transcription activity and the onset of tumor growth. Following this, we combine physics-based molecular docking and multiobjective classification machine learning models to speed up the screening for binders in a larger chemical space able to target these functional multiple binding sites of AR-V7. This step increases the multibinding site hit rate of small molecules by a factor of 17 compared to naive molecular docking. Finally, assessing in atomistic molecular dynamics the effect of a selected binder on AR-V7 dynamics, we find that in the presence of the ChEMBL22003 compound, AR-V7 exhibits less conformational entropy, smaller solvent exposure of phase separation-prone regions, and higher solvent exposure of other protein regions, promoting this compound as a potential AR-V7 phase separation modulator.
Collapse
Affiliation(s)
- Pantelis Karatzas
- School of Chemical Engineering, National Technical University of Athens, 9 Heroon Polytechniou Street, Athens 15780, Greece
| | - Z Faidon Brotzakis
- Institute of Bioinnovation (IBI), Biomedical Science Research Center Alexander Fleming, 34 Fleming Street, Vari 16672, Greece
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Haralambos Sarimveis
- School of Chemical Engineering, National Technical University of Athens, 9 Heroon Polytechniou Street, Athens 15780, Greece
| |
Collapse
|
4
|
Palanisamy B, Mandal AKA. Unlocking the potential: Receptor-mediated targeted drug delivery in cancer therapy. Pathol Res Pract 2025; 270:155955. [PMID: 40209568 DOI: 10.1016/j.prp.2025.155955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/29/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
Receptor-mediated targeted drug delivery has emerged as a pivotal strategy in cancer therapy, offering precision and specificity in combating malignant diseases while minimizing systemic toxicity. This review explores the multifaceted role of receptors in cancer biology, emphasizing their contributions to cancer progression, metastasis, and their potential as therapeutic targets. Ligand-based targeting approaches highlight the utility of small molecules, peptides, and antibodies, as well as the development of novel targeting ligands. A critical focus is placed on engineering receptor-targeted nanoparticles and advanced drug delivery systems. Innovations in dual-targeting strategies and the targeted delivery to the tumour microenvironment (TME) and metastatic niches are discussed, underscoring their potential to enhance therapeutic efficacy. Additionally, receptor-targeted imaging is reviewed for its dual role in diagnosis and real-time treatment monitoring. To address the challenges of side effects and off-target toxicity, strategies that minimize these risks while targeting overexpressed receptors in solid tumours are explored. Finally, the review outlines future directions in receptor-targeted cancer therapy, emphasizing the need for interdisciplinary research to refine these strategies further. This comprehensive analysis aims to provide a roadmap for advancing receptor-based therapeutic approaches, ultimately improving outcomes for cancer patients.
Collapse
Affiliation(s)
- Balaji Palanisamy
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| | - Abul Kalam Azad Mandal
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| |
Collapse
|
5
|
Li C, Cheng S, Yu J, Zheng Q, Yu G, Xu M, Meng X, Zeng X, Liu K, Xu B, Luo H, Xu G. Hit to lead optimization of the 4-trifluoromethylquinoline derivatives as novel SGK1 inhibitors with potent anti-prostate cancer activity. Eur J Med Chem 2025; 287:117336. [PMID: 39908792 DOI: 10.1016/j.ejmech.2025.117336] [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: 10/22/2024] [Revised: 01/19/2025] [Accepted: 01/25/2025] [Indexed: 02/07/2025]
Abstract
Prostate cancer (PCa) remains a significant health concern for males, and serum/glucocorticoid-regulated kinase-1 (SGK1) plays a crucial role in its pathogenesis. This provides a promising target for the development of novel therapies against PCa. Herein, we reported the structural optimization of the hit compound H1, which was discovered in our previous work as an SGK1 inhibitor. Based on docking research for the active binding conformation of compound H1, a series of novel 4-trifluoromethyl quinoline derivatives were developed by replacing the 6-methoxy group in the quinoline skeleton of compound H1 with a larger aryl ring to occupy the hinge region of SGK1. Among them, compound 12f showed the strongest SGK1 inhibitory potency, with an IC50 value of 0.39 μM, representing a 7.8-fold improvement over compound H1. Molecular docking studies revealed that the 6-methoxyphenylamine moiety of compound 12f effectively extends into the hinge region of SGK1, establishing a crucial hydrogen bonding interaction with Glu183 that enhances its biological potency. In vivo, compound 12f effectively suppressed tumor growth in the PC3 xenograft model in BALB/c nude mice without inducing any observable toxicity. Moreover, mechanistic studies showed that compound 12f hindered PC3 cell migration and invasion, improved the thermal stability of SGK1 protein in PC3 cells, decreased SGK1 protein levels in tumor tissues, and effectively inhibited the phosphorylation of SGK1 and its substrates in PC3 cells in a dose- and time-dependent manner. In summary, the results of this study highlight the potential of 12f as a lead compound for further optimization in the development of new therapies against PCa targeting SGK1.
Collapse
Affiliation(s)
- Cheng Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China; Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Sha Cheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Jia Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Qian Zheng
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Gang Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Mei Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Xueling Meng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Xiaoping Zeng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Kun Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Bixue Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China.
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China.
| | - Guangcan Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China.
| |
Collapse
|
6
|
Huang S, Yin H. Multi-Omics Analysis of the Anoikis Gene CASP8 in Prostate Cancer and Biochemical Recurrence (BCR). Biomedicines 2025; 13:661. [PMID: 40149637 PMCID: PMC11939882 DOI: 10.3390/biomedicines13030661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/16/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Prostate cancer, as an androgen-dependent malignant tumor in older men, has attracted the attention of a wide range of clinicians. BCR remains a significant challenge following early prostate cancer treatment. Methods: The specific expression pattern of the Anoikis gene set in prostate cancer cells was first explored by single-cell and spatial transcriptomics analysis. Genes causally associated with prostate cancer were screened using Summary-data-based Mendelian Randomization (SMR). Subsequently, we explored the role and mechanism of CASP8 in prostate cancer cells and defined a new cell type: the CASP8 T cell. We constructed a prediction model that can better predict the BCR of prostate cancer, and explored the differences in various aspects of clinical subgroups, tumor microenvironments, immune checkpoints, drug sensitivities, and tumor-immune circulations between high- and low-risk groups. The results of SMR analysis indicated that CASP8 could increase the risk of prostate cancer. Based on the differential genes of CASP8-positive and -negative T cells, we constructed a four-gene prognostic model with a 5-year AUC of 0.713. Results: The results revealed that high-risk prostate cancer BCR patients had various characteristics such as higher tumor purity, higher BCR rate, downregulated SIRPA immune checkpoints, and unique drug sensitivity. Conclusions: In summary, CASP8 may be a potential biomarker for prostate cancer.
Collapse
Affiliation(s)
- Shan Huang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China;
- Institute of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100034, China
| | - Hang Yin
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China;
- Institute of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100034, China
| |
Collapse
|
7
|
Chahdi A, Jorgez C, Rosenfeld JA, Neetu N, Seth A. Androgen receptor ubiquitination links KCTD13 to genitourinary tract defects. FASEB J 2025; 39:e70406. [PMID: 39968753 DOI: 10.1096/fj.202402072rr] [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: 09/03/2024] [Revised: 01/09/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
The potassium channel tetramerization domain containing 13 (KCTD13) protein is a substrate-specific adapter for cullin3-based E3 ubiquitin ligase. Patients with copy number variants at this locus exhibit genitourinary tract anomalies. In this study, we show that decreased androgen receptor (AR) protein level correlated with increased AR ubiquitination in the testis of Kctd13-deficient mice, suggesting that KCTD13 inhibits AR ubiquitination. KCTD13 increased CUL3-dependent AR ubiquitination but had no effect on CUL3 binding to AR, confirming the role of KCTD13 as an adaptor of CUL3 ligase. Recombinant KCTD13 directly binds to recombinant AR, and the BTB domain of KCTD13 is critical for binding both the N-terminal domain of AR and STUB1. Moreover, KCTD13 dose-dependently decreased STUB1 binding to AR resulting in decreased AR ubiquitination. KCTD13 ΔBTB was unable to bind to AR and subsequently failed to block STUB1-mediated AR ubiquitination, strongly suggesting that reduced AR ubiquitination is dependent on KCTD13 ability to dissociate AR/STUB1 complex. Furthermore, KCTD13 increased the expression of AR target gene, FOXJ1, whereas KCTD13 ΔBTB had no effect. Our data reveal a distinctive mode of action of KCTD13 on AR ubiquitination depending on the E3 ubiquitin ligase involved: (1) KCTD13 increased CUL3-dependent AR ubiquitination but had no effect on CUL3 binding to AR; and (2) KCTD13 decreased STUB1-mediated AR ubiquitination by decreasing STUB1 binding to AR thus preventing AR ubiquitination. We hypothesize that in the testes of Kctd13-deficient mice, the absence of KCTD13 results in increased binding of STUB1 to AR leading to increased AR ubiquitination and degradation.
Collapse
Affiliation(s)
- Ahmed Chahdi
- Department of Surgery, Nemours Children's Health, Orlando, Florida, USA
- College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Carolina Jorgez
- Scott Department of Urology, Baylor College of Medicine, Houston, Texas, USA
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Neetu Neetu
- Department of Biochemistry, Baylor College of Medicine, Houston, Texas, USA
| | - Abhishek Seth
- Department of Surgery, Nemours Children's Health, Orlando, Florida, USA
- College of Medicine, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
8
|
Zheng G, Yan Z, Zou J, Zou X, Chai K, Zhang G. AR and YAP crosstalk: impacts on therapeutic strategies in prostate cancer. Front Oncol 2025; 15:1520808. [PMID: 39963114 PMCID: PMC11830605 DOI: 10.3389/fonc.2025.1520808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Prostate cancer ranks as one of the most common types of cancer affecting men worldwide, and its progression is shaped by a diverse array of influencing factors. The AR signaling pathway plays a pivotal role in the pathogenesis of prostate cancer. While existing anti-androgen treatments show initial efficacy, they ultimately do not succeed in halting the advancement to CRPC. Recent studies have identified alterations in the Hippo-YAP signaling pathway within prostate cancer, highlighting intricate crosstalk with the AR signaling pathway. In this review, we examine the interactions and underlying mechanisms between AR and YAP, the key molecules in these two signaling pathways. AR regulates the stability and function of YAP by modulating its transcription, translation, and phosphorylation status, while YAP exerts both promotional and inhibitory regulatory effects on AR. Based on these findings, this paper investigates their significant roles in the onset, progression, and therapeutic resistance of prostate cancer, and discusses the clinical potential of YAP in prostate cancer treatment.
Collapse
Affiliation(s)
- Guansong Zheng
- First Clinical College, Gannan Medical University, Ganzhou, China
| | - Zhaojie Yan
- First Clinical College, Gannan Medical University, Ganzhou, China
| | - Junrong Zou
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, Gannan Medical University, Ganzhou, China
- Department of Jiangxi Engineering Technology Research Center of Calculi Prevention, Gannan Medical University, Ganzhou, China
| | - Xiaofeng Zou
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Keqiang Chai
- Department of Urology, Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, China
| | - Guoxi Zhang
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
9
|
Ajayi AF, Oyovwi MO, Akano OP, Akanbi GB, Adisa FB. Molecular pathways in reproductive cancers: a focus on prostate and ovarian cancer. Cancer Cell Int 2025; 25:33. [PMID: 39901204 PMCID: PMC11792371 DOI: 10.1186/s12935-025-03658-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Reproductive cancers, including prostate and ovarian cancer, are highly prevalent worldwide and pose significant health challenges. The molecular underpinnings of these cancers are complex and involve dysregulation of various cellular pathways. Understanding these pathways is crucial for developing effective therapeutic strategies. This review aims to provide an overview of the molecular pathways implicated in prostate and ovarian cancers, highlighting key genetic alterations, signaling cascades, and epigenetic modifications. A comprehensive literature search was conducted using databases such as PubMed, Web of Science, and Google Scholar. Articles focusing on molecular pathways in prostate and ovarian cancer were reviewed and analyzed. In prostate cancer, recurrent mutations in genes like AR, TP53, and PTEN drive tumor growth and progression. Androgen signaling plays a significant role, with alterations in the AR pathway contributing to resistance to antiandrogen therapies. In ovarian cancer, high-grade serous carcinomas are characterized by mutations in TP53, BRCA1/2, and homologous recombination repair genes. PI3K and MAPK pathways are frequently activated, promoting cell proliferation and survival. Epigenetic alterations, including DNA methylation and histone modifications, are also prevalent in both cancer types. The molecular pathways involved in prostate and ovarian cancer are diverse and complex. Targeting these pathways with precision medicine approaches holds promise for improving patient outcomes. Further research is needed to elucidate the mechanisms of resistance and identify novel therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Ayodeji Folorunsho Ajayi
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
- Anchor Biomed Research Institute, Ogbomoso, Oyo, Nigeria
- Department of Physiology, Adeleke University, Ede, Osun, Nigeria
| | | | - Oyedayo Phillips Akano
- Department of Physiology, School of Basic Medical Sciences, Babcock University, Ilishan Remo, Ogun, Nigeria
| | - Grace Bosede Akanbi
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
| | - Florence Bukola Adisa
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
| |
Collapse
|
10
|
Hoffmann TJ, Graff RE, Madduri RK, Rodriguez AA, Cario CL, Feng K, Jiang Y, Wang A, Klein RJ, Pierce BL, Eggener S, Tong L, Blot W, Long J, Goss LB, Darst BF, Rebbeck T, Lachance J, Andrews C, Adebiyi AO, Adusei B, Aisuodionoe-Shadrach OI, Fernandez PW, Jalloh M, Janivara R, Chen WC, Mensah JE, Agalliu I, Berndt SI, Shelley JP, Schaffer K, Machiela MJ, Freedman ND, Huang WY, Li SA, Goodman PJ, Till C, Thompson I, Lilja H, Ranatunga DK, Presti J, Van Den Eeden SK, Chanock SJ, Mosley JD, Conti DV, Haiman CA, Justice AC, Kachuri L, Witte JS. Genome-wide association study of prostate-specific antigen levels in 392,522 men identifies new loci and improves prediction across ancestry groups. Nat Genet 2025; 57:334-344. [PMID: 39930085 PMCID: PMC11821537 DOI: 10.1038/s41588-024-02068-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: 10/18/2023] [Accepted: 12/20/2024] [Indexed: 02/14/2025]
Abstract
We conducted a multiancestry genome-wide association study of prostate-specific antigen (PSA) levels in 296,754 men (211,342 European ancestry, 58,236 African ancestry, 23,546 Hispanic/Latino and 3,630 Asian ancestry; 96.5% of participants were from the Million Veteran Program). We identified 318 independent genome-wide significant (P ≤ 5 × 10-8) variants, 184 of which were novel. Most demonstrated evidence of replication in an independent cohort (n = 95,768). Meta-analyzing discovery and replication (n = 392,522) identified 447 variants, of which a further 111 were novel. Out-of-sample variance in PSA explained by our genome-wide polygenic risk scores ranged from 11.6% to 16.6% for European ancestry, 5.5% to 9.5% for African ancestry, 13.5% to 18.2% for Hispanic/Latino and 8.6% to 15.3% for Asian ancestry and decreased with increasing age. Midlife genetically adjusted PSA levels were more strongly associated with overall and aggressive prostate cancer than unadjusted PSA levels. Our study highlights how including proportionally more participants from underrepresented populations improves genetic prediction of PSA levels, offering potential to personalize prostate cancer screening.
Collapse
Affiliation(s)
- Thomas J Hoffmann
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Rebecca E Graff
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Ravi K Madduri
- Data Science and Learning Division, Argonne National Laboratory, Argonne, IL, USA
| | - Alex A Rodriguez
- Data Science and Learning Division, Argonne National Laboratory, Argonne, IL, USA
| | - Clinton L Cario
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Karen Feng
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Yu Jiang
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Anqi Wang
- Center for Genetic Epidemiology, Department of Population and Preventive Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robert J Klein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brandon L Pierce
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Comprehensive Cancer Center, University of Chicago, Chicago, IL, USA
| | - Scott Eggener
- Comprehensive Cancer Center, University of Chicago, Chicago, IL, USA
- Department of Urology, University of Chicago, Chicago, IL, USA
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Lin Tong
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - William Blot
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jirong Long
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Louisa B Goss
- Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Burcu F Darst
- Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Timothy Rebbeck
- Dana Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Joseph Lachance
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Caroline Andrews
- Dana Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Akindele O Adebiyi
- Department of Community Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | | - Oseremen I Aisuodionoe-Shadrach
- College of Health Sciences, University of Abuja, Abuja, Nigeria
- Cancer Science Centre Abuja, Abuja, Nigeria
- University of Abuja Teaching Hospital, Abuja, Nigeria
| | - Pedro W Fernandez
- Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Mohamed Jalloh
- Hospital General Idrissa Pouye, Dakar, Senegal
- Ecole Doctorale, Universite Iba Der Thiam de Thies, Thies, Senegal
| | - Rohini Janivara
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Wenlong C Chen
- Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
| | - James E Mensah
- Korle-Bu Teaching Hospital and University of Ghana Medical School, Accra, Ghana
| | - Ilir Agalliu
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, New York, NY, USA
- Department of Urology, Albert Einstein College of Medicine, New York, NY, USA
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - John P Shelley
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kerry Schaffer
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Shengchao A Li
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Phyllis J Goodman
- SWOG Statistics and Data Management Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Cathee Till
- SWOG Statistics and Data Management Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ian Thompson
- CHRISTUS Santa Rosa Medical Center Hospital, San Antonio, TX, USA
| | - Hans Lilja
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Translational Medicine, Lund University, Malmö, Sweden
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Joseph Presti
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | | | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Jonathan D Mosley
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David V Conti
- Center for Genetic Epidemiology, Department of Population and Preventive Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Christopher A Haiman
- Center for Genetic Epidemiology, Department of Population and Preventive Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Amy C Justice
- Veterans Administration Connecticut Healthcare System, West Haven, CT, USA
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale University School of Public Health, Yale School of Medicine, New Haven, CT, USA
| | - Linda Kachuri
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
| | - John S Witte
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Genetics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
11
|
Safaeesirat A, Taeb H, Tekoglu E, Morova T, Lack NA, Emberly E. Inference of transcriptional regulation from STARR-seq data. Phys Rev E 2025; 111:024402. [PMID: 40103141 DOI: 10.1103/physreve.111.024402] [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: 03/19/2024] [Accepted: 01/15/2025] [Indexed: 03/20/2025]
Abstract
One of the primary regulatory processes in cells is transcription, during which RNA polymerase II (Pol-II) transcribes DNA into RNA. The binding of Pol-II to its site is regulated through interactions with transcription factors (TFs) that bind to DNA at enhancer cis-regulatory elements. Measuring the enhancer activity of large libraries of distinct DNA sequences is now possible using massively parallel reporter assays (MPRAs), and computational methods have been developed to identify the dominant statistical patterns of TF binding within these large datasets. Such methods are global in their approach and may overlook important regulatory sites that function only within the local context. Here we introduce a method for inferring functional regulatory sites (their number, location, and width) within an enhancer sequence based on measurements of its transcriptional activity from an MPRA method such as STARR-seq. The model is based on a mean-field thermodynamic description of Pol-II binding that includes interactions with bound TFs. Our method applied to simulated STARR-seq data for a variety of enhancer architectures shows how data quality impacts the inference and also how it can find local regulatory sites that may be missed in a global approach. We also apply the method to recently measured STARR-seq data on androgen receptor (AR) bound sequences, a TF that plays an important role in the regulation of prostate cancer. The method identifies key regulatory sites within these sequences, which are found to overlap with binding sites of known coregulators of AR.
Collapse
Affiliation(s)
- Amin Safaeesirat
- Simon Fraser University, Department of Physics, Burnaby, British Columbia, Canada VSA1S6
| | - Hoda Taeb
- Simon Fraser University, Department of Physics, Burnaby, British Columbia, Canada VSA1S6
| | - Emirhan Tekoglu
- Koç University, School of Medicine, Istanbul 34450, Turkey
- Koç University, Koç University Research Centre for Translational Medicine (KUTTAM), Istanbul 34010, Turkey
| | - Tunc Morova
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada V6H3Z6
| | - Nathan A Lack
- Koç University, School of Medicine, Istanbul 34450, Turkey
- Koç University, Koç University Research Centre for Translational Medicine (KUTTAM), Istanbul 34010, Turkey
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada V6H3Z6
- University of British Columbia, Department of Urologic Sciences, Vancouver, British Columbia, Canada V6T1Z4
| | - Eldon Emberly
- Simon Fraser University, Department of Physics, Burnaby, British Columbia, Canada VSA1S6
| |
Collapse
|
12
|
Jiang C, Hong Z, Liu S, Hong Z, Dai B. Roles of CDK12 mutations in PCa development and treatment. Biochim Biophys Acta Rev Cancer 2025; 1880:189247. [PMID: 39681197 DOI: 10.1016/j.bbcan.2024.189247] [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: 07/14/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 12/18/2024]
Abstract
Prostate cancer (PCa) is one of the most common cancers in men, and cyclin-dependent kinase 12 (CDK12) is emerging as a novel star player in the PCa tumorigenesis and progression to castration-resistant prostate cancer (CRPC). In PCa, CDK12 alterations are mostly loss-of-function mutations featuring intronic polyadenylation (IPA), focal tandem duplications (FTDs), and R-loops formation and transcription-replication conflicts (TRCs). The occurrence of IPA can result in homologous recombination deficiency (HRD) and androgen receptor (AR) variation. FTDs induce neoantigens and increase the expression of the AR, MYC, and other hotspot- associated genes. R-loops lead to TRCs and influence various cellular processes, including gene expression and genome stability. Due to the poor prognosis of CDK12-mutant PCa patients and the mediocre response to classic standard therapies, HRD and increased neoantigen levels have provided clinicians with new insights into alternative systematic treatments for this novel PCa phenotype. In this review, we summarize the roles of CDK12 mutations in PCa and discuss their clinical value, suggesting that CDK12 potentially represents a target for further research and the development of clinical strategies for PCa.
Collapse
Affiliation(s)
- Chenye Jiang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Genitourinary Cancer Institute, Shanghai 200032, China
| | - Zhe Hong
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Genitourinary Cancer Institute, Shanghai 200032, China.
| | - Shiwei Liu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Genitourinary Cancer Institute, Shanghai 200032, China
| | - Zongyuan Hong
- Laboratory of Quantitative Pharmacology, Wannan Medical College, Wuhu 241002, China
| | - Bo Dai
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Genitourinary Cancer Institute, Shanghai 200032, China.
| |
Collapse
|
13
|
Dahl-Wilkie H, Gomez J, Kelley A, Manjit K, Mansoor B, Kanumuri P, Pardo S, Molleur D, Falah R, Konakalla AR, Omiyale M, Weintraub S, Delk NA. Chronic IL-1-Exposed LNCaP Cells Evolve High Basal p62-KEAP1 Complex Accumulation and NRF2/KEAP1-Dependent and -Independent Hypersensitive Nutrient Deprivation Response. Cells 2025; 14:192. [PMID: 39936983 PMCID: PMC11816438 DOI: 10.3390/cells14030192] [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: 11/09/2024] [Revised: 01/25/2025] [Accepted: 01/26/2025] [Indexed: 02/13/2025] Open
Abstract
Chronic inflammation is a cancer hallmark and chronic exposure to interleukin-1 (IL-1) transforms castration-sensitive prostate cancer (PCa) cells into more fit castration-insensitive PCa cells. p62 is a scaffold protein that protects cells from nutrient deprivation via autophagy and from cytotoxic reactive oxygen via NFκB and NRF2 antioxidant signaling. Herein, we report that the LNCaP PCa cell line acquires high basal accumulation of the p62-KEAP1 complex when chronically exposed to IL-1. p62 promotes non-canonical NRF2 antioxidant signaling by binding and sequestering KEAP1 to the autophagosome for degradation. But despite high basal p62-KEAP1 accumulation, only two of several NRF2-induced genes analyzed, GCLC and HMOX1, showed high basal mRNA levels, suggesting that the high basal p62-KEAP1 accumulation does not result in overall high basal NRF2 activity. Nutrient starvation induces NRF2-dependent GCLC upregulation and HMOX1 repression, and we found that chronic IL-1-exposed LNCaP cells show hypersensitivity to serum starvation-induced GCLC and HMOX1 regulation. Thus, chronic IL-1 exposure affects cell response to nutrient stress. While HMOX1 expression remains NRF2/KEAP1-dependent in chronic IL-1-exposed LNCaP cells, GCLC expression is NRF2/KEAP1-independent. Furthermore, the high basal p62-KEAP1 complex accumulation is not required to regulate GCLC or HMOX1 expression, suggesting cells chronically exposed to IL-1 evolve a novel NRF2-independent role for the p62/KEAP1 axis.
Collapse
Affiliation(s)
- Haley Dahl-Wilkie
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Jessica Gomez
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Anastasia Kelley
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Kirti Manjit
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Basir Mansoor
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Preethi Kanumuri
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Sammy Pardo
- Department of Biochemistry & Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (S.P.); (D.M.); (S.W.)
| | - Dana Molleur
- Department of Biochemistry & Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (S.P.); (D.M.); (S.W.)
| | - Rafah Falah
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Anisha R. Konakalla
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Morolake Omiyale
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Susan Weintraub
- Department of Biochemistry & Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (S.P.); (D.M.); (S.W.)
| | - Nikki A. Delk
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| |
Collapse
|
14
|
Deminami M, Hashimoto M, Takahashi H, Harada N, Minami Y, Kitakaze T, Masuda W, Takenaka S, Inui H, Yamaji R. Androgens suppress the sialyltransferases ST3GAL1 and ST3GAL4 and modulate mucin 10 glycosylation in the submandibular gland, related to sex differences in commensal microbiota composition in mice. Biosci Biotechnol Biochem 2025; 89:241-254. [PMID: 39572079 DOI: 10.1093/bbb/zbae175] [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: 09/18/2024] [Accepted: 11/15/2024] [Indexed: 01/25/2025]
Abstract
Sex differences exist in the commensal microbiota that impact on multiple physiological processes in the host. Here, we examined the mechanism by which the sex differences are formed. In addition to the epithelial ductal cell, the acinar cell mass in the submandibular gland was associated with androgen-androgen receptor (AR) signaling. Sex differences in the formation of submandibular mucin 10 (MUC10) were identified using SDS-PAGE. Neuraminidase treatment, which hydrolyzes terminal sialic acid, influenced the mobility shift of MUC10. Androgen-AR signaling negatively regulated ST3 β-galactoside α-2,3-sialyltransferase 1 (St3gal1) and St3gal4 in the submandibular gland. There was a trend and significant sex differences in α-diversity (Shannon, P = .09) and β-diversity (unweighted UniFrac) in oral microbiota composition, respectively. Some female-preferring bacteria including Akkermansia muciniphila can assimilate mucin by degrading terminal sialic acids. Our results indicate that androgen-AR signaling suppresses ST3GAL1 and ST3GAL4, which can influence sex differences in commensal microbiota composition.
Collapse
Affiliation(s)
- Mana Deminami
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Miku Hashimoto
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Hiroki Takahashi
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan
| | - Naoki Harada
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan
| | - Yukari Minami
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Tomoya Kitakaze
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan
| | - Wataru Masuda
- Department of Nutrition, Faculty of Home Economics, Kyushu Women's University, Kitakyushu, Fukuoka, Japan
| | - Shigeo Takenaka
- Division of Nutrition, Graduate School of Human Life and Ecology, Osaka Metropolitan University, Habikino, Osaka, Japan
| | - Hiroshi Inui
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan
- Department of Health and Nutrition, Otemae University, Osaka, Japan
| | - Ryoichi Yamaji
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan
- Center for Research and Development of Bioresources, Osaka Metropolitan University, Sakai, Osaka, Japan
| |
Collapse
|
15
|
Wang K, Fischer A, Maccio U, Zitzmann K, Robledo M, Lauseker M, Bauer J, Bechmann N, Gahr S, Maurer J, Peischer L, Reul A, Nieß H, Zimmermann P, Ilmer M, Schilbach K, Knösel T, Kroiss M, Fassnacht M, Müller SA, Morand GB, Huber A, Vetter D, Lehmann K, Kulcsar Z, Mohr H, Pellegata NS, Hantel C, Reincke M, Beuschlein F, Pacak K, Grossman AB, Auernhammer CJ, Nölting S. Impact of sex hormones on pheochromocytomas, paragangliomas, and gastroenteropancreatic neuroendocrine tumors. Eur J Endocrinol 2025; 192:46-60. [PMID: 39804847 DOI: 10.1093/ejendo/lvae163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/08/2024] [Indexed: 01/16/2025]
Abstract
OBJECTIVE The effects of sex hormones remain largely unexplored in pheochromocytomas and paragangliomas (PPGLs) and gastroenteropancreatic neuroendocrine tumors (GEP-NETs). METHODS We evaluated the effects of estradiol, progesterone, Dehydroepiandrosterone sulfate (DHEAS), and testosterone on human patient-derived PPGL/GEP-NET primary culture cell viability (n = 38/n = 12), performed next-generation sequencing and immunohistochemical hormone receptor analysis in patient-derived PPGL tumor tissues (n = 36). RESULTS In PPGLs, estradiol and progesterone (1 µm) demonstrated overall significant antitumor effects with the strongest efficacy in PPGLs with NF1 (cluster 2) pathogenic variants. Estrogen receptor alpha (ERα) positivity was detected in 11/36 PPGLs, including 4/4 head-and-neck paragangliomas (HNPGLs). ERα-positive tumors responded with a significant cell viability decrease to estradiol. DHEAS and testosterone (1 µm) displayed no effects, but higher doses of testosterone (10 µm) demonstrated significant antitumor effects, including a pheochromocytoma lung metastasis with strong androgen receptor positivity (30%). Driven by the antitumor effects of estrogen, we evaluated G-protein-coupled estrogen receptor (GPER) agonist G-1 as a potential therapeutic option for PPGLs and found strong significant antitumor potential, with the strongest efficacy in tumors with NF1 pathogenic variants. Moreover, we detected sex-related differences-tumors from male patients showed significantly stronger responsivity to G-1 compared with tumors from female patients. In GEP-NETs, sex hormones showed overall no effects, especially no tumor growth-promoting effects. CONCLUSION We provide novel data on the effects of elevated sex hormone levels, potentially seen during pregnancy or hormone replacement therapy, on PPGL/GEP-NET tumor growth. G-1 might offer a novel therapeutic approach for some PPGLs depending on patient's sex and the individual tumor's genetic/molecular background. All HNPGLs showed ERα positivity.
Collapse
Affiliation(s)
- Katharina Wang
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Alessa Fischer
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | - Umberto Maccio
- Department of Pathology and Molecular Pathology, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | - Kathrin Zitzmann
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, 28029 Madrid, Spain
| | - Michael Lauseker
- Institute for Medical Information Processing, Biometry, and Epidemiology, Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Jana Bauer
- Institute for Medical Information Processing, Biometry, and Epidemiology, Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Simon Gahr
- Department of Internal Medicine II, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Julian Maurer
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Lea Peischer
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Astrid Reul
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | - Hanno Nieß
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Petra Zimmermann
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Matthias Ilmer
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, LMU Munich, 81377 Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS Centre of Excellence), LMU University Hospital, 81377 Munich, Germany
| | - Katharina Schilbach
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Thomas Knösel
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS Centre of Excellence), LMU University Hospital, 81377 Munich, Germany
- Institute of Pathology, Faculty of Medicine, LMU Munich, 80337 Munich, Germany
| | - Matthias Kroiss
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, 80336 Munich, Germany
- Department of Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Martin Fassnacht
- Department of Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Simon A Müller
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich and University of Zurich, CH-8091 Zurich, Switzerland
| | - Gregoire B Morand
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich and University of Zurich, CH-8091 Zurich, Switzerland
| | - Alexander Huber
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich and University of Zurich, CH-8091 Zurich, Switzerland
| | - Diana Vetter
- Department of Visceral and Transplantation Surgery, University Hospital Zurich and University of Zurich, CH-8091 Zurich, Switzerland
| | - Kuno Lehmann
- Department of Visceral and Transplantation Surgery, University Hospital Zurich and University of Zurich, CH-8091 Zurich, Switzerland
| | - Zsolt Kulcsar
- Department of Neuroradiology, University Hospital Zurich and University of Zurich, CH-8091 Zurich, Switzerland
| | - Hermine Mohr
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Natalia S Pellegata
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, 85764 Neuherberg, Germany
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, CH-8091 Zurich, Switzerland
- Department of Internal Medicine III, University Hospital Carl Gustav Carus Dresden, 01307 Dresden, Germany
| | - Martin Reincke
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Felix Beuschlein
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, 80336 Munich, Germany
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, CH-8091 Zurich, Switzerland
- The LOOP Zurich-Medical Research Center, 8044 Zurich, Switzerland
| | - Karel Pacak
- Eunice Kennedy Shriver NICHD, NIH, Bethesda, MD 20892, United States
| | - Ashley B Grossman
- Green Templeton College, University of Oxford, Oxford OX2 6HG, United Kingdom
- NET Unit, ENETS Centre of Excellence, Royal Free Hospital, London NW3 2QG, United Kingdom
| | - Christoph J Auernhammer
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, 80336 Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS Centre of Excellence), LMU University Hospital, 81377 Munich, Germany
| | - Svenja Nölting
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, 80336 Munich, Germany
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, CH-8091 Zurich, Switzerland
| |
Collapse
|
16
|
Zhang X, Zhang M, Sun H, Wang X, Wang X, Sheng W, Xu M. The role of transcription factors in the crosstalk between cancer-associated fibroblasts and tumor cells. J Adv Res 2025; 67:121-132. [PMID: 38309692 PMCID: PMC11725164 DOI: 10.1016/j.jare.2024.01.033] [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: 11/29/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Transcription factors (TFs) fulfill a critical role in the formation and maintenance of different cell types during the developmental process as well as disease. It is believed that cancer-associated fibroblasts (CAFs) are activation status of tissue-resident fibroblasts or derived from form other cell types via transdifferentiation or dedifferentiation. Despite a subgroup of CAFs exhibit anti-cancer effects, most of them are reported to exert effects on tumor progression, further indicating their heterogeneous origin. AIM OF REVIEW This review aimed to summarize and review the roles of TFs in the reciprocal crosstalk between CAFs and tumor cells, discuss the emerging mechanisms, and their roles in cell-fate decision, cellular reprogramming and advancing our understanding of the gene regulatory networks over the period of cancer initiation and progression. KEY SCIENTIFIC CONCEPTS OF REVIEW This manuscript delves into the key contributory factors of TFs that are involved in activating CAFs and maintaining their unique states. Additionally, it explores how TFs play a pivotal and multifaceted role in the reciprocal crosstalk between CAFs and tumor cells. This includes their involvement in processes such as epithelial-mesenchymal transition (EMT), proliferation, invasion, and metastasis, as well as metabolic reprogramming. TFs also have a role in constructing an immunosuppressive microenvironment, inducing resistance to radiation and chemotherapy, facilitating angiogenesis, and even 'educating' CAFs to support the malignancies of tumor cells. Furthermore, this manuscript delves into the current status of TF-targeted therapy and considers the future directions of TFs in conjunction with anti-CAFs therapies to address the challenges in clinical cancer treatment.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Meng Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Hui Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Xu Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Xin Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China.
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China.
| |
Collapse
|
17
|
Chinnapaka S, Bakthavachalam V, Dasari S, Kannan J, Sapkota S, Kumar R, Munirathinam G. Vitamin K3 derivative inhibits androgen receptor signaling in targeting aggressive prostate cancer cells. Biofactors 2025; 51:e2117. [PMID: 39225404 DOI: 10.1002/biof.2117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024]
Abstract
Prostate cancer (PCa) is the second critical cause of cancer-related deaths, with African Americans dying at higher rates in the U.S. The main reasons for the higher mortality rate are ethnic differences and lack of understanding of prostate cancer biology and affordable treatments, as well as the financial burden of African American men to obtain the most effective and safe treatments. The effect of micronutrients, including Vitamin K, on various cancer cell lines has been widely studied, but the potential anticancer effect of VK3-OCH3, an analog of vitamin K3 (Menadione), on African American prostate cancer has not been evaluated. In this study, we compared the anticancer effect of VK3-OCH3 on targeting African American derived PCa cell lines namely RC77-T and MDA-PCa-2b. Our results show that VK3-OCH3 significantly inhibits the proliferation of both RC77-T and MDA-PCa-2b African American PCa cells and promotes apoptosis, and the underlying mechanism of cell death appears to be similar in both the cell lines. Notably, VK3-OCH3 inhibits colony-forming ability and induces apoptosis by blocking the cell cycle at G0 in African American PCa cells. VK3-OCH3 also acts as an anti-metastatic agent by inhibiting the migration ability of the metastatic properties of African American PCa cells. The cell death of African American PCa cells mediated by VK3-OCH3 is associated with the production of free radicals, such as intracellular and mitochondrial reactive oxygen species (ROS). Interestingly, antioxidants such as N-Acetylcysteine (NAC) and Glutathione (GSH) effectively negated the oxidative stress induced by VK3-OCH3 on PCa cell lines derived from African American patients. Of note, VK3-OCH3 reduces androgen receptor and prostate-specific antigen expression in these PCa cells. Furthermore, molecular dynamic studies reiterated that VK3-OCH3 strongly binds to the androgen receptor, suggesting that the androgen receptor is the potential molecular target of VK3-OCH3. In addition, Western blot analysis showed that VK3-OCH3 reduces the expression of androgen receptor, TRX2, and anti-apoptotic signaling molecules such as Bcl-2 and TCTP in the MDA-PCa-2b metastatic PCa cellular model. In conclusion, our results suggested that VK3-OCH3 is a promising anticancer agent that could potentially reduce the mortality rates of African American PCa patients, warranting further preclinical and translational studies.
Collapse
Affiliation(s)
- Somaiah Chinnapaka
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
| | - Velavan Bakthavachalam
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
| | | | - Jhishnuraj Kannan
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
| | - Sworaj Sapkota
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
| | - Raj Kumar
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Solan, Himachal Pradesh, India
| | - Gnanasekar Munirathinam
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
| |
Collapse
|
18
|
Dorado RG, Oceguera Nava EI, Chen G, Zhang Q, Wang G, Chen QH. Licochalcone A-Inspired Chalcones: Synthesis and Their Antiproliferative Potential in Prostate Cancer Cells. Molecules 2024; 29:6023. [PMID: 39770110 PMCID: PMC11679503 DOI: 10.3390/molecules29246023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/30/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Prostate cancer remains a significant global health concern, prompting ongoing exploration of novel therapeutic agents. Licochalcone A, a natural product in the chalcone family isolated from licorice root, is characterized by its enone structure and demonstrates antiproliferative activity in the micromolar range across various cell lines, including prostate cancer. Building on our prior success in enhancing curcumin's antiproliferative potency by replacing the substituted phenol with a 1-alkyl-1H-imizadol-2-yl moiety, we applied a similar approach to design a new class of licochalcone A-inspired chalcones. The synthesis of these target chalcones involved key [3,3]-sigmatropic rearrangement of aryl prenyl ethers and Claisen-Schmidt condensations, yielding three derivative series. These compounds were evaluated for antiproliferative activity in both androgen receptor (AR)-positive and AR-null prostate cancer cell models using WST-1 cell proliferation assay. Systematic evaluation of licochalcone A across four prostate cancer cell lines indicated a modest advantage over enzalutamide, an FDA-approved AR antagonist, in suppressing 22Rv1 cell proliferation. Interestingly, three ester derivatives by replacing the phenol next to the carbonyl with an alkoxide demonstrated similar antiproliferative potency to licochalcone A in both AR-positive and AR-negative prostate cancer cell lines. This suggests that the phenol moiety on licochalcone A may be a promising site for chemical manipulations to enhance anti-prostate cancer activity. Among the synthesized chalcones, nine derivatives showed improved selectivity for AR-positive LNCaP and 22RV1 cells relative to AR-negative PC-3 and DU145 cells, surpassing licochalcone A in selectivity. Additionally, the antiproliferative potency was highly dependent on the R group attached to the imidazole. Most of the derivatives showed antiproliferative potency against androgen receptor-positive LNCaP and 22Rv1 cells, comparable to that of enzalutamide and licochalcone A. These findings suggest that optimization of licochalcone A-inspired chalcones as potential anti-prostate cancer agents warrants further investigation.
Collapse
Affiliation(s)
- Roxana Gonzalez Dorado
- Department of Chemistry & Biochemistry, California State University, Fresno, CA 93740, USA
| | | | - Guanglin Chen
- Department of Chemistry & Biochemistry, California State University, Fresno, CA 93740, USA
| | - Qiang Zhang
- Department of Chemistry and RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Guangdi Wang
- Department of Chemistry and RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Qiao-Hong Chen
- Department of Chemistry & Biochemistry, California State University, Fresno, CA 93740, USA
| |
Collapse
|
19
|
Chen W, Mao Y, Zhan Y, Li W, Wu J, Mao X, Xu B, Shu F. Exosome-delivered NR2F1-AS1 and NR2F1 drive phenotypic transition from dormancy to proliferation in treatment-resistant prostate cancer via stabilizing hormonal receptors. J Nanobiotechnology 2024; 22:761. [PMID: 39695778 DOI: 10.1186/s12951-024-03025-y] [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: 09/18/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
Cancer cells acquire the ability to reprogram their phenotype in response to targeted therapies, yet the transition from dormancy to proliferation in drug-resistant cancers remains poorly understood. In prostate cancer, we utilized high-plasticity mouse models and enzalutamide-resistant (ENZ-R) cellular models to elucidate NR2F1 as a key factor in lineage transition and ENZ resistance. Depletion of NR2F1 drives ENZ-R cells into a relative dormancy state, characterized by reduced proliferation and heightened drug resistance, while NR2F1 overexpression yields contrasting outcomes. Transcriptional sequencing analysis of NR2F1-silenced prostate cancer cells and tissues from the Cancer Genome Atlas-prostate cancer and SU2C cohorts indicated exosomes as the most enriched cell component, with pathways implicated in steroid hormone biosynthesis and drug metabolism. Moreover, NR2F1-AS1 forms a complex with SRSF1 to upregulate NR2F1 expression, facilitating its binding with ESR1 to sustain hormonal receptor expression and enhance proliferation in ENZ-R cells. Furthermore, HnRNPA2B1 interacts with NR2F1 and NR2F1-AS1, assisting their packaging into exosomes, wherein exosomal NR2F1 and NR2F1-AS1 promote the proliferation of dormant ENZ-R cells. Our works offer novel insights into the reawaking of dormant drug-resistant cancer cells governed by NR2F1 upregulation triggered by exosome-derived NR2F1-AS1 and NR2F1, suggesting therapeutic potential for phenotype reversal.
Collapse
Affiliation(s)
- Wenbin Chen
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Yiyou Mao
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - YiYuan Zhan
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenfeng Li
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jun Wu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiangming Mao
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Bin Xu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Fangpeng Shu
- Department of Urology, Guangzhou Women and Children's Medical Center, National Children's Medical Center for South Central Region, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
20
|
Adekoya TO, Smith N, Kothari P, Dacanay MA, Li Y, Richardson RM. CXCR1 Expression in MDA-PCa-2b Cell Upregulates ITM2A to Inhibit Tumor Growth. Cancers (Basel) 2024; 16:4138. [PMID: 39766038 PMCID: PMC11674668 DOI: 10.3390/cancers16244138] [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: 11/01/2024] [Revised: 12/01/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Chemokines, along with their receptors, exert critical roles in tumor development and progression. In prostate cancer (PCa), interleukin-8 (IL-8/CXCL8) was shown to enhance angiogenesis, proliferation, and metastasis. CXCL8 activates two receptors, CXCR1 and CXCR2. While CXCR2 expression was shown to promote PCa growth and metastasis, the role of CXCR1 remains unclear. METHODS In this study, we stably expressed CXCR1 and, as control, CXCR2 in the androgen-dependent PCa cell line MDA-PCa-2b to evaluate the effect of CXCR1 in tumor development. RESULTS MDA-PCa-2b-CXCR1 cells showed decreased cell migration, protein kinase-B (AKT) activation, prostate-specific antigen (PSA) expression, cell proliferation, and tumor development in nude mice, relative to MDA-PCa-2b-Vec and MDA-PCa-2b-CXCR2 cells. MDA-PCa-2b-CXCR1 cells also displayed a significant transition to mesenchymal phenotypes as characterized by decreased E-cadherin expression and a corresponding increased level of N-cadherin and vimentin expression. RNA-seq and Western blot analysis revealed a significant increase in the tumor suppressor integral membrane protein 2A (ITM2A) expression in MDA-PCa-2b-CXCR1 compared to control cells. In prostate adenocarcinoma tissue, ITM2A expression was also shown to be downregulated relative to a normal prostate. Interestingly, the overexpression of ITM2A in MDA-PCa-2b cells (MDA-PCa-2b-ITM2A-GFP) inhibited tumor growth similar to that of MDA-PCa-2b-CXCR1. CONCLUSIONS Taken together, the data suggest that CXCR1 expression in MDA-PCa-2b cells may upregulate ITM2A to abrogate tumor development.
Collapse
Affiliation(s)
- Timothy O. Adekoya
- Cancer Research Program, Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA
| | - Nikia Smith
- Cancer Research Program, Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | - Parag Kothari
- Cancer Research Program, Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | - Monique A. Dacanay
- Cancer Research Program, Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | - Yahui Li
- Cancer Research Program, Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | - Ricardo M. Richardson
- Cancer Research Program, Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA
| |
Collapse
|
21
|
Tyagi A, Chandrasekaran B, Shukla V, Tyagi N, Sharma AK, Damodaran C. Nutraceuticals target androgen receptor-splice variants (AR-SV) to manage castration resistant prostate cancer (CRPC). Pharmacol Ther 2024; 264:108743. [PMID: 39491756 DOI: 10.1016/j.pharmthera.2024.108743] [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: 05/31/2024] [Revised: 09/25/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Every year, prostate cancer is diagnosed in millions of men. The androgen receptor's (AR) unchecked activation is crucial in causing the development and progression of prostate cancer. Second-generation anti-androgen therapies, which primarily focus on targeting the Ligand Binding Domain (LBD) of AR, are effective for most patients. However, the adverse effects pose significant challenges in managing the disease. Furthermore, genetic mutations or the emergence of AR splice variants create an even more complex tumor environment, fostering resistance to these treatments. Natural compounds and their analogs, while showing a lower toxicity profile and a potential for selective AR splice variants inhibition, are constrained by their bioavailability and therapeutic efficacy. Nonetheless, recent breakthroughs in using natural derivatives to target AR and its splice variants have shown promise in treating chemoresistant castration-resistant prostate cancer (CRPC). This review will discuss the role of AR variants, particularly androgen receptor splice variant 7 (AR-V7), in CRPC and investigate the latest findings on how natural compounds and their derivatives target AR and AR splice variants.
Collapse
Affiliation(s)
- Ashish Tyagi
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M University, College Station, TX 77845, United States
| | - Balaji Chandrasekaran
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M University, College Station, TX 77845, United States
| | - Vaibhav Shukla
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M University, College Station, TX 77845, United States
| | - Neha Tyagi
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M University, College Station, TX 77845, United States
| | - Arun K Sharma
- Department of Pharmacology, Penn State Cancer Institute, College of Medicine, Penn State University, Hershey, PA 17033, United States
| | - Chendil Damodaran
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M University, College Station, TX 77845, United States.
| |
Collapse
|
22
|
Pascual G, Sabater A, Bizzotto J, Seniuk R, Sanchis P, Ledesma-Bazan S, Labanca E, Scorticati C, Mazza O, Vazquez E, Toro A, Prada F, Gueron G, Cotignola J. AR (CAG) n Microsatellite and APEX1 c.444T>G (p.Asp148Glu) Polymorphisms as Independent Prognostic Biomarkers in Prostate Cancer: Insights from an Argentinian Cohort. Cancers (Basel) 2024; 16:3815. [PMID: 39594771 PMCID: PMC11592882 DOI: 10.3390/cancers16223815] [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: 10/10/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Prostate cancer (PCa) is the leading malignancy and the third most common cause of cancer-related death in Argentinian men. Predicting outcomes in localized PCa remains difficult due to tumor heterogeneity. In this study, we assessed the impact of AR (CAG)n and APEX1 c.444T>G polymorphisms on biochemical relapse in Argentine patients with localized PCa. METHODS We genotyped blood samples from 123 PCa patients for AR (CAG)n and APEX1 p.Asp148Glu (c.444T>G) polymorphisms. Associations with clinicopathological parameters and biochemical relapse-free survival (BRFS) were assessed. RESULTS AR (CAG)20-23 was associated with a family history of breast/ovarian cancer (p = 0.0469). The combination of AR (CAG)20-23 and APEX1 c.444TT/GG correlated with a 2.89 times higher risk of biochemical relapse (log-rank p = 0.006). Multivariable analysis confirmed AR and APEX1 polymorphisms as independent predictors of biochemical relapse (HR = 3.95, p = 0.002). In patients with PSA levels <10 ng/mL, combined AR (CAG)20-23 and APEX1 c.444TT/GG genotypes were significantly associated with an increased risk of biochemical relapse (HR = 2.61, p = 0.044). Multivariable analysis confirmed the prognostic significance of these genotypes (HR = 3.44, p = 0.02). CONCLUSIONS This study has identified AR (CAG)n and APEX1 c.444T>G polymorphisms as independent predictors of PCa relapse in Argentinian patients, suggesting their potential use in improving prognostic models.
Collapse
Affiliation(s)
- Gaston Pascual
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Agustina Sabater
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires C1073AAO, Argentina
| | - Juan Bizzotto
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires C1073AAO, Argentina
| | - Rocio Seniuk
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Pablo Sanchis
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires C1073AAO, Argentina
| | - Sabrina Ledesma-Bazan
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Estefania Labanca
- Department of Genitourinary Medical Oncology and The David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Carlos Scorticati
- Departamento de Urología, Ciudad Autónoma de Buenos Aires (CABA), Hospital de Clínicas José de San Martín, Universidad de Buenos Aires, Buenos Aires C1120AAF, Argentina
| | - Osvaldo Mazza
- Departamento de Urología, Ciudad Autónoma de Buenos Aires (CABA), Hospital de Clínicas José de San Martín, Universidad de Buenos Aires, Buenos Aires C1120AAF, Argentina
| | - Elba Vazquez
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Ayelen Toro
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Federico Prada
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires C1073AAO, Argentina
| | - Geraldine Gueron
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Javier Cotignola
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| |
Collapse
|
23
|
Israel JS, Marcelin LM, Mehralivand S, Scholze J, Hofmann J, Stope MB, Puhr M, Thomas C, Erb HHH. The impact of androgen-induced translation in modulating androgen receptor activity. Biol Direct 2024; 19:111. [PMID: 39529201 PMCID: PMC11555926 DOI: 10.1186/s13062-024-00550-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION Dysregulated androgen receptor (AR) activity is central to various diseases, particularly prostate cancer (PCa), in which it drives tumour initiation and progression. Consequently, antagonising AR activity via anti-androgens is an indispensable treatment option for metastatic PCa. However, despite initial tumour remission, drug resistance occurs. Therefore, the AR signalling pathway has been intensively investigated. However, the role of AR protein stability in AR signalling and therapy resistance has not yet been deciphered. Therefore, this study aimed to investigate the role of AR protein changes in transactivity and assess its mechanism as a possible target in PCa. METHODS LNCaP, C4-2, and 22Rv1 cells were treated with R1881, enzalutamide, cycloheximide, and Rocaglamide. Mass spectrometry analyses were performed on LNCaP cells to identify the pathways enriched by the treatments. Western blotting was performed to investigate AR protein levels and localisation changes. Changes in AR transactivity were determined by qPCR. RESULTS Mass spectrometry analyses were performed on LNCaP cells to decipher the molecular mechanisms underlying androgen- and antiandrogen-induced alterations in the AR protein. Pathway analysis revealed the enrichment of proteins involved in different pathways that regulate translation. Translational and proteasome inhibitor experiments revealed that these AR protein changes were attributable to modifications in translational activity. Interestingly, the effects on AR protein levels in castration-resistant PCa (CRPC) cells C4-2 or enzalutamide-resistant cells 22Rv1 were less prominent and non-existent. This outcome was similarly observed in the alteration of AR transactivation, which was suppressed in hormone-sensitive prostate cancer (HSPC) LNCaP cells by translational inhibition, akin to the effect of enzalutamide. In contrast, treatment-resistant cell lines showed only a slight change in AR transcription. CONCLUSION This study suggests that in HSPC, AR activation triggers a signalling cascade that increases AR protein levels by enhancing its translation rate, thereby amplifying AR activity. However, this mechanism appears to be dysregulated in castration-resistant PCa cells.
Collapse
Affiliation(s)
- Justus S Israel
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Laura-Maria Marcelin
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Sherif Mehralivand
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Jana Scholze
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Jörg Hofmann
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Matthias B Stope
- German Society of Urology, UroFors Consortium (Natural Scientists in Urological Research), 14163, Berlin, Germany
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, 53127, Bonn, Germany
| | - Martin Puhr
- Department of Urology, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Christian Thomas
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany.
- Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Technischen Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany.
| | - Holger H H Erb
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany.
- German Society of Urology, UroFors Consortium (Natural Scientists in Urological Research), 14163, Berlin, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.
- Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Technischen Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany.
| |
Collapse
|
24
|
Pujana-Vaquerizo M, Bozal-Basterra L, Carracedo A. Metabolic adaptations in prostate cancer. Br J Cancer 2024; 131:1250-1262. [PMID: 38969865 PMCID: PMC11473656 DOI: 10.1038/s41416-024-02762-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: 03/13/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 07/07/2024] Open
Abstract
Prostate cancer is one of the most commonly diagnosed cancers in men and is a major cause of cancer-related deaths worldwide. Among the molecular processes that contribute to this disease, the weight of metabolism has been placed under the limelight in recent years. Tumours exhibit metabolic adaptations to comply with their biosynthetic needs. However, metabolites also play an important role in supporting cell survival in challenging environments or remodelling the tumour microenvironment, thus being recognized as a hallmark in cancer. Prostate cancer is uniquely driven by androgen receptor signalling, and this knowledge has also influenced the paths of cancer metabolism research. This review provides a comprehensive perspective on the metabolic adaptations that support prostate cancer progression beyond androgen signalling, with a particular focus on tumour cell intrinsic and extrinsic pathways.
Collapse
Affiliation(s)
- Mikel Pujana-Vaquerizo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Laura Bozal-Basterra
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain.
| | - Arkaitz Carracedo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain.
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), 28029, Madrid, Spain.
- Traslational Prostate Cancer Research Lab, CIC bioGUNE-Basurto, Biobizkaia Health Research Institute, Baracaldo, Spain.
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Leioa, Spain.
| |
Collapse
|
25
|
Dhawale SA, Bhosle P, Mahajan S, Patil G, Gawale S, Ghodke M, Tapadiya G, Ansari A. Dual targeting in prostate cancer with phytoconstituents as a potent lead: a computational approach for novel drug discovery. J Biomol Struct Dyn 2024; 42:8906-8919. [PMID: 37649379 DOI: 10.1080/07391102.2023.2251059] [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: 05/20/2023] [Accepted: 08/10/2023] [Indexed: 09/01/2023]
Abstract
Prostate Cancer (PCa) is an abnormal cell growth within the prostate. This condition is the second most widespread malignancy in elderly males and one of the most frequently diagnosed life-threatening conditions. The Androgen receptor signaling pathway played a crucial role in the initiation and spread to increase the risk of PCa. Hence, targeting the AR receptor signaling pathway is a key strategy for a therapeutic plan for PCa. Our study focuses on recognizing potential inhibitors for dual targeting in PCa by using the in-silico approach. In this study, we target the two enzymes that are CYP17A1 (3RUK) and 5α-reductase (3G1R) responsible for PCa, with the help of phytoconstituents. The natural plant contains various phytochemical types produced from secondary metabolites and used as a medical treatment. The in-silico investigation of phytoconstituents and enzymes was done by approaching molecular docking, ADMET analysis, and high-level molecular dynamic simulation used to assess the stability and binding affinities of the protein-ligand complex. Some phytoconstituents, such as Peonidin, Pelargonidin, Malvidin and Berberine show complex has good molecular interaction with protein. The reliability of the docking scores was examined using a molecular dynamic simulation, which revealed that the complex remained stable throughout the simulation, which ranged from 0 to 200 ns. The selected hits may be effective against CYP17A1 (3RUK) and 5α-reductase (3G1R) (PCa) using a computer-aided drug design (CADD) method, which further enables researchers for upcoming in-vivo and in-vitro research, according to our in-silico approach.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sachin A Dhawale
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad, India
| | - Pallavi Bhosle
- Pharmacology, Shrinath College of Pharmacy, Aurangabad, India
| | | | - Geetanjali Patil
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad, India
| | - Sachin Gawale
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad, India
| | - Mangesh Ghodke
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad, India
| | - Ganesh Tapadiya
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad, India
| | | |
Collapse
|
26
|
Patke R, Harris AE, Woodcock CL, Thompson R, Santos R, Kumari A, Allegrucci C, Archer N, Gudas LJ, Robinson BD, Persson JL, Fray R, Jeyapalan J, Rutland CS, Rakha E, Madhusudan S, Emes RD, Muyangwa-Semenova M, Alsaleem M, de Brot S, Green W, Ratan H, Mongan NP, Lothion-Roy J. Epitranscriptomic mechanisms of androgen signalling and prostate cancer. Neoplasia 2024; 56:101032. [PMID: 39033689 PMCID: PMC11295630 DOI: 10.1016/j.neo.2024.101032] [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: 05/31/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Prostate cancer (PCa) is the second most common cancer diagnosed in men. While radical prostatectomy and radiotherapy are often successful in treating localised disease, post-treatment recurrence is common. As the androgen receptor (AR) and androgen hormones play an essential role in prostate carcinogenesis and progression, androgen deprivation therapy (ADT) is often used to deprive PCa cells of the pro-proliferative effect of androgens. ADTs act by either blocking androgen biosynthesis (e.g. abiraterone) or blocking AR function (e.g. bicalutamide, enzalutamide, apalutamide, darolutamide). ADT is often effective in initially suppressing PCa growth and progression, yet emergence of castrate-resistant PCa and progression to neuroendocrine-like PCa following ADT are major clinical challenges. For this reason, there is an urgent need to identify novel approaches to modulate androgen signalling to impede PCa progression whilst also preventing or delaying therapy resistance. The mechanistic convergence of androgen and epitranscriptomic signalling offers a potential novel approach to treat PCa. The epitranscriptome involves covalent modifications of mRNA, notably, in the context of this review, the N(6)-methyladenosine (m6A) modification. m6A is involved in the regulation of mRNA splicing, stability, and translation, and has recently been shown to play a role in PCa and androgen signalling. The m6A modification is dynamically regulated by the METTL3-containing methyltransferase complex, and the FTO and ALKBH5 RNA demethylases. Given the need for novel approaches to treat PCa, there is significant interest in new therapies that target m6A that modulate AR expression and androgen signalling. This review critically summarises the potential benefit of such epitranscriptomic therapies for PCa patients.
Collapse
Affiliation(s)
- Rodhan Patke
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Anna E Harris
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Corinne L Woodcock
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Rachel Thompson
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Rute Santos
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Amber Kumari
- Biodiscovery Institute, University of Nottingham, UK
| | - Cinzia Allegrucci
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Nathan Archer
- School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Brian D Robinson
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Jenny L Persson
- Department of Molecular Biology, Umea University, Umea, Sweden
| | - Rupert Fray
- School of Biosciences, University of Nottingham, UK
| | - Jennie Jeyapalan
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Catrin S Rutland
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Emad Rakha
- School of Medicine, University of Nottingham, UK; Nottingham University NHS Trust, Nottingham, UK
| | - Srinivasan Madhusudan
- School of Medicine, University of Nottingham, UK; Nottingham University NHS Trust, Nottingham, UK
| | - Richard D Emes
- Research and Innovation, Nottingham Trent University, UK
| | | | - Mansour Alsaleem
- Biodiscovery Institute, University of Nottingham, UK; Unit of Scientific Research, Applied College, Qassim University, Qassim, Saudi Arabia
| | - Simone de Brot
- Institute of Animal Pathology, University of Bern, Switzerland
| | - William Green
- Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Hari Ratan
- Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Nigel P Mongan
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK; Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA.
| | - Jennifer Lothion-Roy
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK.
| |
Collapse
|
27
|
Parolia A, Eyunni S, Verma BK, Young E, Liu Y, Liu L, George J, Aras S, Das CK, Mannan R, Ur Rasool R, Mitchell-Velasquez E, Mahapatra S, Luo J, Carson SE, Xiao L, Gajjala PR, Venkatesh S, Jaber M, Wang X, He T, Qiao Y, Pang M, Zhang Y, Tien JCY, Louw M, Alhusayan M, Cao X, Su F, Tavana O, Hou C, Wang Z, Ding K, Chinnaiyan AM, Asangani IA. NSD2 is a requisite subunit of the AR/FOXA1 neo-enhanceosome in promoting prostate tumorigenesis. Nat Genet 2024; 56:2132-2143. [PMID: 39251788 PMCID: PMC11525188 DOI: 10.1038/s41588-024-01893-6] [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: 10/31/2023] [Accepted: 08/01/2024] [Indexed: 09/11/2024]
Abstract
Androgen receptor (AR) is a ligand-responsive transcription factor that drives terminal differentiation of the prostatic luminal epithelia. By contrast, in tumors originating from these cells, AR chromatin occupancy is extensively reprogrammed to activate malignant phenotypes, the molecular mechanisms of which remain unknown. Here, we show that tumor-specific AR enhancers are critically reliant on H3K36 dimethyltransferase activity of NSD2. NSD2 expression is abnormally induced in prostate cancer, where its inactivation impairs AR transactivation potential by disrupting over 65% of its cistrome. NSD2-dependent AR sites distinctively harbor the chimeric FOXA1:AR half-motif, which exclusively comprise tumor-specific AR enhancer circuitries defined from patient specimens. NSD2 inactivation also engenders increased dependency on the NSD1 paralog, and a dual NSD1/2 PROTAC degrader is preferentially cytotoxic in AR-dependent prostate cancer models. Altogether, we characterize NSD2 as an essential AR neo-enhanceosome subunit that enables its oncogenic activity, and position NSD1/2 as viable co-targets in advanced prostate cancer.
Collapse
Affiliation(s)
- Abhijit Parolia
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
- Department of Urology, University of Michigan, Ann Arbor, MI, USA.
| | - Sanjana Eyunni
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Molecular and Cellular Pathology Program, University of Michigan, Ann Arbor, MI, USA
| | - Brijesh Kumar Verma
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eleanor Young
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yihan Liu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Cancer Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Lianchao Liu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - James George
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Shweta Aras
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chandan Kanta Das
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rahul Mannan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Reyaz Ur Rasool
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erick Mitchell-Velasquez
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Somnath Mahapatra
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Jie Luo
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Sandra E Carson
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Lanbo Xiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Prathibha R Gajjala
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Sharan Venkatesh
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mustapha Jaber
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Xiaoju Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Tongchen He
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew Pang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuping Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Jean Ching-Yi Tien
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Micheala Louw
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Mohammed Alhusayan
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| | - Fengyun Su
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Omid Tavana
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, MA, USA
| | - Caiyun Hou
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zhen Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
- Department of Urology, University of Michigan, Ann Arbor, MI, USA.
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA.
| | - Irfan A Asangani
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
28
|
Li Q, Wang Y, Chen J, Zeng K, Wang C, Guo X, Hu Z, Hu J, Liu B, Xiao J, Zhou P. Machine learning based androgen receptor regulatory gene-related random forest survival model for precise treatment decision in prostate cancer. Heliyon 2024; 10:e37256. [PMID: 39296076 PMCID: PMC11407950 DOI: 10.1016/j.heliyon.2024.e37256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
Background It has been demonstrated that aberrant androgen receptor (AR) signaling contributes to the pathogenesis of prostate cancer (PCa). To date, the most efficacious strategy for the treatment of PCa remains to target the AR signaling axis. However, numerous PCa patients still face the issue of overtreatment or undertreatment. The establishment of a precise risk prediction model is urgently needed to distinguish patients with high-risk and select appropriate treatment modalities. Methods In this study, a consensus AR regulatory gene-related signature (ARS) was developed by integrating a total of 101 algorithm combinations of 10 machine learning algorithms. We evaluated the value of ARS in predicting patient prognosis and the therapeutic effects of the various treatments. Additionally, we conducted a screening of therapeutic targets and agents for high-risk patients, followed by the verification in vitro and in vivo. Results ARS was an independent risk factor for biochemical recurrence and distant metastasis in PCa patients. The enhanced and consistent prognostic predictive capability of ARS across various platforms was confirmed when compared with 44 previously published signatures. More importantly, PCa patients in the ARShigh group benefit more from PARP inhibitors and immunotherapy, while chemotherapy, radiotherapy, and AR-targeted therapy are more effective for ARSlow patients. The results of in silico screening suggest that AURKB could potentially serve as a promising therapeutic target for ARShigh patients. Conclusions Collectively, this prediction model based on AR regulatory genes holds great clinical translational potential to solve the dilemma of treatment choice and identify potential novel therapeutic targets in PCa.
Collapse
Affiliation(s)
- Qinyu Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yanan Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Junjie Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Kai Zeng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chengwei Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Xiangdong Guo
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhiquan Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jia Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jun Xiao
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Peng Zhou
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
29
|
Le M, Lu W, Tan X, Luo B, Yu T, Sun Y, Guo Z, Huang P, Zhu D, Wu Q, Ganesan A, Wen S. Design, Synthesis, and Biological Evaluation of Potent EZH2/LSD1 Dual Inhibitors for Prostate Cancer. J Med Chem 2024; 67:15586-15605. [PMID: 39196854 DOI: 10.1021/acs.jmedchem.4c01250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
As histone modification enzymes, EZH2 mediates H3K27 trimethylation (H3K27me3), whereas LSD1 removes methyl groups from H3K4me1/2 and H3K9me1/2. Synergistic anticancer effects of combining inhibitors of these two enzymes are observed in leukemia and prostate cancer. Thus, a series of EZH2/LSD1 dual inhibitors are designed and synthesized to evaluate their anticancer activity. After the structure-activity study, one of the best compounds, ML234, displayed excellent antiproliferative capacity against prostate cancer cell lines LNCAP, PC3, and 22RV1. Enzymatic assays ascertained that the anticancer potency of ML234 was mediated through coinhibition of EZH2 and LSD1. Moreover, the accumulation of H3K4me2 and H3K9me2 and the decrease of H3K27me3 induced by ML234 were verified by Western blot analysis. More importantly, the compound remarkably suppressed the tumor growth and enhanced the therapeutic efficacy of clinical drug enzalutamide in the 22RV1 xenograft mouse model, indicating that it may have potential as an anticancer agent in prostate cancer.
Collapse
Affiliation(s)
- Meiling Le
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Wenhua Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xiaozhuo Tan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Bingling Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Tiantian Yu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yameng Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zhirong Guo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Peng Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Daqian Zhu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Qiang Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa 999078, Macau
| | - A Ganesan
- School of Chemistry, Pharmacy & Pharmacology, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| | - Shijun Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
30
|
Jeon J, Ha JS, Lee HS, Jeon S, Hwang HS, Kim D, Kim JS, Kim BS, Kim M, Cho KS. Androgen Deprivation Therapy and the Risk of Newly Developed Dry Eye Syndrome in Patients with Prostate Cancer: A Nationwide Nested Case-Control Study in the Republic of Korea. J Clin Med 2024; 13:5314. [PMID: 39274527 PMCID: PMC11395731 DOI: 10.3390/jcm13175314] [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: 07/11/2024] [Revised: 08/21/2024] [Accepted: 09/06/2024] [Indexed: 09/16/2024] Open
Abstract
Background: We aimed to evaluate the association between androgen deprivation therapy (ADT) and newly developed dry eye syndrome (DES) in patients with prostate cancer. Methods: A nested case-control study was conducted. From the nationwide claims database of the Republic of Korea, 125,005 patients were included in the final analysis. Cases were defined as those newly diagnosed with DES during follow-up, and 12,654 patients were identified. The cases were matched with controls in a ratio of 1:4. Odds ratios (ORs) for newly developed DES associated with ADT were estimated using conditional logistic regression. Results: After matching, 7499 cases and 29,996 controls were selected. ADT was associated with a reduced risk of newly developed DES in patients with prostate cancer compared to no ADT (OR = 0.875; 95% confidence interval, 0.825-0.927; p < 0.0001). An accumulated dose of ADT < 1 year was associated with a reduced risk of incidental DES (OR = 0.811; 95% CI, 0.751-0.875; p < 0.0001), and a duration of 1-2 years was also associated with a reduced risk (OR = 0.890; 95% CI, 0.802-0.986; p = 0.026). No association was observed with an ADT duration of ≥2 years. Conclusions: The use of ADT, especially for shorter durations (<2 years), was associated with a reduced risk of newly developed DES in S. Korean patients with prostate cancer.
Collapse
Affiliation(s)
- Jinhyung Jeon
- Department of Urology, Prostate Cancer Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jee Soo Ha
- Department of Urology, Seoul Hyocheon Foundation Medical Corporation H Plus YangJi Hospital, Seoul 08779, Republic of Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Soyoung Jeon
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ho Sik Hwang
- Department of Ophthalmology, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Daeho Kim
- Department of Urology, Prostate Cancer Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - June Seok Kim
- Department of Urology, Prostate Cancer Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Byeong Seon Kim
- Department of Urology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Min Kim
- Department of Ophthalmology, Institute of Vision Research, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Kang Su Cho
- Department of Urology, Prostate Cancer Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Center of Evidence Based Medicine, Institute of Convergence Science, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
31
|
Lee S, Kim HR, Woo Y, Kim J, Kim HW, Park JY, Suh B, Choi Y, Ahn J, Ryu JH, Roe JS, Song J, Lee SH. UBX-390: A Novel Androgen Receptor Degrader for Therapeutic Intervention in Prostate Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400398. [PMID: 38958553 PMCID: PMC11434238 DOI: 10.1002/advs.202400398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/18/2024] [Indexed: 07/04/2024]
Abstract
The androgen receptor (AR) is an attractive target for treating prostate cancer, considering its role in the development and progression of localized and metastatic prostate cancer. The high global mortality burden of prostate cancer, despite medical treatments such as androgen deprivation or AR antagonist therapy, highlights the need to explore alternative strategies. One strategy involves the use of heterobifunctional degraders, also known as proteolysis-targeting chimeras, which are novel small-molecule therapeutics that inhibit amplified or mutated targets. Here, the study reports a novel cereblon-based AR degrader, UBX-390, and demonstrates its superior activity over established AR degraders, such as ARV-110 or ARCC-4, in prostate cancer cells under short- and long-term treatment conditions. UBX-390 suppresses chromatin binding and gene expression of AR and demonstrates substantial efficacy in the degradation of AR mutants in patients with treatment-resistant prostate cancer. UBX-390 is presented as an optimized AR degrader with remarkable potential for treating castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Soohyun Lee
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hwa-Ryeon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yaejin Woo
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Jiyoung Kim
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Han Wool Kim
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Ji Youn Park
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Beomseon Suh
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Yuri Choi
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Jungmin Ahn
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Je Ho Ryu
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Jae-Seok Roe
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jaewhan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Song Hee Lee
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| |
Collapse
|
32
|
Li H, Fan S, Gong Z, Chan JYK, Tong MCF, Chen GG. Role of hematological and neurological expressed 1 (HN1) in human cancers. Crit Rev Oncol Hematol 2024; 201:104446. [PMID: 38992849 DOI: 10.1016/j.critrevonc.2024.104446] [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: 06/03/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024] Open
Abstract
Hematological and neurological expressed 1 (HN1), also known as Jupiter microtubule associated homolog 1 (JPT1), is a highly conserved protein with widespread expression in various tissues. Ectopic elevation of HN1 has been observed in multiple cancers, highlighting its role in tumorigenesis and progression. Both proteomics and transcriptomics reveal that HN1 is closely associated with severe disease progression, poor prognostic and shorter overall survival. HN1's involvement in cancer cell proliferation and metastasis has been extensively investigated. Overexpression of HN1 is associated with increased tumor growth and disease progression, while its depletion leads to cell cycle arrest and apoptosis. The pivotal role of HN1 in cancer progression, particularly in proliferation, migration, and invasion, underscores its significance in cancer metastasis. Validation of the efficacy and safety of HN1 inhibition, along with the development of diagnostic methods to determine HN1 expression levels in patients, is essential for the translation of HN1-targeted therapies into clinical practice. Overall, HN1 emerges as a valuable prognostic marker and therapeutic target in cancer, and further investigations hold the potential to improve patient outcomes by impeding metastasis and enhancing treatment strategies.
Collapse
Affiliation(s)
- Huangcan Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - Simiao Fan
- Department of Otorhinolaryngology, Head and Neck Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - Zhongqin Gong
- Department of Otorhinolaryngology, Head and Neck Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - Jason Ying Kuen Chan
- Department of Otorhinolaryngology, Head and Neck Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - Michael Chi Fai Tong
- Department of Otorhinolaryngology, Head and Neck Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - George Gong Chen
- Department of Otorhinolaryngology, Head and Neck Surgery, Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
33
|
Thakur N, Singh P, Bagri A, Srivastava S, Dwivedi V, Singh A, Jaiswal SK, Dholpuria S. Therapy resistance in prostate cancer: mechanism, signaling and reversal strategies. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1110-1134. [PMID: 39351434 PMCID: PMC11438573 DOI: 10.37349/etat.2024.00266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/09/2024] [Indexed: 10/04/2024] Open
Abstract
Prostate cancer (PC) depicts a major health challenge all over the globe due to its complexities in the treatment and diverse clinical trajectories. Even in the advances in the modern treatment strategies, the spectrum of resistance to the therapies continues to be a significant challenge. This review comprehensively examines the underlying mechanisms of the therapy resistance occurred in PC, focusing on both the tumor microenvironment and the signaling pathways implicated in the resistance. Tumor microenvironment comprises of stromal and epithelial cells, which influences tumor growth, response to therapy and progression. Mechanisms such as microenvironmental epithelial-mesenchymal transition (EMT), anoikis suppression and stimulation of angiogenesis results in therapy resistance. Moreover, dysregulation of signaling pathways including androgen receptor (AR), mammalian target of rapamycin/phosphoinositide 3 kinase/AKT (mTOR/PI3K/AKT), DNA damage repair and Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathways drive therapy resistance by promoting tumor survival and proliferation. Understanding these molecular pathways is important for developing targeted therapeutic interventions which overcomes resistance. In conclusion, a complete grasp of mechanisms and pathways underlying medication resistance in PC is important for the development of individualized treatment plans and enhancements of clinical outcomes. By studying and understanding the complex mechanisms of signaling pathways and microenvironmental factors contributing to therapy resistance, this study focuses and aims to guide the development of innovative therapeutic approaches to effectively overcome the PC progression and improve the survival rate of patients.
Collapse
Affiliation(s)
- Neha Thakur
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Pallavi Singh
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Aditi Bagri
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Saumya Srivastava
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Vinay Dwivedi
- Amity Institute of Biotechnology, Amity University, Gwalior, Madhya Pradesh 474005, India
| | - Asha Singh
- Amity Institute of Biotechnology, Amity University, Gwalior, Madhya Pradesh 474005, India
| | - Sunil Kumar Jaiswal
- School of Biological and Life Sciences, Galgotias University, Greater Noida, Uttar Pradesh 203201, India
| | - Sunny Dholpuria
- Department of Life Sciences, J. C. Bose University of Science and Technology, YMCA Faridabad, Faridabad, Haryana 121006, India
| |
Collapse
|
34
|
Hoffmann TJ, Graff RE, Madduri RK, Rodriguez AA, Cario CL, Feng K, Jiang Y, Wang A, Klein RJ, Pierce BL, Eggener S, Tong L, Blot W, Long J, Goss LB, Darst BF, Rebbeck T, Lachance J, Andrews C, Adebiyi AO, Adusei B, Aisuodionoe-Shadrach OI, Fernandez PW, Jalloh M, Janivara R, Chen WC, Mensah JE, Agalliu I, Berndt SI, Shelley JP, Schaffer K, Machiela MJ, Freedman ND, Huang WY, Li SA, Goodman PJ, Till C, Thompson I, Lilja H, Ranatunga DK, Presti J, Van Den Eeden SK, Chanock SJ, Mosley JD, Conti DV, Haiman CA, Justice AC, Kachuri L, Witte JS. Genome-wide association study of prostate-specific antigen levels in 392,522 men identifies new loci and improves cross-ancestry prediction. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.10.27.23297676. [PMID: 37961155 PMCID: PMC10635224 DOI: 10.1101/2023.10.27.23297676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
We conducted a multi-ancestry genome-wide association study of prostate-specific antigen (PSA) levels in 296,754 men (211,342 European ancestry; 58,236 African ancestry; 23,546 Hispanic/Latino; 3,630 Asian ancestry; 96.5% of participants were from the Million Veteran Program). We identified 318 independent genome-wide significant (p≤5e-8) variants, 184 of which were novel. Most demonstrated evidence of replication in an independent cohort (n=95,768). Meta-analyzing discovery and replication (n=392,522) identified 447 variants, of which a further 111 were novel. Out-of-sample variance in PSA explained by our genome-wide polygenic risk scores ranged from 11.6%-16.6% in European ancestry, 5.5%-9.5% in African ancestry, 13.5%-18.2% in Hispanic/Latino, and 8.6%-15.3% in Asian ancestry, and decreased with increasing age. Mid-life genetically-adjusted PSA levels were more strongly associated with overall and aggressive prostate cancer than unadjusted PSA. Our study highlights how including proportionally more participants from underrepresented populations improves genetic prediction of PSA levels, offering potential to personalize prostate cancer screening.
Collapse
|
35
|
Eleazar EG, Carrera ARM, Quiambao JIR, Caparanga AR, Tayo LL. QSTR Models in Dioxins and Dioxin-like Compounds Provide Insights into Gene Expression Dysregulation. TOXICS 2024; 12:597. [PMID: 39195699 PMCID: PMC11359467 DOI: 10.3390/toxics12080597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/11/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024]
Abstract
Polychlorinated dibenzo-p-dioxins and polychlorinated dibenzo-p-furans (PCDD/Fs) are a group of organic chemicals containing three-ring structures that can be substituted with one to eight chlorine atoms, leading to 75 dioxin and 135 furan congeners. As endocrine-disrupting chemicals (EDCs), they can alter physiological processes causing a number of disorders. In this study, quantitative structure-toxicity relationship (QSTR) studies were used to determine the correlations between the PCDD/Fs' molecular structures and various toxicity endpoints. Strong QSTR models, with the coefficients of determination (r2) values greater than 0.95 and ANOVA p-values less than 0.0001 were established between molecular descriptors and the endpoints of bioconcentration, fathead minnow LC50, and Daphnia magna LC50. The ability of PCDD/Fs to bind to several nuclear receptors was investigated via molecular docking studies. The results show comparable, and in some instances better, binding affinities of PCDD/Fs toward the receptors relative to their natural agonistic and antagonistic ligands, signifying possible interference with the receptors' natural biological activities. These studies were accompanied by the molecular dynamics simulations of the top-binding PCDD/Fs to show changes in the receptor-ligand complexes during binding and provide insights into these compounds' ability to interfere with transcription and thereby modify gene expression. This introspection of PCDD/Fs at the molecular level provides a deeper understanding of these compounds' toxicity and opens avenues for future studies.
Collapse
Affiliation(s)
- Elisa G. Eleazar
- School of Graduate Studies, Mapua University, Manila 1002, Philippines; (E.G.E.); (A.R.M.C.); (A.R.C.)
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapua University, Manila 1002, Philippines;
| | - Andrei Raphael M. Carrera
- School of Graduate Studies, Mapua University, Manila 1002, Philippines; (E.G.E.); (A.R.M.C.); (A.R.C.)
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapua University, Manila 1002, Philippines;
| | - Janus Isaiah R. Quiambao
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapua University, Manila 1002, Philippines;
| | - Alvin R. Caparanga
- School of Graduate Studies, Mapua University, Manila 1002, Philippines; (E.G.E.); (A.R.M.C.); (A.R.C.)
| | - Lemmuel L. Tayo
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapua University, Manila 1002, Philippines;
- Department of Biology, School of Health Sciences, Mapua University, Makati 1200, Philippines
| |
Collapse
|
36
|
Li M, Zhong K, He G, Yin Y. Changes in immunophenotypes after neoadjuvant endocrine therapy for prostate cancer and their clinical significance. Heliyon 2024; 10:e34864. [PMID: 39170268 PMCID: PMC11336308 DOI: 10.1016/j.heliyon.2024.e34864] [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: 01/18/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024] Open
Abstract
Background To investigate changes in the immunophenotypes of androgen receptor (AR), prostate-specific antigen (PSA), synaptophysin (Syn), chromogranin A (CgA), p53 and Ki-67 after neoadjuvant endocrine therapy (NET) for prostate cancer (PCa) and to analyze their clinical significance. Methods Paired paraffin samples were collected from 40 PCa patients before and after NET, and immunohistochemistry were used to detect AR, PSA, Syn, CgA, p53 and Ki-67 expression. Based on The Cancer Genome Atlas (TCGA), Kaplan‒Meier survival curves were plotted for analysis of PSA and Ki-67 expression in relation to progression-free survival (PFS). Results After NET, the mean scores for PSA and Ki-67 expression in PCa patients were lower than those before NET (P < 0.05), while the mean scores for Syn and CgA expression were higher than those before NET (P < 0.05). The mean Gleason score and WHO/ISUP (World Health Organization/International Society of Urological Pathology) grade after NET were lower than those before NET (P < 0.05). In PCa patients who had not yet received NET, PSA expression correlated positively with Gleason score and WHO/ISUP grade and negatively with Ki-67 expression (P < 0.05); p53 expression correlated negatively with Gleason score and WHO/ISUP grade (P < 0.05). TCGA showed that PFS was lower in PCa patients with high PSA and Ki-67 expression (P < 0.05). Conclusions PSA and Ki-67 protein expressions decreased significantly in PCa patients after NET and can be used as biological markers for prognostic assessment of PCa patients. NETs may induce a neuroendocrine (NE) phenotype in PCa. Monitoring the immunophenotypes of PCa patients after NET may inform assessment of efficacy and prognosis.
Collapse
Affiliation(s)
- Mei Li
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, PR China
- Department of Pathology, NO.2 People’ s Hospital of Fuyang City, Fuyang, Anhui, 236015, PR China
| | - Kun Zhong
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, PR China
| | - Guifang He
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, PR China
| | - Yu Yin
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, PR China
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, PR China
| |
Collapse
|
37
|
Wei L, Mei D, Hu S, Du S. Dual-target EZH2 inhibitor: latest advances in medicinal chemistry. Future Med Chem 2024; 16:1561-1582. [PMID: 39082677 PMCID: PMC11370917 DOI: 10.1080/17568919.2024.2380243] [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: 02/27/2024] [Accepted: 07/09/2024] [Indexed: 09/03/2024] Open
Abstract
Enhancer of zeste homolog 2 (EZH2), a histone methyltransferase, plays a crucial role in tumor progression by regulating gene expression. EZH2 inhibitors have emerged as promising anti-tumor agents due to their potential in cancer treatment strategies. However, single-target inhibitors often face limitations such as drug resistance and side effects. Dual-target inhibitors, exemplified by EZH1/2 inhibitor HH-2853(28), offer enhanced efficacy and reduced adverse effects. This review highlights recent advancements in dual inhibitors targeting EZH2 and other proteins like BRD4, PARP1, and EHMT2, emphasizing rational design, structure-activity relationships, and safety profiles, suggesting their potential in clinical applications.
Collapse
Affiliation(s)
- Lai Wei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Department of Orthodontics, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Dan Mei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Department of Orthodontics, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Sijia Hu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Department of Orthodontics, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Shufang Du
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Department of Orthodontics, Sichuan University, Chengdu, 610041, Sichuan, China
| |
Collapse
|
38
|
Huang B, Deng X, Zhou G, Li K, Feng Y, Xie G, Liu R, Song L, Huang Z, Jia Z. SYT4 binds to SNAP25 to facilitate exosomal secretion and prostate cancer enzalutamide resistance. Cancer Sci 2024; 115:2630-2645. [PMID: 38889208 PMCID: PMC11309949 DOI: 10.1111/cas.16239] [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: 01/12/2024] [Revised: 05/09/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Abstract
Prostate carcinoma represents a predominant malignancy affecting the male population, with androgen deprivation therapy (ADT) serving as a critical therapeutic modality for advanced disease states, but it often leads to the development of resistance. Enzalutamide (Enz), a second-generation antiandrogen drug, initially offers substantial therapeutic benefit, but its efficacy wanes as drug resistance ensues. In this study, we found that synaptotagmin 4 (SYT4) is an upregulated gene in enzalutamide-resistant (EnzR) cell lines. The downregulation of SYT4, in combination with enzalutamide therapy, substantially enhances the antiproliferative effect on resistant prostate cancer cells beyond the capacity of enzalutamide monotherapy. SYT4 promotes vesicle efflux by binding to the synaptosome-associated protein 25 (SNAP25), thereby contributing to cell resistance against enzalutamide. The elevated expression of SYT4 is mediated by bromodomain-containing protein 4 (BRD4), and BRD4 inhibition effectively suppressed the expression of SYT4. Treatment with a therapeutic dose of enzalutamide combined with ASO-1, an antisense oligonucleotide drug targeting SYT4, shows promising results in reversing the resistance of prostate cancer to enzalutamide.
Collapse
Affiliation(s)
- Budeng Huang
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xiyue Deng
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guochao Zhou
- The 947th Army Hospital of the Chinese PLAKashgarChina
| | - Keqiang Li
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yuankang Feng
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guoqing Xie
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Ruoyang Liu
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Liang Song
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhenlin Huang
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhankui Jia
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
39
|
Hinostroza F, Araya-Duran I, Piñeiro A, Lobos I, Pastenes L. Transcription factor roles in the local adaptation to temperature in the Andean Spiny Toad Rhinella spinulosa. Sci Rep 2024; 14:15158. [PMID: 38956427 PMCID: PMC11220030 DOI: 10.1038/s41598-024-66127-5] [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: 05/31/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024] Open
Abstract
Environmental temperature strongly influences the adaptation dynamics of amphibians, whose limited regulation capabilities render them susceptible to thermal oscillations. A central element of the adaptive strategies is the transcription factors (TFs), which act as master regulators that orchestrate stress responses, enabling species to navigate the fluctuations of their environment skillfully. Our study delves into the intricate relationship between TF expression and thermal adaptation mechanisms in the Rhinella spinulosa populations. We sought to elucidate the dynamic modulations of TF expression in prometamorphic and metamorphic tadpoles that inhabit two thermally contrasting environments (Catarpe and El Tatio Geyser, Chile) and which were exposed to two thermal treatments (25 °C vs. 20 °C). Our findings unravel an intriguing dichotomy in response strategies between these populations. First, results evidence the expression of 1374 transcription factors. Regarding the temperature shift, the Catarpe tadpoles show a multifaceted approach by up-regulating crucial TFs, including fosB, atf7, and the androgen receptor. These dynamic regulatory responses likely underpin the population's ability to navigate thermal fluctuations effectively. In stark contrast, the El Tatio tadpoles exhibit a more targeted response, primarily up-regulating foxc1. This differential expression suggests a distinct focus on specific TFs to mitigate the effects of temperature variations. Our study contributes to understanding the molecular mechanisms governing thermal adaptation responses and highlights the resilience and adaptability of amphibians in the face of ever-changing environmental conditions.
Collapse
Affiliation(s)
- Fernando Hinostroza
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, Chile
- Centro de Investigación en Neuropsicología y Neurociencias Cognitivas, Facultad de Ciencias de la Salud, Universidad Católica del Maule, Talca, Chile
- Escuela de Química y Farmacia, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca, Chile
- Centro Para la Investigación Traslacional en Neurofarmacología, Universidad de Valparaíso, Valparaíso, Chile
| | - Ingrid Araya-Duran
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Alejandro Piñeiro
- Laboratorio de Genética y Microevolución, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile
| | - Isabel Lobos
- Laboratorio de Genética y Microevolución, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile
| | - Luis Pastenes
- Laboratorio de Genética y Microevolución, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile.
| |
Collapse
|
40
|
Singh G, Trehan S, Singh A, Goswami K, Farooq A, Antil P, Puri P, Bector G, Jain A, Azhar W. Aryl Hydrocarbon Receptor Signaling in Prostate Cancer Therapy: A Review of Implications for Anti-androgen Treatment Strategies and Resistance. Cureus 2024; 16:e65247. [PMID: 39184676 PMCID: PMC11342139 DOI: 10.7759/cureus.65247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2024] [Indexed: 08/27/2024] Open
Abstract
Prostate cancer is a leading cause of cancer-related morbidity and mortality in men, frequently exhibiting resistance to conventional anti-androgen therapies. This review investigates the emerging significance of the aryl hydrocarbon receptor (AhR) in prostate cancer, focusing on its role in modulating androgen receptor (AR) signaling and its potential as a therapeutic target. AhR, traditionally known for detoxifying harmful compounds, has been increasingly recognized for its dual capacity to either enhance or inhibit AR activity based on cellular context and specific coactivators. Furthermore, AhR influences tumor progression independently of AR by regulating genes involved in cell cycle control and apoptosis. This narrative review synthesizes current research on AhR's multifaceted roles in prostate cancer, evaluates its potential as a biomarker, and discusses the therapeutic implications of targeting AhR, particularly for hormone-refractory prostate cancer. Our findings underscore the necessity for personalized AhR-targeted therapies and advocate for continued clinical research to fully leverage AhR's therapeutic potential.
Collapse
Affiliation(s)
- Gurjot Singh
- Internal Medicine, Southern Illinois University School of Medicine, Springfield, USA
| | - Shubam Trehan
- Internal Medicine, Southern Illinois University School of Medicine, Springfield, USA
| | - Adarshpreet Singh
- Internal Medicine, Southern Illinois University School of Medicine, Springfield, USA
| | - Kanishka Goswami
- Internal Medicine, Southern Illinois University School of Medicine, Springfield, USA
| | - Amna Farooq
- Internal Medicine, Southern Illinois University School of Medicine, Springfield, USA
| | - Priya Antil
- Internal Medicine, Southern Illinois University School of Medicine, Springfield, USA
| | - Piyush Puri
- Internal Medicine, Southern Illinois University School of Medicine, Springfield, USA
| | - Gaurav Bector
- Internal Medicine, Southern Illinois University School of Medicine, Springfield, USA
| | - Aayush Jain
- Internal Medicine, Southern Illinois University School of Medicine, Springfield, USA
| | - Waqas Azhar
- Internal Medicine, Memorial Medical Center, Springfield, USA
- Internal Medicine, Saint John Hospital, Springfield, USA
- Internal Medicine, Southern Illinois University School of Medicine, Springfield, USA
- Hospital Medicine, Springfield Clinic, Springfield, USA
| |
Collapse
|
41
|
Shahi A, Kidane D. Starving cancer cells to enhances DNA damage and immunotherapy response. Oncotarget 2024; 15:392-399. [PMID: 38900609 PMCID: PMC11197973 DOI: 10.18632/oncotarget.28595] [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: 05/10/2024] [Accepted: 06/05/2024] [Indexed: 06/22/2024] Open
Abstract
Prostate cancer (PCa) poses significant challenges in treatment, particularly when it progresses to a metastatic, castrate-resistant state. Conventional therapies, including chemotherapy, radiotherapy, and hormonal treatments, often fail due to toxicities, off-target effects, and acquired resistance. This research perspective defines an alternative therapeutic strategy focusing on the metabolic vulnerabilities of PCa cells, specifically their reliance on non-essential amino acids such as cysteine. Using an engineered enzyme cyst(e)inase to deplete the cysteine/cystine can induce oxidative stress and DNA damage in cancer cells. This depletion elevates reactive oxygen species (ROS) levels, disrupts glutathione synthesis, and enhances DNA damage, leading to cancer cell death. The combinatorial use of cyst(e)inase with agents targeting antioxidant defenses, such as thioredoxins, further amplifies ROS accumulation and cytotoxicity in PCa cells. Overall, in this perspective provides a compressive overview of the previous work on manipulating amino acid metabolism and redox balance modulate the efficacy of DNA repair-targeted and immune checkpoint blockade therapies in prostate cancer.
Collapse
Affiliation(s)
- Aashirwad Shahi
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC 20059, USA
| | - Dawit Kidane
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC 20059, USA
| |
Collapse
|
42
|
Ciaccia PN, Liang Z, Schweitzer AY, Metzner E, Isaacs FJ. Enhanced eMAGE applied to identify genetic factors of nuclear hormone receptor dysfunction via combinatorial gene editing. Nat Commun 2024; 15:5218. [PMID: 38890276 PMCID: PMC11189492 DOI: 10.1038/s41467-024-49365-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: 09/09/2020] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Technologies that generate precise combinatorial genome modifications are well suited to dissect the polygenic basis of complex phenotypes and engineer synthetic genomes. Genome modifications with engineered nucleases can lead to undesirable repair outcomes through imprecise homology-directed repair, requiring non-cleavable gene editing strategies. Eukaryotic multiplex genome engineering (eMAGE) generates precise combinatorial genome modifications in Saccharomyces cerevisiae without generating DNA breaks or using engineered nucleases. Here, we systematically optimize eMAGE to achieve 90% editing frequency, reduce workflow time, and extend editing distance to 20 kb. We further engineer an inducible dominant negative mismatch repair system, allowing for high-efficiency editing via eMAGE while suppressing the elevated background mutation rate 17-fold resulting from mismatch repair inactivation. We apply these advances to construct a library of cancer-associated mutations in the ligand-binding domains of human estrogen receptor alpha and progesterone receptor to understand their impact on ligand-independent autoactivation. We validate that this yeast model captures autoactivation mutations characterized in human breast cancer models and further leads to the discovery of several previously uncharacterized autoactivating mutations. This work demonstrates the development and optimization of a cleavage-free method of genome editing well suited for applications requiring efficient multiplex editing with minimal background mutations.
Collapse
Affiliation(s)
- Peter N Ciaccia
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, 06520, USA
- Systems Biology Institute, Yale University, West Haven, CT, 06516, USA
- Physical and Engineering Biology, Yale University, New Haven, CT, 06520, USA
| | - Zhuobin Liang
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, 06520, USA.
- Systems Biology Institute, Yale University, West Haven, CT, 06516, USA.
- ZL: Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Anabel Y Schweitzer
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, 06520, USA
- Systems Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Eli Metzner
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, 06520, USA
- Systems Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Farren J Isaacs
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, 06520, USA.
- Systems Biology Institute, Yale University, West Haven, CT, 06516, USA.
- Physical and Engineering Biology, Yale University, New Haven, CT, 06520, USA.
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
43
|
Park CG, Adnan KM, Cho H, Ryu CS, Yoon J, Kim YJ. A combined in vitro-in silico method for assessing the androgenic activities of bisphenol A and its analogues. Toxicol In Vitro 2024; 98:105838. [PMID: 38710238 DOI: 10.1016/j.tiv.2024.105838] [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: 09/29/2023] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
Interactions between endocrine-disruptor chemicals (EDCs) and androgen receptor (AR) have adverse effects on the endocrine system, leading to human reproductive dysfunction. Bisphenol A (BPA) is an EDC that can damage both the environment and human health. Although numerous BPA analogues have been produced as substitutes for BPA, few studies have evaluated their endocrine-disrupting abilities. We assessed the (anti)-androgenic activities of BPA and its analogues using a yeast-based reporter assay. The BPA analogues tested were bisphenol S (BPS), 4-phenylphenol (4PP), 4,4'-(9-fluorenyliden)-diphenol (BPFL), tetramethyl bisphenol F (TMBPF), and tetramethyl bisphenol A (TMBPA). We also conducted molecular docking and dynamics simulations to assess the interactions of BPA and its analogues with the ligand-binding domain of human AR (AR-LBD). Neither BPA nor its analogues had androgenic activity; however, all except BPFL exerted robust anti-androgenic effects. Consistent with the in vitro results, anti-androgenic analogues of BPA formed hydrogen bonding patterns with key residues that differed from the patterns of endogenous hormones, indicating that the analogues display in inappropriate orientations when interacting with the binding pocket of AR-LBD. Our findings indicate that BPA and its analogues disrupt androgen signaling by interacting with the AR-LBD. Overall, BPA and its analogues display endocrine-disrupting activity, which is mediated by AR.
Collapse
Affiliation(s)
- Chang Gyun Park
- Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, Saarbrucken 66123, Germany; Division of Experimental Neurosurgery, Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Karim Md Adnan
- Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, Saarbrucken 66123, Germany; Universität des Saarlandes, 66123 Saarbrücken, Germany
| | - Hyunki Cho
- Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, Saarbrucken 66123, Germany; Universität des Saarlandes, 66123 Saarbrücken, Germany
| | - Chang Seon Ryu
- Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, Saarbrucken 66123, Germany
| | - Juyong Yoon
- Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, Saarbrucken 66123, Germany.
| | - Young Jun Kim
- Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, Saarbrucken 66123, Germany.
| |
Collapse
|
44
|
Medić-Milijić N, Jovanić I, Nedeljković M, Marković I, Spurnić I, Milovanović Z, Ademović N, Tomić T, Tanić N, Tanić N. Prognostic and Clinical Significance of PD-L1, EGFR and Androgen Receptor (AR) Expression in Triple-Negative Breast Cancer (TNBC) Patients. Life (Basel) 2024; 14:682. [PMID: 38929666 PMCID: PMC11205209 DOI: 10.3390/life14060682] [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: 03/21/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype and is associated with high recurrence rates, a high incidence of distant metastases and poor overall survival. The aim of this study was to investigate the role of PD-L1, EGFR and AR expression in TNBC promotion and progression. To that end, we analyzed the immunohistochemical expression of these genes in 125 TNBC patients and their relation to clinicopathological parameters and survival. An elevated expression of PD-L1 was significantly correlated with higher tumor and nuclear grade, while a low expression was correlated with loco-regional recurrence without any influence on survival. Contrary to this, the expression of AR showed a positive impact on the DFI and a negative association with tumor grade. Furthermore, PD-L1 and AR demonstrated simultaneous expression, and further co-expression analysis revealed that a positive expression of PD-L1/AR notably correlates with tumor and nuclear grade and has a significant impact on a longer DFI and OS, while a negative PD-L1/AR expression is significantly associated with metastases. Therefore, our results suggest that positive PD-L1/AR expression is beneficial for TNBC patients. In addition, an elevated expression of EGFR contributes to metastases and a worse DFI and OS. In conclusion, we think that low PD-L1/low AR/high EGFR expression followed by high Ki67 expression constitutes a 'high risk' profile of TNBC.
Collapse
Affiliation(s)
- Nataša Medić-Milijić
- Department of Pathology, Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (N.M.-M.); (I.J.); (Z.M.)
| | - Irena Jovanić
- Department of Pathology, Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (N.M.-M.); (I.J.); (Z.M.)
| | - Milica Nedeljković
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia;
| | - Ivan Marković
- Surgical Oncology Clinic, Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (I.M.); (I.S.)
| | - Igor Spurnić
- Surgical Oncology Clinic, Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (I.M.); (I.S.)
| | - Zorka Milovanović
- Department of Pathology, Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (N.M.-M.); (I.J.); (Z.M.)
| | - Nejla Ademović
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 11060 Belgrade, Serbia;
| | - Tijana Tomić
- Department of Radiobiology and Molecular Genetics, Institute of Nuclear Sciences “Vinča”, National Institute of Republic of Serbia, University of Belgrade, Mike Petrocića Alasa 12-14, 11000 Belgrade, Serbia; (T.T.); (N.T.)
| | - Nasta Tanić
- Department of Radiobiology and Molecular Genetics, Institute of Nuclear Sciences “Vinča”, National Institute of Republic of Serbia, University of Belgrade, Mike Petrocića Alasa 12-14, 11000 Belgrade, Serbia; (T.T.); (N.T.)
- Department of Natural Sciences and Mathematics, Field of Biology, State University of Novi Pazar, 36300 Novi Pazar, Serbia
| | - Nikola Tanić
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 11060 Belgrade, Serbia;
| |
Collapse
|
45
|
Fan Z, Yu Q, Deng J, Wang K, Yu H, Fan X, Xie J. Unveiling hormone-stimulated gene mechanisms in prostate cancer: A prognostic model, immune infiltration analysis, and drug sensitivity study. ENVIRONMENTAL TOXICOLOGY 2024; 39:3238-3252. [PMID: 38361268 DOI: 10.1002/tox.24118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/14/2023] [Accepted: 12/25/2023] [Indexed: 02/17/2024]
Abstract
Hormones promote the progression of prostate cancer (PRCA) through the activation of a complex regulatory network. Inhibition of hormones or modulation of specific network nodes alone is insufficient to suppress the entire oncogenic network. Therefore, it is imperative to elucidate the mechanisms underlying the occurrence and development of PRCA in order to identify reliable diagnostic markers and therapeutic targets. To this end, we used publicly available data to analyze the potential mechanisms of hormone-stimulated genes in PRCA, construct a prognostic model, and assess immune infiltration and drug sensitivity. The single-cell RNA-sequencing data of PRCA were subjected to dimensionality reduction clustering and annotation, and the cells were categorized into two groups based on hormone stimulus-related scores. The differentially expressed genes between the two groups were screened and incorporated into the least absolute shrinkage and selection operator machine learning algorithm, and a prognostic model comprising six genes (ZNF862, YIF1A, USP22, TAF7, SRSF3, and SPARC) was constructed. The robustness of the model was validation through multiple methods. Immune infiltration scores in the two risk groups were calculated using three different algorithms. In addition, the relationship between the model genes and immune cell infiltration, and that between risk score and immune cell infiltration were analyzed. Drug sensitivity analysis was performed for the model genes and risk score using public databases to identify potential candidate drugs. Our findings provide novel insights into the mechanisms of hormone-stimulated genes in PRCA progression, prognosis, and drug screening.
Collapse
Affiliation(s)
- Zhongru Fan
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Qianqian Yu
- National Clinical Research Center for Laboratory Medicine, Department of Laboratory Medicine, The First Hospital of China Medical University, Units of Medical Laboratory, Chinese Academy of Medical Sciences, Shenyang, China
| | - Junpeng Deng
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Ke Wang
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Hongqi Yu
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Xin Fan
- Department of Radiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jianjun Xie
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| |
Collapse
|
46
|
Singh K, Kumar P, Singh AK, Singh N, Singh S, Tiwari KN, Agrawal S, Das R, Singh A, Ram B, Tripathi AK, Mishra SK. In silico and network pharmacology analysis of fucosterol: a potent anticancer bioactive compound against HCC. Med Oncol 2024; 41:130. [PMID: 38676780 DOI: 10.1007/s12032-024-02374-w] [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: 01/25/2024] [Accepted: 03/28/2024] [Indexed: 04/29/2024]
Abstract
The Fucaceae family of marine brown algae includes Ascophyllum nodosum. Fucosterol (FSL) is a unique bioactive component that was identified through GC-MS analysis of the hydroalcoholic extract of A. nodosum. Fucosterol's mechanism of action towards hepatocellular cancer was clarified using network pharmacology and docking study techniques. The probable target gene of FSL has been predicted using the TargetNet and SwissTargetPred databases. GeneCards and the DisGNet database were used to check the targeted genes of FSL. By using the web programme Venny 2.1, the overlaps of FSL and HCC disease demonstrated that 18 genes (1.3%) were obtained as targeted genes Via the STRING database, a protein-protein interaction (PPI) network with 18 common target genes was constructed. With the aid of CytoNCA, hub genes were screened using the Cytoscape software, and the targets' hub genes were exported into the ShinyGo online tool for study of KEGG and gene ontology enrichment. Using the software AutoDock, a hub gene molecular docking study was performed. Ten genes, including AR, CYP19A1, ESR1, ESR2, TNF, PPARA, PPARG, HMGCR, SRC, and IGF1R, were obtained. The 10 targeted hubs docked with FSL successfully. The active components FSL of ASD, the FSL, are engaged in fatty liver disease, cancer pathways, and other signalling pathways, which could prove beneficial for the management of HCC.
Collapse
Affiliation(s)
- Kajal Singh
- Department of Biosciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Pradeep Kumar
- Department of Botany, MMV, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Amit Kumar Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, UP, 221005, India
| | - Nancy Singh
- Department of Biosciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Sakshi Singh
- Department of Biotechnology, Parul Institute of Applied Science, Parul University, Vadodara, Gujarat, 391760, India
| | - Kavindra Nath Tiwari
- Department of Botany, MMV, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Shreni Agrawal
- Department of Bioscience and Biotechnology, Banasthali Vidhyapith, Tonk, Rajsthan, India
| | - Richa Das
- Department of Bioscience and Biotechnology, Banasthali Vidhyapith, Tonk, Rajsthan, India
| | - Anuradha Singh
- Department of Biosciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Bhuwal Ram
- Department of Dravyaguna, IMS, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Amit Kumar Tripathi
- School of Basic and Applied Science, Galgotias University, Gautam Buddha Nagar, Greater Noida, Uttar Pradesh, 203201, India
| | - Sunil Kumar Mishra
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, UP, 221005, India.
| |
Collapse
|
47
|
Chen Z, Wang M, Wu D, Zhao L, Metwally H, Jiang W, Wang Y, Bai L, McEachern D, Luo J, Wang M, Li Q, Matvekas A, Wen B, Sun D, Chinnaiyan AM, Wang S. Discovery of CBPD-409 as a Highly Potent, Selective, and Orally Efficacious CBP/p300 PROTAC Degrader for the Treatment of Advanced Prostate Cancer. J Med Chem 2024; 67:5351-5372. [PMID: 38530938 DOI: 10.1021/acs.jmedchem.3c01789] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
CBP/p300 are critical transcriptional coactivators of the androgen receptor (AR) and are promising cancer therapeutic targets. Herein, we report the discovery of highly potent, selective, and orally bioavailable CBP/p300 degraders using the PROTAC technology with CBPD-409 being the most promising compound. CBPD-409 induces robust CBP/p300 degradation with DC50 0.2-0.4 nM and displays strong antiproliferative effects with IC50 1.2-2.0 nM in the VCaP, LNCaP, and 22Rv1 AR+ prostate cancer cell lines. It has a favorable pharmacokinetic profile and achieves 50% of oral bioavailability in mice. A single oral administration of CBPD-409 at 1 mg/kg achieves >95% depletion of CBP/p300 proteins in the VCaP tumor tissue. CBPD-409 exhibits strong tumor growth inhibition and is much more potent and efficacious than two CBP/p300 inhibitors CCS1477 and GNE-049 and the AR antagonist Enzalutamide. CBPD-409 is a promising CBP/p300 degrader for further extensive evaluations for the treatment of advanced prostate cancer and other types of human cancers.
Collapse
Affiliation(s)
- Zhixiang Chen
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mi Wang
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Dimin Wu
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Lijie Zhao
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hoda Metwally
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Wei Jiang
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yu Wang
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Longchuan Bai
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Donna McEachern
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jie Luo
- Michigan Center for Translational Pathology, Department of Pathology, Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Meilin Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Qiuxia Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Aleksas Matvekas
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, Department of Pathology, Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Shaomeng Wang
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
48
|
Chen Z, Wang M, Wu D, Bai L, Xu T, Metwally H, Wang Y, McEachern D, Zhao L, Li R, Takyi-Williams J, Wang M, Wang L, Li Q, Wen B, Sun D, Wang S. Discovery of CBPD-268 as an Exceptionally Potent and Orally Efficacious CBP/p300 PROTAC Degrader Capable of Achieving Tumor Regression. J Med Chem 2024; 67:5275-5304. [PMID: 38477974 DOI: 10.1021/acs.jmedchem.3c02124] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
CBP/p300 proteins are key epigenetic regulators and promising targets for the treatment of castration-resistant prostate cancer and other types of human cancers. Herein, we report the discovery and characterization of CBPD-268 as an exceptionally potent, effective, and orally efficacious PROTAC degrader of CBP/p300 proteins. CBPD-268 induces CBP/p300 degradation in three androgen receptor-positive prostate cancer cell lines, with DC50 ≤ 0.03 nM and Dmax > 95%, leading to potent cell growth inhibition. It has an excellent oral bioavailability in mice and rats. Oral administration of CBPD-268 at 0.3-3 mg/kg resulted in profound and persistent CBP/p300 depletion in tumor tissues and achieved strong antitumor activity in the VCaP and 22Rv1 xenograft tumor models in mice, including tumor regression in the VCaP tumor model. CBPD-268 was well tolerated in mice and rats and displayed a therapeutic index of >10. Taking these results together, CBPD-268 is a highly promising CBP/p300 degrader as a potential new cancer therapy.
Collapse
Affiliation(s)
- Zhixiang Chen
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mi Wang
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Dimin Wu
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Longchuan Bai
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Tianfeng Xu
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hoda Metwally
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yu Wang
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Donna McEachern
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Lijie Zhao
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ruiting Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - John Takyi-Williams
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Meilin Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Lu Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Qiuxia Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Shaomeng Wang
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
49
|
Martin-Caraballo M. Regulation of Molecular Biomarkers Associated with the Progression of Prostate Cancer. Int J Mol Sci 2024; 25:4171. [PMID: 38673756 PMCID: PMC11050209 DOI: 10.3390/ijms25084171] [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: 03/11/2024] [Revised: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Androgen receptor signaling regulates the normal and pathological growth of the prostate. In particular, the growth and survival of prostate cancer cells is initially dependent on androgen receptor signaling. Exposure to androgen deprivation therapy leads to the development of castration-resistant prostate cancer. There is a multitude of molecular and cellular changes that occur in prostate tumor cells, including the expression of neuroendocrine features and various biomarkers, which promotes the switch of cancer cells to androgen-independent growth. These biomarkers include transcription factors (TP53, REST, BRN2, INSM1, c-Myc), signaling molecules (PTEN, Aurora kinases, retinoblastoma tumor suppressor, calcium-binding proteins), and receptors (glucocorticoid, androgen receptor-variant 7), among others. It is believed that genetic modifications, therapeutic treatments, and changes in the tumor microenvironment are contributing factors to the progression of prostate cancers with significant heterogeneity in their phenotypic characteristics. However, it is not well understood how these phenotypic characteristics and molecular modifications arise under specific treatment conditions. In this work, we summarize some of the most important molecular changes associated with the progression of prostate cancers and we describe some of the factors involved in these cellular processes.
Collapse
Affiliation(s)
- Miguel Martin-Caraballo
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA
| |
Collapse
|
50
|
Ramakrishnan S, Cortes-Gomez E, Athans SR, Attwood KM, Rosario SR, Kim SJ, Mager DE, Isenhart EG, Hu Q, Wang J, Woloszynska A. Race-specific coregulatory and transcriptomic profiles associated with DNA methylation and androgen receptor in prostate cancer. Genome Med 2024; 16:52. [PMID: 38566104 PMCID: PMC10988846 DOI: 10.1186/s13073-024-01323-6] [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: 08/23/2023] [Accepted: 03/22/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Prostate cancer is a significant health concern, particularly among African American (AA) men who exhibit higher incidence and mortality compared to European American (EA) men. Understanding the molecular mechanisms underlying these disparities is imperative for enhancing clinical management and achieving better outcomes. METHODS Employing a multi-omics approach, we analyzed prostate cancer in both AA and EA men. Using Illumina methylation arrays and RNA sequencing, we investigated DNA methylation and gene expression in tumor and non-tumor prostate tissues. Additionally, Boolean analysis was utilized to unravel complex networks contributing to racial disparities in prostate cancer. RESULTS When comparing tumor and adjacent non-tumor prostate tissues, we found that DNA hypermethylated regions are enriched for PRC2/H3K27me3 pathways and EZH2/SUZ12 cofactors. Olfactory/ribosomal pathways and distinct cofactors, including CTCF and KMT2A, were enriched in DNA hypomethylated regions in prostate tumors from AA men. We identified race-specific inverse associations of DNA methylation with expression of several androgen receptor (AR) associated genes, including the GATA family of transcription factors and TRIM63. This suggests that race-specific dysregulation of the AR signaling pathway exists in prostate cancer. To investigate the effect of AR inhibition on race-specific gene expression changes, we generated in-silico patient-specific prostate cancer Boolean networks. Our simulations revealed prolonged AR inhibition causes significant dysregulation of TGF-β, IDH1, and cell cycle pathways specifically in AA prostate cancer. We further quantified global gene expression changes, which revealed differential expression of genes related to microtubules, immune function, and TMPRSS2-fusion pathways, specifically in prostate tumors of AA men. Enrichment of these pathways significantly correlated with an altered risk of disease progression in a race-specific manner. CONCLUSIONS Our study reveals unique signaling networks underlying prostate cancer biology in AA and EA men, offering potential insights for clinical management strategies tailored to specific racial groups. Targeting AR and associated pathways could be particularly beneficial in addressing the disparities observed in prostate cancer outcomes in the context of AA and EA men. Further investigation into these identified pathways may lead to the development of personalized therapeutic approaches to improve outcomes for prostate cancer patients across different racial backgrounds.
Collapse
Affiliation(s)
- Swathi Ramakrishnan
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Eduardo Cortes-Gomez
- Department of Bioinformatics and Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Biostatistics, SUNY University at Buffalo, Kimball Tower, Buffalo, NY, 14214, USA
| | - Sarah R Athans
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Kristopher M Attwood
- Department of Bioinformatics and Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Spencer R Rosario
- Department of Bioinformatics and Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Se Jin Kim
- Department of Pharmaceutical Sciences, SUNY University at Buffalo, Buffalo, NY, 14214, USA
| | - Donald E Mager
- Department of Pharmaceutical Sciences, SUNY University at Buffalo, Buffalo, NY, 14214, USA
- Enhanced Pharmacodynamics, LLC, Buffalo, NY, 14203, USA
| | - Emily G Isenhart
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Qiang Hu
- Department of Bioinformatics and Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Jianmin Wang
- Department of Bioinformatics and Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Anna Woloszynska
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|