1
|
Elimam H, Zaki MB, Abd-Elmawla MA, Darwish HA, Hatawsh A, Aborehab NM, Mageed SSA, Moussa R, Mohammed OA, Abdel-Reheim MA, Doghish AS. Natural products and long non-coding RNAs in prostate cancer: insights into etiology and treatment resistance. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6349-6368. [PMID: 39825964 DOI: 10.1007/s00210-024-03736-x] [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: 11/17/2024] [Accepted: 12/14/2024] [Indexed: 01/20/2025]
Abstract
Globally, the incidence and death rates associated with cancer persist in rising, despite considerable advancements in cancer therapy. Although some malignancies are manageable by a mix of chemotherapy, surgery, radiation, and targeted therapy, most malignant tumors either exhibit poor responsiveness to early identification or endure post-treatment survival. The prognosis for prostate cancer (PCa) is unfavorable since it is a perilous and lethal malignancy. The capacity of phytochemical and nutraceutical chemicals to repress oncogenic lncRNAs and activate tumor suppressor lncRNAs has garnered significant attention as a possible strategy to diminish the development, proliferation, metastasis, and invasion of cancer cells. A potential technique to treat cancer and enhance the sensitivity of cancer cells to existing conventional therapies is the use of phytochemicals with anticancer characteristics. Functional studies indicate that lncRNAs modulate drug resistance, stemness, invasion, metastasis, angiogenesis, and proliferation via interactions with tumor suppressors and oncoproteins. Among them, numerous lncRNAs, such as HOTAIR, PlncRNA1, GAS5, MEG3, LincRNA-21, and POTEF-AS1, support the development of PCa through many molecular mechanisms, including modulation of tumor suppressors and regulation of various signal pathways like PI3K/Akt, Bax/Caspase 3, P53, MAPK cascade, and TGF-β1. Other lncRNAs, in particular, MALAT-1, CCAT2, DANCR, LncRNA-ATB, PlncRNA1, LincRNA-21, POTEF-AS1, ZEB1-AS1, SChLAP1, and H19, are key players in regulating the aforementioned processes. Natural substances have shown promising anticancer benefits against PCa by altering essential signaling pathways. The overexpression of some lncRNAs is associated with advanced TNM stage, metastasis, chemoresistance, and reduced survival. LncRNAs possess crucial clinical and transitional implications in PCa, as diagnostic and prognostic biomarkers, as well as medicinal targets. To impede the progression of PCa, it is beneficial to target aberrant long non-coding RNAs using antisense oligonucleotides or small interfering RNAs (siRNAs). This prevents them from transmitting harmful messages. In summary, several precision medicine approaches may be used to rectify dysfunctional lncRNA regulatory circuits, so improving early PCa detection and eventually facilitating the conquest of this lethal disease. Due to their presence in biological fluids and tissues, they may serve as novel biomarkers. Enhancing PCa treatments mitigates resistance to chemotherapy and radiation.
Collapse
Affiliation(s)
- Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt.
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hebatallah A Darwish
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Pharmacology, Toxicology and Biochemistry Department, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt
| | - Abdulrahman Hatawsh
- Biotechnology School, Nile University, 26Th of July Corridor, Sheikh Zayed City, 12588, Giza, Egypt
| | - Nora M Aborehab
- Department of Biochemistry, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Rewan Moussa
- School Faculty of Medicine, Helwan University, Cairo, 11795, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | | | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, , 11829, Cairo, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| |
Collapse
|
2
|
Mohanty D, Padhee S, Priyadarshini A, Kerry RG, Dash B, Sahoo A, Jena S, Panda PC, Khan HA, Nayak S, Ray A. Integrative approach to decipher pharmacological mechanism of Cinnamomum zeylanicum essential oil in prostate cancer. Med Oncol 2025; 42:100. [PMID: 40072751 DOI: 10.1007/s12032-025-02665-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: 12/10/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Prostate cancer has garnered much importance in recent years due to its rising incidence and mortality among men worldwide. The ineffectiveness of existing therapies and adverse events associated with conventional treatment have led patients to turn towards traditional medicine for the management of prostate cancer. Cinnamomum zeylanicum bark essential oil (CZEO) possesses promising anticancer properties, yet the exact mechanism of action of CZEO for the management of prostate cancer remains unclear. Therefore, the current study tried to elucidate the bioactive components and key potential targets through which CZEO may exert its anticancer effect for treating prostate cancer. Fifty-nine constituents were identified by GC-MS, of which 52 were drug-like constituents. A total of 2847 targets related to CZEO and 2283 targets related to prostate cancer were obtained from public databases and the GEO dataset. Twenty-three overlapping targets exist between CZEO and disease targets. Compound-disease-target network analysis revealed camphor, eugenol, methyl eugenol, trans farnesyl acetate and nerol as the core bioactive ingredients of CZEO. The topological screening of the PPI network revealed BCL2, TNF, NFKBIA, CREBBP and IL7R as potential hub targets. These hub targets were validated based on mRNA expression level, pathological stages, overall survival, immune infiltrate and genetic alteration analysis in prostate adenocarcinoma and normal patients. KEGG enrichment analysis proposed that CZEO exhibits its anticancer effect mainly by modulating the PI3-AKT and MAPK signalling pathway. Moreover, molecular docking and dynamics simulation studies revealed a good binding affinity of these core compounds with TNF, NFKBIA and BCL2. CZEO exhibited a remarkable anti-proliferative effect against PC-3 cells with an IC50 value of 13.56 µg/mL. CZEO promoted apoptosis and cell cycle arrest in the G2/M phase in PC-3 cells. CZEO-induced apoptosis was due to loss of mitochondrial membrane potential, increase in reactive oxygen species levels and activation of caspases (caspase 3, caspase 8 and caspase 9). RT-qPCR analysis revealed that CZEO modulated the mRNA expression level of hub genes (BCL2, TNF, NFKBIA, CREBBP, and IL7R, caspase 3, caspase 8 and caspase 9). The present study provides a mechanistic approach of Cinnamomum zeylanicum essential oil against prostate cancer.
Collapse
Affiliation(s)
- Debajani Mohanty
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Sucheesmita Padhee
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Arpita Priyadarshini
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Rout George Kerry
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Biswabhusan Dash
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Ambika Sahoo
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Sudipta Jena
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Pratap Chandra Panda
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Haseeb Ahmad Khan
- Department of Biochemistry, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Sanghamitra Nayak
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Asit Ray
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India.
| |
Collapse
|
3
|
Gyebi GA, Afolabi SO, Ogunyemi OM, Ibrahim IM, Olorundare OE, Adebayo JO, Koketsu M. Apoptotic Potential of Iloneoside from Gongronema latifolium Benth against Prostate Cancer Cells Using In Vitro and In Silico Approach. Cell Biochem Biophys 2025; 83:755-776. [PMID: 39302620 PMCID: PMC11870900 DOI: 10.1007/s12013-024-01507-2] [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] [Accepted: 08/25/2024] [Indexed: 09/22/2024]
Abstract
Prostate cancer is a major cause of cancer-related mortality in men worldwide. The anti-proliferative activity of Gongronema latifolium leaf extracts on some cancer cells has been reported. Herein, we investigated the growth inhibitory effect of the Gongronema latilolium leaf methanol extract and isolated pregnane (iloneoside) against prostate cancer cell lines using the MTT cell proliferation assay, apoptosis quantification, cell cycle analysis using flow cytometry and computational analysis molecular docking, molecular dynamics simulation (MDs), binding free energy computation and cluster analysis. In addition, UPLC-ESI-TOFMS chemical fingerprinting of previously isolated compounds was performed. The extract inhibited the growth of the cell lines with an IC50 of 49.3 µg/ml and 28.4 µg/ml for 24 h and 48 h, respectively, for PC3; and 43.7 µg/ml and 22.3 µg/ml for 24 h and 48 h, respectively, for DU145. Iloneoside demonstrated low inhibitory activities against PC3 and DU145 (IC50 > 80 μM). Apoptotic quantification and cell cycle analysis further showed that iloneoside induced apoptosis in a few cells at a dose of 200 uM. The ensemble-based molecular docking of the iloneoside to BCL-XL and BCL-2 proteins, and docking to MCL-1, BCL-A1 and BFL-1 proteins, respectively, presented binding energies of -7.22 ± 0.5, -8.12 ± 0.55, -7.1, -7.2 and -6.3 kcal/mol, while the MM/PBSA binding free energy was -25.72 ± 7.22 and -27.76 ± 11.32 kcal/mol for BCL-XL and BCL-2 proteins. Furthermore, iloneoside was stable during the 100 ns MDs analysis, while the clustering of the MDs trajectories showed that the interactions were strongly preserved. Iloneoside, in part, or in synergy with other constituents, may be responsible for the antiproliferative activities of the leaf, subject to further investigation.
Collapse
Affiliation(s)
- Gideon A Gyebi
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban, South Africa.
- Natural Products and Structural (Bio-Chem)-informatics Research Laboratory (NpsBC-RL), Department of Biochemistry, Faculty of Science and Technology, Bingham University, Karu, Nigeria.
| | - Saheed O Afolabi
- Biomolecular Modeling and Nutraceuticals Laboratory, Nutritional and Industrial Biochemistry Research Unit, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Oludare M Ogunyemi
- Faculty of Basic Medical Sciences, Department of Pharmacology and Therapeutics, University of Ilorin, Ilorin, Nigeria
| | - Ibrahim M Ibrahim
- Department of Biophysics, Faculty of Sciences, Cairo University, Giza, Egypt
| | - Olufunke E Olorundare
- Faculty of Basic Medical Sciences, Department of Pharmacology and Therapeutics, University of Ilorin, Ilorin, Nigeria
| | - Joseph O Adebayo
- Department of Biochemistry, Faculty of Life Sciences, University of Ilorin, Ilorin, Nigeria
| | - Mamoru Koketsu
- Faculty of Engineering, Department of Chemistry and Biomolecular Science, Gifu University, Gifu, Japan
| |
Collapse
|
4
|
Park MN, Choi J, Maharub Hossain Fahim M, Asevedo EA, Nurkolis F, Ribeiro RIMA, Kang HN, Kang S, Syahputra RA, Kim B. Phytochemical synergies in BK002: advanced molecular docking insights for targeted prostate cancer therapy. Front Pharmacol 2025; 16:1504618. [PMID: 40034825 PMCID: PMC11872924 DOI: 10.3389/fphar.2025.1504618] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Achyranthes japonica (Miq.) Nakai (AJN) and Melandrium firmum (Siebold and Zucc.) Rohrb. (MFR) are medicinal plants recognized for their bioactive phytochemicals, including ecdysteroids, anthraquinones, and flavonoids. This study investigates the anticancer properties of key constituents of these plants, focusing on the BK002 formulation, a novel combination of AJN and MFR. Specifically, the research employs advanced molecular docking and in silico analyses to assess the interactions of bioactive compounds ecdysterone, inokosterone, and 20-hydroxyecdysone (20-HE) with key prostate cancer-related network proteins, including 5α-reductase, CYP17, DNMT1, Dicer, PD-1, and PD-L1. Molecular docking techniques were applied to evaluate the binding affinities contributions of the bioactive compounds in BK002 against prostate cancer-hub network targets. The primary focus was on enzymes like 5α-reductase and CYP17, which are central to androgen biosynthesis, as well as on cancer-related proteins such as DNA methyltransferase 1 (DNMT1), Dicer, programmed death-1 (PD-1), and programmed death ligand-1 (PD-L1). Based on data from prostate cancer patients, key target networks were identified, followed by in silico analysis of the primary bioactive components of BK002.In silico assessments were conducted to evaluate the safety profiles of these compounds, providing insights into their therapeutic potential. The docking studies revealed that ecdysterone, inokosterone, and 20-hydroxyecdysonec demonstrated strong binding affinities to the critical prostate cancer-related enzymes 5α-reductase and CYP17, contributing to a potential reduction in androgenic activity. These compounds also exhibited significant inhibitory interactions with DNMT1, Dicer, PD-1, and PD-L1, suggesting a capacity to interfere with key oncogenic and immune evasion pathways. Ecdysterone, inokosterone, and 20-hydroxyecdysone have demonstrated the ability to target key oncogenic pathways, and their favorable binding affinity profiles further underscore their potential as novel therapeutic agents for prostate cancer. These findings provide a strong rationale for further preclinical and clinical investigations, supporting the integration of BK002 into therapeutic regimens aimed at modulating tumor progression and immune responses.
Collapse
Affiliation(s)
- Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jinwon Choi
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | | | - Estéfani Alves Asevedo
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Experimental Pathology Laboratory, Midwest Campus, Federal University of São João del-Rei, Divinópolis, Brazil
| | - Fahrul Nurkolis
- Department of Biological Sciences, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta, Indonesia
| | | | - Han Na Kang
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Sojin Kang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Rony Abdi Syahputra
- Department of Biological Sciences, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta, Indonesia
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
5
|
Hashemi Karoii D, Bavandi S, Djamali M, Abroudi AS. Exploring the interaction between immune cells in the prostate cancer microenvironment combining weighted correlation gene network analysis and single-cell sequencing: An integrated bioinformatics analysis. Discov Oncol 2024; 15:513. [PMID: 39349877 PMCID: PMC11442730 DOI: 10.1007/s12672-024-01399-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND The rise of treatment resistance and variability across malignant profiles has made precision oncology an imperative in today's medical landscape. Prostate cancer is a prevalent form of cancer in males, characterized by significant diversity in both genomic and clinical characteristics. The tumor microenvironment consists of stroma, tumor cells, and various immune cells. The stromal components and tumor cells engage in mutual communication and facilitate the development of a low-oxygen and pro-cancer milieu by producing cytokines and activating pro-inflammatory signaling pathways. METHODS In order to discover new genes associated with tumor cells that interact and facilitate a hypoxic environment in prostate cancer, we conducted a cutting-edge bioinformatics investigation. This included analyzing high-throughput genomic datasets obtained from the cancer genome atlas (TCGA). RESULTS A combination of weighted gene co-expression network analysis and single-cell sequencing has identified nine dysregulated immune hub genes (AMACR, KCNN3, MME, EGFR, FLT1, GDF15, KDR, IGF1, and KRT7) that are believed to have significant involvement in the biological pathways involved with the advancement of prostate cancer enviriment. In the prostate cancer environment, we observed the overexpression of GDF15 and KRT7 genes, as well as the downregulation of other genes. Additionally, the cBioPortal platform was used to investigate the frequency of alterations in the genes and their effects on the survival of the patients. The Kaplan-Meier survival analysis indicated that the changes in the candidate genes were associated with a reduction in the overall survival of the patients. CONCLUSIONS In summary, the findings indicate that studying the genes and their genomic changes may be used to develop precise treatments for prostate cancer. This approach involves early detection and targeted therapy.
Collapse
Affiliation(s)
- Danial Hashemi Karoii
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Sobhan Bavandi
- Department of Biology, Qaemshahr Branch, Islamic Azad University, Qaemshahr, Iran
| | - Melika Djamali
- Department of Biology, Faculty of Science, Tehran University, Tehran, Iran
| | - Ali Shakeri Abroudi
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
6
|
Rajadnya R, Sharma N, Mahajan A, Ulhe A, Patil R, Hegde M, Mali A. Novel systems biology experimental pipeline reveals matairesinol's antimetastatic potential in prostate cancer: an integrated approach of network pharmacology, bioinformatics, and experimental validation. Brief Bioinform 2024; 25:bbae466. [PMID: 39297880 PMCID: PMC11411774 DOI: 10.1093/bib/bbae466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/21/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024] Open
Abstract
Matairesinol (MAT), a plant lignan renowned for its anticancer properties in hormone-sensitive cancers like breast and prostate cancers, presents a promising yet underexplored avenue in the treatment of metastatic prostate cancer (mPC). To elucidate its specific therapeutic targets and mechanisms, our study adopted an integrative approach, amalgamating network pharmacology (NP), bioinformatics, GeneMANIA-based functional association (GMFA), and experimental validation. By mining online databases, we identified 27 common targets of mPC and MAT, constructing a MAT-mPC protein-protein interaction network via STRING and pinpointing 11 hub targets such as EGFR, AKT1, ERBB2, MET, IGF1, CASP3, HSP90AA1, HIF1A, MMP2, HGF, and MMP9 with CytoHuba. Utilizing DAVID, Gene Ontology (GO) analysis highlighted metastasis-related processes such as epithelial-mesenchymal transition, positive regulation of cell migration, and key Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, including cancer, prostate cancer, PI3K-Akt, and MAPK signaling, while the web resources such as UALCAN and GEPIA2 affirmed the clinical significance of the top 11 hub targets in mPC patient survival analysis and gene expression patterns. Our innovative GMFA enrichment method further enriched network pharmacology findings. Molecular docking analyses demonstrated substantial interactions between MAT and 11 hub targets. Simulation studies confirmed the stable interactions of MAT with selected targets. Experimental validation in PC3 cells, employing quantitative real-time reverse-transcription PCR and various cell-based assays, corroborated MAT's antimetastatic effects on mPC. Thus, this exhaustive NP analysis, complemented by GMFA, molecular docking, molecular dynamics simulations, and experimental validations, underscores MAT's multifaceted role in targeting mPC through diverse therapeutic avenues. Nevertheless, comprehensive in vitro validation is imperative to solidify these findings.
Collapse
Affiliation(s)
- Rama Rajadnya
- Cancer Biology, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Dhankawadi, Pune, Maharashtra 411043, India
| | - Nidhi Sharma
- Cancer Biology, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Dhankawadi, Pune, Maharashtra 411043, India
| | - Akanksha Mahajan
- Cancer Biology, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Dhankawadi, Pune, Maharashtra 411043, India
| | - Amrita Ulhe
- Cancer Biology, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Dhankawadi, Pune, Maharashtra 411043, India
| | - Rajesh Patil
- Department of Pharmaceutical Chemistry, Sinhgad Technical Education Society, Sinhgad College of Pharmacy, Vadgaon (BK), Off Sinhgad Road, Pune, Maharashtra 411041, India
| | - Mahabaleshwar Hegde
- Innovative Nutrition, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Dhankawadi, Pune, Maharashtra 411043, India
| | - Aniket Mali
- Cancer Biology, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Dhankawadi, Pune, Maharashtra 411043, India
| |
Collapse
|
7
|
Ebrahimian Vargahan S, Barati M, Roudbari M, Eini M, Hosseini A. Effect of microRNA-141-3p, E2F3, CDK3, and KAT2B overexpression on histologic tumor grade and metastasis status in untreated breast cancer tissues. BIOIMPACTS : BI 2024; 15:30032. [PMID: 39963567 PMCID: PMC11830142 DOI: 10.34172/bi.30032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 02/20/2025]
Abstract
Introduction Increasing evidence has reported gene expression alterations in breast cancer (BC) tissues, necessitating their investigating to highlight the molecular basis of the disease development or progression. This study investigated the expression of miR-141, E2F3, CDK3, TP53, and KAT2B, and their association with histologic grade and metastasis in BC tissues. Methods The RNA expression level of miR-141, E2F3, CDK3, TP53, and KAT2B genes was analyzed in 23 BC and 23 normal tissue samples by RT-qPCR. The associations of the expression level of these genes with clinicopathological features of the BC tissue samples were evaluated. The study also explored the correlation between RNA levels of genes and miR-141. Results Expression of miR-141, E2F3, CDK3, and KAT2B demonstrated significantly higher levels in BC tumor than normal tissues. TP53 expression showed an increase in tumor compared to normal tissues, although it was insignificant. Moreover, increased RNA expression of miR-141, E2F3, CDK3, and KAT2B corresponded to the advanced stage and regional metastasis of BC. Additionally, the results demonstrated a significant correlation between RNA expression levels of miR-141 with CDK3 and E2F3 with KAT2B. Conclusion Our findings highlighted clinicopathologic indicators that were relevant to aggressive BC. Besides, Correlations between overexpression of miR-141, E2F3, CDK3, and KAT2B in BC tissues suggest regulatory effects. Taken together, it seems results of this study could provide evidence that dysregulation of gene expression contributes significantly to unveiling the underlying molecular basis of BC.
Collapse
Affiliation(s)
- Sepideh Ebrahimian Vargahan
- Department of Medical Biotechnology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmood Barati
- Department of Medical Biotechnology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Roudbari
- Biostatistics Department, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Eini
- Department of Medical Biotechnology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Arshad Hosseini
- Department of Medical Biotechnology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Brown LK, Kanagasabai T, Li G, Celada SI, Rumph JT, Adunyah SE, Stewart LV, Chen Z. Co-targeting SKP2 and KDM5B inhibits prostate cancer progression by abrogating AKT signaling with induction of senescence and apoptosis. Prostate 2024; 84:877-887. [PMID: 38605532 DOI: 10.1002/pros.24706] [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: 11/27/2023] [Revised: 03/08/2024] [Accepted: 03/29/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND Prostate cancer (PCa) is the second-leading cause of cancer mortalities in the United States and is the most commonly diagnosed malignancy in men. While androgen deprivation therapy (ADT) is the first-line treatment option to initial responses, most PCa patients invariably develop castration-resistant PCa (CRPC). Therefore, novel and effective treatment strategies are needed. The goal of this study was to evaluate the anticancer effects of the combination of two small molecule inhibitors, SZL-P1-41 (SKP2 inhibitor) and PBIT (KDM5B inhibitor), on PCa suppression and to delineate the underlying molecular mechanisms. METHODS Human CRPC cell lines, C4-2B and PC3 cells, were treated with small molecular inhibitors alone or in combination, to assess effects on cell proliferation, migration, senescence, and apoptosis. RESULTS SKP2 and KDM5B showed an inverse regulation at the translational level in PCa cells. Cells deficient in SKP2 showed an increase in KDM5B protein level, compared to that in cells expressing SKP2. By contrast, cells deficient in KDM5B showed an increase in SKP2 protein level, compared to that in cells with KDM5B intact. The stability of SKP2 protein was prolonged in KDM5B depleted cells as measured by cycloheximide chase assay. Cells deficient in KDM5B were more vulnerable to SKP2 inhibition, showing a twofold greater reduction in proliferation compared to cells with KDM5B intact (p < 0.05). More importantly, combined inhibition of KDM5B and SKP2 significantly decreased proliferation and migration of PCa cells as compared to untreated controls (p < 0.005). Mechanistically, combined inhibition of KDM5B and SKP2 in PCa cells abrogated AKT activation, resulting in an induction of both cellular senescence and apoptosis, which was measured via Western blot analysis and senescence-associated β-galactosidase (SA-β-Gal) staining. CONCLUSIONS Combined inhibition of KDM5B and SKP2 was more effective at inhibiting proliferation and migration of CRPC cells, and this regimen would be an ideal therapeutic approach of controlling CRPC malignancy.
Collapse
Affiliation(s)
- LaKendria K Brown
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, Tennessee, USA
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA
| | - Thanigaivelan Kanagasabai
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, Tennessee, USA
| | - Guoliang Li
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA
| | - Sherly I Celada
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA
| | - Jelonia T Rumph
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, Tennessee, USA
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, USA
| | - Samuel E Adunyah
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA
| | - LaMonica V Stewart
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, Tennessee, USA
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA
| | - Zhenbang Chen
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA
| |
Collapse
|
9
|
Kumar A, BharathwajChetty B, Manickasamy MK, Unnikrishnan J, Alqahtani MS, Abbas M, Almubarak HA, Sethi G, Kunnumakkara AB. Natural compounds targeting YAP/TAZ axis in cancer: Current state of art and challenges. Pharmacol Res 2024; 203:107167. [PMID: 38599470 DOI: 10.1016/j.phrs.2024.107167] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024]
Abstract
Cancer has become a burgeoning global healthcare concern marked by its exponential growth and significant economic ramifications. Though advancements in the treatment modalities have increased the overall survival and quality of life, there are no definite treatments for the advanced stages of this malady. Hence, understanding the diseases etiologies and the underlying molecular complexities, will usher in the development of innovative therapeutics. Recently, YAP/TAZ transcriptional regulation has been of immense interest due to their role in development, tissue homeostasis and oncogenic transformations. YAP/TAZ axis functions as coactivators within the Hippo signaling cascade, exerting pivotal influence on processes such as proliferation, regeneration, development, and tissue renewal. In cancer, YAP is overexpressed in multiple tumor types and is associated with cancer stem cell attributes, chemoresistance, and metastasis. Activation of YAP/TAZ mirrors the cellular "social" behavior, encompassing factors such as cell adhesion and the mechanical signals transmitted to the cell from tissue structure and the surrounding extracellular matrix. Therefore, it presents a significant vulnerability in the clogs of tumors that could provide a wide window of therapeutic effectiveness. Natural compounds have been utilized extensively as successful interventions in the management of diverse chronic illnesses, including cancer. Owing to their capacity to influence multiple genes and pathways, natural compounds exhibit significant potential either as adjuvant therapy or in combination with conventional treatment options. In this review, we delineate the signaling nexus of YAP/TAZ axis, and present natural compounds as an alternate strategy to target cancer.
Collapse
Affiliation(s)
- Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Bandari BharathwajChetty
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Mukesh Kumar Manickasamy
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Jyothsna Unnikrishnan
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia; BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester LE1 7RH, United Kingdom
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
| | - Hassan Ali Almubarak
- Division of Radiology, Department of Medicine, College of Medicine and Surgery, King Khalid University, Abha 61421, Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 117699, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India.
| |
Collapse
|
10
|
Nayana P, Gollapalli P, Manjunatha H. Investigating the structural basis of piperine targeting AKT1 against prostate cancer through in vitro and molecular dynamics simulations. J Biomol Struct Dyn 2024:1-15. [PMID: 38529821 DOI: 10.1080/07391102.2024.2331096] [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: 01/09/2024] [Accepted: 03/11/2024] [Indexed: 03/27/2024]
Abstract
AkT1, significantly impacts many tumours cell functions, like transcription, apoptosis, glucose metabolism, cell proliferation, and cell migration. For tumours to develop and spread, aberrant activation of AKT1 is essential. Therefore, a major focus of molecularly targeted PCa treatment is AKT1. The present study investigates the effect of piperine compared to SDF using in-vitro studies, viz colony formation assay, comet assay and AKT1 gene expression studies using human PCa cell line PC3. A cluster of approximately at least 50 cells constitutes a colony. The clonogenic assay showed that the number and size of colonies significantly decreased when treated with compounds (SDF and piperine) in comparison to the untreated cells which effectively proliferated to form more colonies. Piperine treatment showed significant inhibition of colony formation than SDF. Effective genotoxicity was observed in piperine-treated PC3 cells with an increased Tail length of 120 µm and it was moderately observed in SDF with a Tail length of 30 µm treated on PC3 cells. The control group did not show any considerable genotoxicity with a Tail length of 6 µm. Our data, both in vitro and in silico, suggested that piperine would be a good starting point for developing novel drugs for the treatment of PCa. The downstream functions of Akt1 may be inhibited by these effects, which could impede the proliferation of PCa cells. High stability of the piperine-AKT1 complex was found by the MD simulation. Higher hydrophilic residues like Lys268 and Ser205 at the active pocket may be the cause of the binding stability. Overall, the observed results confirmed the anti-PCa effect of piperine by causing effective DNA damage and proved to be genotoxic in nature against the human PCa. These effects may impede the downstream activities of Akt1 and result in PCa cell growth regression.
Collapse
Affiliation(s)
- Prakash Nayana
- Department of PG Studies and Research in Biotechnology, Kuvempu University, Shivamogga, Karnataka, India
| | - Pavan Gollapalli
- Center for Bioinformatics and Biostatistics, Nitte (Deemed to be University), Mangalore, Karnataka, India
| | | |
Collapse
|
11
|
Chen Q, Sun Y, Wang S, Xu J. New prospects of cancer therapy based on pyroptosis and pyroptosis inducers. Apoptosis 2024; 29:66-85. [PMID: 37943371 DOI: 10.1007/s10495-023-01906-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2023] [Indexed: 11/10/2023]
Abstract
Pyroptosis is a gasdermin-mediated programmed cell death (PCD) pathway. It differs from apoptosis because of the secretion of inflammatory molecules. Pyroptosis is closely associated with various malignant tumors. Recent studies have demonstrated that pyroptosis can either inhibit or promote the development of malignant tumors, depending on the cell type (immune or cancer cells) and duration and severity of the process. This review summarizes the molecular mechanisms of pyroptosis, its relationship with malignancies, and focuses on current pyroptosis inducers and their significance in cancer treatment. The molecules involved in the pyroptosis signaling pathway could serve as therapeutic targets for the development of novel drugs for cancer therapy. In addition, we analyzed the potential of combining pyroptosis with conventional anticancer techniques as a promising strategy for cancer treatment.
Collapse
Affiliation(s)
- Qiaoyun Chen
- China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, 210008, China
- Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yuxiang Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225000, China
| | - Siliang Wang
- China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, 210008, China.
- Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Jingyan Xu
- China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, 210008, China.
- Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
12
|
Lekhak N, Bhattarai HK. Phytochemicals in Cancer Chemoprevention: Preclinical and Clinical Studies. Cancer Control 2024; 31:10732748241302902. [PMID: 39629692 PMCID: PMC11615997 DOI: 10.1177/10732748241302902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/11/2024] [Accepted: 11/11/2024] [Indexed: 12/08/2024] Open
Abstract
Phytochemicals, chemicals from plants, have garnered huge attention for their potential ability to prevent cancer. In vivo and preclinical models show that they do so often by affecting the hallmarks of cancer. Phytochemicals affect key pathways involved in the survival, genome maintenance, proliferation, senescence, and transendothelial migration of cancer cells. Some phytochemicals, namely antioxidants, can scavenge and quench reactive oxygen species (ROS) to prevent lipid peroxidation and DNA damage. They also trigger apoptosis by stopping the cell cycle at checkpoints to initiate the DNA damage response. Numerous in vitro and in vivo studies suggest that phytochemicals hinder cancer onset and progression by modifying major cell signaling pathways such as JAK/STAT, PI3K/Akt, Wnt, NF-kB, TGF-β, and MAPK. It is a well-known fact that the occurrence of cancer is in itself a very intricate process involving multiple mechanisms concurrently. Cancer prevention using phytochemicals is also an equally complex process that requires investigation and understanding of a myriad of processes going on in the cells and tissues. While many in vitro and preclinical studies have established that phytochemicals may be potential chemopreventive agents of cancer, their role in clinical randomized control trials needs to be established. This paper aims to shed light on the dynamics of chemoprevention using phytochemicals.
Collapse
Affiliation(s)
- Nitish Lekhak
- Department of Biotechnology, Kathmandu University, Dhulikhel, Nepal
| | | |
Collapse
|
13
|
Singh V, Shirbhate E, Kore R, Mishra A, Johariya V, Veerasamy R, Tiwari AK, Rajak H. Dietary Plant Metabolites Induced Epigenetic Modification as a Novel Strategy for the Management of Prostate Cancer. Mini Rev Med Chem 2024; 24:1409-1426. [PMID: 38385496 DOI: 10.2174/0113895575283895240207065454] [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: 09/24/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 02/23/2024]
Abstract
Prostate cancer is a widespread malignancy among men, with a substantial global impact on morbidity and mortality. Despite advances in conventional therapies, the need for innovative and less toxic treatments remains a priority. Emerging evidence suggests that dietary plant metabolites possess epigenetic-modifying properties, making them attractive candidates for prostate cancer treatment. The present work reviews the epigenetic effects of dietary plant metabolites in the context of prostate cancer therapy. We first outline the key epigenetic mechanisms involved in prostate cancer pathogenesis, including histone modifications, DNA methylation, and miRNA or Long Noncoding RNA (lncRNA) dysregulation. Next, we delve into the vast array of dietary plant metabolites that have demonstrated promising anti-cancer effects through epigenetic regulation. Resveratrol, minerals, isothiocyanates, curcumin, tea polyphenols, soy isoflavones and phytoestrogens, garlic compounds, anthocyanins, lycopene, and indoles are among the most extensively studied compounds. These plant-derived bioactive compounds have been shown to influence DNA methylation patterns, histone modifications, and microRNA expression, thereby altering the gene expression allied with prostate cancer progression, cell proliferation, and apoptosis. We also explore preclinical and clinical studies investigating the efficacy of dietary plant metabolites as standalone treatments or in combination with traditional treatments for people with prostate cancer. The present work highlights the potential of dietary plant metabolites as epigenetic modulators to treat prostate cancer. Continued research in this field may pave the way for personalized and precision medicine approaches, moving us closer to the goal of improved prostate cancer management.
Collapse
Affiliation(s)
- Vaibhav Singh
- Department of Pharmacy, Guru Ghasidash Vishwavidyalaya University, Bilaspur-495 009, (C.G.), India
| | - Ekta Shirbhate
- Department of Pharmacy, Guru Ghasidash Vishwavidyalaya University, Bilaspur-495 009, (C.G.), India
| | - Rakesh Kore
- Department of Pharmacy, Guru Ghasidash Vishwavidyalaya University, Bilaspur-495 009, (C.G.), India
| | - Aditya Mishra
- Department of Pharmacy, Guru Ghasidash Vishwavidyalaya University, Bilaspur-495 009, (C.G.), India
| | - Varsha Johariya
- Department of Pharmacy, Guru Ghasidash Vishwavidyalaya University, Bilaspur-495 009, (C.G.), India
| | - Ravichandran Veerasamy
- Departement of Pharmaceutical chemistry, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Amit K Tiwari
- UAMS College of Pharmacy, College of Pharmacy and Pharmaceutical Sciences, UAMS - University of Arkansas for Medical Sciences, Arkansas, (AR) USA
| | - Harish Rajak
- Department of Pharmacy, Guru Ghasidash Vishwavidyalaya University, Bilaspur-495 009, (C.G.), India
| |
Collapse
|
14
|
Kassem NM, Abdelmegid YA, El-Sayed MK, Sayed RS, Abdel-Aalla MH, Kassem HA. Nutrigenomics and microbiome shaping the future of personalized medicine: a review article. J Genet Eng Biotechnol 2023; 21:134. [PMID: 37993702 PMCID: PMC10665279 DOI: 10.1186/s43141-023-00599-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 11/09/2023] [Indexed: 11/24/2023]
Abstract
The relationship between nutrition and genes has long been hinted at and sometimes plainly associated with certain diseases. Now, after many years of research and coincidental findings, it is believed that this relationship, termed "Nutrigenomics," is certainly a factor of major importance in various conditions. In this review article, we discuss nutrigenomics, starting with basics definitions and enzymatic functions and ending with its palpable association with cancer. Now, diet is basically what we eat on a daily basis. Everything that enters through our alimentary tract ends up broken down to minute molecules and amino acids. These molecules interact with our microbiome and genome in discreet ways. For instance, we demonstrate how proper intake of probiotics enhances beneficial bacteria and may alleviate IBS and prevent colorectal cancer on the long term. We also show how a diet rich in folic acid is essential for methylenetetrahydrofolate reductase (MTHFR) function, which lowers risk of colorectal cancer. Also, we discuss how certain diets were associated with development of certain cancers. For example, red and processed meat are highly associated with colorectal and prostate cancer, salty diets with stomach cancer, and obesity with breast cancer. The modification of these diets significantly lowered the risk and improved prognosis of these cancers among many others. We also examined how micronutrients had a role in cancer prevention, as vitamin A and C exert anti-carcinogenic effects through their function as antioxidants. In addition, we show how folic acid prevent DNA mutations by enhancing protein methylation processes. Finally, after a systematic review of myriad articles on the etiology and prevention of cancer, we think that diet should be a crucial feature in cancer prevention and treatment programs. In the future, healthy diets and micronutrients may even be able to successively alter the liability to genetic mutations that result in cancer. It also will play a role in boosting treatment and improving prognosis of diagnosed cancers.
Collapse
Affiliation(s)
- Neemat M Kassem
- Clinical and Chemical Pathology Department, Kasr Al Ainy Centre of Clinical Oncology & Nuclear Medicine, School of Medicine, Cairo University, Cairo, Egypt
| | | | - Mahmoud K El-Sayed
- Faculty of Medicine, Kasr Al-Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Rana S Sayed
- Faculty of Medicine, Kasr Al-Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Mahmoud H Abdel-Aalla
- Faculty of Medicine, Kasr Al-Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Hebatallah A Kassem
- Clinical and Chemical Pathology Department, Kasr Al Ainy Centre of Clinical Oncology & Nuclear Medicine, School of Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
15
|
Yang J, Xin C, Yin G, Li J. Taraxasterol suppresses the proliferation and tumor growth of androgen-independent prostate cancer cells through the FGFR2-PI3K/AKT signaling pathway. Sci Rep 2023; 13:13072. [PMID: 37567936 PMCID: PMC10421874 DOI: 10.1038/s41598-023-40344-w] [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: 01/03/2023] [Accepted: 08/09/2023] [Indexed: 08/13/2023] Open
Abstract
Prostate cancer (PCa) is prevalent among older men and difficult to survive after metastasis. It is urgent to find new drugs and treatments. Several studies show that taraxasterol (TAX) has important anti-inflammatory, anti-oxidative and anti-tumor effects. However, the function and mechanisms of TAX in PCa remain unclear. Here, we found that TAX could significantly suppress the viability and growth of androgen-independent PCa cells and down-regulate the expression of c-Myc and cyclin D1 in vitro. Mechanistically, PI3K/AKT signaling pathway was weakened and the expression of FGFR2 was reduced after TAX treatment in androgen-independent PCa cells. Moreover, TAX evidently inhibited the tumor growth in nude mice and the expression of c-Myc, cyclin D1, p-AKT and FGFR2 were down-regulated in xenograft tumor. These results indicate that TAX suppresses the proliferation of androgen-independent PCa cells via inhibiting the activation of PI3K/AKT signaling pathway and the expression of FGFR2, which means TAX may be a novel anti-tumor agent for later PCa treatment.
Collapse
Affiliation(s)
- Jinqiu Yang
- School of Clinical Medicine, Dali University, Dali, 671013, Yunnan, China
| | - Chulin Xin
- School of Basic Medical Sciences, Dali University, 22 Wanhua Road, Dali, 671013, Yunnan, China
| | - Guangfen Yin
- The First Affiliated Hospital of Dali University, Dali, 671013, Yunnan, China
| | - Juan Li
- School of Basic Medical Sciences, Dali University, 22 Wanhua Road, Dali, 671013, Yunnan, China.
| |
Collapse
|
16
|
Tram VTN, Khoa Ta HD, Anuraga G, Dung PVT, Xuan DTM, Dey S, Wang CY, Liu YN. Dysbindin Domain-Containing 1 in Prostate Cancer: New Insights into Bioinformatic Validation of Molecular and Immunological Features. Int J Mol Sci 2023; 24:11930. [PMID: 37569304 PMCID: PMC10418609 DOI: 10.3390/ijms241511930] [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: 06/14/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 08/13/2023] Open
Abstract
Prostate cancer (PCa) is one of the most prevalent cancers in men, yet its pathogenic pathways remain poorly understood. Transcriptomics and high-throughput sequencing can help uncover cancer diagnostic targets and understand biological circuits. Using prostate adenocarcinoma (PRAD) datasets of various web-based applications (GEPIA, UALCAN, cBioPortal, SR Plot, hTFtarget, Genome Browser, and MetaCore), we found that upregulated dysbindin domain-containing 1 (DBNDD1) expression in primary prostate tumors was strongly correlated with pathways involving the cell cycle, mitotic in KEGG, WIKI, and REACTOME database, and transcription factor-binding sites with the DBNDD1 gene in prostate samples. DBNDD1 gene expression was influenced by sample type, cancer stage, and promoter methylation levels of different cancers, such as PRAD, liver hepatocellular carcinoma (LIHC), and lung adenocarcinoma (LUAD). Regulation of glycogen synthase kinase (GSK)-3β in bipolar disorder and ATP/ITP/GTP/XTP/TTP/CTP/UTP metabolic pathways was closely correlated with the DBNDD1 gene and its co-expressed genes in PCa. DBNDD1 gene expression was positively associated with immune infiltration of B cells, Myeloid-derived suppressor cell (MDSC), M2 macrophages, andneutrophil, whereas negatively correlated with CD8+ T cells, T follicular helper cells, M1 macrophages, and NK cells in PCa. These findings suggest that DBNDD1 may serve as a viable prognostic marker not only for early-stage PCa but also for immunotherapies.
Collapse
Affiliation(s)
- Van Thi Ngoc Tram
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Medical Laboratory, University Medical Center Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Hoang Dang Khoa Ta
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; (H.D.K.T.); (G.A.); (P.V.T.D.); (D.T.M.X.); (S.D.)
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan
| | - Gangga Anuraga
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; (H.D.K.T.); (G.A.); (P.V.T.D.); (D.T.M.X.); (S.D.)
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan
- Department of Statistics, Faculty of Science and Technology, Universitas PGRI Adi Buana, Surabaya 60234, Indonesia
| | - Phan Vu Thuy Dung
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; (H.D.K.T.); (G.A.); (P.V.T.D.); (D.T.M.X.); (S.D.)
| | - Do Thi Minh Xuan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; (H.D.K.T.); (G.A.); (P.V.T.D.); (D.T.M.X.); (S.D.)
| | - Sanskriti Dey
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; (H.D.K.T.); (G.A.); (P.V.T.D.); (D.T.M.X.); (S.D.)
| | - Chih-Yang Wang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; (H.D.K.T.); (G.A.); (P.V.T.D.); (D.T.M.X.); (S.D.)
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan
| | - Yen-Nien Liu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; (H.D.K.T.); (G.A.); (P.V.T.D.); (D.T.M.X.); (S.D.)
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
17
|
Liu J, Zhang R, Su T, Zhou Q, Gao L, He Z, Wang X, Zhao J, Xing Y, Sun F, Cai W, Wang X, Han J, Qin R, Désaubry L, Han B, Chen W. Targeting PHB1 to inhibit castration-resistant prostate cancer progression in vitro and in vivo. J Exp Clin Cancer Res 2023; 42:128. [PMID: 37210546 DOI: 10.1186/s13046-023-02695-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/01/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND Castration-resistant prostate cancer (CRPC) is currently the main challenge for prostate cancer (PCa) treatment, and there is an urgent need to find novel therapeutic targets and drugs. Prohibitin (PHB1) is a multifunctional chaperone/scaffold protein that is upregulated in various cancers and plays a pro-cancer role. FL3 is a synthetic flavagline drug that inhibits cancer cell proliferation by targeting PHB1. However, the biological functions of PHB1 in CRPC and the effect of FL3 on CRPC cells remain to be explored. METHODS Several public datasets were used to analyze the association between the expression level of PHB1 and PCa progression as well as outcome in PCa patients. The expression of PHB1 in human PCa specimens and PCa cell lines was examined by immunohistochemistry (IHC), qRT-PCR, and Western blot. The biological roles of PHB1 in castration resistance and underlying mechanisms were investigated by gain/loss-of-function analyses. Next, in vitro and in vivo experiments were conducted to investigate the anti-cancer effects of FL3 on CRPC cells as well as the underlying mechanisms. RESULTS PHB1 expression was significantly upregulated in CRPC and was associated with poor prognosis. PHB1 promoted castration resistance of PCa cells under androgen deprivation condition. PHB1 is an androgen receptor (AR) suppressive gene, and androgen deprivation promoted the PHB1 expression and its nucleus-cytoplasmic translocation. FL3, alone or combined with the second-generation anti-androgen Enzalutamide (ENZ), suppressed CRPC cells especially ENZ-sensitive CRPC cells both in vitro and in vivo. Mechanically, we demonstrated that FL3 promoted trafficking of PHB1 from plasma membrane and mitochondria to nucleus, which in turn inhibited AR signaling as well as MAPK signaling, yet promoted apoptosis in CRPC cells. CONCLUSION Our data indicated that PHB1 is aberrantly upregulated in CRPC and is involved in castration resistance, as well as providing a novel rational approach for treating ENZ-sensitive CRPC.
Collapse
Affiliation(s)
- Junmei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ranran Zhang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tong Su
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qianqian Zhou
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lin Gao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zongyue He
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xin Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jian Zhao
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Yuanxin Xing
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong First Medical University, Jinan, China
| | - Feifei Sun
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenjie Cai
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xinpei Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingying Han
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ruixi Qin
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, China
| | - Laurent Désaubry
- INSERM, UMR 1260, Regenerative Nanomedicine, University of Strasbourg, FMTS (Fédération de Médecine Translationnelle de L'Université de Strasbourg), Strasbourg, France
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, China.
| | - Weiwen Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
18
|
Omoboyede V, Ibrahim O, Umar HI, Oke GA, Onile OS, Chukwuemeka PO. Computer-aided analysis of quercetin mechanism of overcoming docetaxel resistance in docetaxel-resistant prostate cancer. J Genet Eng Biotechnol 2023; 21:47. [PMID: 37099169 PMCID: PMC10133427 DOI: 10.1186/s43141-023-00498-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 04/20/2023] [Indexed: 04/27/2023]
Abstract
BACKGROUND Prostate cancer (PC) is a silent but potent killer among men. In 2018, PC accounted for more than 350, 000 death cases while more than 1.2 million cases were diagnosed. Docetaxel, a chemotherapeutic drug belonging to the taxane family of drugs, is one of the most potent drugs in combating advanced PC. However, PC cells often evolve resistance against the regimen. Hence, necessitating the search for complementary and alternative therapies. Quercetin, a ubiquitous phytocompound with numerous pharmacological properties, has been reported to reverse docetaxel resistance (DR) in docetaxel-resistant prostate cancer (DRPC). Therefore, this study aimed to explore the mechanism via which quercetin reverses DR in DRPC using an integrative functional network and exploratory cancer genomic data analyses. RESULTS The putative targets of quercetin were retrieved from relevant databases, while the differentially expressed genes (DEGs) in docetaxel-resistant prostate cancer (DRPC) were identified by analysing microarray data retrieved from the Gene Expression Omnibus (GEO) database. Subsequently, the protein-protein interaction (PPI) network of the overlapping genes between the DEGs and quercetin targets was retrieved from STRING, while the hub genes, which represent the key interacting genes of the network, were identified using the CytoHubba plug-in of Cytoscape. The hub genes were further subjected to a comprehensive analysis aimed at identifying their contribution to the immune microenvironment and overall survival (OS) of PC patients, while their alterations in PC patients were also revealed. The biological roles played by the hub genes in chemotherapeutic resistance include the positive regulation of developmental process, positive regulation of gene expression, negative regulation of cell death, and epithelial cell differentiation among others. CONCLUSION Further analysis revealed epidermal growth factor receptor (EGFR) as the most pertinent target of quercetin in reversing DR in DRPC, while molecular docking simulation revealed an effective interaction between quercetin and EGFR. Ultimately, this study provides a scientific rationale for the further exploration of quercetin as a combinational therapy with docetaxel.
Collapse
Affiliation(s)
- Victor Omoboyede
- Department of Biochemistry, School of Life Sciences (SLS), Federal University of Technology Akure, Akure, P.M.B 704, Nigeria.
- Computer-Aided Therapeutics Laboratory (CATL), School of Life Sciences (SLS), Federal University of Technology Akure, Akure, P.M.B 704, Nigeria.
- Computer Aided Therapeutics Discovery and Design (CATDD) Group, School of Life Sciences (SLS), Federal University of Technology Akure, Akure, P.M.B 704, Nigeria.
| | - Ochapa Ibrahim
- Computer-Aided Therapeutics Laboratory (CATL), School of Life Sciences (SLS), Federal University of Technology Akure, Akure, P.M.B 704, Nigeria
- Computer Aided Therapeutics Discovery and Design (CATDD) Group, School of Life Sciences (SLS), Federal University of Technology Akure, Akure, P.M.B 704, Nigeria
- Department of Food Science and Technology, School of Agriculture and Agricultural Technology (SAAT), Federal University of Technology Akure, Akure, P.M.B 704, Nigeria
| | - Haruna Isiyaku Umar
- Department of Biochemistry, School of Life Sciences (SLS), Federal University of Technology Akure, Akure, P.M.B 704, Nigeria
- Computer Aided Therapeutics Discovery and Design (CATDD) Group, School of Life Sciences (SLS), Federal University of Technology Akure, Akure, P.M.B 704, Nigeria
| | - Grace Ayomide Oke
- Department of Food Science and Technology, School of Agriculture and Agricultural Technology (SAAT), Federal University of Technology Akure, Akure, P.M.B 704, Nigeria
| | - Olugbenga Samson Onile
- Biotechnology Programme, Department of Biological Sciences, Elizade University, P.M.B, 002 Ilara-Mokin, Ilara-Mokin, 340271, Nigeria
| | - Prosper Obed Chukwuemeka
- Computer-Aided Therapeutics Laboratory (CATL), School of Life Sciences (SLS), Federal University of Technology Akure, Akure, P.M.B 704, Nigeria
- Computer Aided Therapeutics Discovery and Design (CATDD) Group, School of Life Sciences (SLS), Federal University of Technology Akure, Akure, P.M.B 704, Nigeria
- Department of Biotechnology, School of Life Sciences (SLS), Federal University of Technology Akure, Akure, P.M.B 704, Nigeria
| |
Collapse
|
19
|
Krause W. Resistance to prostate cancer treatments. IUBMB Life 2022; 75:390-410. [PMID: 35978491 DOI: 10.1002/iub.2665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/09/2022] [Indexed: 12/14/2022]
Abstract
A review of the current treatment options for prostate cancer and the formation of resistance to these regimens has been compiled including primary, acquired, and cross-resistance. The diversification of the pathways involved and the escape routes the tumor is utilizing have been addressed. Whereas early stages of tumor can be cured, there is no treatment available after a point of no return has been reached, leaving palliative treatment as the only option. The major reasons for this outcome are the heterogeneity of tumors, both inter- and intra-individually and the nearly endless number of escape routes, which the tumor can select to overcome the effects of treatment. This means that more focus should be applied to the individualization of both diagnosis and therapy of prostate cancer. In addition to current treatment options, novel drugs and ongoing clinical trials have been addressed in this review.
Collapse
|
20
|
Marchetti C. Calcium signaling in prostate cancer cells of increasing malignancy. Biomol Concepts 2022; 13:156-163. [PMID: 35334188 DOI: 10.1515/bmc-2022-0012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/04/2022] [Indexed: 12/16/2022] Open
Abstract
Calcium signaling controls a large variety of cell functions, including proliferation and apoptosis, and plays a major role in neoplastic transformation. Prostate cancer (PCa) is one of the most common malignancies in men. The transition to castration-resistant prostate cancer (CRPC), a lethal form that is still lacking an effective cure, could be influenced by fine tuning intracellular calcium ([Ca2+]i) homeostasis. This study investigates [Ca2+]i dynamics in metastatic PCa cell lines that mimic the progression of PCa to CRPC: (i) well differentiated LNCaP cells that require androgen for survival, and (ii) poorly differentiated, highly aggressive androgen-insensitive prostate cancer (AIPC) PC3 and DU145 cells. In AIPC cells, ATP induces a fast rise in [Ca2+]i, due to release from intracellular stores and sensitive to phospholipase C inhibitors, while LNCaP cells do not respond to ATP challenge. Moreover, AIPC cells showed a reduced capacity to store Ca2+ in thapsigargin-sensitive stores and limited store-operated calcium entry, with respect to androgen-dependent LNCaP cells. Finally, green tea extract causes [Ca2+]i elevation and inhibits proliferation in PC3 and DU145 cells, but is ineffective in LNCaP cells. The consequences of these differences are discussed and interpreted in this study with reference to previously proposed models for Ca2+ dependence of prostate carcinogenesis.
Collapse
Affiliation(s)
- Carla Marchetti
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, via De Marini, 6, 16149 Genova, Italy
| |
Collapse
|
21
|
Liang X, Wang Y, Pei L, Tan X, Dong C. Identification of Prostate Cancer Risk Genetics Biomarkers Based on Intergraded Bioinformatics Analysis. Front Surg 2022; 9:856446. [PMID: 35372462 PMCID: PMC8967941 DOI: 10.3389/fsurg.2022.856446] [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: 01/17/2022] [Accepted: 02/09/2022] [Indexed: 12/04/2022] Open
Abstract
Background Prostate cancer (PCa) is one of the most popular cancer types in men. Nevertheless, the pathogenic mechanisms of PCa are poorly understood. Hence, we aimed to identify the potential genetic biomarker of PCa in the present study. Methods High-throughput data set GSE46602 was obtained from the comprehensive gene expression database (GEO) for screening differentially expressed genes (DEGs). The common DEGs were further screened out using The Cancer Genome Atlas (TCGA) dataset. Functional enrichment analysis includes Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) to study related mechanisms. The Cox and Lasso regression analyses were carried out to compress the target genes and construct the high-risk and low-risk gene model. Survival analyses were performed based on the gene risk signature model. The CIBERSORT algorithm was performed to clarify the correlation of the high- and low-risk gene model in risk and infiltration of immune cells in PCa. Results A total of 385 common DEGs were obtained. The results of functional enrichment analysis show that common DEGs play an important role in PCa. A three-gene signature model (KCNK3, AK5, and ARHGEF38) was established, and the model was significantly associated with cancer-related pathways, overall survival (OS), and tumor microenvironment (TME)-related immune cells in PCa. Conclusion This new risk model may contribute to further investigation in the immune-related pathogenesis in progression of PCa.
Collapse
Affiliation(s)
| | | | | | | | - Chunhui Dong
- Department of Urology, The Fourth Hospital of Hebei Medical University, Shijiahzuang, China
| |
Collapse
|
22
|
Habrowska-Górczyńska DE, Kozieł MJ, Kowalska K, Piastowska-Ciesielska AW. FOXO3a and Its Regulators in Prostate Cancer. Int J Mol Sci 2021; 22:ijms222212530. [PMID: 34830408 PMCID: PMC8625444 DOI: 10.3390/ijms222212530] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 01/01/2023] Open
Abstract
Forkhead box O3 (FOXO3a) is a member of a subfamily of forkhead transcription factors involved in the basic processes within a cell, including proliferation, apoptosis, cell cycle regulation, and DNA damage. As a transcription factor, FOXO3a is involved in the response to cellular stress, UV radiation, or oxidative stress. Its regulation is based on the modification of proteins as well as regulation by other proteins, e.g., growth factors. FOXO3a is commonly deregulated in cancer cells, and its inactivation is associated with initiation and progression of tumorigenesis, suggesting its role as a tumor suppressor; however, its role is still disputed and seems to be dependent on upstream signaling. Nevertheless, FOXO3a serves as an interesting potential target in therapies as it is regulated during treatment with very common anti-cancer drugs such as paclitaxel, cisplatin, docetaxel, and doxorubicin. This review aims to update the reported role of FOXO3a in prostate cancer (PCa), with a focus on its regulators that might serve as potential therapeutic agents in PCa therapy.
Collapse
|