1
|
Wang W, Jiang Q, Tao J, Zhang Z, Liu G, Qiu B, Hu Q, Zhang Y, Xie C, Song J, Jiang G, Zhong H, Zou Y, Li J, Lv S. A structure-based approach to discover a potential isomerase Pin1 inhibitor for cancer therapy using computational simulation and biological studies. Comput Biol Chem 2025; 114:108290. [PMID: 39586226 DOI: 10.1016/j.compbiolchem.2024.108290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 10/03/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Peptidyl-prolyl cis/trans isomerase Pin1 occupies a prominent role in preventing the development of certain malignant tumors. Pin1 is considered a target for the treatment of related malignant tumors, so the identification of novel Pin1 inhibitors is particularly urgent. In this study, we preliminarily predicted eight candidates from FDA-approved drug database as the potential Pin1 inhibitors through virtual screening combined with empirical screening. Therefore, we selected these eight candidates and tested their binding affinity and inhibitory activity against Pin1 using fluorescence titration and PPIase activity assays, respectively. Subsequently, we found that four FDA-approved drugs showed good binding affinities and inhibition effects. In addition, we also observed that bexarotene can reduce cell viability in a dose-dependent and time-dependent manner and induce apoptosis. Finally, we inferred that residues K63, R68 and R69 are important in the binding process between bexarotene and Pin1. All in all, repurposing of FDA-approved drugs to inhibit Pin1 may provide a promising insight into the identification and development of new treatments for certain malignant tumors.
Collapse
Affiliation(s)
- Wang Wang
- School of Basic Medicine, Nanchang Medical College, Nanchang 330006, PR China; Key Laboratory of Pharmacodynamics and Quality Evaluation on ant-inflammatory Chinese Herbs, Jiangxi Administration of Traditional Chinese Medicine, Nanchang 330006, PR China; Key Laboratory of Pharmacodynamics and Safety Evaluation, Health Commission of Jiangxi Province, Nanchang 330006, PR China
| | - Qizhou Jiang
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Jiaxin Tao
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Zhenxian Zhang
- School of Laboratory Medicine, Nanchang Medical College, Nanchang 330006, PR China
| | - GuoPing Liu
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Binxuan Qiu
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Qingyang Hu
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Yuxi Zhang
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Chao Xie
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Jiawen Song
- School of Laboratory Medicine, Nanchang Medical College, Nanchang 330006, PR China
| | - GuoZhen Jiang
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Hui Zhong
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Yanling Zou
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Jiaqi Li
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Shaoli Lv
- School of Basic Medicine, Nanchang Medical College, Nanchang 330006, PR China.
| |
Collapse
|
2
|
Liu S, Wang J. Recent Progress of Glutathione Peroxidase 4 Inhibitors in Cancer Therapy. Mini Rev Med Chem 2025; 25:42-57. [PMID: 38879766 DOI: 10.2174/0113895575308546240607073310] [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: 02/04/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 01/31/2025]
Abstract
Ferroptosis is a novel type of programmed cell death that relies on the build-up of intracellular iron and leads to an increase in toxic lipid peroxides. Glutathione Peroxidase 4 (GPX4) is a crucial regulator of ferroptosis that uses glutathione as a cofactor to detoxify cellular lipid peroxidation. Targeting GPX4 in cancer could be a promising strategy to induce ferroptosis and kill drugresistant cancers effectively. Currently, research on GPX4 inhibitors is of increasing interest in the field of anti-tumor agents. Many reviews have summarized the regulation and ferroptosis induction of GPX4 in human cancer and disease. However, insufficient attention has been paid to GPX4 inhibitors. This article outlines the molecular structures and development prospects of GPX4 inhibitors as novel anticancer agents.
Collapse
Affiliation(s)
- Shangde Liu
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorous Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China
| | - Jian Wang
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorous Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China
| |
Collapse
|
3
|
Liu C, Dan L, Li Q, Bajinka O, Yuan X. The mechanisms of Pin1 as targets for cancer therapy. Front Immunol 2024; 15:1482088. [PMID: 39624096 PMCID: PMC11609185 DOI: 10.3389/fimmu.2024.1482088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 10/25/2024] [Indexed: 01/03/2025] Open
Abstract
Targeted therapy has considerable promise for the effective eradication of cancer at the primary tumor site prior to subsequent metastasis. Using this therapeutic approach, gaining an understanding of mechanistic cancer models is essential for facilitating the inhibition or suppression of tumor growth. Among different oncogenes and proteins, the protein interacting with never-in-mitosis kinase-1 (Pin1) is particularly important. The interaction between Pin1 and phosphorylated threonine-proline motifs results in significant alterations in protein structure and function. In this review, we provide a comprehensive summary of the processes involving Pin1 and its mechanisms in the context of cancer therapy. Pin1 enhances signaling pathways in a number of different human cancers and plays a pivotal role in the suppressive mechanisms relevant to cancer treatment. It is essential for the regulation of proline-directed phosphorylation and for modulating tumor suppressors. Inhibitors of Pin1, particularly naturally occurring substances, have been found to inhibit the carcinogenic activity of Pin1, and consequently this protein could represent an excellent candidate for novel cancer treatment strategies, offering a valuable therapeutic target in carcinogenesis and treatment resistance.
Collapse
Affiliation(s)
- Chuanfeng Liu
- Department of Pulmonary and Critical Care Medicine, Lishui Hospital of Traditional Chinese Medicine, Lishui, China
| | - Lingying Dan
- Department of Endocrinology, Lishui Hospital of Traditional Chinese Medicine, Lishui, China
| | - Quan Li
- Department of Pulmonary and Critical Care Medicine, Lishui Hospital of Traditional Chinese Medicine, Lishui, China
| | - Ousman Bajinka
- School of Medicine and Allied Health Sciences, University of The Gambia, Banjul, Gambia
| | - Xingxing Yuan
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
4
|
Zhang Q, Li E, Zhang Y, Chen Y, Wang D, Wang S. Aggregation-Induced Emission-Active Organic Nanoagent with High Photothermal Conversion Efficiency for Near-Infrared Imaging-Guided Tumor Photothermal Therapy. ACS Biomater Sci Eng 2024; 10:6210-6217. [PMID: 39253844 DOI: 10.1021/acsbiomaterials.4c00959] [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] [Indexed: 09/11/2024]
Abstract
Photothermal therapy (PTT) provides a great prospect for noninvasive cancer therapy. However, it is still highly challenging to construct photothermal agents (PTAs) with the desired performances for imaging-guided PTT applications. Herein, a D-A-D-type naphthalene diamine (NDI)-based photothermal nano-PTAs NDS-BPN NP with near-infrared region (NIR) emission at 822 nm, aggregation-induced emission (AIE), high photothermal conversion efficiency (55.05%), and excellent photothermal stability is successfully designed and prepared through a simple two-step engineering method by using a new AIE molecule NDS-BPN and DSPE-PEG2000 as precursors. The prepared PTT nanoagents NDS-BPN NPs have been further applied for efficient photothermal ablation of cancer cells in vitro and also achieved the NIR fluorescent image-guided PTT tumor therapy in vivo with satisfactory results. We believe that this work provides an attractive NIR AIE NDI-based nano-PTA for the phototherapy of tumors as well as develops the construction strategy of NDI molecular-based photothermal nanoagents with desired performances for imaging-guided PTT.
Collapse
Affiliation(s)
- Qiang Zhang
- Anhui Province Engineering Research Center for Dental Materials and Application, Institute of Synthesis and Application of Medical Materials, Department of Pharmacy, Wannan Medical College, Wuhu 241002, P.R. China
| | - Enqi Li
- Anhui Province Engineering Research Center for Dental Materials and Application, Institute of Synthesis and Application of Medical Materials, Department of Pharmacy, Wannan Medical College, Wuhu 241002, P.R. China
| | - Youwei Zhang
- Anhui Province Engineering Research Center for Dental Materials and Application, Institute of Synthesis and Application of Medical Materials, Department of Pharmacy, Wannan Medical College, Wuhu 241002, P.R. China
| | - Yunyan Chen
- Anhui Province Engineering Research Center for Dental Materials and Application, Institute of Synthesis and Application of Medical Materials, Department of Pharmacy, Wannan Medical College, Wuhu 241002, P.R. China
| | - Dongmei Wang
- School of Laboratory Medicine, Wannan Medical College, Wuhu 241002, P.R. China
| | - Shaozhen Wang
- Anhui Province Engineering Research Center for Dental Materials and Application, Institute of Synthesis and Application of Medical Materials, Department of Pharmacy, Wannan Medical College, Wuhu 241002, P.R. China
| |
Collapse
|
5
|
Li Z, Zhang J, Wang J. Access to Chiral β-Boryl δ-Lactones via NHC-Catalyzed [4 + 2] Annulation. Org Lett 2024; 26:1965-1969. [PMID: 38418377 DOI: 10.1021/acs.orglett.4c00426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2024]
Abstract
We report a carbene-catalyzed [4 + 2] annulation of activated esters and β-borate enones, providing an efficient method to build enantioenriched organoborones with two consecutive stereogenic centers. It is worth noting that this protocol represents a new organocatalytic manner to generate chiral β-C-B bonds. Moreover, it also greatly enriches the structural diversity of the chiral organoboron compounds.
Collapse
Affiliation(s)
- Zhipeng Li
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorous Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, P. R. China
| | - Jingyang Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorous Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, P. R. China
| | - Jian Wang
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorous Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
6
|
Li Y, Yuan Z, Wang L, Yang J, Pu P, Le Y, Chen X, Wang C, Gao Y, Liu Y, Wang J, Gao X, Li Y, Wang H, Zou C. Prolyl isomerase Pin1 sculpts the immune microenvironment of colorectal cancer. Cell Signal 2024; 115:111041. [PMID: 38199598 DOI: 10.1016/j.cellsig.2024.111041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/14/2023] [Accepted: 01/06/2024] [Indexed: 01/12/2024]
Abstract
Pin1, a peptide prolyl cis-trans isomerase, is overexpressed and/or overactivated in many human malignancies. However, whether Pin1 regulates the immunosuppressive TME has not been well defined. In this study, we detected the effect of Pin1 on immune cells and immune checkpoint PD-L1 in the TME of CRC and explored the anti-tumor efficacy of Pin1 inhibitor ATRA combined with PD-1 antibody. We found that Pin1 facilitated the immunosuppressive TME by raising the proportion of myeloid-derived suppressor cells (MDSCs) and declining the percentage of CD8+ T cells and CD4+ T cells. Pin1 restrained PD-L1 protein expression in CRC cells and the effect was tempered by endoplasmic reticulum (ER) stress inducers. Mechanically, Pin1 overexpression decreased the stability of PD-L1 and promoted its degradation by mitigating ER stress. Silencing or inhibiting Pin1 promoted PD-L1 protein expression by inducing ER stress. Hence, Pin1 inhibitor ATRA enhanced the anti-tumor efficacy of PD-1 antibody in the CRC allograft by upregulating PD-L1. Our results reveal the critical and pleiotropic effects of Pin1 on managing the immune cells and immune checkpoint PD-L1 in the TME of CRC, providing a new promising candidate for combination with immunotherapy.
Collapse
Affiliation(s)
- Yang Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Zhongnan Yuan
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Linlin Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Jing Yang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Pei Pu
- The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Yunting Le
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - XianWei Chen
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Chongyang Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Yating Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Yi Liu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Jialin Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medicine Sciences, Harbin 150081, China; Key Laboratory of Cardiovascular Medicine Research of Harbin Medical University, Ministry of Education, Harbin 150081, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Yanze Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China.
| | - Hefei Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China.
| | - Chaoxia Zou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medicine Sciences, Harbin 150081, China.
| |
Collapse
|
7
|
Alzain AA, Elbadwi FA, Shoaib TH, Sherif AE, Osman W, Ashour A, Mohamed GA, Ibrahim SRM, Roh EJ, Hassan AHE. Integrating computational methods guided the discovery of phytochemicals as potential Pin1 inhibitors for cancer: pharmacophore modeling, molecular docking, MM-GBSA calculations and molecular dynamics studies. Front Chem 2024; 12:1339891. [PMID: 38318109 PMCID: PMC10839060 DOI: 10.3389/fchem.2024.1339891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Pin1 is a pivotal player in interactions with a diverse array of phosphorylated proteins closely linked to critical processes such as carcinogenesis and tumor suppression. Its axial role in cancer initiation and progression, coupled with its overexpression and activation in various cancers render it a potential candidate for the development of targeted therapeutics. While several known Pin1 inhibitors possess favorable enzymatic profiles, their cellular efficacy often falls short. Consequently, the pursuit of novel Pin1 inhibitors has gained considerable attention in the field of medicinal chemistry. In this study, we employed the Phase tool from Schrödinger to construct a structure-based pharmacophore model. Subsequently, 449,008 natural products (NPs) from the SN3 database underwent screening to identify compounds sharing pharmacophoric features with the native ligand. This resulted in 650 compounds, which then underwent molecular docking and binding free energy calculations. Among them, SN0021307, SN0449787 and SN0079231 showed better docking scores with values of -9.891, -7.579 and -7.097 kcal/mol, respectively than the reference compound (-6.064 kcal/mol). Also, SN0021307, SN0449787 and SN0079231 exhibited lower free binding energies (-57.12, -49.81 and -46.05 kcal/mol, respectively) than the reference ligand (-37.75 kcal/mol). Based on these studies, SN0021307, SN0449787, and SN0079231 showed better binding affinity that the reference compound. Further the validation of these findings, molecular dynamics simulations confirmed the stability of the ligand-receptor complex for 100 ns with RMSD ranging from 0.6 to 1.8 Å. Based on these promising results, these three phytochemicals emerge as promising lead compounds warranting comprehensive biological screening in future investigations. These compounds hold great potential for further exploration regarding their efficacy and safety as Pin1 inhibitors, which could usher in new avenues for combating cancer.
Collapse
Affiliation(s)
- Abdulrahim A. Alzain
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - Fatima A. Elbadwi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - Tagyedeen H. Shoaib
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - Asmaa E. Sherif
- Department of Pharmacognosy, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Wadah Osman
- Department of Pharmacognosy, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - Ahmed Ashour
- Department of Pharmacognosy, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Gamal A. Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sabrin R. M. Ibrahim
- Preparatory Year Program, Department of Chemistry, Batterjee Medical College, Jeddah, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Eun Joo Roh
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, University of Science and Technology, Daejeon, Republic of Korea
| | - Ahmed H. E. Hassan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|