1
|
Deniz C, Gedik T, Yüksel B, Cebeci E, Şahin F. Anti-cancer effect of sodium pentaborate in combination with cisplatin on lung cancer cell lines. Mol Biol Rep 2024; 52:28. [PMID: 39612011 DOI: 10.1007/s11033-024-10119-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Despite the development of novel therapeutic modalities, lung cancer persists as the leading cause of cancer-related mortality. Platinum-based treatments represent the most prominent treatment option, with cisplatin being the most frequently utilized chemotherapeutic agent. However, cisplatin has several serious side effects. A substantial body of evidence has emerged in recent years indicating that boron compounds exhibit anti-cancer properties when administered as monotherapy or in combination with chemotherapy agents. The objective of this study is to examine the anti-cancer effects of Cisplatin (Cis) and sodium pentaborate pentahydrate (NaB), both individually and in combination, on non-small cell lung cancer (NSCLC) cell line A-549 cells and small cell lung cancer (SCLC) cell line DMS-114 cells under in vitro conditions. METHODS AND RESULTS The effects of cisplatin and NAB on cell survival, apoptosis, the cell cycle, and the expression levels of apoptotic, anti-apoptotic, and tumor suppressor genes were determined by an MTS assay, an Annexin-V assay, a cell cycle analysis, and real-time PCR (qPCR). It was found that the IC-50 value of cisplatin, which was 10 µM when used alone, decreased to 2.5 µM when combined with a non-toxic dose of NaB on the A-549 cell line. BAX and TP53 gene expression levels were elevated by the Nab-Cisplatin combination in the A-549 cell line. The combination was observed to result in an approximately 19-fold increase in CDK2 gene expression in the A-549 cell line and an approximately 6-fold increase in the DMS-114 line, which resulted in S phase and/or G2 phase arrest on the cell cycle. Gene expression levels of Survivin and Ki-67 were decreased by the combination on both cell lines when compared with cisplatin alone. The findings demonstrate that NaB exerts an anti-cancer effect on the A-549 and DMS-114 cell lines. Moreover, when combined with cisplatin, it produces a synergistic anti-cancer effect on the A-549 cell line, whereby apoptosis is activated and cell proliferation is inhibited. CONCLUSION The combination of NaB and cisplatin represents a novel approach to the treatment of NSCLC. This is due to the fact that it reduces the IC-50 value of cisplatin and also results in a greater inhibition of cell division and a stronger induction of cell death when used in the context of a combined treatment. Further insight into the effects of the NaB-cisplatin combination will be gained from in vivo experiments and clinical studies.
Collapse
Affiliation(s)
- Cansın Deniz
- Faculty of Engineering, Department of Genetics and Bioengineering, Yeditepe University, Kayışdağı, Istanbul, 34755, Turkey
| | - Talar Gedik
- Faculty of Engineering, Department of Genetics and Bioengineering, Yeditepe University, Kayışdağı, Istanbul, 34755, Turkey
| | - Büşra Yüksel
- Faculty of Engineering, Department of Genetics and Bioengineering, Yeditepe University, Kayışdağı, Istanbul, 34755, Turkey
| | - Emre Cebeci
- Faculty of Engineering, Department of Genetics and Bioengineering, Yeditepe University, Kayışdağı, Istanbul, 34755, Turkey
| | - Fikrettin Şahin
- Faculty of Engineering, Department of Genetics and Bioengineering, Yeditepe University, Kayışdağı, Istanbul, 34755, Turkey.
| |
Collapse
|
2
|
Kaszubski J, Gagat M, Grzanka A, Wawrzyniak A, Niklińska W, Łapot M, Żuryń A. Cyclin-Dependent Kinase Inhibitors in the Rare Subtypes of Melanoma Therapy. Molecules 2024; 29:5239. [PMID: 39598629 PMCID: PMC11596694 DOI: 10.3390/molecules29225239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Melanoma occurs in various forms and body areas, not only in the cutis, but also in mucous membranes and the uvea. Rarer subtypes of that cancer differ in genomic aberrations, which cause their minor sensibility to regular cutaneous melanoma therapies. Therefore, it is essential to discover new strategies for treating rare forms of melanoma. In recent years, interest in applying CDK inhibitors (CDKIs) in cancer therapy has grown, as they are able to arrest the cell cycle and inhibit cell proliferation. Current studies highlight selective CDK4/6 inhibitors, like palbociclib or abemaciclib, as a very promising therapeutic option, since they were accepted by the FDA for advanced breast cancer treatment. However, cells of every subtype of melanoma do not react to CDKIs the same way, which is partly because of the genetic differences between them. Herein, we discuss the past and current research relevant to targeting various CDKs in mucosal, uveal and acral melanomas. We also briefly describe the issue of amelanotic and desmoplastic types of melanoma and the need to do more research to discover cell cycle dysregulations, which cause the growth of the mentioned forms of cancer.
Collapse
Affiliation(s)
- Jonatan Kaszubski
- Vascular Biology Student Research Club, Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-067 Bydgoszcz, Poland;
| | - Maciej Gagat
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-067 Bydgoszcz, Poland; (A.G.); (A.Ż.)
- Faculty of Medicine, Collegium Medicum, Mazovian Academy in Płock, 09-402 Płock, Poland;
| | - Alina Grzanka
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-067 Bydgoszcz, Poland; (A.G.); (A.Ż.)
| | - Agata Wawrzyniak
- Department of Histology and Embryology, Institute of Medical Sciences, College of Medical Sciences of the University of Rzeszow, University of Rzeszów, 35-310 Rzeszów, Poland;
| | - Wiesława Niklińska
- Department of Histology and Embryology, Medical University of Bialystok, 15-269 Białystok, Poland;
| | - Magdalena Łapot
- Faculty of Medicine, Collegium Medicum, Mazovian Academy in Płock, 09-402 Płock, Poland;
| | - Agnieszka Żuryń
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-067 Bydgoszcz, Poland; (A.G.); (A.Ż.)
| |
Collapse
|
3
|
Liu Y, Deng Y, Yang C, Naranmandura H. Double-Faced Immunological Effects of CDK4/6 Inhibitors on Cancer Treatment: Challenges and Perspectives. Bioengineering (Basel) 2024; 11:1084. [PMID: 39593745 PMCID: PMC11591775 DOI: 10.3390/bioengineering11111084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Cyclin-dependent kinases (CDKs) are generally involved in the progression of cell cycle and cell division in normal cells, while abnormal activations of CDKs are deemed to be a driving force for accelerating cell proliferation and tumorigenesis. Therefore, CDKs have become ideal therapeutic targets for cancer treatment. The U.S FDA has approved three CDK4/6 inhibitors (CDK4/6is) for the treatment of patients with hormone receptor-positive (HR+) or human epidermal growth factor receptor 2-negative (HER2-) advanced or metastatic breast cancer, and these drugs showed impressive results in clinics. Besides cell-cycle arrest, there is growing evidence that CDK4/6is exert paradoxical roles on cancer treatment by altering the immune system. Indeed, clinical data showed that CDK4/6is could change the immune system to exert antitumor effects, while these changes also caused tumor resistance to CDK4/6i. However, the molecular mechanism for the regulation of the immune system by CDK4/6is is unclear. In this review, we comprehensively discuss the paradoxical immunological effects of CDK4/6is in cancer treatment, elucidating their anticancer mechanisms through immunomodulatory activity and induction of acquired drug resistance by dysregulating the immune microenvironment. More importantly, we suggest a few strategies including combining CDK4/6is with immunotherapy to overcome drug resistance.
Collapse
Affiliation(s)
- Yongqin Liu
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Hematology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yiying Deng
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Hematology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chang Yang
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Hematology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hua Naranmandura
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Hematology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
4
|
Lu C, Li X, Fang C, Li C, Xu Y, Guo Y. Pretreatment of artesunate promoted hepatocyte proliferation by activating the PI3K/Akt/mTOR signaling pathway in mice. Acta Cir Bras 2024; 39:e394324. [PMID: 39476067 PMCID: PMC11506702 DOI: 10.1590/acb394324] [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: 03/25/2024] [Accepted: 06/05/2024] [Indexed: 11/02/2024] Open
Abstract
PURPOSE Artesunate (ART) has been implicated in regulating the many processes of liver injury, but its roles in liver regeneration still need to be illustrated. METHODS In the present study, ART was used to pretreat hepatocyte cell line NCTC1469 to study the effect of ART on hepatocyte proliferation in vitro. Furthermore, the potency of ART as a regimen to promote liver regeneration and restore liver function was evaluated following partial hepatectomy (PH) on C57BL/6 mice. RESULTS ART concentration-dependently promoted hepatocyte proliferation and reduced apoptosis. Cell cycle and Ki-67 immunohistochemical analyses demonstrated that ART supplementation promoted the proliferation of hepatocytes and accelerated liver regeneration. Our results provided evidence that ART improved liver function in a dose-dependent manner, as indicated by decreased serum alanine aminotransferase, aspartate aminotransferase, and increased albumin, and hepatocyte growth factor levels in PH mice. Meanwhile, ART promoted the PI3K/Akt/mTOR signaling in NCTC1469 cells and liver tissue of PH mice. In addition, PI3K inhibitor LY294002 blocked the promotion effect of ART on hepatocyte proliferation and cell cycle progression. CONCLUSION ART promoted hepatocyte proliferation via activation of the PI3K/Akt/mTOR pathway, which was beneficial to liver regeneration of PH-induced liver injury.
Collapse
Affiliation(s)
- Changyou Lu
- The Affliated Traditional Chinese Medicine Hospital, Southwest Medical University – Department of Hepatobiliary and Pancreatic Surgery – Luzhou (Sichuan) – China
| | - Xinkai Li
- The Affliated Traditional Chinese Medicine Hospital, Southwest Medical University – Department of Hepatobiliary and Pancreatic Surgery – Luzhou (Sichuan) – China
| | - Chao Fang
- The Affliated Traditional Chinese Medicine Hospital, Southwest Medical University – Department of Hepatobiliary and Pancreatic Surgery – Luzhou (Sichuan) – China
| | - Chuntao Li
- The Affliated Traditional Chinese Medicine Hospital, Southwest Medical University – Department of Hepatobiliary and Pancreatic Surgery – Luzhou (Sichuan) – China
| | - Yunke Xu
- The Affliated Traditional Chinese Medicine Hospital, Southwest Medical University – Department of Hepatobiliary and Pancreatic Surgery – Luzhou (Sichuan) – China
| | - Yong Guo
- The Affliated Traditional Chinese Medicine Hospital, Southwest Medical University – Department of Hepatobiliary and Pancreatic Surgery – Luzhou (Sichuan) – China
| |
Collapse
|
5
|
Hou X, Shi W, Luo W, Luo Y, Huang X, Li J, Ji N, Chen Q. FUS::DDIT3 Fusion Protein in the Development of Myxoid Liposarcoma and Possible Implications for Therapy. Biomolecules 2024; 14:1297. [PMID: 39456230 PMCID: PMC11506083 DOI: 10.3390/biom14101297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
The FUS::DDIT3 fusion protein, formed by the chromosomal translocation t (12;16) (q13;p11), is found in over 90% of myxoid liposarcoma (MLS) cases and is a crucial protein in its development. Many studies have explored the role of FUS::DDIT3 in MLS, and the prevailing view is that FUS::DDIT3 inhibits adipocyte differentiation and promotes MLS growth and invasive migration by functioning as an aberrant transcription factor that affects gene expression and regulates its downstream molecules. As fusion proteins are gradually showing their potential as targets for precision cancer therapy, FUS::DDIT3 has also been investigated as a therapeutic target. Drugs that target FUS::DDIT3 and its downstream molecules for treating MLS are widely utilized in both clinical practice and experimental studies, and some of them have demonstrated promising results. This article reviews the findings of relevant research, providing an overview of the oncogenic mechanisms of the FUS::DDIT3 fusion protein in MLS, as well as recent advancements in its therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ning Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (X.H.); (W.S.); (W.L.); (Y.L.); (X.H.); (J.L.); (Q.C.)
| | | |
Collapse
|
6
|
de Brevern AG. Special Issue: "Molecular Dynamics Simulations and Structural Analysis of Protein Domains". Int J Mol Sci 2024; 25:10793. [PMID: 39409122 PMCID: PMC11477144 DOI: 10.3390/ijms251910793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
The 3D protein structure is the basis for all their biological functions [...].
Collapse
Affiliation(s)
- Alexandre G. de Brevern
- DSIMB Bioinformatics Team, BIGR, INSERM, Université Paris Cité, F-75015 Paris, France; ; Tel.: +33-1-4449-3000
- DSIMB Bioinformatics Team, BIGR, INSERM, Université de la Réunion, F-97715 Saint Denis, France
| |
Collapse
|
7
|
Hawash M. Advances in Cancer Therapy: A Comprehensive Review of CDK and EGFR Inhibitors. Cells 2024; 13:1656. [PMID: 39404419 PMCID: PMC11476325 DOI: 10.3390/cells13191656] [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: 08/23/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
Protein kinases have essential responsibilities in controlling several cellular processes, and their abnormal regulation is strongly related to the development of cancer. The implementation of protein kinase inhibitors has significantly transformed cancer therapy by modifying treatment strategies. These inhibitors have received substantial FDA clearance in recent decades. Protein kinases have emerged as primary objectives for therapeutic interventions, particularly in the context of cancer treatment. At present, 69 therapeutics have been approved by the FDA that target approximately 24 protein kinases, which are specifically prescribed for the treatment of neoplastic illnesses. These novel agents specifically inhibit certain protein kinases, such as receptor protein-tyrosine kinases, protein-serine/threonine kinases, dual-specificity kinases, nonreceptor protein-tyrosine kinases, and receptor protein-tyrosine kinases. This review presents a comprehensive overview of novel targets of kinase inhibitors, with a specific focus on cyclin-dependent kinases (CDKs) and epidermal growth factor receptor (EGFR). The majority of the reviewed studies commenced with an assessment of cancer cell lines and concluded with a comprehensive biological evaluation of individual kinase targets. The reviewed articles provide detailed information on the structural features of potent anticancer agents and their specific activity, which refers to their ability to selectively inhibit cancer-promoting kinases including CDKs and EGFR. Additionally, the latest FDA-approved anticancer agents targeting these enzymes were highlighted accordingly.
Collapse
Affiliation(s)
- Mohammed Hawash
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus P.O. Box 7, Palestine
| |
Collapse
|
8
|
Jiang L, Zhang Z, Luo Z, Li L, Yuan S, Cui M, He K, Xiao J. Rupatadine inhibits colorectal cancer cell proliferation through the PIP5K1A/Akt/CDK2 pathway. Biomed Pharmacother 2024; 176:116826. [PMID: 38838507 DOI: 10.1016/j.biopha.2024.116826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/22/2024] [Accepted: 05/26/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Phosphatidylinositol-4-phosphate 5-kinase type 1 alpha (PIP5K1A) acts upstream of the Akt regulatory pathway and is abnormally expressed in many types of malignancies. However, the role and mechanism of PIP5K1A in colorectal cancer (CRC) have not yet been reported. In this study, we aimed to determine the association between PIP5K1A and progression of CRC and assess the efficacy and mechanism by which rupatadine targets PIP5K1A. METHODS Firstly, expression and function of PIP5K1A in CRC were investigated by human colon cancer tissue chip analysis and cell proliferation assay. Next, rupatadine was screened by computational screening and cytotoxicity assay and interactions between PIP5K1A and rupatadine assessed by kinase activity detection assay and bio-layer interferometry analysis. Next, rupatadine's anti-tumor effect was evaluated by in vivo and in vitro pharmacodynamic assays. Finally, rupatadine's anti-tumor mechanism was explored by quantitative real-time reverse-transcription polymerase chain reaction, western blot, and immunofluorescence. RESULTS We found that PIP5K1A exerts tumor-promoting effects as a proto-oncogene in CRC and aberrant PIP5K1A expression correlates with CRC malignancy. We also found that rupatadine down-regulates cyclin-dependent kinase 2 and cyclin D1 protein expression by inhibiting the PIP5K1A/Akt/GSK-3β pathway, induces cell cycle arrest, and inhibits CRC cell proliferation in vitro and in vivo. CONCLUSIONS PIP5K1A is a potential drug target for treating CRC. Rupatadine, which targets PIP5K1A, could serve as a new option for treating CRC, its therapeutic mechanism being related to regulation of the Akt/GSK-3β signaling pathway.
Collapse
Affiliation(s)
- Lei Jiang
- China Pharmaceutical University, Nanjing 210000, China; Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Zhibo Zhang
- China Pharmaceutical University, Nanjing 210000, China; Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Zhaofeng Luo
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Luan Li
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China
| | - Shengtao Yuan
- China Pharmaceutical University, Nanjing 210000, China
| | - Min Cui
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China.
| | - Ke He
- Minimally Invasive Tumor Therapies Center, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510310, China.
| | - Jing Xiao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China; Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China.
| |
Collapse
|
9
|
Liang JD, Zhang YE, Qin F, Chen WN, Jiang WM, Fang Z, Liang XL, Zhang Q, Li J. Molecular docking and MD simulation studies of 4-thiazol-N-(pyridin-2-yl)pyrimidin-2-amine derivatives as novel inhibitors targeted to CDK2/4/6. J Cancer Res Clin Oncol 2024; 150:302. [PMID: 38856753 PMCID: PMC11164762 DOI: 10.1007/s00432-024-05818-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 05/22/2024] [Indexed: 06/11/2024]
Abstract
PURPOSE Nowadays, cyclin-dependent kinase 4/6 (CDK4/6) inhibitors have been approved for treating metastatic breast cancer and have achieved inspiring curative effects. But some discoveries have indicated that CDK 4/6 are not the requisite factors in some cell types because CDK2 partly compensates for the inhibition of CDK4/6. Thus, it is urgent to design CDK2/4/6 inhibitors for significantly enhancing their potency. This study aims to explore the mechanism of the binding of CDK2/4/6 kinases and their inhibitors to design novel CDK2/4/6 inhibitors for significantly enhancing their potency in different kinds of cancers. MATERIALS AND METHODS A series of 72 disparately functionalized 4-substituted N-phenylpyrimidin-2-amine derivatives exhibiting potent inhibitor activities against CDK2, CDK4 and CDK6 were collected to apply to this research. The total set of these derivatives was divided into a training set (54 compounds) and a test set (18 compounds). The derivatives were constructed through the sketch molecule module in SYBYL 6.9 software. A Powell gradient algorithm and Tripos force field were used to calculate the minimal structural energy and the minimized structure was used as the initial conformation for molecular docking. By the means of 3D-QSAR models, partial least squares (PLS) analysis, molecular dynamics (MD) simulations and binding free energy calculations, we can find the relationship between structure and biological activity. RESULTS In this study, we used molecular docking, 3D-QSAR and molecular dynamics simulation methods to comprehensively analyze the interaction and structure-activity relationships of 72 new CDK2/4/6 inhibitors. We used detailed statistical data to reasonably verify the constructed 3D-QSAR models for three receptors (q2 of CDK2 = 0.714, R2pred = 0.764, q2 = 0.815; R2pred of CDK4 = 0.681, q2 = 0.757; R2pred of CDK6 = 0.674). MD simulations and decomposition energy analysis validated the reasonability of the docking results and identified polar interactions as crucial factors that influence the different bioactivities of the studied inhibitors of CDK2/4/6 receptors, especially the electrostatic interactions of Lys33/35/43 and Asp145/158/163. The nonpolar interaction with Ile10/12/19 was also critical for the differing potencies of the CDK2/4/6 inhibitors. We concluded that the following probably enhanced the bioactivity against CDK2/4/6 kinases: (1) electronegative groups at the N1-position and electropositive and moderate-sized groups at ring E; (2) electrogroups featured at R2; (3) carbon atoms at the X-position or ring C replaced by a benzene ring; and (4) an electrogroup as R4. CONCLUSION Previous studies, to our knowledge, only utilized a single approach of 3D-QSAR and did not integrate this method with other sophisticated techniques such as molecular dynamics simulations to discover new potential inhibitors of CDK2, CDK4, or CDK6. So we applied the intergenerational technology, such as 3D-QSAR technology, molecular docking simulation techniques, molecular dynamics simulations and MMPBSA19/MMGBSA20-binding free energy calculations to statistically explore the correlations between the structure with biological activities. The constructed 3D-QSAR models of the three receptors were reasonable and confirmed by the excellent statistical data. We hope the results obtained from this work will provide some useful references for the development of novel CDK2/4/6 inhibitors.
Collapse
Affiliation(s)
- Jia-Dong Liang
- Department of Head and Neck Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510000, People's Republic of China
| | - Yu-E Zhang
- Department of Pharmacy, The Affiliated Jiangmen TCM Hospital of Jinan University, No. 30 Huayuan East Road, Jiangmen, 529000, People's Republic of China
| | - Fei Qin
- Department of Nursing, The Linyi Mental Health Center, Linyi, People's Republic of China
| | - Wan-Na Chen
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510000, People's Republic of China
| | - Wen-Mei Jiang
- Department of Head and Neck Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Zeng Fang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510000, People's Republic of China
| | - Xiao-Li Liang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510000, People's Republic of China
| | - Quan Zhang
- Department of Head and Neck Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Jie Li
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, Guangzhou, 510623, Guangdong, People's Republic of China.
| |
Collapse
|
10
|
Zhao YQ, Ren YF, Li BB, Wei C, Yu B. The mysterious association between adiponectin and endometriosis. Front Pharmacol 2024; 15:1396616. [PMID: 38813109 PMCID: PMC11133721 DOI: 10.3389/fphar.2024.1396616] [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: 03/06/2024] [Accepted: 04/29/2024] [Indexed: 05/31/2024] Open
Abstract
Adiponectin is a pleiotropic cytokine predominantly derived from adipose tissue. In addition to its role in regulating energy metabolism, adiponectin may also be related to estrogen-dependent diseases, and many studies have confirmed its involvement in mediating diverse biological processes, including apoptosis, autophagy, inflammation, angiogenesis, and fibrosis, all of which are related to the pathogenesis of endometriosis. Although many researchers have reported low levels of adiponectin in patients with endometriosis and suggested that it may serve as a protective factor against the development of the disease. Therefore, the purpose of this review was to provide an up-to-date summary of the roles of adiponectin and its downstream cytokines and signaling pathways in the aforementioned biological processes. Further systematic studies on the molecular and cellular mechanisms of action of adiponectin may provide novel insights into the pathophysiology of endometriosis as well as potential therapeutic targets.
Collapse
Affiliation(s)
| | | | - Bing-Bing Li
- College of Integrated Chinese and Western Medicine, Jining Medical University, Jining, Shandong Province, China
| | | | | |
Collapse
|
11
|
Wei X, Xu H, Zhou M, Zhou Q, Li M, Liu Y. Chemically modified microRNA delivery via DNA tetrahedral frameworks for dental pulp regeneration. J Nanobiotechnology 2024; 22:150. [PMID: 38575923 PMCID: PMC11318316 DOI: 10.1186/s12951-024-02393-9] [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: 01/28/2024] [Accepted: 03/10/2024] [Indexed: 04/06/2024] Open
Abstract
Dental pulp regeneration is a promising strategy for addressing tooth disorders. Incorporating this strategy involves the fundamental challenge of establishing functional vascular networks using dental pulp stem cells (DPSCs) to support tissue regeneration. Current therapeutic approaches lack efficient and stable methods for activating DPSCs. In the study, we used a chemically modified microRNA (miRNA)-loaded tetrahedral-framework nucleic acid nanostructure to promote DPSC-mediated angiogenesis and dental pulp regeneration. Incorporating chemically modified miR-126-3p into tetrahedral DNA nanostructures (miR@TDNs) represents a notable advancement in the stability and efficacy of miRNA delivery into DPSCs. These nanostructures enhanced DPSC proliferation, migration, and upregulated angiogenesis-related genes, enhancing their paracrine signaling effects on endothelial cells. This enhanced effect was substantiated by improvements in endothelial cell tube formation, migration, and gene expression. Moreover, in vivo investigations employing matrigel plug assays and ectopic dental pulp transplantation confirmed the potential of miR@TDNs in promoting angiogenesis and facilitating dental pulp regeneration. Our findings demonstrated the potential of chemically modified miRNA-loaded nucleic acid nanostructures in enhancing DPSC-mediated angiogenesis and supporting dental pulp regeneration. These results highlighted the promising role of chemically modified nucleic acid-based delivery systems as therapeutic agents in regenerative dentistry and tissue engineering.
Collapse
Affiliation(s)
- Xiaoling Wei
- Shanghai Stomatological Hospital and School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Huaxing Xu
- Shanghai Stomatological Hospital and School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Mengqi Zhou
- Shanghai Stomatological Hospital and School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Qiangqiang Zhou
- Shanghai Stomatological Hospital and School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Mingqiang Li
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Yuehua Liu
- Shanghai Stomatological Hospital and School of Stomatology, Fudan University, Shanghai, 200001, China.
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China.
| |
Collapse
|
12
|
Liu X, Wang LL, Duan CY, Rong YR, Liang YQ, Zhu QX, Hao GP, Wang FZ. Daurisoline inhibits proliferation, induces apoptosis, and enhances TRAIL sensitivity of breast cancer cells by upregulating DR5. Cell Biol Int 2024. [PMID: 38563483 DOI: 10.1002/cbin.12162] [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: 11/25/2023] [Revised: 02/27/2024] [Accepted: 03/17/2024] [Indexed: 04/04/2024]
Abstract
Daurisoline (DS) is an isoquinoline alkaloid that exerts anticancer activities in various cancer cells. However, the underlying mechanisms through which DS affects the survival of breast cancer cells remain poorly understood. Therefore, the present study was undertaken to investigate the potential anticancer effect of DS on breast cancer cells and reveal the mechanism underlying the enhanced tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis by DS. Cell counting kit-8 (CCK-8) and 5-ethynyl-2-deoxyuridine (EdU) assay were used to evaluate the ability of cell proliferation. Flow cytometry was selected to examine the cell cycle distribution. TUNEL assay was used to detect the cell apoptosis. The protein expression was measured by Western blot analysis. DS was found to reduce the cell viability and suppress the proliferation of MCF-7 and MDA-MB-231 cells by causing G1 phase cell cycle arrest. DS could trigger apoptosis by promoting the cleavage of caspase-8 and PARP. The phosphorylation of ERK, JNK, and p38MAPK was upregulated clearly following DS treatment. Notably, SP600125 (JNK inhibitor) pretreatment significantly abrogated DS-induced PARP cleavage. DS inactivated Akt/mTOR and Wnt/β-catenin signaling pathway and upregulated the expression of ER stress-related proteins. Additionally, DS amplified TRAIL-caused viability reduction and apoptosis in breast cancer cells. Mechanismly, DS upregulated the protein level of DR4 and DR5, and knockdown of DR5 attenuated the cotreatment-induced cleavage of PARP. Inhibition of JNK could block DS-induced upregulation of DR5. This study provides valuable insights into the mechanisms of DS inhibiting cell proliferation, triggering apoptosis, and enhancing TRAIL sensitivity of breast cancer cells.
Collapse
Affiliation(s)
- Xin Liu
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Lin-Lin Wang
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Cun-Yu Duan
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Yan-Ru Rong
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Ya-Qi Liang
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Qing-Xiang Zhu
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Gang-Ping Hao
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Feng-Ze Wang
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
- Center Laboratory, The Second Affiliated Hospital of Shandong First Medical University, Taian, People's Republic of China
| |
Collapse
|
13
|
Khameneh SC, Sari S, Razi S, Yousefi AM, Bashash D. Inhibition of PI3K/AKT signaling using BKM120 reduced the proliferation and migration potentials of colorectal cancer cells and enhanced cisplatin-induced cytotoxicity. Mol Biol Rep 2024; 51:420. [PMID: 38483663 DOI: 10.1007/s11033-024-09339-2] [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] [Received: 07/08/2023] [Accepted: 02/07/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Although extensive efforts have been made to improve the treatment of colorectal cancer (CRC) patients, the prognosis for these patients remains poor. A wide range of anti-cancer agents has been applied to ameliorate the clinical management of CRC patients; however, drug resistance develops in nearly all patients. Based on the prominent role of PI3K/AKT signaling in the development of CRC and current interest in the application of PI3K inhibitors, we aimed to disclose the exact mechanism underlying the efficacy of BKM120, a well-known pan-class I PI3K inhibitor, in CRC-derived SW480 cells. MATERIALS AND METHODS The effects of BKM120 on SW480 cells were studied using MTT assay, cell cycle assay, Annexin V/PI apoptosis tests, and scratch assay. In the next step, qRT-PCR was used to investigate the underlying molecular mechanisms by which the PI3K inhibitor could suppress the survival of SW480 cells. RESULT The results of the MTT assay showed that BKM120 could decrease the metabolic activity of SW480 cells in a concentration and time-dependent manner. Investigating the exact mechanism of BKM120 showed that this PI3K inhibitor induces its anti-survival effects through a G2/M cell cycle arrest and apoptosis-mediated cell death. Moreover, the scratch assay demonstrated that PI3K inhibition led to the inhibition of cancer invasion and inhibition of PI3K/AKT signaling remarkably sensitized SW480 cells to Cisplatin. CONCLUSION Based on our results, inhibition of PI3K/AKT signaling can be a promising approach, either as a single modality or in combination with Cisplatin. However, further clinical studies should be performed to improve our understanding.
Collapse
Affiliation(s)
- Sepideh Chodary Khameneh
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Soyar Sari
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sara Razi
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Amir-Mohammad Yousefi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Horvath RM, Brumme ZL, Sadowski I. Small molecule inhibitors of transcriptional cyclin-dependent kinases impose HIV-1 latency, presenting "block and lock" treatment strategies. Antimicrob Agents Chemother 2024; 68:e0107223. [PMID: 38319085 PMCID: PMC10923280 DOI: 10.1128/aac.01072-23] [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: 08/17/2023] [Accepted: 01/09/2024] [Indexed: 02/07/2024] Open
Abstract
Current antiretroviral therapy for HIV-1 infection does not represent a cure for infection as viral rebound inevitably occurs following discontinuation of treatment. The "block and lock" therapeutic strategy is intended to enforce proviral latency and durably suppress viremic reemergence in the absence of other intervention. The transcription-associated cyclin-dependent protein kinases (tCDKs) are required for expression from the 5´ HIV-1 long-terminal repeat, but the therapeutic potential of inhibiting these kinases for enforcing HIV-1 latency has not been characterized. Here, we expanded previous observations to directly compare the effect of highly selective small molecule inhibitors of CDK7 (YKL-5-124), CDK9 (LDC000067), and CDK8/19 (Senexin A), and found each of these prevented HIV-1 provirus expression at concentrations that did not cause cell toxicity. Inhibition of CDK7 caused cell cycle arrest, whereas CDK9 and CDK8/19 inhibitors did not, and could be continuously administered to establish proviral latency. Upon discontinuation of drug administration, HIV immediately rebounded in cells that had been treated with the CDK9 inhibitor, while proviral latency persisted for several days in cells that had been treated with CDK8/19 inhibitors. These results identify the mediator kinases CDK8/CDK19 as potential "block and lock" targets for therapeutic suppression of HIV-1 provirus expression.
Collapse
Affiliation(s)
- Riley M. Horvath
- Department of Biochemistry and Molecular Biology Molecular Epigenetics Group, LSI, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zabrina L. Brumme
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | - Ivan Sadowski
- Department of Biochemistry and Molecular Biology Molecular Epigenetics Group, LSI, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
15
|
Wang B, Wang M, Li K, Wang C, Liu X, Rao Q, Song J, Hang Y, Liu S, Wen M, Huang L, Li Y. Calothrixin B derivatives induce apoptosis and cell cycle arrest on HEL cells through the ERK/Ras/Raf/MEK pathway. Biomed Pharmacother 2024; 171:116179. [PMID: 38278023 DOI: 10.1016/j.biopha.2024.116179] [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: 11/08/2023] [Revised: 01/06/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND Acute erythroleukemia (AEL) is acute myeloid leukemia characterized by malignant erythroid proliferation. AEL has a low survival rate, which has seriously threatened the health of older adults. Calothrixin B is a carbazole alkaloid isolated from the cyanobacteria Calothrix and exhibits anti-cancer activity. To discover more potential anti-erythroleukemia compounds, we used calothrixin B as the structural skeleton to synthesize a series of new compounds. METHODS In the cell culture model, we evaluated apoptosis and cell cycle arrest using MTT assay, flow cytometry analysis, JC-1 staining, Hoechst 33258 staining, and Western blot. Additionally, assessing the curative effect in the animal model included observation of the spleen, HE staining, flow cytometry analysis, and detection of serum biochemical indexes. RESULTS Among the Calothrixin B derivatives, H-107 had the best activity against leukemic cell lines. H-107 significantly inhibited the proliferation of HEL cells with an IC50 value of 3.63 ± 0.33 μM. H-107 induced apoptosis of HEL cells by damaging mitochondria and activating the caspase cascade and arrested HEL cells in the G0/G1 phase. Furthermore, H-107 downregulated the protein levels Ras, p-Raf, p-MEK, p-ERK and c-Myc. Pretreatment with ERK inhibitor (U0126) increased H-107-induced apoptosis. Thus, H-107 inhibited the proliferation of HEL cells by the ERK /Ras/Raf/MEK signal pathways. Interestingly, H-107 promoted erythroid differentiation into the maturation of erythrocytes and effectively activated the immune cells in erythroleukemia mice. CONCLUSION Overall, our findings suggest that H-107 can potentially be a novel chemotherapy for erythroleukemia.
Collapse
Affiliation(s)
- Bo Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China; College of Basic Medical, Guizhou Medical University, Guizhou 550004, China
| | - Ming Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China; College of Pharmacy, Guizhou Medical University, Guizhou 550004, China
| | - Ke Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Chaoyan Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China; College of Pharmacy, Guizhou Medical University, Guizhou 550004, China
| | - Xiang Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China; College of Basic Medical, Guizhou Medical University, Guizhou 550004, China
| | - Qing Rao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Jingrui Song
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Yubing Hang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Sheng Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China.
| | - Min Wen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guizhou 550004, China; College of Basic Medical, Guizhou Medical University, Guizhou 550004, China; College of Pharmacy, Guizhou Medical University, Guizhou 550004, China.
| | - Lei Huang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China.
| | - Yanmei Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China.
| |
Collapse
|
16
|
Almalki SG. The pathophysiology of the cell cycle in cancer and treatment strategies using various cell cycle checkpoint inhibitors. Pathol Res Pract 2023; 251:154854. [PMID: 37864989 DOI: 10.1016/j.prp.2023.154854] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/23/2023] [Accepted: 10/02/2023] [Indexed: 10/23/2023]
Abstract
The cell cycle is the series of events that occur in a cell leading to its division and duplication. It can be divided into two main stages: interphase and mitosis. Interphase is the longest stage of the cell cycle and can be further divided into three phases: G1, S, and G2. During G1, the cell grows and prepares for DNA synthesis. In the S phase, DNA synthesis occurs, leading to the replication of the genetic material. In G2, the cell continues to grow and prepares for mitosis. After mitosis, the cell enters the final stage of the cell cycle, called cytokinesis, during which the cytoplasm is divided, resulting in two separate daughter cells. The cell cycle then begins again with interphase. Cell cycle dysregulation is a hallmark of cancer, and it can have several consequences that contribute to the development and progression of cancer. Cyclin inhibitors and checkpoint activators have shown promise in the treatment of cancer, particularly in combination with other therapies.
Collapse
Affiliation(s)
- Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia.
| |
Collapse
|
17
|
Kargbo RB. Cyclin-Dependent Kinase Degradation via E3 Ligase Binding for Potential Disease Modulation in Cancer Treatment. ACS Med Chem Lett 2023; 14:1035-1037. [PMID: 37583814 PMCID: PMC10424305 DOI: 10.1021/acsmedchemlett.3c00275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Indexed: 08/17/2023] Open
Abstract
Cyclin-dependent kinases (CDKs) are crucial regulators of cell cycle progression and transcription. Their dysregulation has been implicated in various diseases, including cancer. This Patent Highlight focuses on recent advances in the development of CDK inhibitor-E3 ligase binding moiety conjugates for targeted CDK degradation and their potential applications in treating diseases modulated by CDKs. The exemplary PROTAC compounds exhibit a broad range of pharmacological activities associated with degradation of the CDK protein target for the treatment of cancer.
Collapse
|
18
|
Zhou Y, Li X, Luo P, Chen H, Zhou Y, Zheng X, Yin Y, Wei H, Liu H, Xia W, Shi M, Li X. Identification of abemaciclib derivatives targeting cyclin-dependent kinase 4 and 6 using molecular dynamics, binding free energy calculation, synthesis, and pharmacological evaluation. Front Pharmacol 2023; 14:1154654. [PMID: 37234717 PMCID: PMC10206264 DOI: 10.3389/fphar.2023.1154654] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
CDK4/6 plays a crucial role in various cancers and is an effective anticancer drug target. However, the gap between clinical requirements and approved CDK4/6 drugs is unresolved. Thus, there is an urgent need to develop selective and oral CDK4/6 inhibitors, particularly for monotherapy. Here, we studied the interaction between abemaciclib and human CDK6 using molecular dynamics simulations, binding free energy calculations, and energy decomposition. V101 and H100 formed stable hydrogen bonds with the amine-pyrimidine group, and K43 interacted with the imidazole ring via an unstable hydrogen bond. Meanwhile, I19, V27, A41, and L152 interacted with abemaciclib through π-alkyl interactions. Based on the binding model, abemaciclib was divided into four regions. With one region modification, 43 compounds were designed and evaluated using molecular docking. From each region, three favorable groups were selected and combined with each other to obtain 81 compounds. Among them, C2231-A, which was obtained by removing the methylene group from C2231, showed better inhibition than C2231. Kinase profiling revealed that C2231-A showed inhibitory activity similar to that of abemaciclib; additionally, C2231-A inhibited the growth of MDA-MB-231 cells to a greater extent than did abemaciclib. Based on molecular dynamics simulation, C2231-A was identified as a promising candidate compound with considerable inhibitory effects on human breast cancer cell lines.
Collapse
Affiliation(s)
- Yanting Zhou
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiandeng Li
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Peifang Luo
- Department of Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Huiting Chen
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yan Zhou
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Xueting Zheng
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Yuan Yin
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Haoche Wei
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongji Liu
- Department of Ophthalmology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Wen Xia
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Mingsong Shi
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Xiaoan Li
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| |
Collapse
|
19
|
Pandey P, Khan F, Upadhyay TK, Sharangi AB. Deciphering the Immunomodulatory Role of Cyclin-Dependent Kinase 4/6 Inhibitors in the Tumor Microenvironment. Int J Mol Sci 2023; 24:ijms24032236. [PMID: 36768557 PMCID: PMC9916547 DOI: 10.3390/ijms24032236] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Cancer is characterized by persistent cell proliferation driven by aberrant cell cycle regulation and stimulation of cyclin-dependent kinases (CDKs). A very intriguing and potential approach for the development of antitumor medicines is the suppression of CDKs that lead to induction of apoptosis and cell cycle arrest. The shift of the cell cycle from the G0/G1 phase to the S phase, which is characterized by active transcription and synthesis, depends on the development of the cyclin D-CDK4/6 complex. A precise balance between anticancer activity and general toxicity is demonstrated by CDK inhibitors, which can specifically block CDK4/6 and control the cell cycle by reducing the G1 to S phase transition. CDK4/6 inhibitors have recently been reported to exhibit significant cell growth inhibition via modulating the tumour microenvironment in cancerous cells. One significant new understanding is that these inhibitors serve important functions in the interaction among tumour cells and the host immune system in addition to being cytostatic. Herein, we discuss the biological significance of CDK4/6 inhibitors in cancer therapeutics, as well as their biological impact on T cells and other important immune cells. Furthermore, we explore the integration of preclinical findings of these pharmaceuticals' ability to enhance antitumor immunity.
Collapse
Affiliation(s)
- Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida 201306, India
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering and Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida 201306, India
- Correspondence:
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Centre of Research for Development, Parul University, Vadodara 391760, India
| | - Amit Baran Sharangi
- Department of Plantation Spices Medicinal and Aromatic Crops, Bidhan Chandra Krishi Viswavidyalaya, Mohanpur 741252, India
| |
Collapse
|
20
|
Zhao S, Wang H, Hu Z, Sahlu BW, Heng N, Gong J, Wang H, Zhu H. Identification of spermatogenesis-related lncRNA in Holstein bull testis after sexual maturity based on transcriptome analysis. Anim Reprod Sci 2022; 247:107146. [DOI: 10.1016/j.anireprosci.2022.107146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/29/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
|
21
|
P62/SQSTM1 mediates the autophagy-lysosome degradation of CDK2 protein undergoing PI3Kα/AKT T308 inhibition. Biochem Biophys Res Commun 2022; 627:5-11. [PMID: 36007335 DOI: 10.1016/j.bbrc.2022.08.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 11/22/2022]
Abstract
CDK2 forms a complex with cyclin A and cyclin E to promote the progress of cell cycle, but when cyclin A and cyclin E are dissociated from the complex and degraded by the ubiquitin proteasome pathway, the fate of the inactive CDK2 is unclear. In this study, we found that the inactive CDK2 protein was degraded by autophagy-lysosome pathway. In the classic model of G0/G1 phase arrest induced by serum starvation, we found that the mRNA level in CDK2 did not change but the protein level decreased. Subsequently, using PI3K and AKT inhibitors and gene knockout methods, it was found that CDK2 degradation was mediated by the inhibition of PI3Kα/AKTT308. In addition, P62/SQSTM1 was found to bind to the inactivated CDK2 protein to help it enter autophagy-lysosome degradation in a CTSB-dependent manner. Taken together, these results confirm that the PI3Kα/AKTT308 inhibition leads to degradation of CDK2 protein in the autophagy-lysosome pathway. These data reveal a new molecular mechanism of CDK2 protein degradation and provide a new strategy and method for regulating CDK2 protein.
Collapse
|