1
|
Shenoy AG, Ravi V, Vishwakarma R, Varghese S, Subair S, Vaswani R, Raju R, Revikumar A, Rehman N. Prostate Cancer and Tea: CYP17A1 Inhibition by Phytochemicals from Tea Plant Camellia sinensis L. and Implications for Anti-androgenic Effect. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2025. [PMID: 40391648 DOI: 10.1089/omi.2025.0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Camellia sinensis L., commonly known as the tea plant, produces derivatives such as green tea, which are among the most extensively consumed beverages worldwide. Green tea is rich in polyphenolic compounds, such as epigallocatechin-3-gallate (EGCG) and gallocatechin gallate. These phytochemicals have drawn particular attention as antioxidants, especially in relation to their potential to reduce the risks for prostate cancer (PC) among other common human diseases. However, the molecular evidence base needs to be strengthened before large-scale controlled clinical trials with C. sinensis L. and/or specific phytochemicals are pursued. We investigated cytochrome P45017A1 (CYP17A1), a key enzyme in androgen biosynthesis, as a molecular target for the green tea phytochemicals. In this study, molecular docking, pharmacokinetic and toxicity evaluations, molecular dynamics (MD) simulations, and post-MD simulation analyses were performed to assess the binding potential of green tea phytochemicals with the CYP17A1 enzyme. A library of 92 green tea-derived phytochemicals, along with the reference inhibitor abiraterone, was docked against the CYP17A1 enzyme. MD simulations validated the stability and enhanced binding affinity of the CYP17A1-EGCG complex compared with the abiraterone complex, as further confirmed by post-MD simulation analyses. Collectively, these findings suggest that EGCG inhibits CYP17A1, potentially reducing androgen biosynthesis and thereby highlighting green tea as a promising natural source for PC therapeutics. Further preclinical and translational studies are warranted to substantiate the clinical applicability of green tea phytochemicals.
Collapse
Affiliation(s)
- Aparna G Shenoy
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, India
| | - Vishal Ravi
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, India
| | - Riya Vishwakarma
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, India
| | - Susmi Varghese
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, India
| | - Suhail Subair
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, India
| | - Ravi Vaswani
- Department of Internal Medicine, Yenepoya Medical College, Yenepoya (Deemed to be University), Mangalore, India
| | - Rajesh Raju
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, India
| | - Amjesh Revikumar
- Kerala Genome Data Centre, Kerala Development and Innovation Strategic Council, Vazhuthacaud, Thiruvananthapuram, India
| | - Niyas Rehman
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, India
| |
Collapse
|
2
|
Matore BW, Murmu A, Banjare P, Vishvakarma NK, Roy PP, Singh J. Discovery of newer 1,3,4-Oxadiazole clubbed Isoindoline-1,3-dione derivatives as potential anticancer agents: Design, machine learning, synthesis, molecular docking, ADMET, DFT and MD simulation. Comput Biol Chem 2025; 118:108492. [PMID: 40306097 DOI: 10.1016/j.compbiolchem.2025.108492] [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: 02/12/2025] [Revised: 04/22/2025] [Accepted: 04/22/2025] [Indexed: 05/02/2025]
Abstract
Cancer remains to be the second leading cause of death, since the available drugs and therapies may get failure due to the early-stage drug resistance, metastasis, poor pharmacokinetics, and toxic effects. This gap can be fulfilled by designing potential anticancer agent with the Phthalimide as a prime scaffold. The robust and reliable pharmacophore model was used for the designing of newer Phthalimide derivatives. Additionally, we clubbed 1,3,4-Oxadiazole with Phthalimide to fulfil these features. The predicted IC50 for all the designed compounds are in µM range and DFT study also confirmed the reactive nature of these molecules. The designed compounds were synthesized and characterized by FT-IR, 1H NMR, 13C NMR and Mass spectroscopy. The in-vitro anticancer evaluation was carried out by performing MTT assay on MCF-7 and HCT-116 cancer cell lines. All compounds showed moderate to potent anticancer activity. The compound B19 was found to be the most potent against both the MCF-7 and HCT-116 with IC50 of 3.468 and 4.508 µM respectively. All the compounds showed good docking score in terms of binding affinity, lib dock score, CDOCKER interaction and binding free energy. MD Simulation study reviled good stability, compactness and rigidity of potent compound throughout the 100 ns run. ADMET results supports the good pharmacokinetics and lower toxicity. In conclusion, we suggest the compound B19 is potential drug-like candidate can be utilized in anticancer treatment on further confirmations. This study is widely useful for the medicinal chemists and scientific community.
Collapse
Affiliation(s)
- Balaji Wamanrao Matore
- Laboratory of Drug Discovery and Ecotoxicology, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur 495009, India
| | - Anjali Murmu
- Laboratory of Drug Discovery and Ecotoxicology, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur 495009, India
| | - Purusottam Banjare
- Laboratory of Drug Discovery and Ecotoxicology, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur 495009, India
| | - Naveen Kumar Vishvakarma
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur 495009, India
| | - Partha Pratim Roy
- Laboratory of Drug Discovery and Ecotoxicology, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur 495009, India
| | - Jagadish Singh
- Laboratory of Drug Discovery and Ecotoxicology, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur 495009, India.
| |
Collapse
|
3
|
Ogbodo UC, Salimat S, Bodun DS, Balogun TA, Omoboyowa DA. Design of small molecules for CDK-2 inhibition in colorectal cancer based on substructure search. J Biomol Struct Dyn 2025; 43:1305-1315. [PMID: 38088360 DOI: 10.1080/07391102.2023.2291546] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/23/2023] [Indexed: 01/16/2025]
Abstract
The global frequency of colorectal cancer motivates extensive drug discovery efforts. CDK2, a key member of the CDK family, has been linked to tumor progression, unregulated cell proliferation, and growth promotion. Water-soluble flavonoids with a fast metabolism called anthocyanins have been shown to have a variety of pharmacological properties, including anti-cancer properties. This study aims to find possible CDK2 inhibitors from Anthocyanin-like molecules. Anthocyanins sourced from PubChem were screened using a virtual screening approach that included a KNIME workflow, QSAR-model, Pharmacophore hypothesis, and a structure-based screening to identify compounds with a better binding affinity and predicted bioactivity compared to the standard, Sorafenib. The top compounds were subjected to a 100 ns MD simulation to confirm their stability at the active site. Compounds 1-5 were shown to have higher binding affinity and bioactivity in this study. These substances interacted with the critical amino acids (LEU 83, ASP 145 and LYS 89) at CDK2's active site. Compared to the reference with a pIC50 value of 6.003 nM, the top compounds listed have superior predicted bioactivity ranging from 6.539 to 6.36 nM. Also, ADMET predictions predicted that Compounds 1-5 were not carcinogenic and not a p-glycoprotein substrate. MD simulation also validated Compound 1's stability at the active site compared to the standard. This study uncovers potential CDK2 inhibitors with good binding affinities, shedding light on their interactions with the target protein. While promising, further in vivo and in vitro investigations are essential to validate the anticancer potential of these compounds.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Uchechukwu C Ogbodo
- Department of Applied Biochemistry, Faculty of Biosciences, Nnamdi Azikiwe University, Awka, Nigeria
| | - Sofela Salimat
- Department of Chemistry, University of Lagos, Lagos, Nigeria
| | - Damilola S Bodun
- Phyto-Medicine and Computational Biology Laboratory, Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Toheeb A Balogun
- Phyto-Medicine and Computational Biology Laboratory, Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Damilola A Omoboyowa
- Phyto-Medicine and Computational Biology Laboratory, Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| |
Collapse
|
4
|
Pandey SK, Sabharwal U, Tripathi S, Mishra A, Yadav N, Dwivedi-Agnihotri H. Androgen Signaling in Prostate Cancer: When a Friend Turns Foe. Endocr Metab Immune Disord Drug Targets 2025; 25:37-56. [PMID: 38831575 DOI: 10.2174/0118715303313528240523101940] [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: 02/29/2024] [Revised: 04/17/2024] [Accepted: 05/02/2024] [Indexed: 06/05/2024]
Abstract
Androgen (AR) signaling is the main signaling for the development of the prostate and its normal functioning. AR is highly specific for testosterone and dihydrotestosterone, significantly contributing to prostate development, physiology, and cancer. All these receptors have emerged as crucial therapeutic targets for PCa. In the year 1966, the Noble prize was awarded to Huggins and Hodge for their groundbreaking discovery of AR. As it is a pioneer transcription factor, it belongs to the steroid hormone receptor family and consists of domains, including DNA binding domain (DBD), hormone response elements (HRE), C-terminal ligand binding domain (LBD), and N-terminal regulatory domains. Structural variations in AR, such as AR gene amplification, LBD mutations, alternative splicing of exons, hypermethylation of AR, and co- regulators, are major contributors to PCa. It's signaling is crucial for the development and functioning of the prostate gland, with the AR being the key player. The specificity of AR for testosterone and dihydrotestosterone is important in prostate physiology. However, when it is dysregulated, AR contributes significantly to PCa. However, the structural variations in AR, such as gene amplification, mutations, alternative splicing, and epigenetic modifications, drive the PCa progression. Therefore, understanding AR function and dysregulation is essential for developing effective therapeutic strategies. Thus, the aim of this review was to examine how AR was initially pivotal for prostate development and how it turned out to show both positive and detrimental implications for the prostate.
Collapse
Affiliation(s)
- Swaroop Kumar Pandey
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, 281406, India
| | - Usha Sabharwal
- P. G. Department of Biosciences, Centre of Advanced Studies, Satellite Campus, Sardar Patel Maidan, 388120, Gujarat, India
| | - Swati Tripathi
- Section of Electron Microscopy, Supportive Centre for Brain Research, National Institute for Physiological Sciences (NIPS) Okazaki, 444-8787, Japan
| | - Anuja Mishra
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, 281406, India
| | - Neha Yadav
- Department of Biophysics, University of Delhi, South Campus, New Delhi, 110021, India
| | | |
Collapse
|
5
|
Irfandi R, Raya I, Ahmad A, Fudholi A, Riswandi, Santi S, Azalea WP, Putri SE, Alam MN, Supratman U, Olubode SO, Abdalrazaq EA, Kandeel M, Soekamto NH, Natsir H, Maming, Ramlawati. Design anticancer potential of Zn(II)isoleucinedithiocarbamate complex on MCF-7 cell lines: synthesis, characterization, molecular docking, molecular dynamic, ADMET, and in-vitro studies. Mol Divers 2024; 28:3199-3214. [PMID: 37884781 DOI: 10.1007/s11030-023-10747-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023]
Abstract
Cisplatin is a cancer medication widely used today, but it still poses some problems due to its toxic properties in the body. To overcome this issue, a new complex has been developed as a potential anticancer drug prospect by minimizing its toxic consequences. A novel Zn(II)IleDTC complex containing isoleucine dithiocarbamate ligands has been produced and analyzed using a range of analytical and spectroscopic methods. The Zn(II) IleDTC complex were characterized using various methods, including UV-Vis spectroscopy, FT-IR, determination of melting point, conductivity, and HOMO-LUMO analysis. Furthermore, computational NMR spectrum analysis was conducted in this study. Molecular docking studies was conducted to evaluate the potential of Zn(II) isoleucine dithiocarbamate as an HIF1 inhibitor. The results showed that the Zn complex exhibited a good docking score of -6.6 and formed hydrogen bonds with ARG 17, VAL264, and GLU15, alkyl bonds with TRP27 and LEU32, and Pi-Alkyl bonds with PRO41 and ARG44. This suggests that the Zn(II) isoleucine dithiocarbamate complex could be a promising candidate for cancer treatment with potential HIF1 inhibition properties. To assess the dynamic stability and efficacy of protein-ligand interactions over time, molecular dynamics simulations was conducted for both individual proteins and protein complexes. The cytotoxicity evaluation of Zn(II) isoleucine dithiocarbamate against MCF-7 cells obtained an IC50 value of 362.70 µg/mL indicating moderate cytotoxicity and morphological changes of cancer cells causing cancer cells to undergo apoptosis. The Zn(II) isoleucine dithiocarbamate complex may have promising potential as an anticancer compound due to its significant inhibitory effect on the breast cancer cell line (MCF7). According to the ADMET study, the complex exhibits drug-like characteristics with low toxicity, further supporting its potential as a viable drug candidate.
Collapse
Affiliation(s)
- Rizal Irfandi
- Doctoral Program, Department of Chemistry, Faculty of Mathematics, and Natural Science, Hasanuddin University, Makassar, 90245, Indonesia
- Department of Biology Education, Faculty of Teacher Training and Education, Universitas Puangrimaggalatung, Sengkang, 90915, Indonesia
| | - Indah Raya
- Department of Chemistry, Faculty of Mathematics, and Natural Science, Hasanuddin University, Makassar, 90245, Indonesia.
| | - Ahyar Ahmad
- Department of Chemistry, Faculty of Mathematics, and Natural Science, Hasanuddin University, Makassar, 90245, Indonesia
| | - Ahmad Fudholi
- Solar Energy Research Institute, Universiti Kebangsaan Malaysia, UKM, 43600, Bangi, Selangor, Malaysia
- Research Centre for Electrical Power and Mechatronics, Institute of Science (LIPI), Bandung, Indonesia
| | - Riswandi
- Department of Chemistry, Faculty of Mathematics, and Natural Science, Hasanuddin University, Makassar, 90245, Indonesia
| | - Santi Santi
- Medical Laboratory Technology, Faculty of Health Technology, Megarezky University, Makassar, 90234, Indonesia
| | - Wynda Puspa Azalea
- District Health Office, Faculty of Pharmacy, Pancasila University, Jakarta, 12620, Indonesia
| | - Suriati Eka Putri
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Negeri Makassar, Makassar, Jalan Daeng Tata Raya, Makassar, 90244, Indonesia
| | - Muhammad Nur Alam
- Doctoral Program, Department of Chemistry, Faculty of Mathematics, and Natural Science, Hasanuddin University, Makassar, 90245, Indonesia
| | - Unang Supratman
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| | - Samuel Olawale Olubode
- Department of Biochemistry, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Eid A Abdalrazaq
- Department of Chemistry, Faculty of Science, Al Hussein Bin Talal University, Ma'an, Jordan
| | - Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Nunuk Hariani Soekamto
- Department of Chemistry, Faculty of Mathematics, and Natural Science, Hasanuddin University, Makassar, 90245, Indonesia
| | - Hasnah Natsir
- Department of Chemistry, Faculty of Mathematics, and Natural Science, Hasanuddin University, Makassar, 90245, Indonesia
| | - Maming
- Department of Chemistry, Faculty of Mathematics, and Natural Science, Hasanuddin University, Makassar, 90245, Indonesia
| | - Ramlawati
- Department of Natural Science Education, Faculty of Mathematics and Natural Science, Universitas Negeri Makassar, Makassar, Indonesia
| |
Collapse
|
6
|
Wang J, Chen X, Sun W, Tang W, Chen J, Zhang Y, Li R, Wang Y. Expression of GLOD4 in the Testis of the Qianbei Ma Goat and Its Effect on Leydig Cells. Animals (Basel) 2024; 14:2611. [PMID: 39272396 PMCID: PMC11393997 DOI: 10.3390/ani14172611] [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: 08/07/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
The expression pattern of GLOD4 in the testis and its regulatory effect on testicular cells was explored in goats to enhance our understanding of spermatogenesis and improve reproduction in breeding rams. In this study, we demonstrated the localization of GLOD4 in testicular cells using immunohistochemistry and subcellular localization analyses. Subsequently, we analyzed the GLOD4 expression pattern in four age-based groups (0, 6, 12, and 18 months old) using real-time quantitative polymerase chain reaction (qRT-PCR) and protein blotting. Finally, we performed GLOD4 silencing and overexpression studies in Leydig cells (LCs) and explored the effects on cell proliferation, the cell cycle, steroid hormone secretion and the expression of candidate testosterone hormone-regulated genes. GLOD4 was mainly expressed in Leydig cells, and the subcellular localization results showed that the GLOD4 protein was mainly localized in the cytoplasm and nucleus. Silencing of GLOD4 significantly suppressed the mRNA expression levels of the testosterone secretion-related genes CYP11A1, 3β-HSD, and CYP17A1 and the mRNA expression levels of cell cycle-related genes CDK6, PCNA, and Cyclin E. Moreover, the cell cycle was blocked at the G2/M phase after GLOD4 silencing, which significantly suppressed testosterone secretion. In contrast, GLOD4 overexpression significantly increased the mRNA expression levels of the testosterone secretion-related genes CYP11A1, 3β-HSD, and CYP17A1 and increased the expression of the cell cycle-related genes CDK6, PCNA, and Cyclin E. Moreover, GLOD4 overexpression promoted the cell cycle from G0/G1 phases to enter the S phase and G2/M phases, promoted the secretion of testosterone. Taken together, our experimental results indicate that GLOD4 may affect the development of cells in Qianbei Ma goats of different ages by influencing the cell cycle, cell proliferation, and testosterone hormone synthesis. These findings enhance our understanding of the functions of GLOD4 in goats.
Collapse
Affiliation(s)
- Jinqian Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Xiang Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Wei Sun
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou 225009, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Wen Tang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Jiajing Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Yuan Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Ruiyang Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Yanfei Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
7
|
Song S, Tai L, Xu Y, Jiang J, Zhou L, Zhao J. Lathyrol reduces the RCC invasion and incidence of EMT via affecting the expression of AR and SPHK2 in RCC mice. Discov Oncol 2024; 15:264. [PMID: 38965120 PMCID: PMC11224167 DOI: 10.1007/s12672-024-01130-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/27/2024] [Indexed: 07/06/2024] Open
Abstract
OBJECTIVE To investigate the effects of Lathyrol on the expression of androgen receptor (AR) and sphingosine kinase 2 (SPHK2) in renal cell carcinoma (RCC) mice and to further explore the mechanism by which Lathyrol inhibits the invasion and incidence of epithelial-mesenchymal transition (EMT). METHODS An RCC xenograft mouse model was constructed, and the mice were randomly divided into a model group, an experiment group and a negative control group. The experiment group was intragastrically gavaged with Lathyrol solution (20 mg/kg), the model group was intragastrically gavaged with 0.9% NaCl (same volume as that used in the experiment group), and the negative control group was injected intraperitoneally with 2 mg/kg cisplatin aqueous solution. Changes in the body weight and tumor volume of the mice were recorded. Western blot (WB) was used to assess the protein expression levels of AR, p-AR, CYP17A1, PARP1, E-cadherin, N-cadherin, vimentin, α-SMA, β-catenin, and ZO-1. Protein expression levels of SPHK2, metal matrix protease 2 (MMP2), MMP9 and urokinase-type plasminogen activator (uPA) in tumor tissues were assessed by immunohistochemistry (IHC). AR expression in tumor tissues was assessed after immunofluorescence (IF) staining. RESULTS After 14 days of drug administration, compared with that in the model group, the tumor volumes in the negative control and experiment groups were lower; the difference in tumor volume among the model, control and experiment groups was statistically significant (P < 0.05). The differences in body weight among the three groups were not statistically significant (P > 0.05). In the model group, the protein expression levels of AR, p-AR, CYP17A1, SPHK2, and PARP1 were relatively increased, the protein expression levels of E-cadherin and ZO-1 were relatively reduced (P < 0.05), and the protein expression levels of N-cadherin, β-catenin, vimentin, and α-SMA were relatively increased (P < 0.05). In the negative control and experiment groups, the protein expression levels of AR, p-AR, CYP17A1, SPHK2, and PARP1 were relatively decreased (P < 0.05), the protein expression levels of E-cadherin and ZO-1 were relatively increased (P < 0.05), and the protein expression levels of N-cadherin, β-catenin, vimentin and α-SMA were relatively decreased (P < 0.05). CONCLUSION Lathyrol and cisplatin inhibit the proliferation of RCC xenografts, reduce the protein expression levels of AR, CYP17A1, SPHK2, PARP1, E-cadherin, and ZO-1 in tumor tissues (P < 0.05), and promote the protein expression levels of N-cadherin, β-catenin, vimentin and α-SMA (P < 0.05). Therefore, Lathyrol reduces RCC invasion and EMT by affecting the expression of AR and SPHK2 in RCC mice.
Collapse
Affiliation(s)
- Shengyou Song
- Department of Urology, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450002, Henan, China
| | - Lunwei Tai
- Department of Urology, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450002, Henan, China
| | - Yuqi Xu
- Department of Urology, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450002, Henan, China
| | - Junling Jiang
- Department of Urology, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Dongfeng Road 6#, Zhengzhou, 450002, Henan, China
| | - Lei Zhou
- Department of Urology, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Dongfeng Road 6#, Zhengzhou, 450002, Henan, China
| | - Junfeng Zhao
- Department of Urology, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Dongfeng Road 6#, Zhengzhou, 450002, Henan, China.
| |
Collapse
|
8
|
Ilesanmi A, Dairo G, Salimat S, Bodun DS, Awoyale B, Balogun TA. Identification of bioactive compounds from Vaccinium vitis-idaea L. (Lingonberry) as inhibitors for treating KRAS-associated cancer: a computational approach. In Silico Pharmacol 2023; 11:32. [PMID: 37915613 PMCID: PMC10616029 DOI: 10.1007/s40203-023-00165-1] [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: 07/29/2023] [Accepted: 10/04/2023] [Indexed: 11/03/2023] Open
Abstract
Lung cancer is the cancer of the lung's epithelial cells typically characterized by difficulty breathing, chest pain, blood-stained coughs, headache, and weight loss. If left unmanaged, lung cancer can spread to other body parts. While several treatment methods exist for managing lung cancer, exploring natural plant sources for developing therapeutics offers great potential in complementing other treatment approaches. In this study, we evaluated the bioactive compounds in Vaccinium vitis-idaea for treating KRAS-associated lung cancer types. In this study, we concentrated on inhibiting the mutated Kirsten rat sarcoma viral oncogene homolog (KRAS) by targeting an associated protein (Phosphodiesterase 6δ) to which KRAS form complexes. We evaluated bioactive compounds from Lingonberry (Vaccinium vitis-idaea L.), adopting computational approaches such as molecular docking, molecular dynamics simulation, molecular mechanics/generalized Born surface area (MM/GBSA) calculations, and pharmacokinetics analysis. A total of 26 out of 39 bioactive compounds of Vaccinium vitis-idaea L. had a higher binding affinity to the target receptor than an approved drug, Sotorasib. Also, further analyses of all lead/top compounds in this study identified (+)-Catechin (Cianidanol), Arbutin, Resveratrol, and Sinapic acid, to be potential drug candidates that could be pursued. In sum, Arbutin, (+)-Catechin, and Sinapic acid are predicted to be the top compound of Vaccinium vitis-idaea L. because of their pharmacokinetic properties and drug-likeness attributes. Also, their stability to the target receptor makes them a potential drug candidate that could be explored for treating KRAS mutation-associated lung cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-023-00165-1.
Collapse
Affiliation(s)
- Ayooluwa Ilesanmi
- Department of Sciences and Mathematics, Mississippi University for Women, Columbus, USA
| | - Gbenga Dairo
- Department of Biological Sciences, Western Illinois University, Macomb, IL USA
| | - Sofela Salimat
- Department of Chemistry, University of Lagos, Lagos, Nigeria
| | - Damilola S. Bodun
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Bibiire Awoyale
- Department of Chemistry, University of Ilorin, Ilorin, Nigeria
| | - Toheeb A. Balogun
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
- Department of Biological Sciences, University of California, San Diego, La Jolla, USA
| |
Collapse
|
9
|
Bodun DS, Omoboyowa DA, Omotuyi OI, Olugbogi EA, Balogun TA, Ezeh CJ, Omirin ES. QSAR-based virtual screening of traditional Chinese medicine for the identification of mitotic kinesin Eg5 inhibitors. Comput Biol Chem 2023; 104:107865. [PMID: 37062146 DOI: 10.1016/j.compbiolchem.2023.107865] [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: 02/06/2023] [Revised: 03/31/2023] [Accepted: 04/01/2023] [Indexed: 04/18/2023]
Abstract
Cell division is a crucial process for the growth and development of all living organisms. Unfortunately, uncontrolled cell division and growth is a hallmark of cancer, leading to the formation of tumors. The Human Eg5 protein, also known as the mitotic kinesin Eg5, plays a vital role in the regulation of cell division and its dysfunction has been linked to cancer development. This study aimed to identify new inhibitors of the Human Eg5 protein. Over 2000 Traditional Chinese Medicine (TCM) compounds were screened through a combination of virtual and structure-based screening methods. The top five compounds (Compounds 1-5) showed improved binding affinity to Human Eg5 compared to the standard drug Monastrol, as demonstrated by docking and MMGBSA scores, as well as interactions with key amino acids GLY 116 and GLY 118. The potential absorption and bioactivity of these compounds were also predicted through ADMET properties and a QSAR model, respectively, and showed improved results compared to the standard. Further quantum mechanics docking confirmed the better binding affinity of the lead compound, Compound 1. Our findings highlight Compound 1-5 as promising hits for inhibiting Human Eg5 and the need for experimental validation of their potential in treating cancer.
Collapse
Affiliation(s)
- Damilola S Bodun
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Ondo State, Nigeria.
| | - Damilola A Omoboyowa
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Ondo State, Nigeria
| | - Olaposi I Omotuyi
- Department of Pharmacology and Toxicology, College of Pharmacy, Afe Babalola University, Ado Ekiti, Nigeria
| | - Ezekiel A Olugbogi
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Ondo State, Nigeria
| | - Toheeb A Balogun
- Department of Biological Sciences, University of California, San Diego, San Diego, CA, United States
| | - Chiamaka J Ezeh
- Department of Biochemistry, Micheal Okpara University of Agriculture, Umudike, Abia State, Nigeria
| | - Emmanuel S Omirin
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Ondo State, Nigeria
| |
Collapse
|
10
|
Olubode SO, Bankole MO, Akinnusi PA, Adanlawo OS, Ojubola KI, Nwankwo DO, Edjebah OE, Adebesin AO, Ayodele AO. Molecular Modeling Studies of Natural Inhibitors of Androgen Signaling in Prostate Cancer. Cancer Inform 2022; 21:11769351221118556. [PMID: 35983016 PMCID: PMC9379963 DOI: 10.1177/11769351221118556] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
Prostate cancer is the second most common disease in men and the sixth leading cause of death from cancer globally, with 20 million men expected to be affected by 2024 thus considered as chronic illness which requires immediate attention. As an androgen-dependent illness that relies on the androgen receptor for development and progression, inhibition of the androgen receptor can lead to a therapeutic solution, hence serving as a vital therapeutic target. This study focused on the computational analysis of the inhibitory potentials of Vitis vinifera, a reported plant with anti-cancer properties, against androgen receptor employing molecular docking, ADMET studies, Binding energy study, pharmacophore modeling, and molecular dynamics simulation approaches. After the investigation, it was determined that 5 compounds: cis-piceid, cis-astrigin, gallocatechin, phlorizin, and trans-polydatin, might be possible androgen receptor inhibitors since they had higher docking scores and ADMET qualities than compared standards, with cis-piceid being the best-predicted inhibitor.
Collapse
Affiliation(s)
- Samuel O Olubode
- Department of Biochemistry, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Mutolib O Bankole
- Department of Chemical Sciences, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Precious A Akinnusi
- Department of Biochemistry, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Olayinka S Adanlawo
- Department of Chemical Sciences, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Kehinde I Ojubola
- Department of Chemical Sciences, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Daniel O Nwankwo
- Department of Biochemistry, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Onome E Edjebah
- Department of Pharmacology and Therapeutics, Delta State University, Delta State, Nigeria
| | - Ayomide O Adebesin
- Department of Biochemistry, Cancer Genomics Lab, Covenant University, Ota, Nigeria
| | - Abigail O Ayodele
- Center for Genomics Research and Innovation, National Biotechnology Development Agency, Abuja, Nigeria
| |
Collapse
|