1
|
Nemr MTM, Elshewy A, Ibrahim ML, El Kerdawy AM, Halim PA. Design, synthesis, antineoplastic activity of new pyrazolo[3,4-d]pyrimidine derivatives as dual CDK2/GSK3β kinase inhibitors; molecular docking study, and ADME prediction. Bioorg Chem 2024; 150:107566. [PMID: 38896936 DOI: 10.1016/j.bioorg.2024.107566] [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: 04/16/2024] [Revised: 06/06/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024]
Abstract
In the current study, novel pyrazolo[3,4-d]pyrimidine derivatives 5a-h were designed and synthesized as targeted anti-cancer agents through dual CDK2/GSK-3β inhibition. The designed compounds demonstrated moderate to potent activity on the evaluated cancer cell lines (MCF-7 and T-47D). Compounds 5c and 5 g showed the most promising cytotoxic activity against the tested cell lines surpassing that of the used reference standard; staurosporine. On the other hand, both compounds showed good safety and tolerability on normal fibroblast cell line (MCR5). The final compounds 5c and 5 g showed a promising dual CDK2/GSK-3β inhibitory activity with IC50 of 0.244 and 0.128 μM, respectively, against CDK2, and IC50 of 0.317 and 0.160 μM, respectively, against GSK-3β. Investigating the effect of compounds 5c and 5 g on CDK2 and GSK-3β downstream cascades showed that they reduced the relative cellular content of phosphorylated RB1 and β-catenin compared to that in the untreated MCF-7 cells. Moreover, compounds 5c and 5 g showed a reasonable selective inhibition against the target kinases CDK2/GSK-3β in comparison to a set of seven off-target kinases. Furthermore, the most potent compound 5 g caused cell cycle arrest at the S phase in MCF-7 cells preventing the cells' progression to G2/M phase inducing cell apoptosis. Molecular docking studies showed that the final pyrazolo[3,4-d]pyrimidine derivatives have analogous binding modes in the target kinases interacting with the hinge region key amino acids. Molecular dynamics simulations confirmed the predicted binding mode by molecular docking. Moreover, in silico predictions indicated their favorable physicochemical and pharmacokinetic properties in addition to their promising cytotoxic activity.
Collapse
Affiliation(s)
- Mohamed T M Nemr
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, P.O. Box 11562, Egypt
| | - Ahmed Elshewy
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, P.O. Box 11562, Egypt; Department of Medicinal Chemistry, Faculty of Pharmacy, Galala University, New Galala 43713, Egypt.
| | - Mohammed L Ibrahim
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, P.O. Box 11562, Egypt
| | - Ahmed M El Kerdawy
- School of Pharmacy, College of Health and Science, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln, United Kingdom; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, P.O. Box 11562, Egypt
| | - Peter A Halim
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, P.O. Box 11562, Egypt
| |
Collapse
|
2
|
Düster R, Anand K, Binder SC, Schmitz M, Gatterdam K, Fisher RP, Geyer M. Structural basis of Cdk7 activation by dual T-loop phosphorylation. Nat Commun 2024; 15:6597. [PMID: 39097586 PMCID: PMC11297931 DOI: 10.1038/s41467-024-50891-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 07/24/2024] [Indexed: 08/05/2024] Open
Abstract
Cyclin-dependent kinase 7 (Cdk7) is required in cell-cycle and transcriptional regulation owing to its function as both a CDK-activating kinase (CAK) and part of transcription factor TFIIH. Cdk7 forms active complexes by associating with Cyclin H and Mat1, and is regulated by two phosphorylations in the activation segment (T loop): the canonical activating modification at T170 and another at S164. Here we report the crystal structure of the human Cdk7/Cyclin H/Mat1 complex containing both T-loop phosphorylations. Whereas pT170 coordinates basic residues conserved in other CDKs, pS164 nucleates an arginine network unique to the ternary Cdk7 complex, involving all three subunits. We identify differential dependencies of kinase activity and substrate recognition on the individual phosphorylations. CAK function is unaffected by T-loop phosphorylation, whereas activity towards non-CDK substrates is increased several-fold by T170 phosphorylation. Moreover, dual T-loop phosphorylation stimulates multisite phosphorylation of the RNA polymerase II (RNAPII) carboxy-terminal domain (CTD) and SPT5 carboxy-terminal repeat (CTR) region. In human cells, Cdk7 activation is a two-step process wherein S164 phosphorylation precedes, and may prime, T170 phosphorylation. Thus, dual T-loop phosphorylation can regulate Cdk7 through multiple mechanisms, with pS164 supporting tripartite complex formation and possibly influencing processivity, while pT170 enhances activity towards key transcriptional substrates.
Collapse
Affiliation(s)
- Robert Düster
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kanchan Anand
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Sophie C Binder
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Maximilian Schmitz
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Karl Gatterdam
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Robert P Fisher
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Matthias Geyer
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
3
|
Gong Y, Li H. CDK7 in breast cancer: mechanisms of action and therapeutic potential. Cell Commun Signal 2024; 22:226. [PMID: 38605321 PMCID: PMC11010440 DOI: 10.1186/s12964-024-01577-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: 11/07/2023] [Accepted: 03/19/2024] [Indexed: 04/13/2024] Open
Abstract
Cyclin-dependent kinase 7 (CDK7) serves as a pivotal regulator in orchestrating cellular cycle dynamics and gene transcriptional activity. Elevated expression levels of CDK7 have been ubiquitously documented across a spectrum of malignancies and have been concomitantly correlated with adverse clinical outcomes. This review delineates the biological roles of CDK7 and explicates the molecular pathways through which CDK7 exacerbates the oncogenic progression of breast cancer. Furthermore, we synthesize the extant literature to provide a comprehensive overview of the advancement of CDK7-specific small-molecule inhibitors, encapsulating both preclinical and clinical findings in breast cancer contexts. The accumulated evidence substantiates the conceptualization of CDK7 as a propitious therapeutic target in breast cancer management.
Collapse
Affiliation(s)
- Ying Gong
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Huiping Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| |
Collapse
|
4
|
Düster R, Anand K, Binder SC, Schmitz M, Gatterdam K, Fisher RP, Geyer M. Structural basis of Cdk7 activation by dual T-loop phosphorylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.14.580246. [PMID: 38405971 PMCID: PMC10888979 DOI: 10.1101/2024.02.14.580246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Cyclin-dependent kinase 7 (Cdk7) occupies a central position in cell-cycle and transcriptional regulation owing to its function as both a CDK-activating kinase (CAK) and part of the general transcription factor TFIIH. Cdk7 forms an active complex upon association with Cyclin H and Mat1, and its catalytic activity is regulated by two phosphorylations in the activation segment (T loop): the canonical activating modification at T170 and another at S164. Here we report the crystal structure of the fully activated human Cdk7/Cyclin H/Mat1 complex containing both T-loop phosphorylations. Whereas pT170 coordinates a set of basic residues conserved in other CDKs, pS164 nucleates an arginine network involving all three subunits that is unique to the ternary Cdk7 complex. We identify differential dependencies of kinase activity and substrate recognition on individual phosphorylations within the Cdk7 T loop. The CAK function of Cdk7 is not affected by T-loop phosphorylation, whereas activity towards non-CDK substrates is increased several-fold by phosphorylation at T170. Moreover, dual T-loop phosphorylation at both T170 and S164 stimulates multi-site phosphorylation of transcriptional substrates-the RNA polymerase II (RNAPII) carboxy-terminal domain (CTD) and the SPT5 carboxy-terminal repeat (CTR) region. In human cells, Cdk7-regulatory phosphorylation is a two-step process in which phosphorylation of S164 precedes, and may prime, T170 phosphorylation. Thus, dual T-loop phosphorylation can regulate Cdk7 through multiple mechanisms, with pS164 supporting tripartite complex formation and possibly influencing Cdk7 processivity, while the canonical pT170 enhances kinase activity towards critical substrates involved in transcription.
Collapse
Affiliation(s)
- Robert Düster
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kanchan Anand
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Sophie C. Binder
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Maximilian Schmitz
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Karl Gatterdam
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Robert P. Fisher
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthias Geyer
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
5
|
Eldehna WM, Maklad RM, Almahli H, Al-Warhi T, Elkaeed EB, Abourehab MAS, Abdel-Aziz HA, El Kerdawy AM. Identification of 3-(piperazinylmethyl)benzofuran derivatives as novel type II CDK2 inhibitors: design, synthesis, biological evaluation, and in silico insights. J Enzyme Inhib Med Chem 2022; 37:1227-1240. [PMID: 35470754 PMCID: PMC9126595 DOI: 10.1080/14756366.2022.2062337] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the current work, a hybridisation strategy was adopted between the privileged building blocks, benzofuran and piperazine, with the aim of designing novel CDK2 type II inhibitors. The hybrid structures were linked to different aromatic semicarbazide, thiosemicarbazide, or acylhydrazone tails to anchor the designed inhibitors onto the CDK2 kinase domain. The designed compounds showed promising CDK2 inhibitory activity. Compounds 9h, 11d, 11e and 13c showed potent inhibitory activity (IC50 of 40.91, 41.70, 46.88, and 52.63 nM, respectively) compared to staurosporine (IC50 of 56.76 nM). Moreover, benzofurans 9e, 9h, 11d, and 13b showed promising antiproliferative activities towards different cancer cell lines, and non-significant cytotoxicity on normal lung fibroblasts MRC-5 cell line. Furthermore, a cell cycle analysis as well as Annexin V-FITC apoptosis assay on Panc-1 cell line were performed. Molecular docking simulations were performed to explore the ability of target benzofurans to adopt the common binding pattern of CDK2 type II inhibitors.
Collapse
Affiliation(s)
- Wagdy M Eldehna
- School of Biotechnology, Badr University in Cairo, Badr City, Egypt.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Raed M Maklad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt.,Institute of Drug Discovery and Development, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Hadia Almahli
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Tarfah Al-Warhi
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Eslam B Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Hatem A Abdel-Aziz
- Department of Applied Organic Chemistry, National Research Center, Dokki, Egypt
| | - Ahmed M El Kerdawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt.,Department of Pharmaceutical Chemistry, School of Pharmacy, NewGiza University (NGU), Cairo, Egypt
| |
Collapse
|
6
|
Eldehna WM, Al-Rashood ST, Al-Warhi T, Eskandrani RO, Alharbi A, El Kerdawy AM. Novel oxindole/benzofuran hybrids as potential dual CDK2/GSK-3β inhibitors targeting breast cancer: design, synthesis, biological evaluation, and in silico studies. J Enzyme Inhib Med Chem 2021; 36:270-285. [PMID: 33327806 PMCID: PMC7751407 DOI: 10.1080/14756366.2020.1862101] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 02/08/2023] Open
Abstract
The serine/threonine protein kinases CDK2 and GSK-3β are key oncotargets in breast cancer cell lines, therefore, in the present study three series of oxindole-benzofuran hybrids were designed and synthesised as dual CDK2/GSK-3β inhibitors targeting breast cancer (5a-g, 7a-h, and 13a-b). The N1 -unsubstituted oxindole derivatives, series 5, showed moderate to potent activity on both MCF-7 and T-47D breast cancer cell lines. Compounds 5d-f showed the most potent cytotoxic activity with IC50 of 3.41, 3.45 and 2.27 μM, respectively, on MCF-7 and of 3.82, 4.53 and 7.80 μM, respectively, on T-47D cell lines, in comparison to the used reference standard (staurosporine) IC50 of 4.81 and 4.34 μM, respectively. On the other hand, the N1 -substituted oxindole derivatives, series 7 and 13, showed moderate to weak cytotoxic activity on both breast cancer cell lines. CDK2 and GSK-3β enzyme inhibition assay of series 5 revealed that compounds 5d and 5f are showing potent dual CDK2/GSK-3β inhibitory activity with IC50 of 37.77 and 52.75 nM, respectively, on CDK2 and 32.09 and 40.13 nM, respectively, on GSK-3β. The most potent compounds 5d-f caused cell cycle arrest in the G2/M phase in MCF-7 cells inducing cell apoptosis because of the CDK2/GSK-3β inhibition. Molecular docking studies showed that the newly synthesised N1 -unsubstituted oxindole hybrids have comparable binding patterns in both CDK2 and GSK-3β. The oxindole ring is accommodated in the hinge region interacting through hydrogen bonding with the backbone CO and NH of the key amino acids Glu81 and Leu83, respectively, in CDK2 and Asp133 and Val135, respectively, in GSK-3β. Whereas, in series 7 and 13, the N1 -substitutions on the oxindole nucleus hinder the compounds from achieving these key interactions with hinge region amino acids what rationalises their moderate to low anti-proliferative activity.
Collapse
Affiliation(s)
- Wagdy M. Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - Sara T. Al-Rashood
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Tarfah Al-Warhi
- Department of Chemistry, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Razan O. Eskandrani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Amal Alharbi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed M. El Kerdawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, New Giza University, Cairo, Egypt
| |
Collapse
|
7
|
Schneidewind T, Brause A, Pahl A, Burhop A, Mejuch T, Sievers S, Waldmann H, Ziegler S. Morphological Profiling Identifies a Common Mode of Action for Small Molecules with Different Targets. Chembiochem 2020; 21:3197-3207. [PMID: 32618075 PMCID: PMC7754162 DOI: 10.1002/cbic.202000381] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/02/2020] [Indexed: 12/24/2022]
Abstract
Unbiased morphological profiling of bioactivity, for example, in the cell painting assay (CPA), enables the identification of a small molecule's mode of action based on its similarity to the bioactivity of reference compounds, irrespective of the biological target or chemical similarity. This is particularly important for small molecules with nonprotein targets as these are rather difficult to identify with widely employed target-identification methods. We employed morphological profiling using the CPA to identify compounds that are biosimilar to the iron chelator deferoxamine. Structurally different compounds with different annotated cellular targets provoked a shared physiological response, thereby defining a cluster based on their morphological fingerprints. This cluster is based on a shared mode of action and not on a shared target, that is, cell-cycle modulation in the S or G2 phase. Hierarchical clustering of morphological fingerprints revealed subclusters that are based on the mechanism of action and could be used to predict target-related bioactivity.
Collapse
Affiliation(s)
- Tabea Schneidewind
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
- Technical University DortmundFaculty of Chemistry and Chemical BiologyOtto-Hahn-Strasse 6Dortmund44227Germany
| | - Alexandra Brause
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
| | - Axel Pahl
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
| | - Annina Burhop
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
| | - Tom Mejuch
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
| | - Sonja Sievers
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
| | - Herbert Waldmann
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
- Technical University DortmundFaculty of Chemistry and Chemical BiologyOtto-Hahn-Strasse 6Dortmund44227Germany
| | - Slava Ziegler
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
| |
Collapse
|
8
|
Lapcik P, Pospisilova A, Janacova L, Grell P, Fabian P, Bouchal P. How Different Are the Molecular Mechanisms of Nodal and Distant Metastasis in Luminal A Breast Cancer? Cancers (Basel) 2020; 12:E2638. [PMID: 32947901 PMCID: PMC7563588 DOI: 10.3390/cancers12092638] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 11/17/2022] Open
Abstract
Lymph node status is one of the best prognostic factors in breast cancer, however, its association with distant metastasis is not straightforward. Here we compare molecular mechanisms of nodal and distant metastasis in molecular subtypes of breast cancer, with major focus on luminal A patients. We analyze a new cohort of 706 patients (MMCI_706) as well as an independent cohort of 836 primary tumors with full gene expression information (SUPERTAM_HGU133A). We evaluate the risk of distant metastasis, analyze targetable molecular mechanisms in Gene Set Enrichment Analysis and identify relevant inhibitors. Lymph node positivity is generally associated with NF-κB and Src pathways and is related to high risk (OR: 5.062 and 2.401 in MMCI_706 and SUPERTAM_HGU133A, respectively, p < 0.05) of distant metastasis in luminal A patients. However, a part (≤15%) of lymph node negative tumors at the diagnosis develop the distant metastasis which is related to cell proliferation control and thrombolysis. Distant metastasis of lymph node positive patients is mostly associated with immune response. These pro-metastatic mechanisms further vary in other molecular subtypes. Our data indicate that the management of breast cancer and prevention of distant metastasis requires stratified approach based on targeted strategies.
Collapse
Affiliation(s)
- Petr Lapcik
- Department of Biochemistry, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic; (P.L.); (A.P.); (L.J.)
| | - Anna Pospisilova
- Department of Biochemistry, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic; (P.L.); (A.P.); (L.J.)
| | - Lucia Janacova
- Department of Biochemistry, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic; (P.L.); (A.P.); (L.J.)
| | - Peter Grell
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, 65653 Brno, Czech Republic;
| | - Pavel Fabian
- Department of Oncological Pathology, Masaryk Memorial Cancer Institute, 65653 Brno, Czech Republic;
| | - Pavel Bouchal
- Department of Biochemistry, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic; (P.L.); (A.P.); (L.J.)
| |
Collapse
|
9
|
Zhang X, Zhang B, Zhang P, Lian L, Li L, Qiu Z, Qian K, Chen A, Liu Q, Jiang Y, Cui J, Qi B. Norcantharidin regulates ERα signaling and tamoxifen resistance via targeting miR-873/CDK3 in breast cancer cells. PLoS One 2019; 14:e0217181. [PMID: 31120927 PMCID: PMC6532885 DOI: 10.1371/journal.pone.0217181] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/07/2019] [Indexed: 12/11/2022] Open
Abstract
MiR-873/CDK3 has been shown to play a critical role in ERα signaling and tamoxifen resistance. Thus, targeting this pathway may be a potential therapeutic approach for the treatment of ER positive breast cancer especially tamoxifen resistant subtype. Here we report that Norcantharidin (NCTD), currently used clinically as an ani-cancer drug in China, regulates miR-873/CDK3 axis in breast cancer cells. NCTD decreases the transcriptional activity of ERα but not ERβ through the modulation of miR-873/CDK3 axis. We also found that NCTD inhibits cell proliferation and tumor growth and miR-873/CDK3 axis mediates cell proliferation suppression of NCTD. More important, we found that NCTD sensitizes resistant cells to tamoxifen. NCTD inhibits tamoxifen induced the transcriptional activity as well ERα downstream gene expressions in tamoxifen resistant breast cancer cells. In addition, we found that NCTD restores tamoxifen induced recruitments of ERα co-repressors N-CoR and SMRT. Knockdown of miR-873 and overexpression of CDK3 diminish the effect of NCTD on tamoxifen resistance. Our data shows that NCTD regulates ERα signaling and tamoxifen resistance by targeting miR-873/CDK3 axis in breast cancer cells. This study may provide an alternative therapy strategy for tamoxifen resistant breast cancer.
Collapse
Affiliation(s)
- Xiumei Zhang
- The Center for Translational Medicine, Yichun University, Yichun, Jiangxi, P.R. China
- College of Chemistry and Bio-engineering, Yichun University, Yichun, Jiangxi, P.R. China
| | - Bingfeng Zhang
- College of Chemistry and Bio-engineering, Yichun University, Yichun, Jiangxi, P.R. China
| | - Panhong Zhang
- The Center for Translational Medicine, Yichun University, Yichun, Jiangxi, P.R. China
- College of Chemistry and Bio-engineering, Yichun University, Yichun, Jiangxi, P.R. China
| | - Lihui Lian
- Department of Cell Biology, College of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, P.R. China
| | - Lianlian Li
- The Center for Translational Medicine, Yichun University, Yichun, Jiangxi, P.R. China
| | - Zhihong Qiu
- The Center for Translational Medicine, Yichun University, Yichun, Jiangxi, P.R. China
| | - Kai Qian
- The Center for Translational Medicine, Yichun University, Yichun, Jiangxi, P.R. China
| | - An Chen
- The Center for Translational Medicine, Yichun University, Yichun, Jiangxi, P.R. China
| | - Qiongqing Liu
- The Center for Translational Medicine, Yichun University, Yichun, Jiangxi, P.R. China
- College of Chemistry and Bio-engineering, Yichun University, Yichun, Jiangxi, P.R. China
| | - Yinjie Jiang
- The Center for Translational Medicine, Yichun University, Yichun, Jiangxi, P.R. China
| | - Jiajun Cui
- The Center for Translational Medicine, Yichun University, Yichun, Jiangxi, P.R. China
- * E-mail: (JC); (BQ)
| | - Bing Qi
- Department of Cell Biology, College of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, P.R. China
- * E-mail: (JC); (BQ)
| |
Collapse
|
10
|
Wood RA, Barbour MJ, Gould GW, Cunningham MR, Plevin RJ. Conflicting evidence for the role of JNK as a target in breast cancer cell proliferation: Comparisons between pharmacological inhibition and selective shRNA knockdown approaches. Pharmacol Res Perspect 2018; 6. [PMID: 29417765 PMCID: PMC5817830 DOI: 10.1002/prp2.376] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/01/2017] [Indexed: 11/11/2022] Open
Abstract
As a target, the JNK pathway has been implicated in roles including cell death, proliferation, and inflammation in variety of contexts which span cardiovascular disease, neurodegenerative pathologies, and cancer. JNK1 and JNK2 have recently been demonstrated to function independently, highlighting a new parameter in the study of the JNK pathway. In order for JNK1 and JNK2-specific roles to be defined, better tools need to be employed. Previous studies have relied upon the broad spectrum JNK inhibitor, SP600125, to characterize the role of JNK signaling in a number of cell lines, including the breast cancer cell line MCF-7. In line with previous literature, our study has demonstrated that SP600125 treatment inhibited c-Jun and JNK phosphorylation and MCF-7 proliferation. However, in addition to targeting JNK1, JNK2, and JNK3, SP600125 has been previously demonstrated to suppress the activity of a number of other serine/threonine kinases, making SP600125 an inadequate tool for JNK isoform-specific roles to be determined. In this study, lentiviral shRNA was employed to selectively knockdown JNK1, JNK2, and JNK1/2 in MCF-7 cells. Using this approach, JNK phosphorylation was fully inhibited following stable knockdown of respective JNK isoforms. Interestingly, despite suppression of JNK phosphorylation, MCF-7 cell proliferation, cell cycle progression, or cell death remained unaffected. These findings raise the question of whether JNK phosphorylation really is pivotal in MCF-7 cell growth and death or if suppression of these events is a result of one of the many off-targets cited for SP600125.
Collapse
Affiliation(s)
- Rachel A Wood
- Strathclyde Institute for Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, UK
| | - Mark J Barbour
- Strathclyde Institute for Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, UK
| | - Gwyn W Gould
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Margaret R Cunningham
- Strathclyde Institute for Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, UK
| | - Robin J Plevin
- Strathclyde Institute for Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, UK
| |
Collapse
|
11
|
Targeting p53 as a promising therapeutic option for cancer by re-activating the wt or mutant p53’s tumor suppression. Future Med Chem 2018; 10:755-777. [DOI: 10.4155/fmc-2017-0175] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
p53 protein, a product of the TP53 tumor suppressor gene, controls the cellular genome’s integrity and is an important regulator of cell cycling, proliferation, apoptosis and metabolism. Mutations of TP53 or inactivation of its gene product are among the first events initiating malignant transformation. The consequent loss of control over the cell cycle, resulting in accelerated cell proliferation and facilitating metabolic reprogramming, gives the initiated (premalignant) cells numerous advantages over healthy cells. Interestingly, p53 status is not only an important marker in cancer diagnosis; it has also become a promising target of personalized therapy. Depending on the TP53 status different therapeutic options have been developed. (Re)-activation of p53 functionality in cancer cells offers promising new alternatives to existing oncological therapies.
Collapse
|
12
|
Sahni JM, Keri RA. Targeting bromodomain and extraterminal proteins in breast cancer. Pharmacol Res 2018; 129:156-176. [PMID: 29154989 PMCID: PMC5828951 DOI: 10.1016/j.phrs.2017.11.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 11/10/2017] [Accepted: 11/13/2017] [Indexed: 12/13/2022]
Abstract
Breast cancer is a collection of distinct tumor subtypes that are driven by unique gene expression profiles. These transcriptomes are controlled by various epigenetic marks that dictate which genes are expressed and suppressed. During carcinogenesis, extensive restructuring of the epigenome occurs, including aberrant acetylation, alteration of methylation patterns, and accumulation of epigenetic readers at oncogenes. As epigenetic alterations are reversible, epigenome-modulating drugs could provide a mechanism to silence numerous oncogenes simultaneously. Here, we review the impact of inhibitors of the Bromodomain and Extraterminal (BET) family of epigenetic readers in breast cancer. These agents, including the prototypical BET inhibitor JQ1, have been shown to suppress a variety of oncogenic pathways while inducing minimal, if any, toxicity in models of several subtypes of breast cancer. BET inhibitors also synergize with multiple approved anti-cancer drugs, providing a greater response in breast cancer cell lines and mouse models than either single agent. The combined findings of the studies discussed here provide an excellent rationale for the continued investigation of the utility of BET inhibitors in breast cancer.
Collapse
Affiliation(s)
- Jennifer M Sahni
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Ruth A Keri
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, United States; Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States; Department of General Medical Sciences-Oncology, Case Western Reserve University, Cleveland, OH 44106, United States.
| |
Collapse
|
13
|
Berrak O, Arisan ED, Obakan-Yerlikaya P, Coker-Gürkan A, Palavan-Unsal N. mTOR is a fine tuning molecule in CDK inhibitors-induced distinct cell death mechanisms via PI3K/AKT/mTOR signaling axis in prostate cancer cells. Apoptosis 2018; 21:1158-78. [PMID: 27484210 DOI: 10.1007/s10495-016-1275-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Purvalanol and roscovitine are cyclin dependent kinase (CDK) inhibitors that induce cell cycle arrest and apoptosis in various cancer cells. We further hypothesized that co-treatment of CDK inhibitors with rapamycin, an mTOR inhibitor, would be an effective combinatory strategy for the inhibition of prostate cancer regard to androgen receptor (AR) status due to inhibition of proliferative pathway, PI3K/AKT/mTOR, and induction of cell death mechanisms. Androgen responsive (AR+), PTEN(-/-) LNCaP and androgen independent (AR-), PTEN(+/-) DU145 prostate cancer cells were exposed to purvalanol (20 µM) and roscovitine (30 µM) with or without rapamycin for 24 h. Cell viability assay, immunoblotting, flow cytometry and fluorescence microscopy was used to define the effect of CDK inhibitors with or without rapamycin on proliferative pathway and cell death mechanisms in LNCaP and DU145 prostate cancer cells. Co-treatment of rapamycin modulated CDK inhibitors-induced cytotoxicity and apoptosis that CDK inhibitors were more potent to induce cell death in AR (+) LNCaP cells than AR (-) DU145 cells. CDK inhibitors in the presence or absence of rapamycin induced cell death via modulating upstream PI3K/AKT/mTOR signaling pathway in LNCaP cells, exclusively only treatment of purvalanol have strong potential to inhibit both upstream and downstream targets of mTOR in LNCaP and DU145 cells. However, co-treatment of rapamycin with CDK inhibitors protects DU145 cells from apoptosis via induction of autophagy mechanism. We confirmed that purvalanol and roscovitine were strong apoptotic and autophagy inducers that based on regulation of PI3K/AKT/mTOR signaling pathway. Co-treatment of rapamycin with purvalanol and roscovitine exerted different effects on cell survival and death mechanisms in LNCaP and DU145 cell due to their AR receptor status. Our studies show that co-treatment of rapamycin with CDK inhibitors inhibit prostate cancer cell viability more effectively than either agent alone, in part, by targeting the mTOR signaling cascade in AR (+) LNCaP cells. In this point, mTOR is a fine-tuning player in purvalanol and roscovitine-induced apoptosis and autophagy via regulation of PI3K/AKT and the downstream targets, which related with cell proliferation.
Collapse
Affiliation(s)
- Ozge Berrak
- Molecular Biology and Genetics Department, Science and Literature Faculty, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey
| | - Elif Damla Arisan
- Molecular Biology and Genetics Department, Science and Literature Faculty, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey.
| | - Pinar Obakan-Yerlikaya
- Molecular Biology and Genetics Department, Science and Literature Faculty, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey
| | - Ajda Coker-Gürkan
- Molecular Biology and Genetics Department, Science and Literature Faculty, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey
| | - Narçin Palavan-Unsal
- Molecular Biology and Genetics Department, Science and Literature Faculty, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey
| |
Collapse
|
14
|
Zheng L, Meng X, Li X, Zhang Y, Li C, Xiang C, Xing Y, Xia Y, Xi T. miR-125a-3p inhibits ERα transactivation and overrides tamoxifen resistance by targeting CDK3 in estrogen receptor-positive breast cancer. FASEB J 2018; 32:588-600. [PMID: 28939591 DOI: 10.1096/fj.201700461rr] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tamoxifen (TAM) is a major adjuvant therapy for patients who are diagnosed with estrogen receptor-α (ER)-positive breast cancer; however, TAM resistance occurs often during treatment and the underlying mechanism is unclear. Here, we report that miR-125a-3p inhibits ERα transcriptional activity and, thus, ER+ breast cancer cell proliferation, which causes cell-cycle arrest at the G1/S stage, inducing apoptosis and suppressing tumor growth by targeting cyclin-dependent kinase 3 (CDK3) in vitro and in vivo. In addition, CDK3 and miR-125a-3p expression levels were measured in 37 cancerous tissues paired with noncancerous samples, and their expression levels were negatively associated with miR-125a-3p level. Of interest, miR-125a-3p level is down-regulated in MCF-7 TAM-resistant (TamR) cells. Of more importance, up-regulation of miR-125a-3p resensitizes MCF-7 TamR cells to TAM, which is dependent on CDK3 expression. These results suggest that miR-125a-3p can function as a novel tumor suppressor in ER+ breast cancer by targeting CDK3, which may be a potential therapeutic approach for TamR breast cancer therapy.-Zheng, L., Meng, X., Li, X., Zhang, Y., Li, C., Xiang, C., Xing, Y., Xia, Y., Xi, T. miR-125a-3p inhibits ERα transactivation and overrides tamoxifen resistance by targeting CDK3 in estrogen receptor-positive breast cancer.
Collapse
Affiliation(s)
- Lufeng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China.,Department of Pharmacology of Chinese Materia Medica, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Xia Meng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Xiaoman Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Cheng Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Chenxi Xiang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Yufeng Xia
- Department of Pharmacology of Chinese Materia Medica, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
15
|
Cascioferro S, Parrino B, Spanò V, Carbone A, Montalbano A, Barraja P, Diana P, Cirrincione G. 1,3,5-Triazines: A promising scaffold for anticancer drugs development. Eur J Med Chem 2017; 142:523-549. [PMID: 29046238 DOI: 10.1016/j.ejmech.2017.09.035] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/16/2017] [Accepted: 09/18/2017] [Indexed: 12/18/2022]
Abstract
This review covering literature reports from the beginning of this century to 2016 describes the synthetic pathways, the antitumor activity, the structure-activity relationship and, whenever reported, the possible mechanism of action of 1,3,5-triazine derivatives as well as of their hetero-fused compounds. Many 1,3,5-triazine derivatives, both uncondensed and hetero-fused, have shown remarkable antitumor activities and some of them reached clinical development.
Collapse
Affiliation(s)
- Stella Cascioferro
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Barbara Parrino
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Virginia Spanò
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Anna Carbone
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Alessandra Montalbano
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Paola Barraja
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Patrizia Diana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Girolamo Cirrincione
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy.
| |
Collapse
|
16
|
Sheldon LA. Inhibition of E2F1 activity and cell cycle progression by arsenic via retinoblastoma protein. Cell Cycle 2017; 16:2058-2072. [PMID: 28880708 DOI: 10.1080/15384101.2017.1338221] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The regulation of cell cycle progression by steroid hormones and growth factors is important for maintaining normal cellular processes including development and cell proliferation. Deregulated progression through the G1/S and G2/M cell cycle transitions can lead to uncontrolled cell proliferation and cancer. The transcription factor E2F1, a key cell cycle regulator, targets genes encoding proteins that regulate cell cycle progression through the G1/S transition as well as proteins important in DNA repair and apoptosis. E2F1 expression and activity is inhibited by inorganic arsenic (iAs) that has a dual role as a cancer therapeutic and as a toxin that leads to diseases including cancer. An understanding of what underlies this dichotomy will contribute to understanding how to use iAs as a more effective therapeutic and also how to treat cancers that iAs promotes. Here, we show that quiescent breast adenocarcinoma MCF-7 cells treated with 17-β estradiol (E2) progress through the cell cycle, but few cells treated with E2 + iAs progress from G1 into S-phase due to a block in cell cycle progression. Our data support a model in which iAs inhibits the dissociation of E2F1 from the tumor suppressor, retinoblastoma protein (pRB) due to changes in pRB phosphorylation which leads to decreased E2F1 transcriptional activity. These findings present an explanation for how iAs can disrupt cell cycle progression through E2F1-pRB and has implications for how iAs acts as a cancer therapeutic as well as how it may promote tumorigenesis through decreased DNA repair.
Collapse
Affiliation(s)
- Lynn A Sheldon
- a Geisel School of Medicine at Dartmouth, Department of Molecular and Systems Biology , Hanover , NH , USA
| |
Collapse
|
17
|
Roscovitine Protects From Arterial Injury by Regulating the Expressions of c-Jun and p27 and Inhibiting Vascular Smooth Muscle Cell Proliferation. J Cardiovasc Pharmacol 2017; 69:161-169. [DOI: 10.1097/fjc.0000000000000453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
18
|
Why (multi)targeting of cyclin-dependent kinases is a promising therapeutic option for hormone-positive breast cancer and beyond. Future Med Chem 2015; 8:55-72. [PMID: 26692095 DOI: 10.4155/fmc.15.155] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Estrogens, via induction of their specific receptors (e.g., ER-α), regulate cell proliferation, differentiation and morphogenesis in mammary epithelium. Cell-cycle progression is driven by activation of complexes consisting of cyclin-dependent kinases (CDKs) and cyclins, which also modulate the activity of ER-α. Loss of control over the cell-cycle results in accelerated cell division and malignant transformation. Thus, a reciprocal relation exists between estrogen signaling and cell proliferation. Based on these findings, a new concept was developed to reduce ER-α activity and bring the cell cycle in transformed cells to heel. Prevention of ER-α activation and control over the deregulated cell cycle was achieved by supplementation with pharmacological CDK inhibitors alone or in combination with selective antiestrogens.
Collapse
|
19
|
Kolodziej M, Goetz C, Di Fazio P, Montalbano R, Ocker M, Strik H, Quint K. Roscovitine has anti-proliferative and pro-apoptotic effects on glioblastoma cell lines: A pilot study. Oncol Rep 2015; 34:1549-56. [PMID: 26151768 DOI: 10.3892/or.2015.4105] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/03/2015] [Indexed: 02/07/2023] Open
Abstract
Purine analogue roscovitine, a cyclin-dependent kinase (CDK) inhibitor, has shown strong anti-proliferative and pro-apoptotic effects in solid and hematologic cancers such as non small-cell lung cancer and lymphomas. It targets CDK2, 7 and 9 preferentially, which are also overexpressed in glioblastoma. Τherefore, the biological effects of roscovitine in glioblastoma cell lines were investigated. Glioblastoma A172 and G28 cell lines were incubated with serial concentrations of roscovitine for 24-120 h. Proliferation was measured using the xCELLigence Real-Time Cell Analyzer, an impedance‑based cell viability system. Cell cycle distribution was assessed by flow cytometry and gene expression was quantified by quantitative RT-PCR and western blot analysis. Roscovitine exhibited a clear dose-dependent anti‑proliferative and pro‑apoptotic effect in the A172 cell line, while G28 cells showed a anti-proliferative effect only at 100 µM. The results of the flow cytometric (FACS) analysis revealed a dose-dependent increase of the G2/M and sub-G1 fractions in A172 cells, while G28 cells responded with an elevated sub-G1 fraction only at the highest concentration. Roscovitine led to a dose‑dependent decrease of transcripts of p53, CDK 7 and cyclins A and E and an increase of >4-fold of p21 in A172 cells. In G28 cells, a dose‑dependent induction of CDK2, p21 and cyclin D was observed between 10 and 50 µM roscovitine after 72 h, however, at the highest concentration of 100 µM, all investigated genes were downregulated. Roscovitine exerted clear dose-dependent anti-proliferative and pro-apoptotic effects in A172 cells and less distinct effects on G28 cells. In A172 cells, roscovitine led to G2/M arrest and induced apoptosis, an effect accompanied by induced p21 and a reduced expression of CDK2, 7 and 9 and cyclins A and E. These effects requre further studies on a larger scale to confirm whether roscovitine can be used as a therapeutic agent against glioblastoma.
Collapse
Affiliation(s)
- M Kolodziej
- Department of Neurosurgery, University Hospital Giessen, Giessen, Germany
| | - C Goetz
- Institute for Surgical Research, University of Marburg, Marburg, Germany
| | - P Di Fazio
- Institute for Surgical Research, University of Marburg, Marburg, Germany
| | - R Montalbano
- Institute for Surgical Research, University of Marburg, Marburg, Germany
| | - M Ocker
- Institute for Surgical Research, University of Marburg, Marburg, Germany
| | - H Strik
- Department of Neurology, Thoracic and Vascular Surgery, University Hospital Marburg, Marburg, Germany
| | - K Quint
- Institute for Surgical Research, University of Marburg, Marburg, Germany
| |
Collapse
|
20
|
Cicenas J, Kalyan K, Sorokinas A, Stankunas E, Levy J, Meskinyte I, Stankevicius V, Kaupinis A, Valius M. Roscovitine in cancer and other diseases. ANNALS OF TRANSLATIONAL MEDICINE 2015. [PMID: 26207228 DOI: 10.3978/j.issn.2305-5839.2015.03.61] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Roscovitine [CY-202, (R)-Roscovitine, Seliciclib] is a small molecule that inhibits cyclin-dependent kinases (CDKs) through direct competition at the ATP-binding site. It is a broad-range purine inhibitor, which inhibits CDK1, CDK2, CDK5 and CDK7, but is a poor inhibitor for CDK4 and CDK6. Roscovitine is widely used as a biological tool in cell cycle, cancer, apoptosis and neurobiology studies. Moreover, it is currently evaluated as a potential drug to treat cancers, neurodegenerative diseases, inflammation, viral infections, polycystic kidney disease and glomerulonephritis. This review focuses on the use of roscovitine in the disease model as well as clinical model research.
Collapse
Affiliation(s)
- Jonas Cicenas
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Karthik Kalyan
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Aleksandras Sorokinas
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Edvinas Stankunas
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Josh Levy
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Ingrida Meskinyte
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Vaidotas Stankevicius
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Algirdas Kaupinis
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Mindaugas Valius
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| |
Collapse
|
21
|
Zhou Y, Han C, Li D, Yu Z, Li F, Li F, An Q, Bai H, Zhang X, Duan Z, Kan Q. Cyclin-dependent kinase 11(p110) (CDK11(p110)) is crucial for human breast cancer cell proliferation and growth. Sci Rep 2015; 5:10433. [PMID: 25990212 PMCID: PMC4438429 DOI: 10.1038/srep10433] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 04/13/2015] [Indexed: 11/17/2022] Open
Abstract
Cyclin-dependent kinases (CDKs) play important roles in the development of many types of cancers by binding with their paired cyclins. However, the function of CDK11 larger protein isomer, CDK11(p110), in the tumorigenesis of human breast cancer remains unclear. In the present study, we explored the effects and molecular mechanisms of CDK11(p110) in the proliferation and growth of breast cancer cells by determining the expression of CDK11(p110) in breast tumor tissues and examining the phenotypic changes of breast cancer cells after CDK11(p110) knockdown. We found that CDK11(p110) was highly expressed in breast tumor tissues and cell lines. Tissue microarray analysis showed that elevated CDK11(p110) expression in breast cancer tissues significantly correlated with poor differentiation, and was also associated with advanced TNM stage and poor clinical prognosis for breast cancer patients. In vitro knockdown of CDK11(p110) by siRNA significantly inhibited cell growth and migration, and dramatically induced apoptosis in breast cancer cells. Flow cytometry demonstrated that cells were markedly arrested in G1 phase of the cell cycle after CDK11(p110) downregulation. These findings suggest that CDK11(p110) is critical for the proliferation and growth of breast cancer cells, which highlights CDK11(p110) may be a promising therapeutic target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yubing Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Chao Han
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Duolu Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Zujiang Yu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Fengmei Li
- Department of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, 195 Tongbai Road, Zhengzhou 450007, China
| | - Feng Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Qi An
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Huili Bai
- Department of Pathology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, 195 Tongbai Road, Zhengzhou 450007, China
| | - Xiaojian Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Zhenfeng Duan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
- Sarcoma Molecular Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, USA
| | - Quancheng Kan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| |
Collapse
|
22
|
Cui J, Yang Y, Li H, Leng Y, Qian K, Huang Q, Zhang C, Lu Z, Chen J, Sun T, Wu R, Sun Y, Song H, Wei X, Jing P, Yang X, Zhang C. MiR-873 regulates ERα transcriptional activity and tamoxifen resistance via targeting CDK3 in breast cancer cells. Oncogene 2014; 34:3895-907. [PMID: 25531331 DOI: 10.1038/onc.2014.430] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 11/19/2014] [Accepted: 11/21/2014] [Indexed: 12/21/2022]
Abstract
miRNAs (microRNAs) are frequently and aberrantly expressed in many cancers. MiR-873 has been revealed to be downregulated in colorectal cancer and glioblastoma. However, its function remains unclear. Here we report that miR-873 is downregulated in breast tumor compared with normal tissue. Enforced expression of miR-873 decreases the transcriptional activity of ER (estrogen receptor)-α but not ERβ through the modulation of ERα phosphorylation in ER-positive breast cancer cells. We also found that miR-873 inhibits breast cancer cell proliferation and tumor growth in nude mice. Reporter gene assays revealed cyclin-dependent kinase 3 (CDK3) as a direct target of miR-873. CDK3 was shown to be overexpressed in breast cancer and phosphorylate ERα at Ser104/116 and Ser118. Furthermore, we found that Mir-873 inhibits ER activity and cell growth via targeting CDK3. Interestingly, miR-873 was observed to be downregulated in tamoxifen-resistant MCF-7/TamR cells, while CDK3 is overexpressed in these cells. More importantly, re-expression of miR-873 reversed tamoxifen resistance in MCF-7/TamR cells. Our data demonstrate that miR-873 is a novel tumor suppressor in ER-positive breast cancer and a potential therapeutic approach for treatment of tamoxifen-resistant breast cancer.
Collapse
Affiliation(s)
- J Cui
- Institute of Disease Control and Prevention, Chinese Academy of Military Medical Sciences, Beijing, China
| | - Y Yang
- Beijing Institute for Neuroscience, Capital Medical University, Beijing, China
| | - H Li
- Department of Molecular & Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Y Leng
- The Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - K Qian
- The Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - Q Huang
- Department of Animal Sciences and Technology, Jilin Agriculture University, Changchun, China
| | - C Zhang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Z Lu
- Institute of Disease Control and Prevention, Chinese Academy of Military Medical Sciences, Beijing, China
| | - J Chen
- Urology Department, the First Hospital of Nanchang University, Nanchang, China
| | - T Sun
- Urology Department, the First Hospital of Nanchang University, Nanchang, China
| | - R Wu
- Institute of Disease Control and Prevention, Chinese Academy of Military Medical Sciences, Beijing, China
| | - Y Sun
- Institute of Disease Control and Prevention, Chinese Academy of Military Medical Sciences, Beijing, China
| | - H Song
- Institute of Disease Control and Prevention, Chinese Academy of Military Medical Sciences, Beijing, China
| | - X Wei
- Department of Applied Chemistry, College of Chemistry & Molecular Engineering, Peking University, Beijing, China
| | - P Jing
- Department of Chemistry, College of Arts and Sciences, Indiana University-Purdue University Fort Wayne Fort Wayne, IN, USA
| | - X Yang
- Institute of Health Sciences, Anhui University, Hefei, China
| | - C Zhang
- Institute of Disease Control and Prevention, Chinese Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
23
|
He X, Xiang H, Zong X, Yan X, Yu Y, Liu G, Zou D, Yang H. CDK2-AP1 inhibits growth of breast cancer cells by regulating cell cycle and increasing docetaxel sensitivity in vivo and in vitro. Cancer Cell Int 2014; 14:130. [PMID: 25550687 PMCID: PMC4279590 DOI: 10.1186/s12935-014-0130-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 11/17/2014] [Indexed: 11/10/2022] Open
Abstract
Background Cell cycle regulatory pathway is a well-established pathway mainly dependent on cyclin-dependent kinases (CDKs), which are regulated positively by cyclins and negatively by cyclin-dependent kinase inhibitors(CKIs). Cyclin-dependent kinase 2 associate protein 1(CDK2-AP1) is a specific negative regulatory protein for CDK2, is important in the cancer cell cycle. However, the function of CDK2-AP1 in breast cancer remains unclear. We designed therefore explored the effects of CDK2-AP1 on breast cancer growth and its chemo-sensitivity. Methods Expression of CDK2-AP1, CDK2 and CyclinD1 in 209 cases of pathological specimens using IHC staining was measured. Lost-of-function and Gain-of-function assays were used in vivo and in vitro relating to the specific role of CDK2-AP1 in breast cancer. We analyzed in vivo and in vitro the impact of CDK2-AP1 on chemotherapy sensitivity in breast cancer. Results The positive ratio of CDK2-AP1 expression was reduced successively in normal breast tissue, DCIS, invasive breast cancer and relapsed breast cancer, however, with CDK2 and CyclinD1 it was suggested that CDK2-AP1 was correlated closely with the tumorigenesis and progress, and might work as a tumor suppressor. After down-regulating CDK2-AP1 in breast cancer cells, the cell cycle was accelerated and cell proliferation enhanced. The cell cycle was arrested in G0/G1 phase and G2/M phase after up-regulating CDK2-AP1 in breast cancer cells, inhibiting cell proliferation. The expression of CDK2 and CyclinD1 changed accordingly after downregulation or upregulation of CDK2-AP1 by western blot, suggesting a role of the CDK2-AP1/CDK2/CyclinD1 cell cycle pathway in the initiation and progression of breast cancer. Similar results were obtained in animal assays. The data indicates that CDK2-AP1 can induce sensitivity to docetaxel treatment in breast cancer cells. Conclusions CDK2-AP1 affects tumorigenesis, tumor growth and chemo-sensitivity by cell cycle regulation, which can potentially to be a therapeutical agent in breast cancer.
Collapse
Affiliation(s)
- Xiangming He
- Department of Breast Surgery, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, 310022 China
| | - Hua Xiang
- Department of Pathology, First Affiliated Hospital of College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 China
| | - Xiangyun Zong
- Department of Breast Surgery, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, 310022 China ; Department of Breast Surgery, Shanghai Jiao Tong University affiliated Shanghai sixth Hospital, 600 Yishan Road, Shanghai, 200233 China
| | - Xuebing Yan
- Department of Breast Surgery, Shanghai Jiao Tong University affiliated Shanghai sixth Hospital, 600 Yishan Road, Shanghai, 200233 China
| | - Yang Yu
- Department of Breast Surgery, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, 310022 China
| | - Guan Liu
- Department of Radiotherapy, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, 310022 China
| | - Dehong Zou
- Department of Breast Surgery, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, 310022 China
| | - Hongjian Yang
- Department of Breast Surgery, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, 310022 China
| |
Collapse
|
24
|
Kabadi SV, Faden AI. Selective CDK inhibitors: promising candidates for future clinical traumatic brain injury trials. Neural Regen Res 2014; 9:1578-80. [PMID: 25368642 PMCID: PMC4211197 DOI: 10.4103/1673-5374.141779] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2014] [Indexed: 01/15/2023] Open
Abstract
Traumatic brain injury induces secondary injury that contributes to neuroinflammation, neuronal loss, and neurological dysfunction. One important injury mechanism is cell cycle activation which causes neuronal apoptosis and glial activation. The neuroprotective effects of both non-selective (Flavopiridol) and selective (Roscovitine and CR-8) cyclin-dependent kinase inhibitors have been shown across multiple experimental traumatic brain injury models and species. Cyclin-dependent kinaseinhibitors, administered as a single systemic dose up to 24 hours after traumatic brain injury, provide strong neuroprotection-reducing neuronal cell death, neuroinflammation and neurological dysfunction. Given their effectiveness and long therapeutic window, cyclin-dependent kinase inhibitors appear to be promising candidates for clinical traumatic brain injury trials.
Collapse
Affiliation(s)
- Shruti V Kabadi
- Department of Anesthesiology, Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan I Faden
- Department of Anesthesiology, Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
25
|
Huber RJ. The cyclin-dependent kinase family in the social amoebozoan Dictyostelium discoideum. Cell Mol Life Sci 2014; 71:629-39. [PMID: 23974243 PMCID: PMC11113532 DOI: 10.1007/s00018-013-1449-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 07/24/2013] [Accepted: 08/05/2013] [Indexed: 10/26/2022]
Abstract
Cyclin-dependent kinases (Cdk) are a family of serine/threonine protein kinases that regulate eukaryotic cell cycle progression. Their ability to modulate the cell cycle has made them an attractive target for anti-cancer therapies. Cdk protein function has been studied in a variety of Eukaryotes ranging from yeast to humans. In the social amoebozoan Dictyostelium discoideum, several homologues of mammalian Cdks have been identified and characterized. The life cycle of this model organism is comprised of a feeding stage where single cells grow and divide mitotically as they feed on their bacterial food source and a multicellular developmental stage that is induced by starvation. Thus it is a valuable system for studying a variety of cellular and developmental processes. In this review I summarize the current knowledge of the Cdk protein family in Dictyostelium by highlighting the research efforts focused on the characterization of Cdk1, Cdk5, and Cdk8 in this model Eukaryote. Accumulated evidence indicates that each protein performs distinct functions during the Dictyostelium life cycle with Cdk1 being required for growth and Cdk5 and Cdk8 being required for processes that occur during development. Recent studies have shown that Dictyostelium Cdk5 shares attributes with mammalian Cdk5 and that the mammalian Cdk inhibitor roscovitine can be used to inhibit Cdk5 activity in Dictyostelium. Together, these results show that Dictyostelium can be used as a model system for studying Cdk protein function.
Collapse
Affiliation(s)
- Robert J Huber
- Center for Human Genetic Research, Massachusetts General Hospital, Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, MA, 02114, USA,
| |
Collapse
|
26
|
Rocca A, Farolfi A, Bravaccini S, Schirone A, Amadori D. Palbociclib (PD 0332991) : targeting the cell cycle machinery in breast cancer. Expert Opin Pharmacother 2013; 15:407-20. [PMID: 24369047 DOI: 10.1517/14656566.2014.870555] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The cyclin D-cyclin-dependent kinases 4 and 6 (CDK4/6)-retinoblastoma (Rb) pathway, governing the cell cycle restriction point, is frequently altered in breast cancer and is a potentially relevant target for anticancer therapy. Palbociclib (PD 0332991) , a potent and selective inhibitor of CDK4 and CDK6, inhibits proliferation of several Rb-positive cancer cell lines and xenograft models. AREAS COVERED The basic features and abnormalities of the cell cycle in breast cancer are described, along with their involvement in estrogen signaling and endocrine resistance. The pharmacological features of palbociclib, its activity in preclinical models of breast cancer and the potential determinants of response are then illustrated, and its clinical development in breast cancer described. A literature search on the topic was conducted through PubMed and the proceedings of the main cancer congresses of recent years. EXPERT OPINION The combination of palbociclib with endocrine agents is a very promising treatment and Phase III clinical trials are ongoing to confirm its efficacy. Further, potentially useful combinations are those with drugs targeting mitogenic signaling pathways, such as HER2- and PI3K-inhibitors. Combination with chemotherapy seems more problematic, as antagonism has been reported in preclinical models. The identification of predictive factors, already explored in preclinical studies, must be further refined and validated in clinical trials.
Collapse
Affiliation(s)
- Andrea Rocca
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Department of Medical Oncology , Meldola , Italy +39 0543 739100 ; +39 0543 739151 ;
| | | | | | | | | |
Collapse
|
27
|
Gürkan AC, Arisan ED, Obakan P, Palavan-Ünsal N. Inhibition of polyamine oxidase prevented cyclin-dependent kinase inhibitor-induced apoptosis in HCT 116 colon carcinoma cells. Apoptosis 2013; 18:1536-47. [PMID: 23892915 DOI: 10.1007/s10495-013-0885-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Roscovitine and purvalanol are novel cyclin-dependent kinase (CDK) inhibitors that prevent cell proliferation and induce apoptotic cell death in various cancer cell lines. Although a number of studies have demonstrated the potential apoptotic role of roscovitine, there is limited data about the therapeutic efficiency of purvalanol on cancer cells. The natural polyamines (PAs) putrescine, spermidine, and spermine have essential roles in the regulation of cell differentiation, growth, and proliferation, and increased levels of these compounds have been associated with cancer progression. Recently, depletion of intracellular PA levels because of modulation of PA catabolic enzymes was shown to be an indicator of the efficacy of chemotherapeutic agents. In this study, our aim was to investigate the potential role of PA catabolic enzymes in CDK inhibitor-induced apoptosis in HCT 116 colon carcinoma cells. Exposure of cells to roscovitine or purvalanol decreased cell viability in a dose- and time-dependent manner. The selected concentrations of roscovitine and purvalanol inhibited cell viability by 50 % compared with control cells and induced apoptosis by activating the mitochondria-mediated pathway in a caspase-dependent manner. However, the apoptotic effect of purvalanol was stronger than that of roscovitine in HCT 116 cells. In addition, we found that CDK inhibitors decreased PA levels and significantly upregulated expression of key PA catabolic enzymes such as polyamine oxidase (PAO) and spermine oxidase (SMO). MDL-72,527, a specific inhibitor of PAO and SMO, decreased apoptotic potential of CDK inhibitors on HCT 116 cells. Moreover, transient silencing of PAO was also reduced prevented CDK inhibitor-induced apoptosis in HCT 116 cells. We conclude that the PA catabolic pathway, especially PAO, is a critical target for understanding the molecular mechanism of CDK inhibitor-induced apoptosis.
Collapse
Affiliation(s)
- Ajda Coker Gürkan
- Molecular Biology and Genetics Department, Science and Literature Faculty, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey
| | | | | | | |
Collapse
|
28
|
Role of adipokines and cytokines in obesity-associated breast cancer: therapeutic targets. Cytokine Growth Factor Rev 2013; 24:503-13. [PMID: 24210902 DOI: 10.1016/j.cytogfr.2013.10.001] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 10/10/2013] [Accepted: 10/11/2013] [Indexed: 02/06/2023]
Abstract
Obesity is the cause of a large proportion of breast cancer incidences and mortality in post-menopausal women. In obese people, elevated levels of various growth factors such as insulin and insulin-like growth factors (IGFs) are found. Elevated insulin level leads to increased secretion of estrogen by binding to the circulating sex hormone binding globulin (SHBG). The increased estrogen-mediated downstream signaling favors breast carcinogenesis. Obesity leads to altered expression profiles of various adipokines and cytokines including leptin, adiponectin, IL-6, TNF-α and IL-1β. The increased levels of leptin and decreased adiponectin secretion are directly associated with breast cancer development. Increased levels of pro-inflammatory cytokines within the tumor microenvironment promote tumor development. Efficacy of available breast cancer drugs against obesity-associated breast cancer is yet to be confirmed. In this review, we will discuss different adipokine- and cytokine-mediated molecular signaling pathways involved in obesity-associated breast cancer, available therapeutic strategies and potential therapeutic targets for obesity-associated breast cancer.
Collapse
|
29
|
Cui C, Wang Y, Wang Y, Zhao M, Peng S. Exploring the Relationship between the Inhibition Selectivity and the Apoptosis of Roscovitine-Treated Cancer Cells. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2013; 2013:389390. [PMID: 23691435 PMCID: PMC3649549 DOI: 10.1155/2013/389390] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 03/18/2013] [Indexed: 06/02/2023]
Abstract
THE ANTITUMOR ACTIVITY OF ROSCOVITINE WAS TESTED IN FOUR CERVICAL CARCINOMA CELLS: C33A, HCE-1, HeLa, and SiHa. The effects of roscovitine on ATP Lite assay, cell cycle, and apoptosis were assessed. The Sub-G1 DNA content occurred great increasing, and this indicates that apoptosis was induced quickly in HeLa cells, but slowly in the other cells. The morphological observation results showed that roscovitine induced apoptosis and cell death in the cervical carcinoma cells. Results revealed that roscovitine exhibited selective cytotoxicity towards 4 cervical carcinoma cells, and the cells showed different morphologic and apoptotic changes at the same concentration. It was estimated that cervical carcinoma cells responded differently to roscovitine because of differences in apoptotic and genetic background in different cervical carcinoma cells. This study suggested that roscovitine had the potential to be a chemotherapeutic agent against cervical carcinoma.
Collapse
Affiliation(s)
- Chunying Cui
- College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Yaonan Wang
- Medical Experiment and Test Center, Capital Medical University, Beijing 100069, China
| | - Yuji Wang
- College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Ming Zhao
- College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Shiqi Peng
- College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| |
Collapse
|
30
|
Bogush T, Dudko E, Bogush E, Polotsky B, Tjulandin S, Davydov M. Tamoxifen non-estrogen receptor mediated molecular targets. Oncol Rev 2012; 6:e15. [PMID: 25992213 PMCID: PMC4419624 DOI: 10.4081/oncol.2012.e15] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 06/18/2012] [Accepted: 07/05/2012] [Indexed: 01/09/2023] Open
Abstract
Recent experimental studies revealing new biological effects of tamoxifen on tumor cells both expressing and not expressing different types of estrogen receptors (ERα and ERβ) show new aspects of a seemingly well known agent. This review describes tamoxifen targets, the blocking of which leads to inhibition of tumor cell growth and angiogenesis, stimulation of programmed cell death (apoptosis, autophagia and necrosis), inhibition of multidrug resistance, invasion and metastasis. Since outcomes of tamoxifen action on cells are prognostically good from the point of view of both tumor growth/metastasis inhibition and tumor response to drug therapy, the authors believe this is an extremely important addition to tamoxifen antiestrogenic effect. Arguments are provided to consider the strategy of long-term tamoxifen treatment proposed by Professor Craig V. Jordan in the 1970s that is also applicable to the treatment of other tumors. This is, first of all, the fact that expression of estrogen receptor-beta that can also be targeted by tamoxifen therapy in solid tumors of practically all known sites and histologies. The authors believe that molecular biological screening of patients with respect to expression of tamoxifen cellular targets other than ERα and ERβ is needed to use to the full all tamoxifen biological activities other than modulation of estrogen receptors during long-term adjuvant therapy for cancers of various sites.
Collapse
Affiliation(s)
- Tatiana Bogush
- N.N. Blokhin Russian Cancer Research Center under the Russian Academy of Medical Sciences, Russian Federation, Moscow
| | - Evgeny Dudko
- N.N. Blokhin Russian Cancer Research Center under the Russian Academy of Medical Sciences, Russian Federation, Moscow
| | - Elena Bogush
- N.N. Blokhin Russian Cancer Research Center under the Russian Academy of Medical Sciences, Russian Federation, Moscow
| | - Boris Polotsky
- N.N. Blokhin Russian Cancer Research Center under the Russian Academy of Medical Sciences, Russian Federation, Moscow
| | - Sergei Tjulandin
- N.N. Blokhin Russian Cancer Research Center under the Russian Academy of Medical Sciences, Russian Federation, Moscow
| | - Mikhail Davydov
- N.N. Blokhin Russian Cancer Research Center under the Russian Academy of Medical Sciences, Russian Federation, Moscow
| |
Collapse
|
31
|
Huber RJ, O'Day DH. The cyclin-dependent kinase inhibitor roscovitine inhibits kinase activity, cell proliferation, multicellular development, and Cdk5 nuclear translocation in Dictyostelium discoideum. J Cell Biochem 2012; 113:868-76. [PMID: 22234985 DOI: 10.1002/jcb.23417] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Roscovitine, a cyclin-dependent kinase (Cdk) inhibitor, inhibited kinase activity and the axenic growth of Dictyostelium discoideum at micromolar concentrations. Growth was almost fully rescued in 50 µM and ≈ 50% rescued in 100 µM roscovitine-treated cultures by the over-expression of Cdk5-GFP. This supports the importance of Cdk5 function during cell proliferation in Dictyostelium and indicates that Cdk5 is a primary target of the drug. Roscovitine did not affect the expression of Cdk5 protein during axenic growth but did inhibit its nuclear translocation. This novel result suggests that the effects of roscovitine could be due in part to altering Cdk5 translocation in other systems as well. Kinase activity was inhibited by roscovitine in assays using AX3 whole cell lysates, but not in assays using lysates from Cdk5-GFP over-expressing cells. At higher concentrations, roscovitine impaired slug and fruiting body formation. Fruiting bodies that did form were small and produced relatively fewer spores many of which were round. However, roscovitine did not affect stalk cell differentiation. Together with previous findings, these data reveal that roscovitine inhibits Cdk5 during growth and as yet undefined Cdks during mid-late development.
Collapse
Affiliation(s)
- Robert J Huber
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON, Canada
| | | |
Collapse
|
32
|
Caldon CE, Sergio CM, Kang J, Muthukaruppan A, Boersma MN, Stone A, Barraclough J, Lee CS, Black MA, Miller LD, Gee JM, Nicholson RI, Sutherland RL, Print CG, Musgrove EA. Cyclin E2 Overexpression Is Associated with Endocrine Resistance but not Insensitivity to CDK2 Inhibition in Human Breast Cancer Cells. Mol Cancer Ther 2012; 11:1488-99. [DOI: 10.1158/1535-7163.mct-11-0963] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
33
|
Kabadi SV, Stoica BA, Hanscom M, Loane DJ, Kharebava G, Murray II MG, Cabatbat RM, Faden AI. CR8, a selective and potent CDK inhibitor, provides neuroprotection in experimental traumatic brain injury. Neurotherapeutics 2012; 9:405-21. [PMID: 22167461 PMCID: PMC3324621 DOI: 10.1007/s13311-011-0095-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Traumatic brain injury (TBI) induces secondary injury mechanisms, including cell cycle activation (CCA), that leads to neuronal death and neurological dysfunction. We recently reported that delayed administration of roscovitine, a relatively selective cyclin-dependent kinase (CDK) inhibitor, inhibits CCA and attenuates neurodegeneration and functional deficits following controlled cortical impact (CCI) injury in mice. Here we evaluated the neuroprotective potential of CR8, a more potent second-generation roscovitine analog, using the mouse CCI model. Key CCA markers (cyclin A and B1) were significantly up-regulated in the injured cortex following TBI, and phosphorylation of CDK substrates was increased. Central administration of CR8 after TBI, at a dose 20 times less than previously required for roscovitine, attenuated CCA pathways and reduced post-traumatic apoptotic cell death at 24 h post-TBI. Central administration of CR8, at 3 h after TBI, significantly attenuated sensorimotor and cognitive deficits, decreased lesion volume, and improved neuronal survival in the cortex and dentate gyrus. Moreover, unlike roscovitine treatment in the same model, CR8 also attenuated post-traumatic neurodegeneration in the CA3 region of the hippocampus and thalamus at 21 days. Furthermore, delayed systemic administration of CR8, at a dose 10 times less than previously required for roscovitine, significantly improved cognitive performance after CCI. These findings further demonstrate the neuroprotective potential of cell cycle inhibitors following experimental TBI. Given the increased potency and efficacy of CR8 as compared to earlier purine analog types of CDK inhibitors, this drug should be considered as a candidate for future clinical trials of TBI.
Collapse
Affiliation(s)
- Shruti V. Kabadi
- Department of Anesthesiology, Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Bogdan A. Stoica
- Department of Anesthesiology, Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Marie Hanscom
- Department of Anesthesiology, Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - David J. Loane
- Department of Anesthesiology, Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Giorgi Kharebava
- Department of Anesthesiology, Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Michael G. Murray II
- Department of Anesthesiology, Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Rainier M. Cabatbat
- Department of Anesthesiology, Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Alan I. Faden
- Department of Anesthesiology, Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201 USA
| |
Collapse
|
34
|
The impact of CDK inhibition in human malignancies associated with pronounced defects in apoptosis: advantages of multi-targeting small molecules. Future Med Chem 2012; 4:395-424. [DOI: 10.4155/fmc.12.12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Malignant cells in chronic lymphocytic leukemia (CLL) and related diseases are heterogeneous and consist primarily of long-lived resting cells in the periphery and a minor subset of dividing cells in proliferating centers. Both cell populations have different molecular signatures that play a major role in determining their sensitivity to therapy. Contemporary approaches to treating CLL are heavily reliant on cytotoxic chemotherapeutics. However, none of the current treatment regimens can be considered curative. Pharmacological CDK inhibitors have extended the repertoire of potential drugs for CLL. Multi-targeted CDK inhibitors affect CDKs involved in regulating both cell cycle progression and transcription. Their interference with transcriptional elongation represses anti-apoptotic proteins and, thus, promotes the induction of apoptosis. Importantly, there is evidence that treatment with CDK inhibitors can overcome resistance to therapy. The pharmacological CDK inhibitors have great potential for use in combination with other therapeutics and represent promising tools for the development of new curative treatments for CLL.
Collapse
|
35
|
Węsierska-Gądek J, Kramer MP. The impact of multi-targeted cyclin-dependent kinase inhibition in breast cancer cells: clinical implications. Expert Opin Investig Drugs 2011; 20:1611-28. [PMID: 22017180 DOI: 10.1517/13543784.2011.628985] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The progression of the mammalian cell cycle is driven by the transient activation of complexes consisting of cyclins and cyclin-dependent kinases (CDKs). Loss of control over the cell cycle results in accelerated cell division and malignant transformation and can be caused by the upregulation of cyclins, the aberrant activation of CDKs or the inactivation of cellular CDK inhibitors. For these reasons, cell cycle regulators are regarded as very promising therapeutic targets for the treatment of human malignancies. AREAS COVERED This review covers the structures and anti-breast cancer activity of selected pharmacological pan-specific CDK inhibitors. Multi-targeted CDK inhibitors affect CDKs involved in the regulation of both cell cycle progression and transcriptional control. The inhibition of CDK7/CDK9 has a serious impact on the activity of RNA polymerase II; when its carboxy-terminal domain is unphosphorylated, it is unable to recruit the cofactors required for transcriptional elongation, resulting in a global transcriptional block. Multi-targeted inhibition of CDKs represses anti-apoptotic proteins and thus promotes the induction of apoptosis. Moreover, the inhibition of CDK7 in estrogen receptor (ER)-positive breast cancer cells prevents activating phosphorylation of ER-α. EXPERT OPINION These diverse modes of action make multi-targeted CDK inhibitors promising drugs for the treatment of breast cancers.
Collapse
Affiliation(s)
- Józefa Węsierska-Gądek
- Medical University of Vienna, Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center, Cell Cycle Regulation Group, Borschkegasse 8a, 1090 Vienna, Austria.
| | | |
Collapse
|
36
|
Nair BC, Vallabhaneni S, Tekmal RR, Vadlamudi RK. Roscovitine confers tumor suppressive effect on therapy-resistant breast tumor cells. Breast Cancer Res 2011; 13:R80. [PMID: 21834972 PMCID: PMC3218960 DOI: 10.1186/bcr2929] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 06/28/2011] [Accepted: 08/11/2011] [Indexed: 12/21/2022] Open
Abstract
Introduction Current clinical strategies for treating hormonal breast cancer involve the use of anti-estrogens that block estrogen receptor (ER)α functions and aromatase inhibitors that decrease local and systemic estrogen production. Both of these strategies improve outcomes for ERα-positive breast cancer patients, however, development of therapy resistance remains a major clinical problem. Divergent molecular pathways have been described for this resistant phenotype and interestingly, the majority of downstream events in these resistance pathways converge upon the modulation of cell cycle regulatory proteins including aberrant activation of cyclin dependent kinase 2 (CDK2). In this study, we examined whether the CDK inhibitor roscovitine confers a tumor suppressive effect on therapy-resistant breast epithelial cells. Methods Using various in vitro and in vivo assays, we tested the effect of roscovitine on three hormonal therapy-resistant model cells: (a) MCF-7-TamR (acquired tamoxifen resistance model); (b) MCF-7-LTLTca (acquired letrozole resistance model); and (c) MCF-7-HER2 that exhibit tamoxifen resistance (ER-growth factor signaling cross talk model). Results Hormonal therapy-resistant cells exhibited aberrant activation of the CDK2 pathway. Roscovitine at a dose of 20 μM significantly inhibited the cell proliferation rate and foci formation potential of all three therapy-resistant cells. The drug treatment substantially increased the proportion of cells in G2/M cell cycle phase with decreased CDK2 activity and promoted low cyclin D1 levels. Interestingly, roscovitine also preferentially down regulated the ERα isoform and ER-coregulators including AIB1 and PELP1. Results from xenograft studies further showed that roscovitine can attenuate growth of therapy-resistant tumors in vivo. Conclusions Roscovitine can reduce cell proliferation and survival of hormone therapy-resistant breast cancer cells. Our results support the emerging concept that inhibition of CDK2 activity has the potential to abrogate growth of hormonal therapy-resistant cells.
Collapse
Affiliation(s)
- Binoj C Nair
- Department of Obstetrics and Gynecology, CTRC at UT Health Science Center, San Antonio, Texas 78229, USA
| | | | | | | |
Collapse
|
37
|
Węsierska-Gądek J, Gritsch D, Zulehner N, Komina O, Maurer M. Interference with ER-α enhances the therapeutic efficacy of the selective CDK inhibitor roscovitine towards ER-positive breast cancer cells. J Cell Biochem 2011; 112:1103-17. [PMID: 21308739 DOI: 10.1002/jcb.23024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In recent years many risk factors for the development of breast cancer that are linked to estrogens have been identified, and roscovitine (ROSC), a selective cyclin-dependent kinase (CDK) inhibitor, has been shown to be an efficient inhibitor of the proliferation of human breast cancer cells. Therefore, we have examined the possibility that interference with estrogen signaling pathways, using tamoxifen (TAM), a selective estrogen receptor modulator (SERM), could modulate the efficacy of treatment with ROSC. In conjunction with TAM, ROSC exhibited enhanced anti-proliferative activity and CDK inhibition, particularly in estrogen-dependent MCF-7 cells. The interaction between both drugs was synergistic. However, in ER-α-negative cells the interaction was antagonistic. Exposure of MCF-7 cells to ROSC abolished the activating phosphorylation of CDK2 and CDK7 at Ser(164/170). This in turn prevented the phosphorylation of the carboxyl-terminal repeat domain of RNA Polymerase II and ER-α at Ser(118), resulting in the down-regulation of the latter. Concomitantly, wt p53 was strongly activated by phosphorylation at Ser(46). Our results demonstrate that ROSC negatively affects the functional status of ER-α, making it potentially useful in the treatment of estrogen-dependent breast cancer cells.
Collapse
Affiliation(s)
- Józefa Węsierska-Gądek
- Cell Cycle Regulation Group, Institute of Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| | | | | | | | | |
Collapse
|